1
|
Khurshid S, Venkataramany AS, Montes M, Kipp JF, Roberts RD, Wein N, Rigo F, Wang PY, Cripe TP, Chandler DS. Employing splice-switching oligonucleotides and AAVrh74.U7 snRNA to target insulin receptor splicing and cancer hallmarks in osteosarcoma. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200908. [PMID: 39720325 PMCID: PMC11666956 DOI: 10.1016/j.omton.2024.200908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/10/2024] [Accepted: 11/21/2024] [Indexed: 12/26/2024]
Abstract
Patients with osteosarcoma (OS), a debilitating pediatric bone malignancy, have limited treatment options to combat aggressive disease. OS thrives on insulin growth factor (IGF)-mediated signaling that can facilitate cell proliferation. Previous efforts to target IGF-1R signaling were mostly unsuccessful, likely due to compensatory signaling through alternative splicing of the insulin receptor (IR) to the proliferative IR-A isoform. Here, we leverage splice-switching oligonucleotides (SSOs) to mitigate IR splicing toward the IR-B isoform. We show that SSOs can modulate cancer cell hallmarks and anoikis-resistant growth. Furthermore, we engineered the SSO sequence in an U7 snRNA packaged in an adeno-associated virus (AAV) to test the feasibility of viral vector-mediated gene therapy delivery. We noted modest increases in IR-B isoform levels after virus transduction, which prompted us to investigate the role of combinatorial treatments with dalotuzumab, an anti-IGF-1R monoclonal antibody. After observing additive impacts on phosphoprotein phosphorylation and anoikis-resistant growth with the dalotuzumab and SSO combination, we treated OS cells with dalotuzumab and the AAVrh74.U7 snRNA IR virus, which significantly slowed OS cell proliferation. While these viruses require further optimization, we highlight the potential for SSO therapy and viral vector delivery, as it may offer new treatment avenues for OS patients and be translated to other cancers.
Collapse
Affiliation(s)
- Safiya Khurshid
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Akila S. Venkataramany
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA
- Medical Scientist Training Program, The Ohio State University, Columbus, OH 43210, USA
| | - Matias Montes
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - John F. Kipp
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Ryan D. Roberts
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Division of Hematology, Oncology and Blood and Marrow Transplant, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43215, USA
| | - Nicolas Wein
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA 92010, USA
| | - Pin-Yi Wang
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Timothy P. Cripe
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Division of Hematology, Oncology and Blood and Marrow Transplant, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43215, USA
| | - Dawn S. Chandler
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Molecular, Cellular and Developmental Biology Graduate Program and The Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
2
|
Fatema K, Luelling S, Kirkham M, Pavek A, Heyneman AL, Barrott J. Epigenetics and precision medicine in bone and soft tissue sarcomas. EPIGENETICS IN PRECISION MEDICINE 2022:147-191. [DOI: 10.1016/b978-0-12-823008-4.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
3
|
Shi Y, Ye Z, Lu G, Yang N, Zhang J, Wang L, Cui J, del Pozo MA, Wu Y, Xia D, Shen HM. Cholesterol-enriched membrane micro-domaindeficiency induces doxorubicin resistancevia promoting autophagy in breast cancer. Mol Ther Oncolytics 2021; 23:311-329. [PMID: 34786475 PMCID: PMC8573103 DOI: 10.1016/j.omto.2021.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/26/2021] [Accepted: 10/07/2021] [Indexed: 10/27/2022] Open
Abstract
Drug resistance has become one of the largest challenges for cancer chemotherapies. Under certain conditions, cancer cells hijack autophagy to cope with therapeutic stress, which largely undermines the chemo-therapeutic efficacy. Currently, biomarkers indicative of autophagy-derived drug resistance remain largely inclusive. Here, we report a novel role of lipid rafts/cholesterol-enriched membrane micro-domains (CEMMs) in autophagosome biogenesis and doxorubicin resistance in breast tumors. We showed that CEMMs are required for the interaction of VAMP3 with syntaxin 6 (STX6, a cholesterol-binding SNARE protein). Upon disruption of CEMM, VAMP3 is released from STX6, resulting in the trafficking of ATG16L1-containing vesicles to recycling endosomes and subsequent autophagosome biogenesis. Furthermore, we found that CEMM marker CAV1 is decreased in breast cancer patients and that the CEMM deficiency-induced autophagy is related to doxorubicin resistance, which is overcome by autophagy inhibition. Taken together, we propose a novel model whereby CEMMs in recycling endosomes support the VAMP3 and STX6 interaction and function as barriers to limit the activity of VAMP3 in autophagic vesicle fusion, thus CEMM deficiency promotes autophagosome biogenesis and doxorubicin resistance in breast tumors.
