51
|
|
52
|
Rieke DT, Ochsenreither S, Klinghammer K, Seiwert TY, Klauschen F, Tinhofer I, Keilholz U. Methylation of RAD51B, XRCC3 and other homologous recombination genes is associated with expression of immune checkpoints and an inflammatory signature in squamous cell carcinoma of the head and neck, lung and cervix. Oncotarget 2018; 7:75379-75393. [PMID: 27683114 PMCID: PMC5342748 DOI: 10.18632/oncotarget.12211] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/07/2016] [Indexed: 12/29/2022] Open
Abstract
Immune checkpoints are emerging treatment targets, but mechanisms underlying checkpoint expression are poorly understood. Since alterations in DNA repair genes have been connected to the efficacy of checkpoint inhibitors, we investigated associations between methylation of DNA repair genes and CTLA4 and CD274 (PD-L1) expression.A list of DNA repair genes (179 genes) was selected from the literature, methylation status and expression of inflammation-associated genes (The Cancer Genome Atlas data) was correlated in head and neck squamous cell carcinoma (HNSCC), cervical and lung squamous cell carcinoma.A significant positive correlation of the methylation status of 15, 3 and 2 genes with checkpoint expression was identified, respectively. RAD51B methylation was identified in all cancer subtypes. In HNSCC and cervical cancer, there was significant enrichment for homologous recombination genes. Methylation of the candidate genes was also associated with expression of other checkpoints, ligands, MHC- and T-cell associated genes as well as an interferon-inflammatory immune gene signature, predictive for the efficacy of PD-1 inhibition in HNSCC.Homologous recombination deficiency might therefore be mediated by DNA repair gene hypermethylation and linked to an immune-evasive phenotype in SCC. The methylation status of these genes could represent a new predictive biomarker for immune checkpoint inhibition.
Collapse
Affiliation(s)
- Damian T Rieke
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Ochsenreither
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Hematology and Medical Oncology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Konrad Klinghammer
- Department of Hematology and Medical Oncology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tanguy Y Seiwert
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Frederick Klauschen
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Inge Tinhofer
- Department of Radiooncology and Radiotherapy, Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Research Center Heidelberg (DKFZ)/German Cancer Consortium (DKTK) partner site Berlin, Berlin, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Hematology and Medical Oncology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
53
|
Qian Y, Sang Y, Wang FXC, Hong B, Wang Q, Zhou X, Weng T, Wu Z, Zheng M, Zhang H, Yao H. Prognostic significance of B7-H4 expression in matched primary pancreatic cancer and liver metastases. Oncotarget 2018; 7:72242-72249. [PMID: 27750217 PMCID: PMC5342158 DOI: 10.18632/oncotarget.12665] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/10/2016] [Indexed: 01/21/2023] Open
Abstract
Liver metastasis development in pancreatic cancer patients is common and confers a poor prognosis. Clinical relevance of biomarker analysis in metastatic tissue is necessary. B7-H4 has an inhibitory effect on T cell mediated response and may be involved in tumor development. Although B7-H4 expression has been detected in pancreatic cancer, its expression in liver metastases from pancreatic cancer is still unknown. In this study, overall 43 pancreatic cancer liver metastases (with matched primaries in 15/43 cases) and 57 pancreatic cancer cases without liver metastases or other distant metastases were analyzed for their expression of B7-H4 by immunohistochemistry. Survival curves and log-rank tests were used to test the association of B7-H4 expression with survival. B7-H4 was highly expressed in 28 (65.1%) of the 43 liver metastases and 9 (60.0%) of the 15 matched primary tumors. The expression of B7-H4 in liver metastases was significantly higher than in the matched primary tumors (p < 0.05). Patients with high B7-H4 expression in their primary pancreatic cancer had higher risk of developing liver metastases (p < 0.05). In univariate analysis, B7-H4 expression was significantly associated with the risk of death (p < 0.05). And the multivariate analysis identified that B7-H4 was an independent prognostic indicator (p < 0.05). Our results revealed B7-H4 to be associated with poor prognosis in patients with pancreatic cancer liver metastasis. B7-H4 may promote pancreatic cancer metastasis and was promising to be a potential prognostic indicator of pancreatic cancer.
Collapse
Affiliation(s)
- Yun Qian
- Department of Clinical Laboratory, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yiwen Sang
- Department of Clinical Laboratory, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Frederick X C Wang
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Texas 75080, USA
| | - Bo Hong
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Qi Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xinhui Zhou
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Tianhao Weng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhigang Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Min Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hong Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hangping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
54
|
Feucht J, Kayser S, Gorodezki D, Hamieh M, Döring M, Blaeschke F, Schlegel P, Bösmüller H, Quintanilla-Fend L, Ebinger M, Lang P, Handgretinger R, Feuchtinger T. T-cell responses against CD19+ pediatric acute lymphoblastic leukemia mediated by bispecific T-cell engager (BiTE) are regulated contrarily by PD-L1 and CD80/CD86 on leukemic blasts. Oncotarget 2018; 7:76902-76919. [PMID: 27708227 PMCID: PMC5363558 DOI: 10.18632/oncotarget.12357] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 09/02/2016] [Indexed: 01/22/2023] Open
Abstract
T-cell immunotherapies are promising options in relapsed/refractory B-precursor acute lymphoblastic leukemia (ALL). We investigated the effect of co-signaling molecules on T-cell attack against leukemia mediated by CD19/CD3-bispecific T-cell engager. Primary CD19+ ALL blasts (n≥10) and physiologic CD19+CD10+ bone marrow precursors were screened for 20 co-signaling molecules. PD-L1, PD-1, LAG-3, CD40, CD86, CD27, CD70 and HVEM revealed different stimulatory and inhibitory profiles of pediatric ALL compared to physiologic cells, with PD-L1 and CD86 as most prominent inhibitory and stimulatory markers. PD-L1 was increased in relapsed ALL patients (n=11) and in ALLs refractory to Blinatumomab (n=5). Exhaustion markers (PD-1, TIM-3) were significantly higher on patients' T cells compared to physiologic controls. T-cell proliferation and effector function was target-cell dependent and correlated to expression of co-signaling molecules. Blockade of inhibitory PD-1-PD-L and CTLA-4-CD80/86 pathways enhanced T-cell function whereas blockade of co-stimulatory CD28-CD80/86 interaction significantly reduced T-cell function. Combination of Blinatumomab and anti-PD-1 antibody was feasible and induced an anti-leukemic in vivo response in a 12-year-old patient with refractory ALL. In conclusion, ALL cells actively regulate T-cell function by expression of co-signaling molecules and modify efficacy of therapeutic T-cell attack against ALL. Inhibitory interactions of leukemia-induced checkpoint molecules can guide future T-cell therapies.
Collapse
Affiliation(s)
- Judith Feucht
- Department of General Pediatrics, Hematology and Oncology, Children's University Hospital Tübingen, Tübingen, Germany.,Memorial Sloan Kettering Cancer Center, Center for Cell Engineering, New York, NY, USA
| | - Simone Kayser
- Department of General Pediatrics, Hematology and Oncology, Children's University Hospital Tübingen, Tübingen, Germany
| | - David Gorodezki
- Department of General Pediatrics, Hematology and Oncology, Children's University Hospital Tübingen, Tübingen, Germany
| | - Mohamad Hamieh
- Memorial Sloan Kettering Cancer Center, Center for Cell Engineering, New York, NY, USA
| | - Michaela Döring
- Department of General Pediatrics, Hematology and Oncology, Children's University Hospital Tübingen, Tübingen, Germany.,Dr. von Hauner Children's Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Franziska Blaeschke
- Dr. von Hauner Children's Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Patrick Schlegel
- Department of General Pediatrics, Hematology and Oncology, Children's University Hospital Tübingen, Tübingen, Germany
| | - Hans Bösmüller
- Institute of Pathology, University Hospital Tübingen, Tübingen, Germany
| | | | - Martin Ebinger
- Department of General Pediatrics, Hematology and Oncology, Children's University Hospital Tübingen, Tübingen, Germany
| | - Peter Lang
- Department of General Pediatrics, Hematology and Oncology, Children's University Hospital Tübingen, Tübingen, Germany
| | - Rupert Handgretinger
- Department of General Pediatrics, Hematology and Oncology, Children's University Hospital Tübingen, Tübingen, Germany
| | - Tobias Feuchtinger
- Department of General Pediatrics, Hematology and Oncology, Children's University Hospital Tübingen, Tübingen, Germany.,Dr. von Hauner Children's Hospital, Ludwig Maximilians University Munich, Munich, Germany
| |
Collapse
|
55
|
Xu W, Hiếu T, Malarkannan S, Wang L. The structure, expression, and multifaceted role of immune-checkpoint protein VISTA as a critical regulator of anti-tumor immunity, autoimmunity, and inflammation. Cell Mol Immunol 2018; 15:438-446. [PMID: 29375120 DOI: 10.1038/cmi.2017.148] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/08/2017] [Accepted: 11/09/2017] [Indexed: 12/26/2022] Open
Abstract
Among various immunoregulatory molecules, the B7 family of immune-checkpoint receptors consists of highly valuable targets for cancer immunotherapy. Antibodies targeting two B7 family co-inhibitory receptors, CTLA-4 and PD-1, have elicited long-term clinical outcomes in previously refractory cancer types and are considered a breakthrough in cancer therapy. Despite the success, the relatively low response rate (20-30%) warrants efforts to identify and overcome additional immune-suppressive pathways. Among the expanding list of T cell inhibitory regulators, V domain immunoglobulin suppressor of T cell activation (VISTA) is a unique B7 family checkpoint that regulates a broad spectrum of immune responses. Here, we summarize recent advances that highlight the structure, expression, and multi-faceted immunomodulatory mechanisms of VISTA in the context of autoimmunity, inflammation, and anti-tumor immunity.
Collapse
Affiliation(s)
- Wenwen Xu
- Department of Microbiology and Immunology, Milwaukee, WI 53226, USA
| | - TạMinh Hiếu
- Department of Microbiology and Immunology, Milwaukee, WI 53226, USA
| | - Subramaniam Malarkannan
- Department of Microbiology and Immunology, Milwaukee, WI 53226, USA.,Department of Medicine, Milwaukee, WI 53226, USA.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Blood Research Institute, 53226, Milwaukee, WI, USA
| | - Li Wang
- Department of Microbiology and Immunology, Milwaukee, WI 53226, USA.
| |
Collapse
|
56
|
Li G, Quan Y, Che F, Wang L. B7-H3 in tumors: friend or foe for tumor immunity? Cancer Chemother Pharmacol 2018; 81:245-253. [PMID: 29299639 DOI: 10.1007/s00280-017-3508-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/21/2017] [Indexed: 12/11/2022]
Abstract
B7-H3 is a type I transmembrane co-stimulatory molecule of the B7 family. B7-H3 mRNA is widely distributed in most tissues; however, B7-H3 protein is not constitutively expressed. Few molecules have been shown to mediate the regulation of B7-H3 expression, and their regulatory mechanisms remain unexplored. Recently, TREM-like transcript 2 (TLT-2) has been identified as a potential receptor of B7-H3. However, TLT-2 may not be the only receptor of B7-H3, as B7-H3 has many contradictory roles. As a co-stimulatory molecule, B7-H3 increases the proliferation of both CD4+ and CD8+ T-cells and enhances cytotoxic T-cell activity. However, greatly increased T-cell proliferation and IL-2 levels have been observed in the absence of B7-H3. Thus far, it has been shown that various tumors test positive for B7-H3 expression and that B7-H3 levels correlate with tumor growth, invasion, metastasis, malignant stage, and recurrence rate. Furthermore, transfection of cells with a B7-H3 plasmid and treatment with monoclonal antibodies to block B7-H3 are the main immunotherapeutic strategies for cancer treatment. Several groups have generated anti-B7-H3 antibodies and observed tumor growth suppression in vitro and in vivo. Therefore, it is likely that B7-H3 plays an important role in cancer diagnosis and treatment, aside from its role as a co-stimulatory molecule.
