11801
|
Abstract
Although immunotherapy has been at the forefront of cancer therapy for the last several years, better clinical responses are still desired. Interleukin-33 is perhaps one of the most overlooked antitumor cytokines. Its ability to promote type 1 immune responses, which control tumor growth in preclinical animal models is overshadowed by its association with type 2 immunity and poor prognosis in some human cancers. Accumulating evidence shows that IL-33 is a powerful new tool for restoring and enhancing the body's natural antitumor immunity cycle. Furthermore, the antitumor mechanisms of IL-33 are two-fold, as it can directly boost CD8+ T cell function and restore dendritic cell dysfunction in vivo. Mechanistic studies have identified a novel pathway induced by IL-33 and its receptor ST2 in which dendritic cells avoid dysfunction and retain cross-priming abilities in tumor-bearing conditions. Here, we also comment on IL-33 data in human cancers and explore the idea that endogenous IL-33 may not deserve its reputation for promoting tumor growth. In fact, tumors may hijack the IL-33/ST2 axis to avoid immune surveillance and escape antitumor immunity.
Collapse
Affiliation(s)
- Donye Dominguez
- Robert H. Lurie Comprehensive Cancer Center, Department of Medicine–Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bin Zhang
- Robert H. Lurie Comprehensive Cancer Center, Department of Medicine–Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
11802
|
Advances in esophageal cancer: A new perspective on pathogenesis associated with long non-coding RNAs. Cancer Lett 2018; 413:94-101. [DOI: 10.1016/j.canlet.2017.10.046] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/07/2017] [Accepted: 10/31/2017] [Indexed: 12/20/2022]
|
11803
|
|
11804
|
Liu J, Wang D, Zhang C, Zhang Z, Chen X, Lian J, Liu J, Wang G, Yuan W, Sun Z, Wang W, Song M, Wang Y, Wu Q, Cao L, Wang D, Zhang Y. Identification of liver metastasis-associated genes in human colon carcinoma by mRNA profiling. Chin J Cancer Res 2018; 30:633-646. [PMID: 30700932 PMCID: PMC6328509 DOI: 10.21147/j.issn.1000-9604.2018.06.08] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective Liver metastasis, which contributes substantially to high mortality, is the most common recurrent mode of colon carcinoma. Thus, it is necessary to identify genes implicated in metastatic colonization of the liver in colon carcinoma. Methods We compared mRNA profiling in 18 normal colon mucosa (N), 20 primary tumors (T) and 19 liver metastases (M) samples from the dataset GSE49355 and GSE62321 of Gene Expression Omnibus (GEO) database. Gene ontology (GO) and pathways of the identified genes were analyzed. Co-expression network and protein-protein interaction (PPI) network were employed to identify the interaction relationship. Survival analyses based on The Cancer Genome Atlas (TCGA) database were used to further screening. Then, the candidate genes were validated by our data. Results We identified 22 specific genes related to liver metastasis and they were strongly associated with cell migration, adhesion, proliferation and immune response. Simultaneously, the results showed that C-X-C motif chemokine ligand 14 (CXCL14) might be a favorable prediction factor for survival of patients with colon carcinoma. Importantly, our validated data further suggested that lower CXCL14 represented poorer outcome and contributed to metastasis. Gene set enrichment analysis (GSEA) showed that CXCL14 was negatively related to the regulation of stem cell proliferation and epithelial to mesenchymal transition (EMT). Conclusions CXCL14 was identified as a crucial anti-metastasis regulator of colon carcinoma for the first time, and might provide novel therapeutic strategies for colon carcinoma patients to improve prognosis and prevent metastasis.
Collapse
Affiliation(s)
- Jianling Liu
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Dan Wang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Chaoqi Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhen Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xinfeng Chen
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jingyao Lian
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jinbo Liu
- Department of Anorectal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Guixian Wang
- Department of Anorectal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Weitang Yuan
- Department of Anorectal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhenqiang Sun
- Department of Anorectal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Weijia Wang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Mengjia Song
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yaping Wang
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou 450052, China
| | - Qian Wu
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Ling Cao
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Dong Wang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yi Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
11805
|
Shi X, Shiao SL. The role of macrophage phenotype in regulating the response to radiation therapy. Transl Res 2018; 191:64-80. [PMID: 29175267 PMCID: PMC6018060 DOI: 10.1016/j.trsl.2017.11.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 10/23/2017] [Accepted: 11/11/2017] [Indexed: 12/14/2022]
Abstract
Increasing experimental and clinical evidence has revealed a critical role for myeloid cells in the development and progression of cancer. The ability of monocytes and macrophages to regulate inflammation allows them to manipulate the tumor microenvironment to support the growth and development of malignant cells. Recent studies have shown that macrophages can exist in several functional states depending on the microenvironment they encounter in the tissue. These functional phenotypes influence not only the genesis and propagation of tumors, but also the efficacy of cancer therapies, particularly radiation. Early classification of the macrophage phenotypes, or "polarization states," identified 2 major states, M1 and M2, that have cytotoxic and wound repair capacity, respectively. In the context of tumors, classically activated or M1 macrophages driven by interferon-gamma support antitumor immunity while alternatively activated or M2 macrophages generated in part from interleukin-4 exposure hinder antitumor immunity by suppressing cytotoxic responses against a tumor. In this review, we discuss the role that the functional phenotype of a macrophage population plays in tumor development. We will then focus specifically on how macrophages and myeloid cells regulate the tumor response to radiation therapy.
Collapse
Affiliation(s)
- Xiaoshan Shi
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Stephen L Shiao
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA.
| |
Collapse
|
11806
|
Bendriss-Vermare N, Gourdin N, Vey N, Faget J, Sisirak V, Labidi-Galy I, Le Mercier I, Goutagny N, Puisieux I, Ménétrier-Caux C, Caux C. Plasmacytoid DC/Regulatory T Cell Interactions at the Center of an Immunosuppressive Network in Breast and Ovarian Tumors. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
11807
|
Panjwani PK, Charu V, DeLisser M, Molina-Kirsch H, Natkunam Y, Zhao S. Programmed death-1 ligands PD-L1 and PD-L2 show distinctive and restricted patterns of expression in lymphoma subtypes. Hum Pathol 2018; 71:91-99. [DOI: 10.1016/j.humpath.2017.10.029] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/22/2017] [Accepted: 10/25/2017] [Indexed: 12/13/2022]
|
11808
|
Xiang T, Bai JY, She C, Yu DJ, Zhou XZ, Zhao TL. Bromodomain protein BRD4 promotes cell proliferation in skin squamous cell carcinoma. Cell Signal 2018; 42:106-113. [DOI: 10.1016/j.cellsig.2017.10.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/16/2017] [Accepted: 10/16/2017] [Indexed: 12/27/2022]
|
11809
|
Backer RA, Hombrink P, Helbig C, Amsen D. The Fate Choice Between Effector and Memory T Cell Lineages: Asymmetry, Signal Integration, and Feedback to Create Bistability. Adv Immunol 2018; 137:43-82. [DOI: 10.1016/bs.ai.2017.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
11810
|
Göpfert C, Andreas N, Weber F, Häfner N, Yakovleva T, Gaestel M, Kamradt T, Drube S. The p38-MK2/3 Module Is Critical for IL-33-Induced Signaling and Cytokine Production in Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2017; 200:1198-1206. [PMID: 29288203 DOI: 10.4049/jimmunol.1700727] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 11/30/2017] [Indexed: 12/20/2022]
Abstract
IL-33 is an IL-1 cytokine superfamily member. Binding of IL-33 to the IL-33R induces activation of the canonical NF-κB signaling and activation of MAPKs. In bone marrow-derived dendritic cells, IL-33 induces the production of IL-6, IL-13, and TNF-α. However, the signaling pathways resulting in IL-33-induced effector functions of dendritic cells are unknown. In this article, we show that the IL-33-induced cytokine production is only partly dependent on p65. Thereby, p65 mediates the production of IL-6, but not of IL-13, whereas the p38-Mapk-activated protein kinases 2/3 (MK2/3) signaling module mediates the IL-13, but not the IL-6, production. In addition, GM-CSF, which is critical for the differentiation and proliferation of bone marrow-derived dendritic cells, potentiates the p65-dependent IL-6 and the p38-MK2/3-dependent IL-13 production. Furthermore, we found that effective TNF-α production is only induced in the presence of GM-CSF and IL-33 via the p38-MK2/3 signaling module. Taken together, we found that the p38-MK2/3 signaling module is essential to mediate IL-33-induced cytokine production in dendritic cells.
Collapse
Affiliation(s)
| | - Nico Andreas
- Institute of Immunology, Jena University Hospital, 07743 Jena, Germany
| | - Franziska Weber
- Institute of Immunology, Jena University Hospital, 07743 Jena, Germany
| | - Norman Häfner
- Department of Gynecology, Jena University Hospital, Friedrich-Schiller University, 07743 Jena, Germany; and
| | - Tatiana Yakovleva
- Institute for Cell Biochemistry, Medical School Hannover, 30625 Hannover, Germany
| | - Matthias Gaestel
- Institute for Cell Biochemistry, Medical School Hannover, 30625 Hannover, Germany
| | - Thomas Kamradt
- Institute of Immunology, Jena University Hospital, 07743 Jena, Germany
| | - Sebastian Drube
- Institute of Immunology, Jena University Hospital, 07743 Jena, Germany;
| |
Collapse
|
11811
|
Prognostic significance of programmed death-1 and programmed death-ligand 1 expression in patients with esophageal squamous cell carcinoma. Oncotarget 2017; 7:30772-80. [PMID: 27120796 PMCID: PMC5058716 DOI: 10.18632/oncotarget.8956] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 04/02/2016] [Indexed: 12/30/2022] Open
Abstract
Aims To evaluate the expression of programmed death-1 (PD-1) and programmed death-ligand 1 (PD-L1) and their clinical and prognostic significance in primary esophageal squamous cell carcinoma (ESCC). Results The expression rate of PD-1 and PD-L1 in ESCC was 33.5% (117/349) and 41.4% (222/536), respectively. PD-L1 expression differed significantly by tumor location, grade, lymph node metastases, and disease stage (P < 0.05). Moreover, its expression was associated with the disease free survival (DFS). Patients with positive PD-L1 expression had reduced risk for disease relapse compared to those without PD-L1 expression (Hazard ratio [HR] = 0.75, 95% confidence interval [CI]: 0.56–1.00, P = 0.048). Kaplan-Meier curves showed the similar result, P = 0.047. However, there was no significant correlation between PD-1 expression and clinicopathological factors or outcome in ESCC (P > 0.05). Methods The expression of PD-1 and PD-L1 was assessed by immunohistochemistry on tissue microarrays from 536 primary ESCC who underwent surgery during January 2008 and April 2012 in Zhejiang Cancer Hospital. Chi-square test and Cox proportional hazards regression were employed to analyze the associations between their expressions and clinicopathological variables and survival. Conclusions Our results suggested that PD-L1 could be a favorable indicator of prognosis in ESCC.