Collapse
Affiliation(s)
- Yin Shi
- Department of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore 119077, Singapore
| | - Zu Ye
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore 119077, Singapore
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston 77030, USA
| | - Guang Lu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore 119077, Singapore
| | - Naidi Yang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore 119077, Singapore
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University, Nanjing, Jiangsu Province 211800, China
| | - Jianbin Zhang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore 119077, Singapore
| | - Liming Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore 119077, Singapore
- School of Biomedical Science, Hunan University, Changsha, Hunan, China
| | - Jianzhou Cui
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore 119077, Singapore
| | - Miguel A. del Pozo
- Integrin Signaling Laboratory, Vascular Biology and Inflammation Department, Centro Nacional de Investigaciones Cardiovasculares, Madrid 28029, Spain
| | - Yihua Wu
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Dajing Xia
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore 119077, Singapore
- Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| |
Collapse
|
4
|
Prospects for NK Cell Therapy of Sarcoma. Cancers (Basel) 2020; 12:cancers12123719. [PMID: 33322371 PMCID: PMC7763692 DOI: 10.3390/cancers12123719] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Sarcomas are a group of aggressive tumors originating from mesenchymal tissues. Patients with advanced disease have poor prognosis due to the ineffectiveness of current treatment protocols. A subset of lymphocytes called natural killer (NK) cells is capable of effective surveillance and clearance of sarcomas, constituting a promising tool for immunotherapeutic treatment. However, sarcomas can cause impairment in NK cell function, associated with enhanced tumor growth and dissemination. In this review, we discuss the molecular mechanisms of sarcoma-mediated suppression of NK cells and their implications for the design of novel NK cell-based immunotherapies against sarcoma. Abstract Natural killer (NK) cells are innate lymphoid cells with potent antitumor activity. One of the most NK cell cytotoxicity-sensitive tumor types is sarcoma, an aggressive mesenchyme-derived neoplasm. While a combination of radical surgery and radio- and chemotherapy can successfully control local disease, patients with advanced sarcomas remain refractory to current treatment regimens, calling for novel therapeutic strategies. There is accumulating evidence for NK cell-mediated immunosurveillance of sarcoma cells during all stages of the disease, highlighting the potential of using NK cells as a therapeutic tool. However, sarcomas display multiple immunoevasion mechanisms that can suppress NK cell function leading to an uncontrolled tumor outgrowth. Here, we review the current evidence for NK cells’ role in immune surveillance of sarcoma during disease initiation, promotion, progression, and metastasis, as well as the molecular mechanisms behind sarcoma-mediated NK cell suppression. Further, we apply this basic understanding of NK–sarcoma crosstalk in order to identify and summarize the most promising candidates for NK cell-based sarcoma immunotherapy.
Collapse
|
5
|
Natural Killer Cell Immunotherapy for Osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1257:141-154. [PMID: 32483737 DOI: 10.1007/978-3-030-43032-0_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Natural killer (NK) cells are lymphocytes of the innate immune system that have the ability to recognize malignant cells through balanced recognition of cell-surface indicators of stress and danger. Once activated through such recognition, NK cells release cytokines and induce target cell lysis through multiple mechanisms. NK cells are increasingly recognized for their role in controlling tumor progression and metastasis and as important mediators of immunotherapeutic modalities such as cytokines, antibodies, immunomodulating drugs, and stem cell transplantation. Recent advances in manipulating NK cell number, function, and genetic modification have caused renewed interest in their potential for adoptive immunotherapies, which are actively being tested in clinical trials. Here, we summarize the evidence for NK cell recognition of osteosarcoma, discuss immune therapies that are directly or indirectly dependent on NK cell function, and describe potential approaches for manipulating NK cell number and function to enhance therapy against osteosarcoma.