Collapse
Affiliation(s)
- Gen Li
- Department of Neurology, Clinical Medicine College, Weifang Medical University, No. 7166, Baotong West Street, Weifang city, Shandong Province, 261053, China.,Central Laboratory, Linyi People's Hospital, Shandong University, NB27, Eastern Part of Jiefang Road, Lanshan District, Linyi city, Shandong Province, 276000, China
| | - Yanchun Quan
- Central Laboratory, Linyi People's Hospital, Shandong University, NB27, Eastern Part of Jiefang Road, Lanshan District, Linyi city, Shandong Province, 276000, China
| | - Fengyuan Che
- Central Laboratory, Linyi People's Hospital, Shandong University, NB27, Eastern Part of Jiefang Road, Lanshan District, Linyi city, Shandong Province, 276000, China. .,Department of Neurology, Linyi People's Hospital, Shandong University, NB27, Eastern Part of Jiefang Road, Lanshan District, Linyi city, Shandong Province, 276000, China.
| | - Lijuan Wang
- Central Laboratory, Linyi People's Hospital, Shandong University, NB27, Eastern Part of Jiefang Road, Lanshan District, Linyi city, Shandong Province, 276000, China. .,Department of Hematology, Linyi People's Hospital, Shandong University, NB27, Eastern Part of Jiefang Road, Lanshan District, Linyi city, Shandong Province, 276000, China.
| |
Collapse
|
57
|
Tumor-expressed immune checkpoint B7x promotes cancer progression and antigen-specific CD8 T cell exhaustion and suppressive innate immune cells. Oncotarget 2017; 8:82740-82753. [PMID: 29137299 PMCID: PMC5669925 DOI: 10.18632/oncotarget.21098] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/29/2017] [Indexed: 02/04/2023] Open
Abstract
B7x (B7-H4 or B7S1) is a coinhibitory member of the B7 immune checkpoint ligand family that regulates immune function following ligation with its unknown cognate receptors. B7x has limited expression on normal tissues, but is up-regulated on solid human tumors to inhibit anti-tumor immunity and associates with poor clinical prognosis. We assessed the contribution of cytokine stimuli to induce surface B7x expression on cancer cells and the role of tumor-expressed B7x in a murine pulmonary metastasis model, and finally evaluated the potential interaction between B7x and Neuropilin-1, a suggested potential cognate receptor. We showed that pro-inflammatory and anti-inflammatory cytokines IFNγ, TNFα, and IL-10 did not induce expression of B7x on human or murine cancer cells. Following i.v. injection of CT26, a murine colon cancer cell line in the BALB/c background, we observed a significant increase in tumor burden in the lung of B7x-expressing CT26 mice compared to B7x-negative parental CT26 control mice. This was marked by a significant increase in M2 tumor associated macrophages and antigen-specific CD8 T cell exhaustion. Finally, we found through multiple systems that there was no evidence for B7x and Neuropilin-1 direct interaction. Thus, the B7x pathway has an essential role in modulating the innate and adaptive immune cell infiltrate in the tumor microenvironment with its currently unknown cognate receptor(s).
Collapse
|
58
|
Flem-Karlsen K, Tekle C, Andersson Y, Flatmark K, Fodstad Ø, Nunes-Xavier CE. Immunoregulatory protein B7-H3 promotes growth and decreases sensitivity to therapy in metastatic melanoma cells. Pigment Cell Melanoma Res 2017; 30:467-476. [PMID: 28513992 DOI: 10.1111/pcmr.12599] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/09/2017] [Indexed: 12/26/2022]
Abstract
B7-H3 (CD276) belongs to the B7 family of immunoregulatory proteins and has been implicated in cancer progression and metastasis. In this study, we found that metastatic melanoma cells with knockdown expression of B7-H3 showed modest decrease in proliferation and glycolytic capacity and were more sensitive to dacarbazine (DTIC) chemotherapy and small-molecule inhibitors targeting MAP kinase (MAPK) and AKT/mTOR pathways: vemurafenib (PLX4032; BRAF inhibitor), binimetinib (MEK-162; MEK inhibitor), everolimus (RAD001; mTOR inhibitor), and triciribidine (API-2; AKT inhibitor). Similar effects were observed in melanoma cells in the presence of an inhibitory B7-H3 monoclonal antibody, while the opposite was seen in B7-H3-overexpressing cells. Further, combining B7-H3 inhibition with small-molecule inhibitors resulted in significantly increased antiproliferative effect in melanoma cells, as well as in BRAFV600E mutated cell lines derived from patient biopsies. Our findings indicate that targeting B7-H3 may be a novel alternative to improve current therapy of metastatic melanoma.
Collapse
Affiliation(s)
- Karine Flem-Karlsen
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Christina Tekle
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Yvonne Andersson
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Kjersti Flatmark
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Øystein Fodstad
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Caroline E Nunes-Xavier
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| |
Collapse
|
59
|
Altan M, Pelekanou V, Schalper KA, Toki M, Gaule P, Syrigos K, Herbst RS, Rimm DL. B7-H3 Expression in NSCLC and Its Association with B7-H4, PD-L1 and Tumor-Infiltrating Lymphocytes. Clin Cancer Res 2017; 23:5202-5209. [PMID: 28539467 DOI: 10.1158/1078-0432.ccr-16-3107] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/13/2017] [Accepted: 05/16/2017] [Indexed: 01/08/2023]
Abstract
Purpose: The immune checkpoint PD-1 and its receptor B7-H1 (PD-L1) are successful therapeutic targets in cancer but less is known about other B7 family members. Here, we determined the expression level of B7-H3 protein in non-small cell lung cancer (NSCLC) and evaluated its association with tumor-infiltrating lymphocytes (TIL), PD-L1, B7-H4, and major clinicopathologic characteristics is in 3 NSCLC cohorts.Experimental design: We used multiplexed automated quantitative immunofluorescence (QIF) to assess the levels of B7-H3, PD-L1, B7-H4, and TILs in 634 NSCLC cases with validated antibodies. Associations between the marker levels, major clinicopathologic variables and survival were analyzed.Results: Expression of B7-H3 protein was found in 80.4% (510/634) of the cases. High B7-H3 protein level (top 10 percentile) was associated with poor overall survival (P < 0.05). Elevated B7-H3 was consistently associated with smoking history across the 3 cohorts, but not with sex, age, clinical stage, and histology. Coexpression of B7-H3 and PD-L1 was found in 17.6% of the cases (112/634) and with B7-H4 in 10% (63/634). B7-H4 and PD-L1 were simultaneously detected only in 1.8% of NSCLCs (12/634). The expression of B7-H3 was not associated with the levels of CD3-, CD8-, and CD20-positive TILs.Conclusions: B7-H3 protein is expressed in the majority of NSCLCs and is associated with smoking history. High levels of B7-H3 protein have a negative prognostic impact in lung carcinomas. Coexpression of B7-H3 with PD-L1 and B7-H4 is relatively low, suggesting a nonredundant biological role of these targets. Clin Cancer Res; 23(17); 5202-9. ©2017 AACR.
Collapse
Affiliation(s)
- Mehmet Altan
- Section of Medical Oncology, Yale School of Medicine, New Haven, Connecticut.,Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Vasiliki Pelekanou
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Kurt A Schalper
- Section of Medical Oncology, Yale School of Medicine, New Haven, Connecticut.,Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Maria Toki
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Patricia Gaule
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Konstantinos Syrigos
- Third Department of Medicine, University of Athens, School of Medicine, Sotiria General Hospital, Athens, Greece
| | - Roy S Herbst
- Section of Medical Oncology, Yale School of Medicine, New Haven, Connecticut
| | - David L Rimm
- Section of Medical Oncology, Yale School of Medicine, New Haven, Connecticut. .,Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
60
|
Leung J, St-Onge P, Stagg J, Suh WK. Synergistic effects of host B7-H4 deficiency and gemcitabine treatment on tumor regression and anti-tumor T cell immunity in a mouse model. Cancer Immunol Immunother 2017; 66:491-502. [PMID: 28074226 PMCID: PMC11028495 DOI: 10.1007/s00262-016-1950-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 12/20/2016] [Indexed: 02/08/2023]
Abstract
B7-H4 (B7x/B7S1), a B7 family inhibitor of T cell activity, is expressed in multiple human cancers and correlates with decreased infiltrating lymphocytes and poor prognosis. In murine models, tumor-expressed B7-H4 enhances tumor growth and reduces T cell immunity, and blockade of tumor-B7-H4 rescues T cell activity and lowers tumor burden. This implicates B7-H4 as a target for cancer immunotherapy, yet limits the efficacy of B7-H4 blockade exclusively to patients with B7-H4+ tumors. Given the expression of B7-H4 on host immune cells, we have previously shown that BALB/c mice lacking host B7-H4 have enhanced anti-tumor profiles, yet similar 4T1 tumor growth relative to control. Given that T cell-mediated immunotherapies work best for tumors presenting tumor-associated neoantigens, we further investigated the function of host B7-H4 in the growth of a more immunogenic derivative, 4T1-12B, which is known to elicit strong anti-tumor CD8 T cell responses due to expression of a surrogate tumor-specific antigen, firefly luciferase. Notably, B7-H4 knockout hosts not only mounted greater tumor-associated anti-tumor T cell responses, but also displayed reduced tumors. Additionally, B7-H4-deficiency synergized with gemcitabine to further inhibit tumor growth, often leading to tumor eradication and the generation of protective T cell immunity. These findings imply that inhibition of host B7-H4 can enhance anti-tumor T cell immunity in immunogenic cancers, and can be combined with other anti-cancer therapies to further reduce tumor burden regardless of tumor-B7-H4 positivity.
Collapse
Affiliation(s)
- Joanne Leung
- Institut de recherches cliniques de Montréal (IRCM), 110 avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Philippe St-Onge
- Institut de recherches cliniques de Montréal (IRCM), 110 avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Département de Médecine (Programmes de Biologie Moléculaire), Université de Montréal, Montréal, QC, Canada
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Institut du Cancer de Montréal, Faculté de Pharmacie, Université de Montréal, Québec, Canada
| | - Woong-Kyung Suh
- Institut de recherches cliniques de Montréal (IRCM), 110 avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada.
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada.
- Département de Médecine (Programmes de Biologie Moléculaire), Université de Montréal, Montréal, QC, Canada.
- Département de Médecine, Université de Montréal, Montréal, QC, Canada.
- Département de Microbiologie, Infectiologie, et Immunologie, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
61
|
Smolle MA, Calin HN, Pichler M, Calin GA. Noncoding RNAs and immune checkpoints-clinical implications as cancer therapeutics. FEBS J 2017; 284:1952-1966. [PMID: 28132417 DOI: 10.1111/febs.14030] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/31/2016] [Accepted: 01/25/2017] [Indexed: 12/16/2022]
Abstract
A major mechanism of tumor development and progression is silencing of the patient's immune response to cancer-specific antigens. Defects in the so-called cancer immunity cycle may occur at any stage of tumor development. Within the tumor microenvironment, aberrant expression of immune checkpoint molecules with activating or inhibitory effects on T lymphocytes induces immune tolerance and cellular immune escape. Targeting immune checkpoint molecules such as programmed cell death protein 1 (PD-1) and its ligand PD-L1 with specific antibodies has proven to be a major advance in the treatment of several types of cancer. Another way to therapeutically influence the tumor microenvironment is by modulating the levels of microRNAs (miRNAs), small noncoding RNAs that shuttle bidirectionally between malignant and tumor microenvironmental cells. These small RNA transcripts have two features: (a) their expression is quite specific to distinct tumors, and (b) they are involved in early regulation of immune responses. Consequently, miRNAs may be ideal molecules for use in cancer therapy. Many miRNAs are aberrantly expressed in human cancer cells, opening new opportunities for cancer therapy, but the exact functions of these miRNAs and their interactions with immune checkpoint molecules have yet to be investigated. This review summarizes recently reported findings about miRNAs as modulators of immune checkpoint molecules and their potential application as cancer therapeutics in clinical practice.