Collapse
|
11812
|
Grigor EJM, Fergusson DA, Haggar F, Kekre N, Atkins H, Shorr R, Holt RA, Hutton B, Ramsay T, Seftel M, Jonker D, Daugaard M, Thavorn K, Presseau J, Lalu MM. Efficacy and safety of chimeric antigen receptor T-cell (CAR-T) therapy in patients with haematological and solid malignancies: protocol for a systematic review and meta-analysis. BMJ Open 2017; 7:e019321. [PMID: 29288188 PMCID: PMC5988064 DOI: 10.1136/bmjopen-2017-019321] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/11/2017] [Accepted: 11/01/2017] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Patients with relapsed or refractory malignancies have a poor prognosis. Immunotherapy with chimeric antigen receptor T (CAR-T) cells redirects a patient's immune cells against the tumour antigen. CAR-T cell therapy has demonstrated promise in treating patients with several haematological malignancies, including acute B-cell lymphoblastic leukaemia and B-cell lymphomas. CAR-T cell therapy for patients with other solid tumours is also being tested. Safety is an important consideration in CAR-T cell therapy given the potential for serious adverse events, including death. Previous reviews on CAR-T cell therapy have been limited in scope and methodology. Herein, we present a protocol for a systematic review to identify CAR-T cell interventional studies and examine the safety and efficacy of this therapy in patients with haematology malignancies and solid tumours. METHODS AND ANALYSIS We will search MEDLINE, including In-Process and Epub Ahead of Print, EMBASE and the Cochrane Central Register of Controlled Trials from 1946 to 22 February 2017. Studies will be screened by title, abstract and full text independently and in duplicate. Studies that report administering CAR-T cells of any chimeric antigen receptor construct targeting antigens in patients with haematological malignancies and solid tumours will be eligible for inclusion. Outcomes to be extracted will include complete response rate (primary outcome), overall response rate, overall survival, relapse and adverse events. A meta-analysis will be performed to synthesise the prevalence of outcomes reported as proportions with 95% CIs. The potential for bias within included studies will be assessed using a modified Institute of Health Economics tool. Heterogeneity of effect sizes will be determined using the Cochrane I 2 statistic. ETHICS AND DISSEMINATION The review findings will be submitted for peer-reviewed journal publication and presented at relevant conferences and scientific meetings to promote knowledge transfer. PROSPERO REGISTRATION NUMBER CRD42017075331.
Collapse
Affiliation(s)
- Emma J M Grigor
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Dean A Fergusson
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | - Fatima Haggar
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Natasha Kekre
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Blood and Marrow Transplant Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Harold Atkins
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Blood and Marrow Transplant Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Cancer Therapeutic Program, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Risa Shorr
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Robert A Holt
- British Columbia Cancer Agency, Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Brian Hutton
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Tim Ramsay
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Matthew Seftel
- Department of Hematology, University of Winnipeg, Winnipeg, Manitoba, Canada
| | - Derek Jonker
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Division of Medical Oncology, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Mads Daugaard
- Molecular Pathology & Cell Imaging Core Facility, Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Kednapa Thavorn
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Institute for Clinical Evaluative Sciences, Ottawa, Ontario, Canada
| | - Justin Presseau
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Manoj M Lalu
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
- Regenerative Medicine Program, The Ottawa Health Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
11813
|
Peng X, Liu G, Peng H, Chen A, Zha L, Wang Z. SOX4 contributes to TGF-β-induced epithelial-mesenchymal transition and stem cell characteristics of gastric cancer cells. Genes Dis 2017; 5:49-61. [PMID: 30258935 PMCID: PMC6147107 DOI: 10.1016/j.gendis.2017.12.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/11/2017] [Indexed: 12/17/2022] Open
Abstract
SOX4 is highly expressed in gastric cancer (GC) and is associated with tumor grade, metastasis and prognosis, however the mechanism is not clear. We report herein that SOX4 was upregulated and overexpression of SOX4 was associated with increased expression of the markers of Epithelial–mesenchymal transition (EMT) and stemness in clinic patient samples. In vitro, overexpression of SOX4 promoted the invasion as showed by Transwell assay and stemness of GC cells as assessed by sphere formation assay, which was suppressed by silencing SOX4 with shRNA. Further studies showed that SOX4 up-regulated the expression of EMT transcription factors Twist1, snail1 and zeb1 and stemness transcription factors SOX2 and OCT4, and promoted the nuclear translocation of β-catenin. Moreover, we revealed that TGF-β treatment significantly up-regulated the expression of SOX4 and silencing SOX4 reversed TGF-β induced invasion and sphere formation ability of GC cells. Finally, we showed that SOX4 promoted the lung metastasis and tumor formation ability of gastric cancer cells in nude mice. Our results suggest that SOX4 is a target TGF-β signaling and mediates TGF-β-induced EMT and stem cell characteristics of GC cells, revealing a novel role of TGF-β/SOX4 axis in the regulation of malignant behavior of GC.
Collapse
Affiliation(s)
- Xudong Peng
- Gastrointestinal Surgical Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Guangyi Liu
- Gastrointestinal Surgical Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Hongxia Peng
- Department of General Surgery, The First People's Hospital, Yibin, Sichuan, 644000 China
| | - Anqi Chen
- Gastrointestinal Surgical Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Lang Zha
- Gastrointestinal Surgical Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Ziwei Wang
- Gastrointestinal Surgical Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| |
Collapse
|
11814
|
|
11815
|
Antonelli A, Ferrari SM, Fallahi P. Current and future immunotherapies for thyroid cancer. Expert Rev Anticancer Ther 2017; 18:149-159. [PMID: 29241377 DOI: 10.1080/14737140.2018.1417845] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Cancer immunotherapies were approved in recent years, including immune checkpoint inhibitors. Experience with ipilimumab (CTLA-4 antagonist), nivolumab and pembrolizumab (PD-1 antagonists), and atezolizumab (PD-L1 antagonist) has shown that the impact on overall survival in cancer patients is paramount. Immune checkpoint inhibitors target the immune system and they can be applied across multiple cancers; the response rate is ranging from 20 to 40%. Many studies have shown that thyroid cancer (TC) cells produce cytokines and chemokines, inducing several tumor-promoting effects. Targeting and/or lowering cytokines and chemokines concentrations within the tumor microenvironment would produce a therapeutic benefit. In TC, increased Treg and PD-1+ T cell frequencies are indicative of aggressive disease and PD-L1 expression correlates with a greater risk of recurrence. Area covered: After performing a literature search, a few pioneering studies have evaluated immunotherapy in thyroid cancer. More recently a case has been described involving anaplastic thyroid cancer treated with vemurafenib and nivolumab, with substantial regression and complete radiographic and clinical remission. Expert commentary: The use of immune checkpoint inhibitors in aggressive TC has not yet been extensively investigated and further studies in a large number of TC patients are urgently needed.
Collapse
Affiliation(s)
- Alessandro Antonelli
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | | | - Poupak Fallahi
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| |
Collapse
|
11816
|
Ji L, Zhong B, Jiang X, Mao F, Liu G, Song B, Wang CY, Jiao Y, Wang JP, Xu ZB, Li X, Zhan B. Actein induces autophagy and apoptosis in human bladder cancer by potentiating ROS/JNK and inhibiting AKT pathways. Oncotarget 2017; 8:112498-112515. [PMID: 29348843 PMCID: PMC5762528 DOI: 10.18632/oncotarget.22274] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/20/2017] [Indexed: 02/06/2023] Open
Abstract
Human bladder cancer is a common genitourinary malignant cancer worldwide. However, new therapeutic strategies are required to overcome its stagnated survival rate. Triterpene glycoside Actein (ACT), extracted from the herb black cohosh, suppresses the growth of human breast cancer cells. Our study attempted to explore the role of ACT in human bladder cancer cell growth and to reveal the underlying molecular mechanisms. We found that ACT significantly impeded the bladder cancer cell proliferation via induction of G2/M cycle arrest. Additionally, ACT administration triggered autophagy and apoptosis in bladder cancer cells, proved by the autophagosome formation, LC3B-II accumulation, improved cleavage of Caspases/poly (ADP-ribose) polymerase (PARP). Furthermore, reduction of reactive oxygen species (ROS) and p-c-Jun N-terminal kinase (JNK) could markedly reverse ACT-induced autophagy and apoptosis. In contrast, AKT and mammalian target of rapamycin (mTOR) were greatly de-phosphorylated by ACT, while suppressing AKT and mTOR activity could enhance the effects of ACT on apoptosis and autophagy induction. In vivo, ACT reduced the tumor growth with little toxicity. Taken together, our findings indicated that ACT suppressed cell proliferation, induced autophagy and apoptosis through promoting ROS/JNK activation, and blunting AKT pathway in human bladder cancer, which indicated that ACT might be an effective candidate against human bladder cancer in future.
Collapse
Affiliation(s)
- Lu Ji
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Bing Zhong
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Xi Jiang
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Fei Mao
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Gang Liu
- Department of Orthopaedics, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Bin Song
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Cheng-Yuan Wang
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Yong Jiao
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Jiang-Ping Wang
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Zhi-Bin Xu
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Xing Li
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Bo Zhan
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| |
Collapse
|
11817
|
Lau CH, Suh Y. In vivo genome editing in animals using AAV-CRISPR system: applications to translational research of human disease. F1000Res 2017; 6:2153. [PMID: 29333255 PMCID: PMC5749125 DOI: 10.12688/f1000research.11243.1] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/14/2017] [Indexed: 12/12/2022] Open
Abstract
Adeno-associated virus (AAV) has shown promising therapeutic efficacy with a good safety profile in a wide range of animal models and human clinical trials. With the advent of clustered regulatory interspaced short palindromic repeat (CRISPR)-based genome-editing technologies, AAV provides one of the most suitable viral vectors to package, deliver, and express CRISPR components for targeted gene editing. Recent discoveries of smaller Cas9 orthologues have enabled the packaging of Cas9 nuclease and its chimeric guide RNA into a single AAV delivery vehicle for robust
in vivo genome editing. Here, we discuss how the combined use of small Cas9 orthologues, tissue-specific minimal promoters, AAV serotypes, and different routes of administration has advanced the development of efficient and precise
in vivo genome editing and comprehensively review the various AAV-CRISPR systems that have been effectively used in animals. We then discuss the clinical implications and potential strategies to overcome off-target effects, immunogenicity, and toxicity associated with CRISPR components and AAV delivery vehicles. Finally, we discuss ongoing non-viral-based
ex vivo gene therapy clinical trials to underscore the current challenges and future prospects of CRISPR/Cas9 delivery for human therapeutics.
Collapse
Affiliation(s)
- Cia-Hin Lau
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, SAR, China
| | - Yousin Suh
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA.,Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
11818
|
Long KB, Collier AI, Beatty GL. Macrophages: Key orchestrators of a tumor microenvironment defined by therapeutic resistance. Mol Immunol 2017; 110:3-12. [PMID: 29273393 DOI: 10.1016/j.molimm.2017.12.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/22/2017] [Accepted: 12/02/2017] [Indexed: 12/12/2022]
Abstract
Macrophages have emerged as promising therapeutic targets in cancer. Within tumor tissue, macrophages foster tumor development, invasion, and metastasis. As the phenotype of macrophages is inherently pliable and dependent on cues received from the surrounding microenvironment, macrophages co-evolve with malignant and other non-malignant cells during cancer progression. In doing so, they establish a microenvironment that is therapeutically resistant and thwarts the productivity of T cell immunosuveillance. Strategies designed to deplete, inhibit, or redirect macrophages with anti-tumor activity are being explored to reverse the pro-tumor properties of macrophages that are commonly observed in cancer. In this review, we discuss our current understanding of the mechanisms that regulate macrophage recruitment to tumors, their impact on the tumor microenvironment, and their promise as therapeutic targets for improving the efficacy of cytotoxic- and immune-based therapies.
Collapse
Affiliation(s)
- Kristen B Long
- Department of Biology, Mansfield University, Mansfield, PA 16933, USA
| | - Arthur I Collier
- Department of Biology, Mansfield University, Mansfield, PA 16933, USA
| | - Gregory L Beatty
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
11819
|
Tsou PS, Coit P, Kilian NC, Sawalha AH. EZH2 Modulates the DNA Methylome and Controls T Cell Adhesion Through Junctional Adhesion Molecule A in Lupus Patients. Arthritis Rheumatol 2017; 70:98-108. [PMID: 28973837 DOI: 10.1002/art.40338] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/26/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE EZH2 is an epigenetic regulator that mediates H3K27 trimethylation (H3K27me3) and modulates DNA methylation. The aim of this study was to characterize the role of EZH2 in CD4+ T cells in the pathogenesis of systemic lupus erythematosus. METHODS EZH2 expression levels were determined in CD4+ T cells isolated from lupus patients and healthy controls. The epigenetic effects of EZH2 overexpression in CD4+ T cells were evaluated using a genome-wide DNA methylation approach. Gene expression profiles and microRNAs (miRNAs) were assessed by quantitative polymerase chain reaction, while protein expression was examined by Western blotting. A cell adhesion assay was used to assess adhesion of CD4+ T cells to human microvascular endothelial cells. RESULTS EZH2 and H3K27me3 levels were increased in CD4+ T cells from lupus patients compared to healthy controls. T cell production of EZH2 was down-regulated in the presence of miR-26a and miR-101, and levels of both miRNAs were reduced in lupus CD4+ T cells. Overexpression of EZH2 induced in CD4+ T cells resulted in significant DNA methylation changes. Genes involved in leukocyte adhesion and migration, including F11R (which encodes junctional adhesion molecule A [JAM-A]), became hypomethylated in CD4+ T cells when EZH2 was overexpressed. Overexpression of EZH2 resulted in increases in JAM-A expression and CD4+ T cell adhesion. Preincubation of EZH2-transfected CD4+ T cells with neutralizing antibodies against JAM-A significantly blunted cell adhesion. Similarly, CD4+ T cells from lupus patients overexpressed JAM-A and adhered significantly more to endothelial cells than to T cells from healthy controls. Blocking JAM-A or EZH2 significantly reduced the capacity of lupus CD4+ T cells to adhere to endothelial cells. CONCLUSION The results of this study identify a novel role of EZH2 in T cell adhesion mediated by epigenetic remodeling and up-regulation of JAM-A. Blockade of EZH2 or JAM-A might have therapeutic potential by acting to reduce T cell adhesion, migration, and extravasation in patients with lupus.
Collapse
|
11820
|
Muthukumaran S, Bhuvanasundar R, Umashankar V, Sulochana KN. Insights on ornithine decarboxylase silencing as a potential strategy for targeting retinoblastoma. Biomed Pharmacother 2017; 98:23-28. [PMID: 29241071 DOI: 10.1016/j.biopha.2017.12.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 11/27/2017] [Accepted: 12/07/2017] [Indexed: 01/26/2023] Open
Abstract
Ornithine Decarboxylase (ODC) is a key enzyme involved in polyamine synthesis and is reported to be up regulated in several cancers. However, the effect of ODC gene silencing in retinoblastoma is to be understood for utilization in therapeutic applications. Hence, in this study, a novel siRNA (small interference RNA) targeting ODC was designed and validated in Human Y79 retinoblastoma cells for its effects on intracellular polyamine levels, Matrix Metalloproteinase 2 & 9 activity and Cell cycle. The designed siRNA showed efficient silencing of ODC mRNA expression and protein levels in Y79 cells. It also showed significant reduction of intracellular polyamine levels and altered levels of oncogenic LIN28b expression. By this study, a regulatory loop is proposed, wherein, ODC silencing in Y79 cells to result in decreased polyamine levels, thereby, leading to altered protein levels of Lin28b, MMP-2 and MMP-9, which falls in line with earlier studies in neuroblastoma. Thus, by this study, we propose ODC silencing as a prospective strategy for targeting retinoblastoma.
Collapse
Affiliation(s)
- Sivashanmugam Muthukumaran
- Centre for Bioinformatics, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Vision Research Foundation, Chennai, India; School of Chemical and Biotechnology, SASTRA University, Thanjavur, India
| | - Renganathan Bhuvanasundar
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Vision Research Foundation, Chennai, India
| | - Vetrivel Umashankar
- Centre for Bioinformatics, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Vision Research Foundation, Chennai, India.
| | - K N Sulochana
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Vision Research Foundation, Chennai, India
| |
Collapse
|
11821
|
Huang G, Sun X, Liu D, Zhang Y, Zhang B, Xiao G, Li X, Gao X, Hu C, Wang M, Ren H, Qin S. The efficacy and safety of anti-PD-1/PD-L1 antibody therapy versus docetaxel for pretreated advanced NSCLC: a meta-analysis. Oncotarget 2017; 9:4239-4248. [PMID: 29423118 PMCID: PMC5790535 DOI: 10.18632/oncotarget.23279] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 11/17/2017] [Indexed: 12/26/2022] Open
Abstract
Antibodies against the immune checkpoint proteins PD-1 and PD-L1 are novel therapeutic drugs for the treatment of advanced non-small cell lung cancer (NSCLC). Many clinical trials involving these drugs achieved breakthroughs in patients previously treated for advanced NSCLC. However, the results of these clinical studies are not consistent. In this report, we performed a meta-analysis to assess the efficacy and safety of anti-PD-1/PD-L1 antibodies compared with docetaxel treatment for advanced NSCLC patients from 5 randomized clinical trials. We demonstrated that the patients in anti-PD-1/PD-L1 antibody therapy groups had significantly longer overall survival (OS) (HR = 0.69, 95% CI 0.63-0.75, P < 0.05) and progression-free survival (PFS) (HR = 0.76, 95% CI 0.63-0.92, P < 0.05) than those in chemotherapy groups, especially PD-L1 positive patients. Anti-PD-1/PD-L1 antibodies improved the objective response rate (ORR) compared with docetaxel (OR = 1.64, 95% CI 1.19-2.26, p < 0.05). In addition, the anti-PD-1/PD-L1 antibody therapy had fewer treatment-related adverse events (AEs) (OR = 0.33, 95% CI 0.28-0.39, P < 0.05) than docetaxel, especially the grade ≥3 AEs (OR = 0.18, 95% CI 0.12-0.28, P < 0.001). In conclusion, our study revealed that, compared with docetaxel, anti-PD-1/PD-L1 antibody therapy improved clinical efficacy and safety in previously treated advanced NSCLC patients. This therapy may be a promising treatment for advanced NSCLC patients.
Collapse
Affiliation(s)
- Guanghong Huang
- Department Two of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xin Sun
- Department Two of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Dapeng Liu
- Department Two of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yunfeng Zhang
- Department Two of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Boxiang Zhang
- Department Two of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Guodong Xiao
- Department Two of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiang Li
- Department Two of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiao Gao
- Department Two of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Chenhao Hu
- Department Two of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Meng Wang
- Department Two of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Hong Ren
- Department Two of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Sida Qin
- Department Two of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
11822
|
Majumder S, Thieme K, Batchu SN, Alghamdi TA, Bowskill BB, Kabir MG, Liu Y, Advani SL, White KE, Geldenhuys L, Tennankore KK, Poyah P, Siddiqi FS, Advani A. Shifts in podocyte histone H3K27me3 regulate mouse and human glomerular disease. J Clin Invest 2017; 128:483-499. [PMID: 29227285 DOI: 10.1172/jci95946] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/31/2017] [Indexed: 01/09/2023] Open
Abstract
Histone protein modifications control fate determination during normal development and dedifferentiation during disease. Here, we set out to determine the extent to which dynamic changes to histones affect the differentiated phenotype of ordinarily quiescent adult glomerular podocytes. To do this, we examined the consequences of shifting the balance of the repressive histone H3 lysine 27 trimethylation (H3K27me3) mark in podocytes. Adriamycin nephrotoxicity and subtotal nephrectomy (SNx) studies indicated that deletion of the histone methylating enzyme EZH2 from podocytes decreased H3K27me3 levels and sensitized mice to glomerular disease. H3K27me3 was enriched at the promoter region of the Notch ligand Jag1 in podocytes, and derepression of Jag1 by EZH2 inhibition or knockdown facilitated podocyte dedifferentiation. Conversely, inhibition of the Jumonji C domain-containing demethylases Jmjd3 and UTX increased the H3K27me3 content of podocytes and attenuated glomerular disease in adriamycin nephrotoxicity, SNx, and diabetes. Podocytes in glomeruli from humans with focal segmental glomerulosclerosis or diabetic nephropathy exhibited diminished H3K27me3 and heightened UTX content. Analogous to human disease, inhibition of Jmjd3 and UTX abated nephropathy progression in mice with established glomerular injury and reduced H3K27me3 levels. Together, these findings indicate that ostensibly stable chromatin modifications can be dynamically regulated in quiescent cells and that epigenetic reprogramming can improve outcomes in glomerular disease by repressing the reactivation of developmental pathways.