Collapse
Affiliation(s)
- Maria A Smolle
- Division of Clinical Oncology, Internal Medicine, Medical University of Graz, Graz, Austria.,Department of Orthopaedics and Trauma, Medical University of Graz, Graz, Austria.,Center for Biomarker Research in Medicine, Graz, Austria
| | | | - Martin Pichler
- Division of Clinical Oncology, Internal Medicine, Medical University of Graz, Graz, Austria.,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA.,Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
62
|
Cancer-immune therapy: restoration of immune response in cancer by immune cell modulation. THE NUCLEUS 2017. [DOI: 10.1007/s13237-017-0194-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
63
|
Ueta M, Sawai H, Shingaki R, Kawai Y, Sotozono C, Kojima K, Yoon KC, Kim MK, Seo KY, Joo CK, Nagasaki M, Kinoshita S, Tokunaga K. Genome-wide association study using the ethnicity-specific Japonica array: identification of new susceptibility loci for cold medicine-related Stevens-Johnson syndrome with severe ocular complications. J Hum Genet 2017; 62:485-489. [PMID: 28100913 DOI: 10.1038/jhg.2016.160] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 12/06/2016] [Accepted: 12/07/2016] [Indexed: 01/03/2023]
Abstract
A genome-wide association study (GWAS) for cold medicine-related Stevens-Johnson syndrome (CM-SJS) with severe ocular complications (SOC) was performed in a Japanese population. A recently developed ethnicity-specific array with genome-wide imputation that was based on the whole-genome sequences of 1070 unrelated Japanese individuals was used. Validation analysis with additional samples from Japanese individuals and replication analysis using samples from Korean individuals identified two new susceptibility loci on chromosomes 15 and 16. This study might suggest the usefulness of GWAS using the ethnicity-specific array and genome-wide imputation based on large-scale whole-genome sequences. Our findings contribute to the understanding of genetic predisposition to CM-SJS with SOC.
Collapse
Affiliation(s)
- Mayumi Ueta
- Department of Frontier Medical Science and Technology for Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Human Genetics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Hiromi Sawai
- Department of Human Genetics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Ryosei Shingaki
- Life Science Business Department, Healthcare Medical Business Promotion Division, Toshiba Corporation Healthcare Company, Tokyo, Japan
| | - Yusuke Kawai
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kaname Kojima
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Kyung-Chul Yoon
- Department of Ophthalmology, Chonnam National University, Gwangju, Korea
| | - Mee Kum Kim
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul, Korea
| | - Kyoung Yul Seo
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - Choun-Ki Joo
- Department of Ophthalmology and Visual Science, Seoul St Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Masao Nagasaki
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Shigeru Kinoshita
- Department of Frontier Medical Science and Technology for Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Katsushi Tokunaga
- Department of Human Genetics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
64
|
Johansson H, Andersson R, Bauden M, Hammes S, Holdenrieder S, Ansari D. Immune checkpoint therapy for pancreatic cancer. World J Gastroenterol 2016; 22:9457-9476. [PMID: 27920468 PMCID: PMC5116591 DOI: 10.3748/wjg.v22.i43.9457] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 09/18/2016] [Accepted: 10/19/2016] [Indexed: 02/06/2023] Open
Abstract
Novel treatment modalities are necessary for pancreatic cancer. Immunotherapy with immune checkpoint inhibition has shown effect in other solid tumors, and could have a place in pancreatic cancer treatment. Most available clinical studies on immune checkpoint inhibitors for pancreatic cancer are not yet completed and are still recruiting patients. Among the completed trials, there have been findings of a preliminary nature such as delayed disease progression and enhanced overall survival after treatment with immune checkpoint inhibitors in mono- or combination therapy. However, due to small sample sizes, major results are not yet identifiable. The present article provides a clinical overview of immune checkpoint inhibition in pancreatic cancer. PubMed, ClinicalTrials.gov and American Society of Clinical Oncology's meeting abstracts were systematically searched for relevant clinical studies. Four articles, five abstracts and 25 clinical trials were identified and analyzed in detail.
Collapse
|
65
|
Association of PD-1/PD-L axis expression with cytolytic activity, mutational load, and prognosis in melanoma and other solid tumors. Proc Natl Acad Sci U S A 2016; 113:E7769-E7777. [PMID: 27837027 DOI: 10.1073/pnas.1607836113] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Programmed cell death protein-1 (PD-1)/programmed death ligand-1 (PD-L1) checkpoint blockade has led to remarkable and durable objective responses in a number of different tumor types. A better understanding of factors associated with the PD-1/PD-L axis expression is desirable, as it informs their potential role as prognostic and predictive biomarkers and may suggest rational treatment combinations. In the current study, we analyzed PD-L1, PD-L2, PD-1, and cytolytic activity (CYT) expression, as well as mutational density from melanoma and eight other solid tumor types using The Cancer Genome Atlas database. We found that in some tumor types, PD-L2 expression is more closely linked to Th1/IFNG expression and PD-1 and CD8 signaling than PD-L1 In contrast, mutational load was not correlated with a Th1/IFNG gene signature in any tumor type. PD-L1, PD-L2, PD-1, CYT expression, and mutational density are all positive prognostic features in melanoma, and conditional inference modeling revealed PD-1/CYT expression (i.e., an inflamed tumor microenvironment) as the most impactful feature, followed by mutational density. This study elucidates the highly interdependent nature of these parameters, and also indicates that future biomarkers for anti-PD-1/PD-L1 will benefit from tumor-type-specific, integrated, mRNA, protein, and genomic approaches.
Collapse
|
66
|
Yarbrough WG, Panaccione A, Chang MT, Ivanov SV. Clinical and molecular insights into adenoid cystic carcinoma: Neural crest-like stemness as a target. Laryngoscope Investig Otolaryngol 2016; 1:60-77. [PMID: 28894804 PMCID: PMC5510248 DOI: 10.1002/lio2.22] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/10/2016] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES This review surveys trialed therapies and molecular defects in adenoid cystic carcinoma (ACC), with an emphasis on neural crest-like stemness characteristics of newly discovered cancer stem cells (CSCs) and therapies that may target these CSCs. DATA SOURCES Articles available on Pubmed or OVID MEDLINE databases and unpublished data. REVIEW METHODS Systematic review of articles pertaining to ACC and neural crest-like stem cells. RESULTS Adenoid cystic carcinoma of the salivary gland is a slowly growing but relentless cancer that is prone to nerve invasion and metastases. A lack of understanding of molecular etiology and absence of targetable drivers has limited therapy for patients with ACC to surgery and radiation. Currently, no curative treatments are available for patients with metastatic disease, which highlights the need for effective new therapies. Research in this area has been inhibited by the lack of validated cell lines and a paucity of clinically useful markers. The ACC research environment has recently improved, thanks to the introduction of novel tools, technologies, approaches, and models. Improved understanding of ACC suggests that neural crest-like stemness is a major target in this rare tumor. New cell culture techniques and patient-derived xenografts provide tools for preclinical testing. CONCLUSION Preclinical research has not identified effective targets in ACC, as confirmed by the large number of failed clinical trials. New molecular data suggest that drivers of neural crest-like stemness may be required for maintenance of ACC; as such, CSCs are a target for therapy of ACC.
Collapse
Affiliation(s)
- Wendell G. Yarbrough
- Section of Otolaryngology, Department of Surgery, Yale School of MedicineNew HavenConnecticutUSA
- Yale Cancer CenterNew HavenConnecticutUSA
| | - Alexander Panaccione
- Department of Cancer BiologyVanderbilt University School of MedicineNashvilleTennesseeU.S.A.
| | - Michael T. Chang
- Section of Otolaryngology, Department of Surgery, Yale School of MedicineNew HavenConnecticutUSA
| | - Sergey V. Ivanov
- Section of Otolaryngology, Department of Surgery, Yale School of MedicineNew HavenConnecticutUSA
| |
Collapse
|
67
|
Schalper KA, Carvajal-Hausdorf D, McLaughlin J, Altan M, Velcheti V, Gaule P, Sanmamed MF, Chen L, Herbst RS, Rimm DL. Differential Expression and Significance of PD-L1, IDO-1, and B7-H4 in Human Lung Cancer. Clin Cancer Res 2016; 23:370-378. [PMID: 27440266 DOI: 10.1158/1078-0432.ccr-16-0150] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 07/11/2016] [Accepted: 07/13/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE To determine the expression level, associations, and biological role of PD-L1, IDO-1, and B7-H4 in non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN Using multiplexed quantitative immunofluorescence (QIF), we measured the levels of PD-L1, IDO-1, B7-H4, and different tumor-infiltrating lymphoycte (TIL) subsets in 552 stages I-IV lung carcinomas from two independent populations. Associations between the marker levels, TILs, and major clinicopathologic variables were determined. Validation of findings was performed using mRNA expression data from The Cancer Genome Atlas (TCGA) and in vitro stimulation of lung adenocarcinoma A549 cells with IFNγ and IL10. RESULTS PD-L1 was detected in 16.9% and 21.8% of cases in each population. IDO-1 was expressed in 42.6% and 49.8%; and B7-H4 in 12.8% and 22.6% of cases, respectively. Elevated PD-L1 and IDO-1 were consistently associated with prominent B- and T-cell infiltrates, but B7-H4 was not. Coexpression of the three protein markers was infrequent, and comparable results were seen in the lung cancer TCGA dataset. Levels of PD-L1 and IDO-1 (but not B7-H4) were increased by IFNγ stimulation in A549 cells. Treatment with IL10 upregulated B7-H4 but did not affect PD-L1 and IDO-1 levels. CONCLUSIONS PD-L1, IDO-1, and B7-H4 are differentially expressed in human lung carcinomas and show limited co-expression. While PD-L1 and IDO-1 are associated with increased TILs and IFNγ stimulation, B7-H4 is not. The preferential expression of discrete immune evasion pathways in lung cancer could participate in therapeutic resistance and support design of optimal clinical trials. Clin Cancer Res; 23(2); 370-8. ©2016 AACR.
Collapse
MESH Headings
- A549 Cells
- Aged
- B7-H1 Antigen/genetics
- Biomarkers, Tumor/genetics
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Disease-Free Survival
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Interferon-gamma/pharmacology
- Interleukin-10/pharmacology
- Lymphocytes, Tumor-Infiltrating/pathology
- Middle Aged
- Neoplasm Staging
- RNA, Messenger/drug effects
- RNA, Messenger/genetics
- V-Set Domain-Containing T-Cell Activation Inhibitor 1/genetics
Collapse
Affiliation(s)
- Kurt A Schalper
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut.