Collapse
Affiliation(s)
- Syamantak Majumder
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Karina Thieme
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Sri N Batchu
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Tamadher A Alghamdi
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Bridgit B Bowskill
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - M Golam Kabir
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Youan Liu
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Suzanne L Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Kathryn E White
- Electron Microscopy Research Services, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | | | - Penelope Poyah
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ferhan S Siddiqi
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
11823
|
Vikram B. Research in imaging/biomarkers for precision medicine in lung cancer: National Cancer Institute funding opportunities. Transl Lung Cancer Res 2017; 6:615-616. [PMID: 29218263 DOI: 10.21037/tlcr.2017.09.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Bhadrasain Vikram
- Radiation Research Program/Division of Cancer Treatment & Diagnosis, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
11824
|
Massa C, Seliger B. The tumor microenvironment: Thousand obstacles for effector T cells. Cell Immunol 2017; 343:103730. [PMID: 29249298 DOI: 10.1016/j.cellimm.2017.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 12/04/2017] [Accepted: 12/07/2017] [Indexed: 12/24/2022]
Abstract
The immune system is endowed with the capability to recognize and destroy transformed cells, but even in the presence of an immune infiltrate many tumors do progress. In the last decades new discoveries have shed light into (some of) the underlying mechanisms. Immune effector cells are not only under the influence of immune suppressive cell subsets, but also intrinsically regulated by immune check point molecules that under physiological condition avoid attach of healthy tissue. Moreover, tumor cells are modifying the surrounding microenvironment through secretion of immune modulators as well as via their own metabolism, thus further impairing the development of immune effector functions. Different approaches are currently being evaluated in the clinic to overcome those regulatory mechanisms and to unleash effector T cells.
Collapse
Affiliation(s)
- Chiara Massa
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Barbara Seliger
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
| |
Collapse
|
11825
|
Ono K, Shiozawa E, Ohike N, Fujii T, Shibata H, Kitajima T, Fujimasa K, Okamoto N, Kawaguchi Y, Nagumo T, Tazawa S, Homma M, Yamochi-Onizuka T, Norose T, Yoshida H, Murakami M, Tate G, Takimoto M. Immunohistochemical CD73 expression status in gastrointestinal neuroendocrine neoplasms: A retrospective study of 136 patients. Oncol Lett 2017; 15:2123-2130. [PMID: 29434915 DOI: 10.3892/ol.2017.7569] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 10/25/2017] [Indexed: 12/23/2022] Open
Abstract
The WHO 2010 classification divides gastrointestinal neuroendocrine neoplasms (GI-NENs) into neuroendocrine tumor (NET) G1, NET G2, neuroendocrine carcinoma (NEC) and mixed adenoendocrine carcinoma (MANEC) groups. A total of 136 cases of GI-NENs diagnosed at our hospitals as gastrointestinal carcinoids, endocrine cell carcinomas and NENs over the last 11 years, using the WHO 2010 classification were assessed. Among the 136 cases, 88.2% (120/136) were classified into the NET group (NET G1/G2) and 11.8% (16/136) were classified into the NEC group (NEC/MANEC). The incidences of lymphatic and venous invasions were higher in the NEC group compared with in the NET group (P<0.0001 and P=0.0021, respectively). The immunohistochemical staining of cluster of differentiation 73 (CD73) was evaluated in GI-NENs. CD73 is a potentially useful molecule in tumor immunity. In general, CD73 on the tumor cell membrane converts adenosine monophosphate to adenosine, which restrains the production of interferon-γ and cytocidal activity. Although the association between stem cells of pancreatic NENs and CD73 has been reported, few studies have reported on CD73 expression in GI-NENs. Immunohistochemical CD73 expression on the cytomembrane of neuroendocrine cells was detected in 27.2% (37/136) of the GI-NENs. The positive ratio of CD73 was significantly higher in the NEC group compared with in the NET group (P=0.0015). CD73 is also considered as a potential biomarker of anti-programmed death-1 (PD-1) therapy. The expression of programmed death-ligand 1 (PD-L1) on the cytomembrane of GI-NENs was assessed. The positive ratio of PD-L1 was higher in the NEC group compared with in the NET group (P=0.0011). Furthermore, CD73 expression status was significantly correlated with PD-L1 expression (P<0.0001). These results indicate that CD73 may be an interesting candidate for a biomarker for certain prognostic factors and therapeutics concerning PD-1 therapy.
Collapse
Affiliation(s)
- Kohei Ono
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan.,Department of Gastroenterological and General Surgery, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Eisuke Shiozawa
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Nobuyuki Ohike
- Department of Pathology and Laboratory Medicine, Showa University Fujigaoka Hospital, Aoba-ku, Yokohama 227-8501, Japan
| | - Tomonori Fujii
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan.,Department of Gastroenterological and General Surgery, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Hideki Shibata
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan.,Department of Gastroenterological and General Surgery, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Tetsuya Kitajima
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan.,Department of Gastroenterological and General Surgery, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Koichiro Fujimasa
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan.,Department of Gastroenterological and General Surgery, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Naoko Okamoto
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Yukiko Kawaguchi
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Tasuku Nagumo
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Sakiko Tazawa
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Mayumi Homma
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Toshiko Yamochi-Onizuka
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Tomoko Norose
- Department of Pathology and Laboratory Medicine, Showa University Fujigaoka Hospital, Aoba-ku, Yokohama 227-8501, Japan
| | - Hitoshi Yoshida
- Department of Gastroenterology, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Masahiko Murakami
- Department of Gastroenterological and General Surgery, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Gensyu Tate
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Masafumi Takimoto
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8666, Japan
| |
Collapse
|
11826
|
Qi B, Wang Y, Chen ZJ, Li XN, Qi Y, Yang Y, Cui GH, Guo HZ, Li WH, Zhao S. Down-regulation of miR-30a-3p/5p promotes esophageal squamous cell carcinoma cell proliferation by activating the Wnt signaling pathway. World J Gastroenterol 2017; 23:7965-7977. [PMID: 29259372 PMCID: PMC5725291 DOI: 10.3748/wjg.v23.i45.7965] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/26/2017] [Accepted: 10/26/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the potential role of microRNA-30a (miR-30a) in esophageal squamous cell carcinoma (ESCC).
METHODS Expression of miR-30a-3p/5p was analyzed using microarray data and fresh ESCC tissue samples. Both in vitro and in vivo assays were used to investigate the effects of miR-30a-3p/5p on ESCC cell proliferation. Furthermore, Kyoto Encyclopedia of Genes and Genomes analysis was performed to explore underlying mechanisms involved in ESCC, and then, assays were carried out to verify the potential molecular mechanism of miR-30a in ESCC.
RESULTS Low expression of miR-30a-3p/5p was closely associated with advanced ESCC progression and poor prognosis of patients with ESCC. Knock-down of miR-30a-3p/5p promoted ESCC cell proliferation. Increased miR-30a-3p/5p expression inhibited the Wnt signaling pathway by targeting Wnt2 and Fzd2.
CONCLUSION Down-regulation of miR-30a-3p/5p promotes ESCC cell proliferation by activating the Wnt signaling pathway through inhibition of Wnt2 and Fzd2.
Collapse
Affiliation(s)
- Bo Qi
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan Province, China
| | - Yan Wang
- Periodicals Publishing House, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Zhi-Jun Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan Province, China
| | - Xiang-Nan Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yu Qi
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yang Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Guang-Hui Cui
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Hai-Zhou Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Wei-Hao Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Song Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
11827
|
Li J, Liao Y, Ding T, Wang B, Yu X, Chu Y, Xu J, Zheng L. Tumor-infiltrating macrophages express interleukin-25 and predict a favorable prognosis in patients with gastric cancer after radical resection. Oncotarget 2017; 7:11083-93. [PMID: 26840565 PMCID: PMC4905459 DOI: 10.18632/oncotarget.7095] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 01/17/2016] [Indexed: 12/28/2022] Open
Abstract
Interleukin-25 (IL-25) is a recently identified member of the proinflammatory IL-17 cytokine family; however, its role in human tumors remains largely unknown. The aim of this study was to investigate the cellular source and clinical significance of IL-25 in gastric cancer (GC) in situ. The results demonstrated that macrophages (Mφs) were the primary IL-25-expressing cells (IL-25+) in GC in situ. Moreover, IL-25+ cells were highly enriched in the intra-tumoral (IT) region of GC tissues (p < 0.001). The production of IL-25 in Mφs exposed to culture supernatant from gastric cancer cell line SGC7901 in vitro was induced by transforming growth factor-β1, and their density in the IT region was positively associated with those of other effector immune cells, namely, CD4+ T cells, CD8+ T cells and CD103+T cells (p < 0.01). This suggested that macrophages might produce IL-25 to create an antitumor micromilieu in GC tissues. The level of IL-25+IT cells was positively associated with histological grade (p < 0.001) and found to be an independent predictor of favorable survival (p = 0.024) in patients with GC after radical resection. These findings suggest that IL-25+IT cells may be a novel therapeutic target in those patients.
Collapse
Affiliation(s)
- Jinqing Li
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yuan Liao
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China.,Department of Laboratory Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Tong Ding
- Department of Cell Biology, Nanjing Medical University, Nanjing, P.R. China
| | - Bo Wang
- Department of Urology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Xingjuan Yu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yifan Chu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Jing Xu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Limin Zheng
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China.,Key Laboratory of Gene Engineering of The Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| |
Collapse
|
11828
|
Kim S, Song JH, Kim S, Qu P, Martin BK, Sehareen WS, Haines DC, Lin PC, Sharan SK, Chang S. Loss of oncogenic miR-155 in tumor cells promotes tumor growth by enhancing C/EBP-β-mediated MDSC infiltration. Oncotarget 2017; 7:11094-112. [PMID: 26848978 PMCID: PMC4905460 DOI: 10.18632/oncotarget.7150] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 01/19/2015] [Indexed: 11/25/2022] Open
Abstract
The oncogenic role of microRNA-155 (miR-155) in leukemia is well established but its role in other cancers, especially breast cancer, is gradually emerging. In this study we examined the effect of mir-155 loss in a well-characterized spontaneous breast cancer mouse model where Brca1 and Trp53 are deleted by K14-Cre. miR-155 is known to be up-regulated in BRCA1-deficient tumors. Surprisingly, complete loss of miR-155 (miR-155ko/ko) did not alter the tumor free survival of the mutant mice. However, we found increased infiltration of myeloid derived suppressor cells (MDSCs) in miR-155 deficient tumors. In addition, cytokine/chemokine array analysis revealed altered level of cytokines that are implicated in the recruitment of MDSCs. Mechanistically, we identified C/EBP-β, a known miR-155 target, to regulate the expression of these cytokines in the miR-155-deficient cells. Furthermore, using an allograft model, we showed that inhibition of miR-155 in cancer cells suppressed in vivo growth, which was restored by the loss of miR-155 in the microenvironment. Taken together, we have uncovered a novel tumor suppressive function of miR-155 in the tumor microenvironment, which is also dependent on miR-155 expression in the tumor cells. Because of the oncogenic as well as tumor suppressive roles of miR-155, our findings warrant caution against a systemic inhibition of miR-155 for anticancer therapy.
Collapse
Affiliation(s)
- Sinae Kim
- Department of Biomedical Sciences, Department of Physiology, University of Ulsan School of Medicine, Seoul, South Korea
| | - Jin Hoi Song
- Aging Research Center, Korea Research Institute of Bioscience & Biotechnology, Taejeon, South Korea
| | - Seokho Kim
- Aging Research Center, Korea Research Institute of Bioscience & Biotechnology, Taejeon, South Korea
| | - Peng Qu
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Betty K Martin
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Waheed S Sehareen
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Diana C Haines
- Pathology Histotechnology Laboratory, Leidos Inc., Frederick National Laboratory for Cancer, Frederick, MD, USA
| | - Pengnian C Lin
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Shyam K Sharan
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Suhwan Chang
- Department of Biomedical Sciences, Department of Physiology, University of Ulsan School of Medicine, Seoul, South Korea
| |
Collapse
|
11829
|
Li KP, Fang YP, Liao JQ, Duan JD, Feng LG, Luo XZ, Liang ZJ. Upregulation of miR‑598 promotes cell proliferation and cell cycle progression in human colorectal carcinoma by suppressing INPP5E expression. Mol Med Rep 2017; 17:2991-2997. [PMID: 29257251 PMCID: PMC5783518 DOI: 10.3892/mmr.2017.8207] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 08/18/2017] [Indexed: 01/05/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common types of cancer worldwide. Recently, microRNAs (miRs) have been considered as novel therapeutic targets for the treatment of cancer. miR‑598 is a poorly investigated miR. The underlying mechanism of miR‑598 in CRC cells remains to be elucidated. In the present study, miR‑598 was demonstrated to be significantly upregulated in CRC tissue by analyzing data from The Cancer Genome Atlas and the Gene Expression Omnibus. The results of a polymerase chain reaction demonstrated that miR‑598 expression was significantly upregulated in CRC tissues and cells. Gain of function and loss of function assays demonstrated that miR‑598 significantly promoted cell proliferation and cell cycle progression. miR‑598 was demonstrated to modulate cell functions by regulating 72 kDa inositol polyphosphate‑5‑phosphatase (INPP5E). In addition, knockdown of INPP5E counteracted the growth arrest caused by an miR‑598‑inhibitor. In conclusion, the present study demonstrated that miR‑598 contributed to cell proliferation and cell cycle progression in CRC by targeting INPP5E.
Collapse
Affiliation(s)
- Kun-Ping Li
- Department of General Surgery, Huizhou First Hospital, Huizhou, Guangdong 516000, P.R. China
| | - Yong-Ping Fang
- Department of General Surgery, Huizhou First Hospital, Huizhou, Guangdong 516000, P.R. China
| | - Jin-Qi Liao
- Department of General Surgery, Huizhou First Hospital, Huizhou, Guangdong 516000, P.R. China
| | - Jin-Dong Duan
- Department of General Surgery, Huizhou First Hospital, Huizhou, Guangdong 516000, P.R. China
| | - Li-Guang Feng
- Department of General Surgery, Huizhou First Hospital, Huizhou, Guangdong 516000, P.R. China
| | - Xiao-Zai Luo
- Department of General Surgery, Huizhou First Hospital, Huizhou, Guangdong 516000, P.R. China
| | - Zhi-Jian Liang
- Department of General Surgery, Huizhou First Hospital, Huizhou, Guangdong 516000, P.R. China
| |
Collapse
|
11830
|
Ong CY, Vasanwala FF. Thrombotic Paradox: Ischaemic Stroke in Immune Thrombocytopaenia. A Case Report and Review. Cureus 2017; 9:e1904. [PMID: 29410943 PMCID: PMC5796812 DOI: 10.7759/cureus.1904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Immune thrombocytopaenia (also known as idiopathic thrombocytopaenic purpura) (ITP) is a chronic condition with isolated low platelet counts. Although it is largely perceived that ITP predisposes to bleeding risks, thrombotic events, such as ischaemic strokes, do happen paradoxically in patients with ITP. A 68-year-old lady presented with right upper limb weakness and was diagnosed with an ischaemic stroke and was started on clopidogrel. She had a history of ITP. Two months later, she again had another ischaemic stroke. Prednisolone was added as her platelet count was below 50 x 109/L. Based on this case and recent case studies, we suggest the administration of antiplatelet or anticoagulant agents judiciously if the platelet count is 50 x 109/L or above with monitoring of bleeding risks. As for the management of ITP, we do agree with the general opinion that treatment is rarely required for patients with a platelet count above 50 x 109/L. We recommend a haematology consult for discussion on treatment initiation if the platelet count is below 50 x 109/L.
Collapse
Affiliation(s)
- Chong Yau Ong
- Family Medicine, Sengkang General Hospital, Sengkang Health, Singhealth, Singapore
| | - Farhad F Vasanwala
- Department of General Medicine, Sengkang General Hospital, Sengkang Health, Singhealth, Singapore
| |
Collapse
|
11831
|
Introna M. CIK as therapeutic agents against tumors. J Autoimmun 2017; 85:32-44. [DOI: 10.1016/j.jaut.2017.06.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 06/19/2017] [Accepted: 06/19/2017] [Indexed: 01/26/2023]
|
11832
|
Abstract
Despite the success of anti-programmed cell death protein 1 (PD1), anti-PD1 ligand 1 (PDL1) and anti-cytotoxic T lymphocyte antigen 4 (CTLA4) therapies in advanced cancer, a considerable proportion of patients remain unresponsive to these treatments (known as innate resistance). In addition, one-third of patients relapse after initial response (known as adaptive resistance), which suggests that multiple non-redundant immunosuppressive mechanisms coexist within the tumour microenvironment. A major immunosuppressive mechanism is the adenosinergic pathway, which now represents an attractive new therapeutic target for cancer therapy. Activation of this pathway occurs within hypoxic tumours, where extracellular adenosine exerts local suppression through tumour-intrinsic and host-mediated mechanisms. Preclinical studies in mice with adenosine receptor antagonists and antibodies have reported favourable antitumour immune responses with some definition of the mechanism of action. Currently, agents targeting the adenosinergic pathway are undergoing first-in-human clinical trials as single agents and in combination with anti-PD1 or anti-PDL1 therapies. In this Review, we describe the complex interplay of adenosine and adenosine receptors in the development of primary tumours and metastases and discuss the merits of targeting one or more components that compose the adenosinergic pathway. We also review the early clinical data relating to therapeutic agents inhibiting the adenosinergic pathway.
Collapse
Affiliation(s)
- Dipti Vijayan
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, 4006, Queensland, Australia
| | - Arabella Young
- Diabetes Center, University of California, San Francisco, California 94143, USA
| | - Michele W L Teng
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, 4006, Queensland, Australia
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, 4006, Queensland, Australia
| |
Collapse
|
11833
|
Guo L, Zhang P, Chen Z, Xia H, Li S, Zhang Y, Kobberup S, Zou W, Lin JD. Hepatic neuregulin 4 signaling defines an endocrine checkpoint for steatosis-to-NASH progression. J Clin Invest 2017; 127:4449-4461. [PMID: 29106384 PMCID: PMC5707158 DOI: 10.1172/jci96324] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/26/2017] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is characterized by progressive liver injury, inflammation, and fibrosis; however, the mechanisms that govern the transition from hepatic steatosis, which is relatively benign, to NASH remain poorly defined. Neuregulin 4 (Nrg4) is an adipose tissue-enriched endocrine factor that elicits beneficial metabolic effects in obesity. Here, we show that Nrg4 is a key component of an endocrine checkpoint that preserves hepatocyte health and counters diet-induced NASH in mice. Nrg4 deficiency accelerated liver injury, fibrosis, inflammation, and cell death in a mouse model of NASH. In contrast, transgenic expression of Nrg4 in adipose tissue alleviated diet-induced NASH. Nrg4 attenuated hepatocyte death in a cell-autonomous manner by blocking ubiquitination and proteasomal degradation of c-FLIPL, a negative regulator of cell death. Adeno-associated virus-mediated (AAV-mediated) rescue of hepatic c-FLIPL expression in Nrg4-deficent mice functionally restored the brake for steatosis to NASH transition. Thus, hepatic Nrg4 signaling serves as an endocrine checkpoint for steatosis-to-NASH progression by activating a cytoprotective pathway to counter stress-induced liver injury.
Collapse
Affiliation(s)
- Liang Guo
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Peng Zhang
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Zhimin Chen
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Houjun Xia
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Siming Li
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Yanqiao Zhang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Sune Kobberup
- Metabolic Disease Research, Novo Nordisk, Maaloev, Denmark
| | - Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
11834
|
Xu-Welliver M, Carbone DP. Blood-based biomarkers in lung cancer: prognosis and treatment decisions. Transl Lung Cancer Res 2017; 6:708-712. [PMID: 29218272 DOI: 10.21037/tlcr.2017.09.08] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Despite recent advances, non-small cell lung cancer (NSCLC) remains a devastating disease with overall poor prognosis. Major contributing factors include obstacles to diagnosing the disease early in its course during the asymptomatic stage as well as diversity and complexity of its biology underlying tumorigenesis and tumor progression. Advances in molecularly targeted therapies which drives the development of personalized cancer care require precise and comprehensive understanding of tumor biology, not only at the time of diagnosis but also during treatment course and surveillance. As lung tumor tissue can be difficult to obtain without invasive and potentially risky procedures, it is difficult to monitor treatment response with serial tissue biopsies. Development of non-invasive but reliable blood based tumor markers has become an important research area. In this review, we focus on the following circulating biomarkers that have been identified in recent years: circulating tumor cells (CTCs); circulating cell-free nucleic acids, such as circulating tumor DNA (ctDNA) and microRNA (miR); and other biomarkers such as genomic and proteomic features. These biomarkers not only have prognostic values, but also can help guild treatment decisions by monitoring tumor burden, detecting minimal residual disease and/or recurrent disease, as well as monitoring evolution of genetic alterations throughout the treatment course.