- Medical Oncology Section, Yale School of Medicine, New Haven, Connecticut
- Translational Immuno-oncology Laboratory, Yale Cancer Center, New Haven, Connecticut
| | | | - Joseph McLaughlin
- Medical Oncology Section, Yale School of Medicine, New Haven, Connecticut
| | - Mehmet Altan
- Medical Oncology Section, Yale School of Medicine, New Haven, Connecticut
| | | | - Patricia Gaule
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | | | - Lieping Chen
- Immunobiology, Yale School of Medicine, New Haven, Connecticut
| | - Roy S Herbst
- Medical Oncology Section, Yale School of Medicine, New Haven, Connecticut
| | - David L Rimm
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
- Medical Oncology Section, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
68
|
Gargiulo P, Della Pepa C, Berardi S, Califano D, Scala S, Buonaguro L, Ciliberto G, Brauchli P, Pignata S. Tumor genotype and immune microenvironment in POLE-ultramutated and MSI-hypermutated Endometrial Cancers: New candidates for checkpoint blockade immunotherapy? Cancer Treat Rev 2016; 48:61-8. [PMID: 27362548 DOI: 10.1016/j.ctrv.2016.06.008] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 06/11/2016] [Accepted: 06/13/2016] [Indexed: 12/27/2022]
Abstract
Endometrial Cancer (EC) is still a challenge for gynecological oncologists because the treatment of the advanced disease remains an unmet need for patients. The Cancer Genome Atlas Research Network (TCGA) recently provided a comprehensive genomic and transcriptomic analysis of EC, offering a new classification of the disease, based on genetic features, which defines four subgroups of cancer rather than the two traditionally recognized. In the molecular classification two types of EC, the polymerase epsilon (POLE)-ultramutated and the microsatellite instability (MSI)-hypermutated, seem to present an enhanced immune microenvironment and a high mutation burden. The blockade of the immune checkpoints is an innovative approach that has largely demonstrated to be effective in solid malignancies, such as lung, renal and melanoma; it acts by reducing the cancer-induced immune-suppression through inhibition of the PD-1/PD-L1 (Programmed Death and PD-Ligand) axis. All available evidence supporting an over-expression of the PD-1/PD-L1 pathway in EC has been reviewed. In particular in the POLE and MSI ECs an up-regulation of this pathway was found, aiming to suggest a rationale for testing the PD-1/PD-L1 immunotherapy in these cancer subgroups.
Collapse
Affiliation(s)
- Piera Gargiulo
- Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Effingerstrasse 33, CH-3008 Bern, Switzerland; Department of Urology and Gynecology, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Via Mariano Semmola 52, 80131 Naples, Italy.
| | - Chiara Della Pepa
- Department of Urology and Gynecology, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Via Mariano Semmola 52, 80131 Naples, Italy.
| | - Simona Berardi
- Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Effingerstrasse 33, CH-3008 Bern, Switzerland.
| | - Daniela Califano
- Department of Research-Functional Genomics, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Via Mariano Semmola 52, 80131 Naples, Italy.
| | - Stefania Scala
- Department of Research-Functional Genomics, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Via Mariano Semmola 52, 80131 Naples, Italy.
| | - Luigi Buonaguro
- Molecular Biology and Viral Oncogenesis Unit, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Via Mariano Semmola 52, 80131 Naples, Italy.
| | - Gennaro Ciliberto
- Scientific Directorate, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Via Mariano Semmola 52, 80131 Naples, Italy.
| | - Peter Brauchli
- Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Effingerstrasse 33, CH-3008 Bern, Switzerland.
| | - Sandro Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Via Mariano Semmola 52, 80131 Naples, Italy.
| |
Collapse
|
69
|
Nayama M, Collinet P, Salzet M, Vinatier D. [Immunological aspects of ovarian cancer: Therapeutic perspectives]. ACTA ACUST UNITED AC 2016; 45:1020-1036. [PMID: 27320132 DOI: 10.1016/j.jgyn.2016.05.005] [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: 09/06/2013] [Revised: 05/07/2016] [Accepted: 05/13/2016] [Indexed: 01/09/2023]
Abstract
Ovarian cancer is recognized by the immunological system of its host. Initially, it is effective to destroy and eliminate the cancer. But gradually, resistant tumor cells more aggressive and those able to protect themselves by inducing immune tolerance will be selected. Immunotherapy to be effective should consider both components of immune response with an action on cytotoxic immune effectors and action on tolerance mechanisms. The manipulations of the immune system should be cautious, because the immune effects are not isolated. A theoretically efficient handling may simultaneously cause an adverse effect which was not envisaged and could neutralize the benefits of treatment. Knowledge of tolerance mechanisms set up by the tumor is for the clinician a prerequisite before they prescribe these treatments. For each cancer, the knowledge of its immunological status is a prerequisite to propose adapted immunological therapies.
Collapse
Affiliation(s)
- M Nayama
- Service de gynécologie obstétrique, maternité Issaka-Gazoby, BP 10975, Niamey, Niger
| | - P Collinet
- CHU de Lille, 59000 Lille, France; Département universitaire de gynécologie obstétrique, université Nord-de-France, 59045 Lille cedex, France
| | - M Salzet
- EA 4550, IFR 147, laboratoire PRISM : protéomique, réponse inflammatoire, spectrométrie de Masse, université Lille 1, bâtiment SN3, 1(er) étage, 59655 Villeneuve d'Ascq cedex, France
| | - D Vinatier
- CHU de Lille, 59000 Lille, France; EA 4550, IFR 147, laboratoire PRISM : protéomique, réponse inflammatoire, spectrométrie de Masse, université Lille 1, bâtiment SN3, 1(er) étage, 59655 Villeneuve d'Ascq cedex, France; Département universitaire de gynécologie obstétrique, université Nord-de-France, 59045 Lille cedex, France.
| |
Collapse
|
70
|
Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer 2016; 16:275-87. [PMID: 27079802 PMCID: PMC5381938 DOI: 10.1038/nrc.2016.36] [Citation(s) in RCA: 2055] [Impact Index Per Article: 228.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
With recent approvals for multiple therapeutic antibodies that block cytotoxic T lymphocyte associated antigen 4 (CTLA4) and programmed cell death protein 1 (PD1) in melanoma, non-small-cell lung cancer and kidney cancer, and additional immune checkpoints being targeted clinically, many questions still remain regarding the optimal use of drugs that block these checkpoint pathways. Defining biomarkers that predict therapeutic effects and adverse events is a crucial mandate, highlighted by recent approvals for two PDL1 diagnostic tests. Here, we discuss biomarkers for anti-PD1 therapy based on immunological, genetic and virological criteria. The unique biology of the CTLA4 immune checkpoint, compared with PD1, requires a different approach to biomarker development. Mechanism-based insights from such studies may guide the design of synergistic treatment combinations based on immune checkpoint blockade.
Collapse
Affiliation(s)
- Suzanne L Topalian
- Department of Surgery, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy, 1550 Orleans Street, CRB2 Room 508, Baltimore, Maryland 21287, USA
| | - Janis M Taube
- Department of Dermatology, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy, 1550 Orleans Street, CRB2 Room 508, Baltimore, Maryland 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy, 1550 Orleans Street, CRB2 Room 508, Baltimore, Maryland 21287, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy, 1550 Orleans Street, CRB2 Room 508, Baltimore, Maryland 21287, USA
| | - Robert A Anders
- Department of Pathology, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy, 1550 Orleans Street, CRB2 Room 508, Baltimore, Maryland 21287, USA
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy, 1550 Orleans Street, CRB2 Room 508, Baltimore, Maryland 21287, USA
| |
Collapse
|
71
|
Yoon BR, Chung YH, Yoo SJ, Kawara K, Kim J, Yoo IS, Park CG, Kang SW, Lee WW. Preferential Induction of the T Cell Auxiliary Signaling Molecule B7-H3 on Synovial Monocytes in Rheumatoid Arthritis. J Biol Chem 2016; 291:4048-57. [PMID: 26702052 PMCID: PMC4759181 DOI: 10.1074/jbc.m115.680298] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 11/24/2015] [Indexed: 12/29/2022] Open
Abstract
B7-H3, a newly identified B7 family member, has functional duality as a co-stimulator and co-inhibitor that fine-tunes T cell-mediated immune responses. Given that B7-H3 expression on human monocytes and dendritic cells is enhanced by inflammatory cytokines, its potential inmmunoregulatory role at sites of inflammation has been suggested. Further, monocytes play crucial roles in the pathophysiology of various inflammatory disorders including autoimmune diseases; however, the immunological role of B7-H3 in rheumatoid arthritis (RA) has not been defined. Thus, we aimed to investigate the possible roles of monocyte B7-H3 in the pathogenesis of RA. Synovial monocytes, but not peripheral monocytes, in RA patients predominantly express surface B7-H3. The 4Ig isoform of B7-H3 is exclusively induced on the cell surface, whereas the 2Ig B7-H3 isoform is constitutively expressed in the intracytoplasmic region of both peripheral and synovial monocytes. B7-H3 knockdown experiments reveal that surface B7-H3 has an inhibitory effect on IFN-γ production in CD4 memory cells. Moreover, surface B7-H3 expression on synovial monocytes inversely correlates with RA clinical parameters. Our findings demonstrate that activation-induced B7-H3 expression on synovial monocytes has the potential to inhibit Th1-mediated immune responses and immunomodulatory roles affecting RA pathogenesis.
Collapse
Affiliation(s)
- Bo Ruem Yoon
- From the Departments of Microbiology and Immunology and
| | - Yeon-Ho Chung
- From the Departments of Microbiology and Immunology and the BK21Plus Biomedical Science Project, and the Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Su-Jin Yoo
- the Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
| | - Kenji Kawara
- From the Departments of Microbiology and Immunology and the Tokyo Medical and Dental University School of Medicine, Tokyo 113-8510, Japan, and
| | - Jinhyun Kim
- the Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
| | - In Seol Yoo
- the Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
| | - Chung-Gyu Park
- From the Departments of Microbiology and Immunology and Biomedical Sciences and
| | - Seong Wook Kang
- the Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 301-721, South Korea,
| | - Won-Woo Lee
- From the Departments of Microbiology and Immunology and the BK21Plus Biomedical Science Project, and the Seoul National University College of Medicine, Seoul 110-799, South Korea, Biomedical Sciences and the Cancer Research Institute, Ischemic/Hypoxic Disease Institute, and Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul National University Hospital Biomedical Research Institute, Seoul 110-799, South Korea
| |
Collapse
|
72
|
Khosravi-Maharlooei M, Pakyari M, Jalili RB, Salimi-Elizei S, Lai JCY, Poormasjedi-Meibod M, Kilani RT, Dutz J, Ghahary A. Tolerogenic effect of mouse fibroblasts on dendritic cells. Immunology 2016; 148:22-33. [PMID: 26789277 DOI: 10.1111/imm.12584] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 12/28/2015] [Accepted: 01/11/2016] [Indexed: 12/30/2022] Open
Abstract
There is controversy about the immunomodulatory effect of fibroblasts on dendritic cells (DCs). To clarify this issue, in this study, we have evaluated different features of fibroblast-primed DCs including their ability to express co-inhibitory and co-stimulatory molecules, pro-inflammatory and anti-inflammatory cytokines and their ability to induce T-cell proliferation. We also examined migratory capacity of DCs to lymphatic tissues and present fibroblast-derived antigens after encountering fibroblasts. The results of our in vitro study showed that both co-inhibitory (programmed death ligand 1 and ligand 2 and B7H4) and co-stimulatory (CD86) molecules were up-regulated when DCs were co-cultured with fibroblasts. In an animal model, we showed that intra- peritoneal injection (IP) of both syngeneic and allogeneic fibroblasts significantly increased both total DC count and expression level of co-inhibitory and co-stimulatory molecules on DCs. Priming of DCs with syngeneic and allogeneic fibroblasts reduced the proliferation of CD4(+) and CD8(+) T cells. Even activation of fibroblast- primed DCs failed to restore their ability to induce T-cell proliferation. Likewise, priming of DCs with fibroblasts blocked the ability of ovalbumin-pulsed DCs to induce proliferation of ovalbumin-specific CD4(+) T cells. Compared with non-activated DCs, fibroblast-primed DCs had significantly higher expression levels of interleukin-10 and indoleamine 2, 3 dioxygenase. Fibroblast-primed DCs had a significantly reduced interleukin-12 expression level compared with that of activated DCs. After priming with fibroblasts, DCs were able to migrate to lymphatic tissues and present fibroblast-derived antigens (ovalbumin). In conclusion, after priming with fibroblasts, DCs gain tolerogenic features. This finding suggests the potential role of fibroblasts in the maintenance of immune tolerance.