Collapse
Affiliation(s)
- Meng Xu-Welliver
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, USA
| | - David P Carbone
- Division of Medical Oncology, Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, USA
| |
Collapse
|
11835
|
Huang L, Lian J, Chen X, Qin G, Zheng Y, Zhang Y. WASH overexpression enhances cancer stem cell properties and correlates with poor prognosis of esophageal carcinoma. Cancer Sci 2017; 108:2358-2365. [PMID: 28914471 PMCID: PMC5715296 DOI: 10.1111/cas.13400] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/07/2017] [Accepted: 09/12/2017] [Indexed: 01/13/2023] Open
Abstract
There is increasing evidence that cytoskeleton remodeling is involved in cancer progression. Wiskott-Aldrich syndrome protein (WASP) family represents a key regulator of actin cytoskeleton remodeling. However, the underlying mechanism of the WASP family in cancer progression remains elusive. Here, we studied the role of WASP and SCAR Homolog (WASH), a recently identified WASP family member, in human esophageal squamous cell carcinoma (ESCC). Using three human ESCC cell lines, we found that WASH expression was significantly elevated in cancer stem-like cells enriched by sphere formation assay. WASH knockdown decreased the sphere-forming capacity of esophageal cancer cells whereas WASH over-expression exhibited the opposite effect. Mechanistically, we identified interleukin-8 (IL-8) as a key downstream target of WASH. IL-8 knockdown completely attenuated tumor sphere formation induced by WASH overexpression. WASH knockdown also delayed the growth of human ESCC xenografts in BALB/c nude mice. Importantly, high WASH levels were associated with poor clinical prognosis in a total of 145 human ESCC tissues. Collectively, our results suggest an essential role of the WASH/IL-8 pathway in human ESCC by maintaining the stemness of cancer cells. Hence, targeting this pathway might represent a promising strategy to control human esophageal carcinoma.
Collapse
Affiliation(s)
- Lan Huang
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jingyao Lian
- School of Life SciencesZhengzhou UniversityZhengzhouChina
| | - Xinfeng Chen
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guohui Qin
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yujia Zheng
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yi Zhang
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- School of Life SciencesZhengzhou UniversityZhengzhouChina
| |
Collapse
|
11836
|
Dorfeshan P, Ghaffari Novin M, Salehi M, Masteri Farahani R, Fadaei-Fathabadi F, Sehatti R. The Effects of In Vitro Maturation Technique on The Expression of Genes Involved in Embryonic Genome Activation of Human Embryos. CELL JOURNAL 2017; 20:90-97. [PMID: 29308624 PMCID: PMC5759685 DOI: 10.22074/cellj.2018.4804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 02/14/2017] [Indexed: 01/25/2023]
Abstract
Objective In vitro maturation technique (IVM) is shown to have an effect on full maturation of immature oocytes and
the subsequent embryo development. Embryonic genome activation (EGA) is considered as a crucial and the first
process after fertilization. EGA failure leads to embryo arrest and possible implantation failure. This study aimed to
determine the role of IVM in EGA-related genes expression in human embryo originated from immature oocytes and
recovered from women receiving gonadotrophin treatment for assisted reproduction.
Materials and Methods In this experimental study, germinal vesicle (GV) oocytes were cultured in vitro. After
intracytoplasmic sperm injection of the oocytes, fertilization, cleavage and embryo quality score were assessed in
vitro and in vivo. After 3-4 days, a single blastomere was biopsied from the embryos and then frozen. Afterwards, the
expression of EGA-related genes in embryos was assayed using quantitative reverse transcriptase-polymerase chain
reaction (PCR).
Results The in vitro study showed reduced quality of embryos. No significant difference was found between embryo
quality scores for the two groups (P=0.754). The in vitro group exhibited a relatively reduced expression of the EGA-
related genes, when compared to the in vivo group (all of them showed P=0.0001).
Conclusion Although displaying the normal morphology, the IVM process appeared to have a negative influence on
developmental gene expression levels of human preimplanted embryos. Based on our results, the embryo normal
morphology cannot be considered as an ideal scale for the successful growth of embryo at implantation and downstream
processes.
Collapse
Affiliation(s)
- Parvin Dorfeshan
- Department of Biology and Anatomical Sciences, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marefat Ghaffari Novin
- Department of Biology and Anatomical Sciences, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Infertility and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran. Electronic Address:
| | - Mohammad Salehi
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Masteri Farahani
- Department of Biology and Anatomical Sciences, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Fadaei-Fathabadi
- Department of Biology and Anatomical Sciences, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ronak Sehatti
- Infertility and Reproductive Health Research Center, Aban Hospital, Tehran, Iran
| |
Collapse
|
11837
|
Ramello MC, Haura EB, Abate-Daga D. CAR-T cells and combination therapies: What's next in the immunotherapy revolution? Pharmacol Res 2017; 129:194-203. [PMID: 29203440 DOI: 10.1016/j.phrs.2017.11.035] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapies are dramatically reshaping the clinical management of oncologic patients. For many of these therapies, the guidelines for administration, monitoring, and management of associated toxicities are still being established. This is especially relevant for adoptively transferred, genetically-modified T cells, which have unique pharmacokinetic properties, due to their ability to replicate and persist long-term, following a single administration. Furthermore, in the case of CAR-T cells, the use of synthetic immune receptors may impact signaling pathways involved in T cell function and survival in unexpected ways. We, herein, comment on the most salient aspects of CAR-T cell design and clinical experience in the treatment of solid tumors. In addition, we discuss different possible scenarios for combinations of CAR-T cells and other treatment modalities, with a special emphasis on kinase inhibitors, elaborating on the strategies to maximize synergism. Finally, we discuss some of the technologies that are available to explore the molecular events governing the success of these therapies. The young fields of synthetic and systems biology are likely to be major players in the advancement of CAR-T cell therapies, providing the tools and the knowledge to engineer patients' T lymphocytes into intelligent cancer-fighting micromachines.
Collapse
Affiliation(s)
- Maria C Ramello
- Dept. of Immunology, H. Lee Moffitt Cancer Center and Research Institute. Tampa, FL, United States
| | - Eric B Haura
- Dept. of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, United States
| | - Daniel Abate-Daga
- Dept. of Immunology, H. Lee Moffitt Cancer Center and Research Institute. Tampa, FL, United States; Dept. of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, United States; Dept. of Oncological Sciences, Morsani School of Medicine, University of South Florida, United States
| |
Collapse
|
11838
|
Alexander SP, Christopoulos A, Davenport AP, Kelly E, Marrion NV, Peters JA, Faccenda E, Harding SD, Pawson AJ, Sharman JL, Southan C, Davies JA. THE CONCISE GUIDE TO PHARMACOLOGY 2017/18: G protein-coupled receptors. Br J Pharmacol 2017; 174 Suppl 1:S17-S129. [PMID: 29055040 PMCID: PMC5650667 DOI: 10.1111/bph.13878] [Citation(s) in RCA: 532] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2017/18 provides concise overviews of the key properties of nearly 1800 human drug targets with an emphasis on selective pharmacology (where available), plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. Although the Concise Guide represents approximately 400 pages, the material presented is substantially reduced compared to information and links presented on the website. It provides a permanent, citable, point-in-time record that will survive database updates. The full contents of this section can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.13878/full. G protein-coupled receptors are one of the eight major pharmacological targets into which the Guide is divided, with the others being: ligand-gated ion channels, voltage-gated ion channels, other ion channels, nuclear hormone receptors, catalytic receptors, enzymes and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. The landscape format of the Concise Guide is designed to facilitate comparison of related targets from material contemporary to mid-2017, and supersedes data presented in the 2015/16 and 2013/14 Concise Guides and previous Guides to Receptors and Channels. It is produced in close conjunction with the Nomenclature Committee of the Union of Basic and Clinical Pharmacology (NC-IUPHAR), therefore, providing official IUPHAR classification and nomenclature for human drug targets, where appropriate.
Collapse
Affiliation(s)
- Stephen Ph Alexander
- School of Life Sciences, University of Nottingham Medical School, Nottingham, NG7 2UH, UK
| | - Arthur Christopoulos
- Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria 3052, Australia
| | | | - Eamonn Kelly
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Neil V Marrion
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Elena Faccenda
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Simon D Harding
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Adam J Pawson
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Joanna L Sharman
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Christopher Southan
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Jamie A Davies
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD, UK
| |
Collapse
|
11839
|
Affiliation(s)
- Robert Zeiser
- From the Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany (R.Z.); and the Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis (B.R.B.)
| | - Bruce R Blazar
- From the Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany (R.Z.); and the Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis (B.R.B.)
| |
Collapse
|
11840
|
Chuang SY, Lin YK, Lin CF, Wang PW, Chen EL, Fang JY. Elucidating the Skin Delivery of Aglycone and Glycoside Flavonoids: How the Structures Affect Cutaneous Absorption. Nutrients 2017; 9:nu9121304. [PMID: 29189718 PMCID: PMC5748754 DOI: 10.3390/nu9121304] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 11/21/2017] [Accepted: 11/28/2017] [Indexed: 12/14/2022] Open
Abstract
Flavonoids are bioactive phytochemicals that exhibit protective potential against cutaneous inflammation and photoaging. We selected eight flavonoid aglycones or glycosides to elucidate the chemistry behind their skin absorption capability through experimental and computational approaches. The skin delivery was conducted using nude mouse and pig skins mounted on an in vitro Franz cell assembly. The anti-inflammatory activity was examined using the O2•– and elastase inhibition in activated human neutrophils. In the equivalent dose (6 mM) application on nude mouse skin, the skin deposition of naringenin and kaempferol was 0.37 and 0.11 nM/mg, respectively, which was higher than that of the other flavonoids. Both penetrants were beneficial for targeted cutaneous therapy due to their minimal diffusion across the skin. The absorption was generally greater for topically applied aglycones than glycosides. Although naringenin could be classified as a hydrophilic flavonoid, the flexibility of the chiral center in the C ring of this flavanone could lead to better skin transport than the flavonols and flavones with a planar structure. An optimized hydrophilic and lipophilic balance of the flavonoid structure was important for governing the cutaneous delivery. The hydrogen bond acceptor and stratum corneum lipid docking estimated by molecular modeling showed some relationships with the skin deposition. The interaction with cholesteryl sulfate could be a factor for predicting the cutaneous absorption of aglycone flavonoids (correlation coefficient = 0.97). Baicalin (3 µM) showed the highest activity against oxidative burst with an O2•– inhibition percentage of 77%. Although naringenin displayed an inhibition efficiency of only 20%, this compound still demonstrated an impressive therapeutic index because of the high absorption. Our data are advantageous to providing the information on the structure–permeation relationship for topically applied flavonoids.
Collapse
Affiliation(s)
- Shih-Yi Chuang
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan 333, Taiwan.
| | - Yin-Ku Lin
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung 204, Taiwan.
- School of Traditional Chinese Medicine, Chang Gung University, Kweishan, Taoyuan 333, Taiwan.
| | - Chwan-Fwu Lin
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan 333, Taiwan.
- Department of Cosmetic Science, Chang Gung University of Science and Technology, Kweishan, Taoyuan 333, Taiwan.
| | - Pei-Wen Wang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan.
| | - En-Li Chen
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan 333, Taiwan.
| | - Jia-You Fang
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan 333, Taiwan.
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan 333, Taiwan.
- Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Kweishan, Taoyuan 333, Taiwan.
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan 333, Taiwan.
| |
Collapse
|
11841
|
Zhu L, Su F, Xu Y, Zou Q. Network-based method for mining novel HPV infection related genes using random walk with restart algorithm. Biochim Biophys Acta Mol Basis Dis 2017; 1864:2376-2383. [PMID: 29197659 DOI: 10.1016/j.bbadis.2017.11.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/03/2017] [Accepted: 11/26/2017] [Indexed: 12/27/2022]
Abstract
The human papillomavirus (HPV), a common virus that infects the reproductive tract, may lead to malignant changes within the infection area in certain cases and is directly associated with such cancers as cervical cancer, anal cancer, and vaginal cancer. Identification of novel HPV infection related genes can lead to a better understanding of the specific signal pathways and cellular processes related to HPV infection, providing information for the development of more efficient therapies. In this study, several novel HPV infection related genes were predicted by a computation method based on the known genes involved in HPV infection from HPVbase. This method applied the algorithm of random walk with restart (RWR) to a protein-protein interaction (PPI) network. The candidate genes were further filtered by the permutation and association tests. These steps eliminated genes occupying special positions in the PPI network and selected key genes with strong associations to known HPV infection related genes based on the interaction confidence and functional similarity obtained from published databases, such as STRING, gene ontology (GO) terms and KEGG pathways. Our study identified 104 novel HPV infection related genes, a number of which were confirmed to relate to the infection processes and complications of HPV infection, as reported in the literature. These results demonstrate the reliability of our method in identifying HPV infection related genes. This article is part of a Special Issue entitled: Accelerating Precision Medicine through Genetic and Genomic Big Data Analysis edited by Yudong Cai & Tao Huang.
Collapse
Affiliation(s)
- Liucun Zhu
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Fangchu Su
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - YaoChen Xu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Quan Zou
- School of Computer Science and Technology, TianJin University, Tianjin 300350, China.
| |
Collapse
|
11842
|
Rosdah AA, Bond ST, Sivakumaran P, Hoque A, Oakhill JS, Drew BG, Delbridge LMD, Lim SY. Mdivi-1 Protects Human W8B2 + Cardiac Stem Cells from Oxidative Stress and Simulated Ischemia-Reperfusion Injury. Stem Cells Dev 2017; 26:1771-1780. [PMID: 29054138 DOI: 10.1089/scd.2017.0157] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cardiac stem cell (CSC) therapy is a promising approach to treat ischemic heart disease. However, the poor survival of transplanted stem cells in the ischemic myocardium has been a major impediment in achieving an effective cell-based therapy against myocardial infarction. Inhibiting mitochondrial fission has been shown to promote survival of several cell types. However, the role of mitochondrial morphology in survival of human CSC remains unknown. In this study, we investigated whether mitochondrial division inhibitor-1 (Mdivi-1), an inhibitor of mitochondrial fission protein dynamin-related protein-1 (Drp1), can improve survival of a novel population of human W8B2+ CSCs in hydrogen peroxide (H2O2)-induced oxidative stress and simulated ischemia-reperfusion injury models. Mdivi-1 significantly reduced H2O2-induced cell death in a dose-dependent manner. This cytoprotective effect was accompanied by an increased proportion of cells with tubular mitochondria, but independent of mitochondrial membrane potential recovery and reduction of mitochondrial superoxide production. In simulated ischemia-reperfusion injury model, Mdivi-1 given as a pretreatment or throughout ischemia-reperfusion injury significantly reduced cell death. However, the cytoprotective effect of Mdivi-1 was not observed when given at reperfusion. Moreover, the cytoprotective effect of Mdivi-1 in the simulated ischemia-reperfusion injury model was not accompanied by changes in mitochondrial morphology, mitochondrial membrane potential, or mitochondrial reactive oxygen species production. Mdivi-1 also did not affect mitochondrial bioenergetics of intact W8B2+ CSCs. Taken together, these experiments demonstrated that Mdivi-1 treatment of human W8B2+ CSCs enhances their survival and can be employed to improve therapeutic efficacy of CSCs for ischemic heart disease.
Collapse
Affiliation(s)
- Ayeshah A Rosdah
- 1 St Vincent's Institute of Medical Research , Fitzroy, Australia .,2 Department of Physiology, University of Melbourne , Melbourne, Australia .,3 Faculty of Medicine, Universitas Sriwijaya , Palembang, Indonesia
| | - Simon T Bond
- 4 Molecular Metabolism and Ageing Laboratory, Baker Heart and Diabetes Institute , Melbourne, Australia
| | | | - Ashfaqul Hoque
- 1 St Vincent's Institute of Medical Research , Fitzroy, Australia
| | - Jonathan S Oakhill
- 1 St Vincent's Institute of Medical Research , Fitzroy, Australia .,5 Mary MacKillop Institute for Health Research, Australian Catholic University , Melbourne, Australia
| | - Brian G Drew
- 4 Molecular Metabolism and Ageing Laboratory, Baker Heart and Diabetes Institute , Melbourne, Australia
| | - Lea M D Delbridge
- 2 Department of Physiology, University of Melbourne , Melbourne, Australia
| | - Shiang Y Lim
- 1 St Vincent's Institute of Medical Research , Fitzroy, Australia .,6 Department of Surgery, University of Melbourne , Melbourne, Australia
| |
Collapse
|
11843
|
Schubert ML, Hoffmann JM, Dreger P, Müller-Tidow C, Schmitt M. Chimeric antigen receptor transduced T cells: Tuning up for the next generation. Int J Cancer 2017; 142:1738-1747. [PMID: 29119551 DOI: 10.1002/ijc.31147] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/05/2017] [Accepted: 10/23/2017] [Indexed: 12/17/2022]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has recently achieved impressive clinical outcome in patients with CD19-positive hematologic malignancies. Extrapolation of CAR T cell treatment to solid tumors, however, has not yet yielded similar results. This might be due to intrinsic causes, e.g. insufficient CAR T cell activation or CAR toxicity as well as extrinsic factors displaying an unfavorable tumor environment for CAR T cells by raising physical and chemical barriers. In this review, we discuss the advantages as well as major obstacles of CAR T cell therapy, particularly in the context of solid tumors, and focus on efforts and novel strategies in CAR T cell development.
Collapse
Affiliation(s)
- Maria-Luisa Schubert
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Jean-Marc Hoffmann
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
11844
|
miR-155-5p inhibition promotes the transition of bone marrow mesenchymal stem cells to gastric cancer tissue derived MSC-like cells via NF-κB p65 activation. Oncotarget 2017; 7:16567-80. [PMID: 26934326 PMCID: PMC4941335 DOI: 10.18632/oncotarget.7767] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 02/05/2016] [Indexed: 01/01/2023] Open
Abstract
Gastric cancer tissue-derived MSC-like cells (GC-MSC) share similar characteristics to bone marrow MSC (BM-MSC); however, the phenotypical and functional differences and the molecular mechanism of transition between the two cell types remain unclear. Compared to BM-MSC, GC-MSC exhibited the classic phenotype of reactive stroma cells, a stronger gastric cancer promoting capacity and lower expression of miR-155-5p. Inhibition of miR-155-5p by transfecting miRNA inhibitor induced a phenotypical and functional transition of BM-MSC into GC-MSC-like cells, and the reverse experiment deprived GC-MSC of tumor-promoting phenotype and function. NF-kappa B p65 (NF-κB p65) and inhibitor of NF-kappa B kinase subunit epsilon (IKBKE/IKKε) were identified as targets of miR-155-5p and important for miRNA inhibitor activating NF-κB p65 in the transition. Inactivation of NF-κB by pyrrolidine dithiocarbamic acid (PDTC) significantly blocked the effect of miR-155-5p inhibitor on BM-MSC. IKBKE, NF-κB p65 and phospho-NF-κB p65 proteins were highly enriched in MSC-like cells of gastric cancer tissues, and the latter two were correlated with the pathological progression of gastric cancer. In GC-MSC, the expression of miR-155-5p was downregulated and NF-κB p65 protein was increased and activated. NF-κB inactivation by PDTC or knockdown of its downstream cytokines reversed the phenotype and function of GC-MSC. Taken together, our findings revealed that miR-155-5p downregulation induces BM-MSC to acquire a GC-MSC-like phenotype and function depending on NF-κB p65 activation, which suggests a novel mechanism underlying the cancer associated MSC remodeling in the tumor microenvironment and offers an effective target and approach for gastric cancer therapy.
Collapse
|
11845
|
Abstract
Background: Checkpoint inhibitors (CPI) have now been established as standard agents in the management of patients with metastatic renal cell carcinoma (mRCC). Given the unique toxicity profiles of CPIs, a detailed understanding of their incidence rate and characteristics is critical. Objective: To perform a systematic review for the analysis of the incidence rate and characteristics of toxicities in mRCC patients treated with CPIs in published clinical trials. Methods: A systematic search of EMBASE (Ovid) and MEDLINE (Ovid) was conducted as per PRISMA guidelines to identify prospective clinical trials of checkpoint inhibitors in mRCC. The search method involved querying for the terms renal cell carcinoma or kidney carcinoma with any of the following: programmed cell death 1, PD-1, programmed cell death ligand 1, PD-L1, cytotoxic T-lymphocyte antigen 4, CTLA-4, immunotherapy, checkpoint inhibitor, anti-PD-1, or anti-PD-L1. Only prospective clinical trials were included. Results: The systematic review yielded 9,722 records through the MEDLINE (Ovid) and EMBASE (Ovid) databases. Ultimately, five prospective clinical trials with 722 patients were selected for inclusion. The rates of any grade adverse event (AE) and grade (G) 3-4 AEs were 79.9% and 20.9%, respectively. Regarding immune-related AEs (irAEs), the most common system affected by any grade irAE was the skin (30.89%) and the most common grade 3-4 irAE was related to the hepatic system (8.23%). Rates of AEs were similar across the CPI monotherapy clinical trials. Conclusions: The rates of AEs in mRCC patients treated with CPI is similar to rates in other cancers. AEs in mRCC are fairly consistent among monotherapy trials with PD-1 and PD-L1 inhibitors and as one would expect higher when CTLA-4 and PD-1 inhibitors are offered in combination.