Collapse
Affiliation(s)
| | - MohammadReza Pakyari
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
| | - Reza B Jalili
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
| | - Sanam Salimi-Elizei
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
| | - Jacqueline C Y Lai
- Department of Dermatology and Skin Science, Faculty of Medicine, Child and Family Research Institute, The University of British Columbia, Vancouver, BC, Canada
| | | | - Ruhangiz T Kilani
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
| | - Jan Dutz
- Department of Dermatology and Skin Science, Faculty of Medicine, Child and Family Research Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Aziz Ghahary
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
73
|
Xu H, Chen X, Tao M, Chen K, Chen C, Xu G, Li W, Yuan S, Mao Y. B7-H3 and B7-H4 are independent predictors of a poor prognosis in patients with pancreatic cancer. Oncol Lett 2016; 11:1841-1846. [PMID: 26998087 DOI: 10.3892/ol.2016.4128] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 12/21/2015] [Indexed: 11/05/2022] Open
Abstract
B7-H3 and B7-H4 belong to the peripheral membrane protein B7 family and are hypothesized to regulate immunity. These proteins are expressed in human pancreatic cancer (PC), but their prognostic significance is poorly understood. The present study examined the association between B7-H3 and B7-H4 expression and the overall survival time in patients with PC that underwent surgery at the Second Affiliated Hospital to Zhengzhou University between April 2000 and January 2009. Immunohistochemical analysis demonstrated that B7-H3 and B7-H4 were expressed in 35 (88%) and 30 (75%) tumor tissue samples, respectively, which were obtained from 40 patients with PC. Statistical analysis revealed that B7-H3 expression was associated with an early tumor-node-metastasis stage (stage I and II; P<0.01), and B7-H4 expression was associated with tumors located in the body and tail of the pancreas (P<0.01) and lymph node metastasis (P=0.02). In addition, using Spearman's rank correlation coefficient, the present study demonstrated a positive correlation between B7-H3 expression and B7-H4 expression (r=0.37; P=0.02) in tumor samples. B7-H4 expression (P=0.01), tumors located in the pancreatic body and tail (P<0.01), lymph node metastasis (P=0.02) and combined B7-H3 and B7-H4 expression (P<0.01) were indicators of a poor overall survival time. However, solitary B7-H4 expression (P=0.03) and combined B7-H3 and B7-H4 expression (P=0.04) remained significant prognostic factors following adjustment for other prognostic factors in a multivariate Cox's proportional hazards regression model. Therefore, the present results indicate that solitary B7-H4 expression and a combination of B7-H3 and B7-H4 expression are independent predictors of a poor prognosis in patients with PC.
Collapse
Affiliation(s)
- Hong Xu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Xiaoling Chen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China; Department of Hematology and Oncology, Central Hospital of Zhabei District, Shanghai 200070, P.R. China
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Kai Chen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Chen Chen
- Department of Gastroenterology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Gang Xu
- Department of Gastroenterology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Suxu Yuan
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Yixiang Mao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| |
Collapse
|
74
|
Jin Y, Zhang P, Li J, Zhao J, Liu C, Yang F, Yang D, Gao A, Lin W, Ma X, Sun Y. B7-H3 in combination with regulatory T cell is associated with tumor progression in primary human non-small cell lung cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:13987-13995. [PMID: 26823710 PMCID: PMC4713496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 10/22/2015] [Indexed: 06/05/2023]
Abstract
B7-H3 belongs to the co-inhibitory B7 family and plays an important role in the adaptive immune response in regulating T cells. In human malignancies, B7-H3 is reported to be involved in tumor immune evasion. However, the detailed molecular mechanism of B7-H3 in tumor evasion remains unclear, particularly in non-small cell lung cancer (NSCLC). Regulatory T cells (Tregs) are known as a key player in the inhibition of immune mechanisms. The study demonstrated the correlation between B7-H3 on tumor cells and the number of Tregs in the tumor microenvironment in NSCLC. B7-H3 was examined in tumor tissues from 110 patients with NSCLC by immunohistochemical analysis. Forkhead box P3+ (FOXP3+) Tregs in those spencimens were also detected and numbered. Survival curves were drawn using the Kaplan-Meier method and compared by the log-rank test. High B7-H3 expression in tumor cells significantly correlated with male gender, squamous NSCLC, advanced stage and shorter overall survival (OS) (P = 0.035, P = 0.004, P = 0.037, P = 0.014, respectively). Meanwhile, FOXP3 expression in tumor-infiltrating lymphocytes (TILs) was associated with male gender, regional lymph node involvement, advanced stage and worse OS (P = 0.009, P = 0.015, P = 0.014, P = 0.034, respectively). Significant correlation was identified between the expression of B7-H3 and the number of FOXP3+ TILs (P = 0.013). Patients with B7-H3 high/FOXP3 high had poorer OS (P = 0.006), suggesting that B7-H3 and Tregs may play a cooperatively role in tumor immune evasion, leading to poor outcomes for NSCLC patients.
Collapse
Affiliation(s)
- Yingjie Jin
- Department of Oncology, Jinan Central Hospital, Shandong UniversityJinan 250013, Shandong, P. R. China
- Department of Oncology, Zhangqiu HospitalZhangqiu, 250200, Shandong, P. R. China
| | - Pei Zhang
- Department of Oncology, Jinan Central Hospital, Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Juan Li
- Department of Oncology, Jinan Central Hospital, Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Jianqiang Zhao
- Department of Oncology, Jinan Central Hospital, Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Chuanyong Liu
- Department of Oncology, Jinan Central Hospital, Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Fei Yang
- Department of Pathology, Jinan Central Hospital, Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Dong Yang
- Department of Oncology, Jinan Central Hospital, Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Aiqin Gao
- Department of Oncology, Jinan Central Hospital, Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Wenli Lin
- Department of Oncology, Jinan Central Hospital, Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Xiaoxia Ma
- Department of Gastroscopy Center, Jinan Central Hospital, Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Yuping Sun
- Department of Oncology, Jinan Central Hospital, Shandong UniversityJinan 250013, Shandong, P. R. China
| |
Collapse
|
75
|
Karachaliou N, Pilotto S, Teixidó C, Viteri S, González-Cao M, Riso A, Morales-Espinosa D, Molina MA, Chaib I, Santarpia M, Richardet E, Bria E, Rosell R. Melanoma: oncogenic drivers and the immune system. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:265. [PMID: 26605311 PMCID: PMC4630557 DOI: 10.3978/j.issn.2305-5839.2015.08.06] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 08/04/2015] [Indexed: 12/19/2022]
Abstract
Advances and in-depth understanding of the biology of melanoma over the past 30 years have contributed to a change in the consideration of melanoma as one of the most therapy-resistant malignancies. The finding that oncogenic BRAF mutations drive tumor growth in up to 50% of melanomas led to a molecular therapy revolution for unresectable and metastatic disease. Moving beyond BRAF, inactivation of immune regulatory checkpoints that limit T cell responses to melanoma has provided targets for cancer immunotherapy. In this review, we discuss the molecular biology of melanoma and we focus on the recent advances of molecularly targeted and immunotherapeutic approaches.
Collapse
|
76
|
Alegre ML, Chong AS. Costimulatory molecules. Transpl Immunol 2015. [DOI: 10.1002/9781119072997.ch4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
77
|
Liu J, Liu Y, Wang W, Wang C, Che Y. Expression of immune checkpoint molecules in endometrial carcinoma. Exp Ther Med 2015; 10:1947-1952. [PMID: 26640578 DOI: 10.3892/etm.2015.2714] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 08/10/2015] [Indexed: 02/01/2023] Open
Abstract
The main obstacle in the development of an effective tumor vaccine is the inherent ability of tumors to evade immune responses. Tumors often use common immune mechanisms and regulators to evade the immune system. The present study aimed to analyze the expression levels of indoleamine 2,3-dioxygenase (IDO), programmed death-ligand (PD-L) 1, PD-L2, B7-H4, galectin-1 and galectin-3 in tissue samples from patients with endometrial carcinoma, in order to detect the immunosuppressive environment of endometrial carcinomas. The levels of IDO, PD-L1, PD-L2 and B7-H4 were analyzed by immunohistochemical methods, and the levels of galectin-1 and galectin-3 in tumor lysates were determined using ELISA. PD-L2 was expressed at low levels in the majority of tumor samples. IDO expression was detected in 38, 63 and 43% of primary endometrial carcinoma, recurrent endometrial carcinoma, and metastatic endometrial carcinoma specimens, respectively. Positive expression rates for PD-L1 were 83% in primary endometrial carcinoma, 68% in recurrent endometrial carcinoma, and 100% in metastatic endometrial carcinoma, whereas B7-H4 expression was detected in 100% of both primary endometrial carcinoma and recurrent endometrial carcinoma samples, and in 96% of metastatic endometrial carcinoma specimens. The expression levels of galectin-1 and galectin-3 were not significantly different between the normal and tumor specimens. The results of the present study suggest that the interaction between PD-1/PD-L1 and B7-H4 may be a potential target for immune intervention in the treatment of endometrial carcinoma. Furthermore, the results may provide the basis for immunosuppressant therapy in the treatment of patients with uterine cancer.
Collapse
Affiliation(s)
- Jia Liu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Yuling Liu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Wuliang Wang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Chenyang Wang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Yanhong Che
- Department of Gynecology and Obstetrics, Women & Infants Hospital of Zhengzhou, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
78
|
Luo L, Zhu G, Xu H, Yao S, Zhou G, Zhu Y, Tamada K, Huang L, Flies AD, Broadwater M, Ruff W, van Deursen JMA, Melero I, Zhu Z, Chen L. B7-H3 Promotes Pathogenesis of Autoimmune Disease and Inflammation by Regulating the Activity of Different T Cell Subsets. PLoS One 2015; 10:e0130126. [PMID: 26065426 PMCID: PMC4465912 DOI: 10.1371/journal.pone.0130126] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 05/18/2015] [Indexed: 11/18/2022] Open
Abstract
B7-H3 is a cell surface molecule in the immunoglobulin superfamily that is frequently upregulated in response to autoantigens and pathogens during host T cell immune responses. However, B7-H3's role in the differential regulation of T cell subsets remains largely unknown. Therefore, we constructed a new B7-H3 deficient mouse strain (B7-H3 KO) and evaluated the functions of B7-H3 in the regulation of Th1, Th2, and Th17 subsets in experimental autoimmune encephalomyelitis (EAE), experimental asthma, and collagen-induced arthritis (CIA); these mouse models were used to predict human immune responses in multiple sclerosis, asthma, and rheumatoid arthritis, respectively. Here, we demonstrate that B7-H3 KO mice have significantly less inflammation, decreased pathogenesis, and limited disease progression in both EAE and CIA mouse models when compared with littermates; these results were accompanied by a decrease in IFN-γ and IL-17 production. In sharp contrast, B7-H3 KO mice developed severe ovalbumin (OVA)-induced asthma with characteristic infiltrations of eosinophils in the lung, increased IL-5 and IL-13 in lavage fluid, and elevated IgE anti-OVA antibodies in the blood. Our results suggest B7-H3 has a costimulatory function on Th1/Th17 but a coinhibitory function on Th2 responses. Our studies reveal that B7-H3 could affect different T cell subsets which have important implications for regulating pathogenesis and disease progression in human autoimmune disease.
Collapse
MESH Headings
- Animals
- Apoptosis
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Asthma/chemically induced
- Asthma/immunology
- Asthma/pathology
- B7 Antigens/physiology
- Blotting, Western
- Cell Differentiation
- Cell Proliferation
- Cells, Cultured
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Humans
- Immunoenzyme Techniques
- Inflammation/chemically induced
- Inflammation/immunology
- Inflammation/pathology
- Lymphocyte Activation
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Th1 Cells/immunology
- Th17 Cells/immunology
- Th2 Cells/immunology
Collapse
Affiliation(s)
- Liqun Luo
- Institute of Immunotherapy, Sun Yat-Sen University, Guangzhou, Guangdong, China
- * E-mail: (LL); (LC)
| | - Gefeng Zhu
- Department of Immunobiology and Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Haiying Xu
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Sheng Yao
- Department of Immunobiology and Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Gang Zhou
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Yuwen Zhu
- Department of Immunobiology and Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Koji Tamada
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Lanqing Huang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Andrew D. Flies
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Megan Broadwater
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - William Ruff
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jan M. A. van Deursen
- Department of Pediatrics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ignacio Melero
- Department of Medicine, University of Navarra, Pamplona, Spain
| | - Zhou Zhu
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Lieping Chen
- Department of Immunobiology and Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail: (LL); (LC)
| |
Collapse
|
79
|
Abstract
A number of consensuses regarding cancer immunology have recently emerged from both preclinical immunotherapy models and analysis of cancer patients. First and foremost, the natural state of endogenous tumor reactive T cells is characterized by general hyporesponsiveness or anergy. This is likely due to a number of mechanisms that tumors use to induce tolerance as they develop. While many of the newer generation vaccines can effectively transfer antigen to and activate dendritic cells, T-cell tolerance remains a major barrier that is difficult to overcome by vaccination alone. Preclinical models demonstrate that for poorly immunogenic tumors, once tolerance has been established, therapeutic vaccines alone are ineffective at curing animals with a significant established tumor burden. However, combination strategies of vaccination together with inhibitors of immunologic checkpoints and agonists for co-stimulatory pathways are proving capable of overcoming tolerance and generating significant anti-tumor responses even in cases of established metastatic cancer.
Collapse
Affiliation(s)
- Drew Pardoll
- Department of Oncology and Director Cancer Immunology Program, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD.
| |
Collapse
|
80
|
Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 2015; 27:450-61. [PMID: 25858804 PMCID: PMC4400238 DOI: 10.1016/j.ccell.2015.03.001] [Citation(s) in RCA: 3151] [Impact Index Per Article: 315.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/17/2015] [Accepted: 03/03/2015] [Indexed: 02/08/2023]
Abstract
The immune system recognizes and is poised to eliminate cancer but is held in check by inhibitory receptors and ligands. These immune checkpoint pathways, which normally maintain self-tolerance and limit collateral tissue damage during anti-microbial immune responses, can be co-opted by cancer to evade immune destruction. Drugs interrupting immune checkpoints, such as anti-CTLA-4, anti-PD-1, anti-PD-L1, and others in early development, can unleash anti-tumor immunity and mediate durable cancer regressions. The complex biology of immune checkpoint pathways still contains many mysteries, and the full activity spectrum of checkpoint-blocking drugs, used alone or in combination, is currently the subject of intense study.
Collapse
Affiliation(s)
- Suzanne L Topalian
- Department of Surgery, Sidney Kimmel Comprehensive Cancer Center and Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Charles G Drake
- The Brady Urological Institute, Sidney Kimmel Comprehensive Cancer Center and Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center and Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Drew M Pardoll
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center and Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
81
|
B7-H3 expression in donor T cells and host cells negatively regulates acute graft-versus-host disease lethality. Blood 2015; 125:3335-46. [PMID: 25814530 DOI: 10.1182/blood-2014-09-603357] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 03/10/2015] [Indexed: 12/18/2022] Open
Abstract
Members of the B7 family have been shown to be important for regulating immune responses by providing either positive or negative costimulatory signals. The function of B7-H3 has been controversial. We show that B7-H3 is upregulated in graft-versus-host disease (GVHD) target organs, including the colon, liver, and lung. Infusion of allogeneic donor T cells into B7-H3(-/-) vs wild-type (WT) recipients resulted in increased GVHD lethality associated with increased T-cell proliferation, colonic inflammatory cytokines, and destruction of epithelial barriers. Allogeneic B7-H3(-/-) vs WT donor T cells also had increased T-cell proliferation and GVHD lethality associated with increased proliferation and cytokine secretion in the spleen, intraepithelial lymphocyte inflammatory cytokines, and intestinal permeability. Both resting and activated regulatory T cells (Tregs) lack B7-H3 messenger RNA. Consistent with these data, GVHD was augmented in recipients of B7-H3(-/-) Treg-depleted grafts. In two delayed lymphocyte infusion (DLI) models, T cells lacking B7-H3 are capable of providing graft-versus-leukemia (GVL) effects. We conclude that B7-H3 is responsible for providing a negative costimulatory signal. Our studies provide support for developing and testing new therapies directed toward the B7-H3 pathway, including approaches to augment host B7-H3 early after bone marrow transplantation to prevent GVHD and to develop potent antagonistic antibodies later after transplant to facilitate DLI-mediated GVL without GVHD complications.
Collapse
|
82
|
Immunological mechanisms controlling hepatitis C virus infection. ASIAN PACIFIC JOURNAL OF TROPICAL DISEASE 2015. [DOI: 10.1016/s2222-1808(14)60633-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
83
|
Shin DS, Ribas A. The evolution of checkpoint blockade as a cancer therapy: what's here, what's next? Curr Opin Immunol 2015; 33:23-35. [PMID: 25621841 DOI: 10.1016/j.coi.2015.01.006] [Citation(s) in RCA: 259] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 01/09/2015] [Accepted: 01/11/2015] [Indexed: 12/22/2022]
Abstract
Unleashing the immune system to fight cancer has become one of the main treatment modalities since the anti-CTLA-4 antibody, ipilimumab was approved for patients with advanced melanoma in 2011. Pembrolizumab and nivolumab, two anti-PD-1 antibodies recently approved for the treatment of patients with metastatic melanoma, are being actively investigated for the treatment of multiple caners including lung, breast, bladder and renal cancers along with other anti-PD-1/L1 antibodies. Early results of combining of anti-CTLA-4 antibody and anti-PD-1 antibody treatment for advanced melanoma patients are showing impressive response rates with manageable toxicity profiles. There are several other checkpoint molecules that are likely potential inhibitory targets. The outcome of blocking some of these negative immune regulators, such as LAG-3 or TIM-3, is being pursued in the clinic or about to enter clinical development. Blockade of these molecules is demonstrating promising preclinical activity alone or when combined with anti-PD-1/L1. Future studies will define bio-markers of these therapies and how to target them alone or in combination with other immunotherapies, chemotherapy, radiotherapy and small molecule inhibitors.
Collapse
Affiliation(s)
- Daniel Sanghoon Shin
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Department of Molecular, Cellular and Integrative Physiology, UCLA, Los Angeles, CA, USA
| | - Antoni Ribas
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA; Department of Surgery, Division of Surgical-Oncology, UCLA, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center at UCLA, 10833 Le Conte Avenue, Los Angeles, CA 90095-1782, USA; Department of Molecular, Cellular and Integrative Physiology, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
84
|
Rahbar R, Lin A, Ghazarian M, Yau HL, Paramathas S, Lang PA, Schildknecht A, Elford AR, Garcia-Batres C, Martin B, Berman HK, Leong WL, McCready DR, Reedijk M, Done SJ, Miller N, Youngson B, Suh WK, Mak TW, Ohashi PS. B7-H4 expression by nonhematopoietic cells in the tumor microenvironment promotes antitumor immunity. Cancer Immunol Res 2014; 3:184-95. [PMID: 25527357 DOI: 10.1158/2326-6066.cir-14-0113] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The B7 family plays a critical role in both positive and negative regulation of immune responses by engaging a variety of receptors on lymphocytes. Importantly, blocking coinhibitory molecules using antibodies specific for CTLA-4 and PD-1 enhances tumor immunity in a subset of patients. Therefore, it is critical to understand the role of different B7 family members since they may be suitable therapeutic targets. B7-H4 is another member that inhibits T-cell function, and it is also upregulated on a variety of tumors and has been proposed to promote tumor growth. Here, we investigate the role of B7-H4 in tumor development and show that B7-H4 expression inhibits tumor growth in two mouse models. Furthermore, we show that B7-H4 expression is required for antitumor immune responses in a mouse model of mammary tumorigenesis. We found that the expression levels of B7-H4 correlate with MHC class I expression in both mouse and human samples. We show that IFNγ upregulates B7-H4 expression on mouse embryo fibroblasts and that the upregulation of B7-H4 on tumors is dependent on T cells. Notably, patients with breast cancer with increased B7-H4 expression show a prolonged time to recurrence. These studies demonstrate a positive role for B7-H4 in promoting antitumor immunity.
Collapse
Affiliation(s)
- Ramtin Rahbar
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Albert Lin
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Magar Ghazarian
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Helen-Loo Yau
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Sangeetha Paramathas
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Philipp A Lang
- Department of Molecular Medicine II, Heinrich Heine University Dösseldorf, Dösseldorf, Germany
| | - Anita Schildknecht
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Alisha R Elford
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Carlos Garcia-Batres
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Bernard Martin
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Hal K Berman
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Laboratory Medicine Program, University Health Network (UHN), Toronto, Ontario, Canada. Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Wey L Leong
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - David R McCready
- Department of Surgical Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Michael Reedijk
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Susan J Done
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Laboratory Medicine Program, University Health Network (UHN), Toronto, Ontario, Canada. Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Naomi Miller
- Laboratory Medicine Program, University Health Network (UHN), Toronto, Ontario, Canada. Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Bruce Youngson
- Laboratory Medicine Program, University Health Network (UHN), Toronto, Ontario, Canada. Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Woong-Kyung Suh
- Immune Regulation Laboratory, Institut de Recherches Cliniques de Montreal (IRCM), Montreal, Quebec, Canada
| | - Tak W Mak
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Immunology, University of Toronto, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Pamela S Ohashi
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Immunology, University of Toronto, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
85
|
Byers JT, Paniccia A, Kaplan J, Koenig M, Kahn N, Wilson L, Chen L, Schulick RD, Edil BH, Zhu Y. Expression of the Novel Costimulatory Molecule B7-H5 in Pancreatic Cancer. Ann Surg Oncol 2014; 22 Suppl 3:S1574-9. [DOI: 10.1245/s10434-014-4293-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Indexed: 11/18/2022]
|
86
|
Pelham CJ, Pandya AN, Agrawal DK. Triggering receptor expressed on myeloid cells receptor family modulators: a patent review. Expert Opin Ther Pat 2014; 24:1383-95. [PMID: 25363248 DOI: 10.1517/13543776.2014.977865] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Triggering receptor expressed on myeloid cells (TREM) receptors and TREM-like transcript (TLT; or TREML) receptors of the immunoglobulin superfamily are known as key modulators of host immune responses. TREM-1 (CD354) and TREM-2 share the transmembrane adaptor DNAX-activation protein of 12 kDa (DAP12), but they possess separate stimulatory and inhibitory functional roles, especially in myeloid cells. AREAS COVERED This review covers findings related to TREMs and TLTs published in patent applications from their discovery in 2000 to the present. New roles for TREM-1, TREM-2, TLT-1 and TLT-2 in maladies ranging from acute and chronic inflammatory disorders to cardiovascular diseases and cancers are discussed. Putative endogenous ligands and novel synthetic peptide blockers are also discussed. EXPERT OPINION So far, therapeutic use of activators/blockers specific for TREMs and TLTs has been limited to preclinical animal models. TREM-1 is an immediate therapeutic target for acute and chronic inflammatory conditions, especially sepsis. Certain mutations in DAP12 and TREM-2 manifest into a disorder named polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy, and newly identified TREM-2 variants confer a significant increase in risk of developing Alzheimer's disease. This makes TREM-2 an attractive therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Christopher J Pelham
- Creighton University School of Medicine, Department of Biomedical Sciences and Center for Clinical & Translational Science , Omaha, NE 68178 , USA
| | | | | |
Collapse
|
87
|
Yoon BR, Yoo SJ, Choi YH, Chung YH, Kim J, Yoo IS, Kang SW, Lee WW. Functional phenotype of synovial monocytes modulating inflammatory T-cell responses in rheumatoid arthritis (RA). PLoS One 2014; 9:e109775. [PMID: 25329467 PMCID: PMC4201467 DOI: 10.1371/journal.pone.0109775] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 09/11/2014] [Indexed: 12/29/2022] Open
Abstract
Monocytes function as crucial innate effectors in the pathogenesis of chronic inflammatory diseases, including autoimmunity, as well as in the inflammatory response against infectious pathogens. Human monocytes are heterogeneous and can be classified into three distinct subsets based on CD14 and CD16 expression. Although accumulating evidence suggests distinct functions of monocyte subsets in inflammatory conditions, their pathogenic roles in autoimmune diseases remain unclear. Thus, we investigated the phenotypic and functional characteristics of monocytes derived from synovial fluid and peripheral blood in RA patients in order to explore the pathogenic roles of these cells. In RA patients, CD14+CD16+, but not CD14dimCD16+, monocytes are predominantly expanded in synovial fluid and, to a lesser degree, in peripheral blood. Expression of co-signaling molecules of the B7 family, specifically CD80 and CD276, was markedly elevated on synovial monocytes, while peripheral monocytes of RA and healthy controls did not express these molecules without stimulation. To explore how synovial monocytes might gain these unique properties in the inflammatory milieu of the synovial fluid, peripheral monocytes were exposed to various stimuli. CD16 expression on CD14+ monocytes was clearly induced by TGF-β, although co-treatment with IL-1β, TNF-α, or IL-6 did not result in any additive effects. In contrast, TLR stimulation with LPS or zymosan significantly downregulated CD16 expression such that the CD14+CD16+ monocyte subset could not be identified. Furthermore, treatment of monocytes with IFN-γ resulted in the induction of CD80 and HLA-DR expression even in the presence of TGF-β. An in vitro assay clearly showed that synovial monocytes possess the unique capability to promote Th1 as well as Th17 responses of autologous peripheral CD4 memory T cells. Our findings suggest that the cytokine milieu of the synovial fluid shapes the unique features of synovial monocytes as well as their cardinal role in shaping inflammatory T-cell responses in RA.