Collapse
Affiliation(s)
| | - Jorge A Garcia
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA.,Cleveland Clinic Glickman Urological and Kidney Institute, Cleveland, OH, USA
| |
Collapse
|
11846
|
Xue Q, Bettini E, Paczkowski P, Ng C, Kaiser A, McConnell T, Kodrasi O, Quigley MF, Heath J, Fan R, Mackay S, Dudley ME, Kassim SH, Zhou J. Single-cell multiplexed cytokine profiling of CD19 CAR-T cells reveals a diverse landscape of polyfunctional antigen-specific response. J Immunother Cancer 2017; 5:85. [PMID: 29157295 PMCID: PMC5697351 DOI: 10.1186/s40425-017-0293-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/16/2017] [Indexed: 12/25/2022] Open
Abstract
Background It remains challenging to characterize the functional attributes of chimeric antigen receptor (CAR)-engineered T cell product targeting CD19 related to potency and immunotoxicity ex vivo, despite promising in vivo efficacy in patients with B cell malignancies. Methods We employed a single-cell, 16-plex cytokine microfluidics device and new analysis techniques to evaluate the functional profile of CD19 CAR-T cells upon antigen-specific stimulation. CAR-T cells were manufactured from human PBMCs transfected with the lentivirus encoding the CD19-BB-z transgene and expanded with anti-CD3/anti-CD28 coated beads. The enriched CAR-T cells were stimulated with anti-CAR or control IgG beads, stained with anti-CD4 RPE and anti-CD8 Alexa Fluor 647 antibodies, and incubated for 16 h in a single-cell barcode chip (SCBC). Each SCBC contains ~12,000 microchambers, covered with a glass slide that was pre-patterned with a complete copy of a 16-plex antibody array. Protein secretions from single CAR-T cells were captured and subsequently analyzed using proprietary software and new visualization methods. Results We demonstrate a new method for single-cell profiling of CD19 CAR-T pre-infusion products prepared from 4 healthy donors. CAR-T single cells exhibited a marked heterogeneity of cytokine secretions and polyfunctional (2+ cytokine) subsets specific to anti-CAR bead stimulation. The breadth of responses includes anti-tumor effector (Granzyme B, IFN-γ, MIP-1α, TNF-α), stimulatory (GM-CSF, IL-2, IL-8), regulatory (IL-4, IL-13, IL-22), and inflammatory (IL-6, IL-17A) functions. Furthermore, we developed two new bioinformatics tools for more effective polyfunctional subset visualization and comparison between donors. Conclusions Single-cell, multiplexed, proteomic profiling of CD19 CAR-T product reveals a diverse landscape of immune effector response of CD19 CAR-T cells to antigen-specific challenge, providing a new platform for capturing CAR-T product data for correlative analysis. Additionally, such high dimensional data requires new visualization methods to further define precise polyfunctional response differences in these products. The presented biomarker capture and analysis system provides a more sensitive and comprehensive functional assessment of CAR-T pre-infusion products and may provide insights into the safety and efficacy of CAR-T cell therapy. Electronic supplementary material The online version of this article (10.1186/s40425-017-0293-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qiong Xue
- Novartis Pharmaceuticals, 64 Sidney Street, Cambridge, MA, 02139, USA. .,Present Address: Novartis Institute of BioMedical Research, 300 Technology Square, Cambridge, MA, 02139, USA.
| | - Emily Bettini
- IsoPlexis Corporation, 35 NE Industrial Rd, Branford, CT, 06405, USA
| | | | - Colin Ng
- IsoPlexis Corporation, 35 NE Industrial Rd, Branford, CT, 06405, USA
| | - Alaina Kaiser
- IsoPlexis Corporation, 35 NE Industrial Rd, Branford, CT, 06405, USA
| | - Timothy McConnell
- IsoPlexis Corporation, 35 NE Industrial Rd, Branford, CT, 06405, USA
| | - Olja Kodrasi
- Novartis Pharmaceuticals, 64 Sidney Street, Cambridge, MA, 02139, USA.,Present Address: Novartis Institute of BioMedical Research, 64 Sidney street, Cambridge, MA, 02139, USA
| | - Máire F Quigley
- Novartis Pharmaceuticals, 64 Sidney Street, Cambridge, MA, 02139, USA.,Present Address: Novartis Pharmaceuticals, 45 Sidney Street, Cambridge, MA, 02139, USA
| | - James Heath
- NanoSystems Biology Cancer Center, Division of Chemistry, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Sean Mackay
- IsoPlexis Corporation, 35 NE Industrial Rd, Branford, CT, 06405, USA
| | - Mark E Dudley
- Novartis Pharmaceuticals, 64 Sidney Street, Cambridge, MA, 02139, USA.,Present Address: Adaptimmune, 351 Rouse Blvd, Philadelphia, PA, 19112, USA
| | - Sadik H Kassim
- Novartis Pharmaceuticals, 64 Sidney Street, Cambridge, MA, 02139, USA.,Present Address: Mustang Bio, 95 Sawyer Road, Waltham, MA, 02453, USA
| | - Jing Zhou
- IsoPlexis Corporation, 35 NE Industrial Rd, Branford, CT, 06405, USA.
| |
Collapse
|
11847
|
Pierini S, Perales-Linares R, Uribe-Herranz M, Pol JG, Zitvogel L, Kroemer G, Facciabene A, Galluzzi L. Trial watch: DNA-based vaccines for oncological indications. Oncoimmunology 2017; 6:e1398878. [PMID: 29209575 DOI: 10.1080/2162402x.2017.1398878] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 12/16/2022] Open
Abstract
DNA-based vaccination is a promising approach to cancer immunotherapy. DNA-based vaccines specific for tumor-associated antigens (TAAs) are indeed relatively simple to produce, cost-efficient and well tolerated. However, the clinical efficacy of DNA-based vaccines for cancer therapy is considerably limited by central and peripheral tolerance. During the past decade, considerable efforts have been devoted to the development and characterization of novel DNA-based vaccines that would circumvent this obstacle. In this setting, particular attention has been dedicated to the route of administration, expression of modified TAAs, co-expression of immunostimulatory molecules, and co-delivery of immune checkpoint blockers. Here, we review preclinical and clinical progress on DNA-based vaccines for cancer therapy.
Collapse
Affiliation(s)
- Stefano Pierini
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Renzo Perales-Linares
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mireia Uribe-Herranz
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan G Pol
- Université Paris Descartes/Paris V, France.,Université Pierre et Marie Curie/Paris VI, Paris.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France.,Université Pierre et Marie Curie/Paris VI, Paris.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Andrea Facciabene
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
11848
|
Chang SM, Hu WW. Long non-coding RNA MALAT1 promotes oral squamous cell carcinoma development via microRNA-125b/STAT3 axis. J Cell Physiol 2017; 233:3384-3396. [PMID: 28926115 DOI: 10.1002/jcp.26185] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 09/15/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Shi-Min Chang
- Department of Stomatology; Beijing Friendship Hospital; Capital Medical University; Xicheng District Beijing China
| | - Wei-Wei Hu
- Department of Stomatology; Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University; Huai'an China
| |
Collapse
|
11849
|
Stamato MA, Juli G, Romeo E, Ronchetti D, Arbitrio M, Caracciolo D, Neri A, Tagliaferri P, Tassone P, Amodio N. Inhibition of EZH2 triggers the tumor suppressive miR-29b network in multiple myeloma. Oncotarget 2017; 8:106527-106537. [PMID: 29290968 PMCID: PMC5739753 DOI: 10.18632/oncotarget.22507] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/29/2017] [Indexed: 11/25/2022] Open
Abstract
Downregulation of tumor suppressor (TS) microRNAs (miRNAs) commonly occurs in human cancer, including multiple myeloma (MM). We previously demonstrated that miR-29b is a relevant TS miRNA, whose expression in MM cells is inhibited by HDAC4-dependent deacetylation. Here, we provide novel insights into epigenetic mechanisms suppressing miR-29b in MM. In MM patient-derived plasma cells, we found inverse correlation between miR-29b and EZH2 mRNA expression. Both siRNAs and pharmacologic inhibitors of EZH2 led to miR-29b upregulation, and this effect was ascribed to reduced H3K27-trimethylation (H3K27me3) of miR-29a/b-1 promoter regions. Induction of miR-29b upon EZH2 inhibition occurred together with downregulation of major miR-29b pro-survival targets, such as SP1, MCL-1 and CDK6. Knock-down of the EZH2-interacting long non-coding RNA MALAT1 also reduced H3K27me3 of miR-29a/b-1 promoter, along with induction of miR-29b and downregulation of miR-29b targets. Importantly, inhibition of miR-29b by antagomiRs dramatically reduced in vitro anti-MM activity of small molecule EZH2-inhibitors, indicating that functional miR-29b is crucial for the activity of these compounds. Altogether, these results disclose novel epigenetic alterations contributing to the suppression of miR-29b molecular network, which can be instrumental for the development of rationally designed miRNA-based anti-MM therapeutics.
Collapse
Affiliation(s)
- Maria Angelica Stamato
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Giada Juli
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Enrica Romeo
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Domenica Ronchetti
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.,Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Antonino Neri
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.,Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, US
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
11850
|
Delgoffe GM. Filling the Tank: Keeping Antitumor T Cells Metabolically Fit for the Long Haul. Cancer Immunol Res 2017; 4:1001-1006. [PMID: 27908931 DOI: 10.1158/2326-6066.cir-16-0244] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 10/31/2016] [Accepted: 11/02/2016] [Indexed: 02/06/2023]
Abstract
Discoveries in tumor immunology and subsequent clinical advances in cancer immunotherapy have revealed that the immune system is not oblivious to tumor progression but heavily interacts with developing neoplasia and malignancy. A major factor preventing immune destruction is the establishment of a highly immunosuppressive tumor microenvironment (TME), which provides architecture to the tumor, supports indirect means of immunosuppression such as the recruitment of tolerogenic cells like regulatory T cells and myeloid-derived suppressor cells (MDSC), and represents a zone of metabolically dearth conditions. T-cell activation and consequent effector function are cellular states characterized by extreme metabolic demands, and activation in the context of insufficient metabolic substrates results in anergy or regulatory differentiation. Thus, T cells must endure both immunosuppression (co-inhibitory molecule ligation, regulatory T cells, and suppressive cytokines) but also a sort of metabolic suppression in the TME. Here I will review the general features of the TME, identify the metabolic demands of activated effector T cells, discuss the known metabolic checkpoints associated with intratumoral T cells, and propose strategies for generating superior antitumor T cells, whether in vitro for adoptive cell therapy or through in vivo reinvigoration of the existing immune response. Cancer Immunol Res; 4(12); 1001-6. ©2016 AACR.
Collapse
Affiliation(s)
- Greg M Delgoffe
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania. .,Tumor Microenvironment Center, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| |
Collapse
|