Collapse
Affiliation(s)
- Bo Ruem Yoon
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Su-Jin Yoo
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejon, Korea
| | - Yeon ho Choi
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Yeon-Ho Chung
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Jinhyun Kim
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejon, Korea
| | - In Seol Yoo
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejon, Korea
| | - Seong Wook Kang
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejon, Korea
- * E-mail: (WWL); (SWK)
| | - Won-Woo Lee
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Ischemic/Hypoxic Disease Institute, and Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul, Korea
- * E-mail: (WWL); (SWK)
| |
Collapse
|
88
|
Wang L, Le Mercier I, Putra J, Chen W, Liu J, Schenk AD, Nowak EC, Suriawinata AA, Li J, Noelle RJ. Disruption of the immune-checkpoint VISTA gene imparts a proinflammatory phenotype with predisposition to the development of autoimmunity. Proc Natl Acad Sci U S A 2014; 111:14846-14851. [PMID: 25267631 PMCID: PMC4205642 DOI: 10.1073/pnas.1407447111] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
V domain-containing Ig suppressor of T-cell activation (VISTA) is a negative checkpoint regulator that suppresses T cell-mediated immune responses. Previous studies using a VISTA-neutralizing monoclonal antibody show that VISTA blockade enhances T-cell activation. The current study describes a comprehensive characterization of mice in which the gene for VISTA has been deleted. Despite the apparent normal hematopoietic development in young mice, VISTA genetic deficiency leads to a gradual accumulation of spontaneously activated T cells, accompanied by the production of a spectrum of inflammatory cytokines and chemokines. Enhanced T-cell responsiveness was also observed upon immunization with neoantigen. Despite the presence of multiorgan chronic inflammation, aged VISTA-deficient mice did not develop systemic or organ-specific autoimmune disease. Interbreeding of the VISTA-deficient mice with 2D2 T-cell receptor transgenic mice, which are predisposed to the development of experimental autoimmune encephalomyelitis, drastically enhanced disease incidence and intensity. Disease development is correlated with the increase in the activation of encephalitogenic T cells in the periphery and enhanced infiltration into the CNS. Taken together, our data suggest that VISTA is a negative checkpoint regulator whose loss of function lowers the threshold for T-cell activation, allowing for an enhanced proinflammatory phenotype and an increase in the frequency and intensity of autoimmunity under susceptible conditions.
Collapse
Affiliation(s)
- Li Wang
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226;
| | | | | | - Wenna Chen
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Jun Liu
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Austin D Schenk
- Surgery, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | | | | | - Jiannan Li
- Departments of Microbiology and Immunology
| | - Randolph J Noelle
- Departments of Microbiology and Immunology, Medical Research Council Centre of Transplantation, Guy's Hospital, King's College London, King's Health Partners, London SE1 9RT, United Kingdom
| |
Collapse
|
89
|
Guo G, Cao D, Xu H, Ruan Z, Fei L, Xie Z, Wu Y, Chen Y. The characteristic expression of B7-H3 and B7-H4 in liver biopsies from patients with HBV-related acute-on-chronic liver failure. Pathol Int 2014; 62:665-74. [PMID: 23005593 DOI: 10.1111/j.1440-1827.2012.02856.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Hepatitis B virus (HBV) is a major public health problem, and HBV-related acute-on-chronic liver failure (HBV-ACLF) has an extremely poor prognosis due to a lack of effective treatments. B7-H3 and B7-H4 are two novel members of the B7 superfamily that are actively involved in regulating the pathogenesis of infectious diseases. However, the intrahepatic expression of both members in HBV-ACLF patients has yet to be described. In this study, we analyzed the expression of B7-H3 and B7-H4 in HBV-ACLF biopsies by immunohistochemistry. Our results showed that both members were observed in all HBV-ACLF samples, and their expression was chiefly observed on infiltrating inflammatory cells and the damaged bile ducts. Immunofluorescence double staining showed that B7-H4 was expressed chiefly on CD3(+) T cells, CD68(+) macrophages, CK-18(+) bile ducts, and CD31(+) endothelial cells, while B7-H3 was found on all cell types detected. The expression of the programmed death (PD)-1 ligands, PD-L1 and PD-L2, was also detected in these liver tissues and they were found to be co-expressed with B7-H3 and B7-H4. These results suggest that the B7-family signaling is most likely to affect the pathogenesis of this disease, and a clear understanding of their functional roles may further elucidate the disease process.
Collapse
Affiliation(s)
- Guoning Guo
- Department of Emergency, South-West Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Zhang N, Fang P, Gu ZJ. Preparation and Characterization of Monoclonal Antibody Against Human B7-H4 Molecule. Monoclon Antib Immunodiagn Immunother 2014; 33:270-4. [DOI: 10.1089/mab.2013.0082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Ning Zhang
- Department of Immunology, School of Medicine, Soochow University, Suzhou, China
| | - Peng Fang
- Department of Immunology, School of Medicine, Soochow University, Suzhou, China
| | - Zong-Jiang Gu
- Department of Immunology, School of Medicine, Soochow University, Suzhou, China
- Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
91
|
Mapping the immunosuppressive environment in uterine tumors: implications for immunotherapy. Cancer Immunol Immunother 2014; 63:545-57. [PMID: 24658839 PMCID: PMC4024136 DOI: 10.1007/s00262-014-1537-8] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 03/08/2014] [Indexed: 01/22/2023]
Abstract
The major hurdle for cancer vaccines to be effective is posed by tumor immune evasion. Several common immune mechanisms and mediators are exploited by tumors to avoid immune destruction. In an attempt to shed more light on the immunosuppressive environment in uterine tumors, we analyzed the presence of PD-L1, PD-L2, B7-H4, indoleamine 2,3-dioxygenase (IDO), galectin-1, galectin-3, arginase-1 activity and myeloid-derived suppressor cell (MDSC) infiltration. IDO, PD-L1, PD-L2 and B7-H4 were analyzed by immunohistochemistry. PD-L2 was mostly expressed at low levels in these tumors. We found high IDO expression in 21 % of endometrial carcinoma samples and in 14 % of uterine sarcoma samples. For PD-L1 and B7-H4, we found high expression in 92 and 90 % of endometrial cancers, respectively, and in 100 and 92 % of the sarcomas. Galectin-1 and 3 were analyzed in tissue lysates by ELISA, but we did not find an increase in both molecules in tumor lysates compared with benign tissues. We detected expression of galectin-3 by fibroblasts, immune cells and tumor cells in single-cell tumor suspensions. In addition, we noted a highly significant increase in arginase-1 activity in endometrial carcinomas compared with normal endometria, which was not the case for uterine sarcomas. Finally, we could demonstrate MDSC infiltration in fresh tumor suspensions from uterine tumors. These results indicate that the PD-1/PD-L1 interaction and B7-H4 could be possible targets for immune intervention in uterine cancer patients as well as mediation of MDSC function. These observations are another step toward the implementation of inhibitors of immunosuppression in the treatment of uterine cancer patients.
Collapse
|
92
|
VSIG4 expression on macrophages facilitates lung cancer development. J Transl Med 2014; 94:706-15. [PMID: 24862966 DOI: 10.1038/labinvest.2014.73] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 04/17/2014] [Accepted: 04/24/2014] [Indexed: 01/22/2023] Open
Abstract
Tumor-associated macrophages are a prominent component of lung cancer stroma and contribute to tumor progression. The protein V-set and Ig domain-containing 4 (VSIG4), a novel B7 family-related macrophage protein that has the capacity to inhibit T-cell activation, has a potential role in the development of lung cancer. In this study, 10 human non-small-cell lung cancer specimens were collected and immunohistochemically analyzed for VSIG4 expression. Results showed massive VSIG4(+) cell infiltration throughout the samples. Immunofluorescent double staining showed that VSIG4 was present on CD68(+) macrophages, but absent from CD3(+) T cells, CD31(+) endothelial cells, and CK-18(+) epithelial cells. Moreover, VSIG4 was coexpressed on B7-H1(+) and B7-H3(+) cells in these tumor specimens. Transfection of the VSIG4 gene into 293FT cells demonstrated that the VSIG4 signal could inhibit cocultured CD4(+) and CD8(+) T-cell proliferation and cytokine (IL-2 and IFN-γ) production in vitro. Interestingly, in a murine tumor model induced by Lewis lung carcinoma cell line, we found that tumors grown in VSIG4-deficient (VSIG4(-/-)) mice were significantly smaller than those found in wild-type littermates. All of these results demonstrate that macrophage-associated VSIG4 is an activator that facilitates lung carcinoma development. Specific targeting of VSIG4 may prove to be a novel, efficacious strategy for the treatment of this carcinoma.
Collapse
|
93
|
Lines JL, Sempere LF, Broughton T, Wang L, Noelle R. VISTA is a novel broad-spectrum negative checkpoint regulator for cancer immunotherapy. Cancer Immunol Res 2014; 2:510-7. [PMID: 24894088 PMCID: PMC4085258 DOI: 10.1158/2326-6066.cir-14-0072] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the past few years, the field of cancer immunotherapy has made great progress and is finally starting to change the way cancer is treated. We are now learning that multiple negative checkpoint regulators (NCR) restrict the ability of T-cell responses to effectively attack tumors. Releasing these brakes through antibody blockade, first with anti-CTLA4 and now followed by anti-PD1 and anti-PDL1, has emerged as an exciting strategy for cancer treatment. More recently, a new NCR has surfaced called V-domain immunoglobulin (Ig)-containing suppressor of T-cell activation (VISTA). This NCR is predominantly expressed on hematopoietic cells, and in multiple murine cancer models is found at particularly high levels on myeloid cells that infiltrated the tumors. Preclinical studies with VISTA blockade have shown promising improvement in antitumor T-cell responses, leading to impeded tumor growth and improved survival. Clinical trials support combined anti-PD1 and anti-CTLA4 as safe and effective against late-stage melanoma. In the future, treatment may involve combination therapy to target the multiple cell types and stages at which NCRs, including VISTA, act during adaptive immune responses.
Collapse
Affiliation(s)
- J Louise Lines
- Authors' Affiliations: Medical Research Council Centre of Transplantation, Guy's Hospital; Department of Immune Regulation and Intervention, King's College, King's Health Partners, London, United Kingdom; Van Andel Research Institute, Grand Rapids, Michigan; Departments of Medicine and Microbiology and Immunology, and Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire; and Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WisconsinAuthors' Affiliations: Medical Research Council Centre of Transplantation, Guy's Hospital; Department of Immune Regulation and Intervention, King's College, King's Health Partners, London, United Kingdom; Van Andel Research Institute, Grand Rapids, Michigan; Departments of Medicine and Microbiology and Immunology, and Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire; and Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Lorenzo F Sempere
- Authors' Affiliations: Medical Research Council Centre of Transplantation, Guy's Hospital; Department of Immune Regulation and Intervention, King's College, King's Health Partners, London, United Kingdom; Van Andel Research Institute, Grand Rapids, Michigan; Departments of Medicine and Microbiology and Immunology, and Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire; and Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Thomas Broughton
- Authors' Affiliations: Medical Research Council Centre of Transplantation, Guy's Hospital; Department of Immune Regulation and Intervention, King's College, King's Health Partners, London, United Kingdom; Van Andel Research Institute, Grand Rapids, Michigan; Departments of Medicine and Microbiology and Immunology, and Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire; and Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WisconsinAuthors' Affiliations: Medical Research Council Centre of Transplantation, Guy's Hospital; Department of Immune Regulation and Intervention, King's College, King's Health Partners, London, United Kingdom; Van Andel Research Institute, Grand Rapids, Michigan; Departments of Medicine and Microbiology and Immunology, and Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire; and Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Li Wang
- Authors' Affiliations: Medical Research Council Centre of Transplantation, Guy's Hospital; Department of Immune Regulation and Intervention, King's College, King's Health Partners, London, United Kingdom; Van Andel Research Institute, Grand Rapids, Michigan; Departments of Medicine and Microbiology and Immunology, and Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire; and Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Randolph Noelle
- Authors' Affiliations: Medical Research Council Centre of Transplantation, Guy's Hospital; Department of Immune Regulation and Intervention, King's College, King's Health Partners, London, United Kingdom; Van Andel Research Institute, Grand Rapids, Michigan; Departments of Medicine and Microbiology and Immunology, and Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire; and Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WisconsinAuthors' Affiliations: Medical Research Council Centre of Transplantation, Guy's Hospital; Department of Immune Regulation and Intervention, King's College, King's Health Partners, London, United Kingdom; Van Andel Research Institute, Grand Rapids, Michigan; Departments of Medicine and Microbiology and Immunology, and Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire; and Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WisconsinAuthors' Affiliations: Medical Research Council Centre of Transplantation, Guy's Hospital; Department of Immune Regulation and Intervention, King's College, King's Health Partners, London, United Kingdom; Van Andel Research Institute, Grand Rapids, Michigan; Departments of Medicine and Microbiology and Immunology, and Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire; and Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WisconsinAuthors' Affiliations: Medical Research Council Centre of Transplantation, Guy's Hospital; Department of Immune Regulation and Intervention, King's College, King's Health Partners, London, United Kingdom; Van Andel Research Institute, Grand Rapids, Michigan; Departments of Medicine and Microbiology and Immunology, and Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire; and Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WisconsinAuthors' Affiliations: Medical Research Coun
| |
Collapse
|
94
|
Wachsmann MB, Pop LM, Vitetta ES. Pancreatic ductal adenocarcinoma: a review of immunologic aspects. J Investig Med 2014. [PMID: 22406516 DOI: 10.231/jim.0b013e31824a4d79] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
With the continued failures of both early diagnosis and treatment options for pancreatic cancer, it is now time to comprehensively evaluate the role of the immune system on the development and progression of pancreatic cancer. It is important to develop strategies that harness the molecules and cells of the immune system to treat this disease. This review will focus primarily on the role of immune cells in the development and progression of pancreatic ductal adenocarcinoma and to evaluate what is known about the interaction of immune cells with the tumor microenvironment and their role in tumor growth and metastasis. We will conclude with a brief discussion of therapy for pancreatic cancer and the potential role for immunotherapy. We hypothesize that the role of the immune system in tumor development and progression is tissue specific. Our hope is that better understanding of this process will lead to better treatments for this devastating disease.
Collapse
Affiliation(s)
- Megan B Wachsmann
- Masters Program in Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | |
Collapse
|
95
|
Esposito P, Grosjean F, Rampino T, Libetta C, Gregorini M, Fasoli G, Marchi G, Sileno G, Montagna F, Dal Canton A. Costimulatory pathways in kidney transplantation: pathogenetic role, clinical significance and new therapeutic opportunities. Int Rev Immunol 2014; 33:212-233. [PMID: 24127878 DOI: 10.3109/08830185.2013.829470] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2013] [Indexed: 02/05/2023]
Abstract
Costimulatory pathways play a key role in immunity, providing the second signal required for a full activation of adaptive immune response. Different costimulatory families (CD28, TNF-related, adhesion and TIM molecules), characterized by structural and functional analogies, have been described. Costimulatory molecules modulate T cell activation, B cell function, Ig production, cytokine release and many other processes, including atherosclerosis. Patients suffering from renal diseases present significant alterations of the costimulatory pathways, which might make them particularly liable to infections. These alterations are further pronounced in patients undergoing kidney transplantation. In these patients, different costimulatory patterns have been related to distinct clinical features. The importance that costimulation has gained during the last years has led to development of several pharmacological approaches to modulate this critical step in the immune activation. Different drugs, mainly monoclonal antibodies targeting various costimulatory molecules (i.e. anti-CD80, CTLA-4 fusion proteins, anti-CD154, anti-CD40, etc.) were designed and tested in both experimental and clinical studies. The results of these studies highlighted some criticisms, but also some promising findings and now costimulatory blockade is considered a suitable strategy, with belatacept (a CTLA-4 fusion protein) being approved as the first costimulatory blocker for use in renal transplantation. In this review, we summarize the current knowledge on costimulatory pathways in the setting of kidney transplantation. We describe the principal costimulatory molecule families, their role and clinical significance in patients undergoing renal transplantation and the new therapeutic approaches that have been developed to modulate the costimulatory pathways.
Collapse
Affiliation(s)
- Pasquale Esposito
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico S. Matteo and University of Pavia , Pavia , Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Abstract
B7-H4 is a ligand in the B7 costimulatory family, executing suppressive function on the immune system in many diseases, such as cancer, allograft rejection, and autoimmune diseases. The receptor for this molecule has yet to be clarified. The engagement of B7-H4 inhibits proliferation of immune cells by stopping the cell cycle at the G0/G1 phase and leads to apoptosis via the Fas/FasL pathway consequently accelerating tumor progression and alleviating allograft rejection. The pathogenic role of B7-H4 in tumors has been widely established, but few studies have focused on its function in other disorders. Here, we review recent advances in our understanding of B7-H4 biology in disease settings other than tumors and document the beneficial values to treat those diseases by targeting this molecule and related signaling pathways.
Collapse
|
97
|
Bauzon M, Hermiston T. Armed therapeutic viruses - a disruptive therapy on the horizon of cancer immunotherapy. Front Immunol 2014; 5:74. [PMID: 24605114 PMCID: PMC3932422 DOI: 10.3389/fimmu.2014.00074] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 02/11/2014] [Indexed: 12/17/2022] Open
Abstract
For the past 150 years cancer immunotherapy has been largely a theoretical hope that recently has begun to show potential as a highly impactful treatment for various cancers. In particular, the identification and targeting of immune checkpoints have given rise to exciting data suggesting that this strategy has the potential to activate sustained antitumor immunity. It is likely that this approach, like other anti-cancer strategies before it, will benefit from co-administration with an additional therapeutic and that it is this combination therapy that may generate the greatest clinical outcome for the patient. In this regard, oncolytic viruses are a therapeutic moiety that is well suited to deliver and augment these immune-modulating therapies in a highly targeted and economically advantageous way over current treatment. In this review, we discuss the blockade of immune checkpoints, how oncolytic viruses complement and extend these therapies, and speculate on how this combination will uniquely impact the future of cancer immunotherapy.
Collapse
Affiliation(s)
- Maxine Bauzon
- Bayer HealthCare, US Innovation Center, Biologics Research , San Francisco, CA , USA
| | - Terry Hermiston
- Bayer HealthCare, US Innovation Center, Biologics Research , San Francisco, CA , USA
| |
Collapse
|
98
|
Abstract
Melanoma have been shown to escape immune surveillance by different mechanisms such as loss of HLA class I antigens, upregulation of nonclassical HLA-G antigen and Fas, increased secretion of immune suppressive cytokines and metabolites as well as altered expression of co-stimulatory and coinhibitory signals. Recently, an important role of B7-H1 and B7-H4 in the immune escape of melanoma has been described. High mRNA and/or protein expression levels of these coinhibitory molecules were detected in both melanoma cell lines and melanoma lesions when compared to melanocytes. However, their clinical relevance is currently controversially discussed regarding a correlation of B7-H family members with tumor grading and staging as well as survival of patients in melanoma.
Collapse
Affiliation(s)
- Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
99
|
Vasaturo A, Di Blasio S, Peeters DGA, de Koning CCH, de Vries JM, Figdor CG, Hato SV. Clinical Implications of Co-Inhibitory Molecule Expression in the Tumor Microenvironment for DC Vaccination: A Game of Stop and Go. Front Immunol 2013; 4:417. [PMID: 24348481 PMCID: PMC3847559 DOI: 10.3389/fimmu.2013.00417] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 11/15/2013] [Indexed: 12/13/2022] Open
Abstract
The aim of therapeutic dendritic cell (DC) vaccines in cancer immunotherapy is to activate cytotoxic T cells to recognize and attack the tumor. T cell activation requires the interaction of the T cell receptor with a cognate major-histocompatibility complex-peptide complex. Although initiated by antigen engagement, it is the complex balance between co-stimulatory and co-inhibitory signals on DCs that results in T cell activation or tolerance. Even when already activated, tumor-specific T cells can be neutralized by the expression of co-inhibitory molecules on tumor cells. These and other immunosuppressive cues in the tumor microenvironment are major factors currently hampering the application of DC vaccination. In this review, we discuss recent data regarding the essential and complex role of co-inhibitory molecules in regulating the immune response within the tumor microenvironment. In particular, possible therapeutic intervention strategies aimed at reversing or neutralizing suppressive networks within the tumor microenvironment will be emphasized. Importantly, blocking co-inhibitory molecule signaling, often referred to as immune checkpoint blockade, does not necessarily lead to an effective activation of tumor-specific T cells. Therefore, combination of checkpoint blockade with other immune potentiating therapeutic strategies, such as DC vaccination, might serve as a synergistic combination, capable of reversing effector T cells immunosuppression while at the same time increasing the efficacy of T cell-mediated immunotherapies. This will ultimately result in long-term anti-tumor immunity.
Collapse
Affiliation(s)
- Angela Vasaturo
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands
| | - Stefania Di Blasio
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands
| | - Deborah G A Peeters
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands
| | - Coco C H de Koning
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands
| | - Jolanda M de Vries
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands ; Department of Medical Oncology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands
| | - Stanleyson V Hato
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands
| |
Collapse
|
100
|
Wang L, Kang FB, Shan BE. B7-H3-mediated tumor immunology: Friend or foe? Int J Cancer 2013; 134:2764-71. [PMID: 24013874 DOI: 10.1002/ijc.28474] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/29/2013] [Indexed: 12/22/2022]
Affiliation(s)
- Ling Wang
- Cancer Research Institute; The Fourth Hospital of Hebei Medical University; Shijiazhuang Hebei People's Republic of China
| | - Fu-Biao Kang
- Department of Liver Diseases; Bethune International Peace Hospital; Shijiazhuang Hebei People's Republic of China
| | - Bao-En Shan
- Cancer Research Institute; The Fourth Hospital of Hebei Medical University; Shijiazhuang Hebei People's Republic of China
| |
Collapse
|