Collapse
|
6
|
Mollinedo F, Gajate C. Lipid rafts as signaling hubs in cancer cell survival/death and invasion: implications in tumor progression and therapy: Thematic Review Series: Biology of Lipid Rafts. J Lipid Res 2020; 61:611-635. [PMID: 33715811 PMCID: PMC7193951 DOI: 10.1194/jlr.tr119000439] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/17/2020] [Indexed: 12/13/2022] Open
Abstract
Cholesterol/sphingolipid-rich membrane domains, known as lipid rafts or membrane rafts, play a critical role in the compartmentalization of signaling pathways. Physical segregation of proteins in lipid rafts may modulate the accessibility of proteins to regulatory or effector molecules. Thus, lipid rafts serve as sorting platforms and hubs for signal transduction proteins. Cancer cells contain higher levels of intracellular cholesterol and lipid rafts than their normal non-tumorigenic counterparts. Many signal transduction processes involved in cancer development (insulin-like growth factor system and phosphatidylinositol 3-kinase-AKT) and metastasis [cluster of differentiation (CD)44] are dependent on or modulated by lipid rafts. Additional proteins playing an important role in several malignant cancers (e.g., transmembrane glycoprotein mucin 1) are also being detected in association with lipid rafts, suggesting a major role of lipid rafts in tumor progression. Conversely, lipid rafts also serve as scaffolds for the recruitment and clustering of Fas/CD95 death receptors and downstream signaling molecules leading to cell death-promoting raft platforms. The partition of death receptors and downstream signaling molecules in aggregated lipid rafts has led to the formation of the so-called cluster of apoptotic signaling molecule-enriched rafts, or CASMER, which leads to apoptosis amplification and can be pharmacologically modulated. These death-promoting rafts can be viewed as a linchpin from which apoptotic signals are launched. In this review, we discuss the involvement of lipid rafts in major signaling processes in cancer cells, including cell survival, cell death, and metastasis, and we consider the potential of lipid raft modulation as a promising target in cancer therapy.
Collapse
Affiliation(s)
- Faustino Mollinedo
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Cientificas (CSIC), E-28040 Madrid, Spain. mailto:
| | - Consuelo Gajate
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Cientificas (CSIC), E-28040 Madrid, Spain
| |
Collapse
|
7
|
Muraki M. Sensitization to cell death induced by soluble Fas ligand and agonistic antibodies with exogenous agents: A review. AIMS MEDICAL SCIENCE 2020. [DOI: 10.3934/medsci.2020011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
8
|
The Fas/FasL Signaling Pathway: Its Role in the Metastatic Process and as a Target for Treating Osteosarcoma Lung Metastases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1258:177-187. [PMID: 32767242 DOI: 10.1007/978-3-030-43085-6_12] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Understanding how the tumor microenvironment participates in inhibiting or supporting tumor growth is critical for the development of novel therapies. Osteosarcoma (OS) metastasizes almost exclusively to the lung, an organ where Fas ligand (FasL) is constitutively expressed. This chapter focuses on our studies dedicated to the interaction of OS cells with the lung microenvironment. We will summarize our studies conducted over the past 20 years showing the importance of the Fas/FasL signaling pathway to the establishment and progression of OS metastases in the lung. We demonstrated that the FasL+ lung microenvironment eliminates Fas-positive (Fas+) OS cells that metastasize to the lungs, through apoptosis induced by Fas signaling following interaction of Fas on the tumor cell surface with FasL on the lung epithelial cells. Expression of the Fas receptor on OS cells inversely correlated with the ability of OS cells to form lung metastases. Blocking this pathway interferes with this process, allowing Fas+ cells to grow in the lung. By contrast, upregulation of Fas on Fas- OS cells inhibited their ability to metastasize to the lung. We demonstrated how the FasL+ lung microenvironment can be leveraged for therapeutic intent through the upregulation of Fas expression. To this end, we demonstrated that the histone deacetylase inhibitor entinostat upregulated Fas expression on OS cells, reduced their ability to form lung metastases, and induced regression of established micrometastases. Fas expression in OS cells is regulated epigenetically by the microRNA miR-20a. We showed that expressions of Fas and miR-20a are inversely correlated, and that delivery of anti-miR-20a in vivo to mice with established osteosarcoma lung metastases resulted in upregulation of Fas and tumor regression. Therefore, targeting the Fas signaling pathway may present therapeutic opportunities, which target the lung microenvironment for elimination of OS lung metastases. We have also shown that in addition to being critically involved in the metastatic potential, the Fas signaling pathway may also contribute to the efficacy of chemotherapy. We demonstrated that the chemotherapeutic agent gemcitabine (GCB) increased Fas expression in both human and mouse OS cells in vitro. In vivo, aerosol GCB therapy induced upregulation of Fas expression and the regression of established osteosarcoma lung metastases. The therapeutic efficacy of GCB was contingent upon a FasL+ lung microenvironment as aerosol GCB had no effect in FasL-deficient mice. Manipulation of Fas expression and the Fas pathway should be considered, as this concept may provide additional novel therapeutic approaches for treating patients with OS lung metastases.
Collapse
|
9
|
Wang Z, Wang Z, Li B, Wang S, Chen T, Ye Z. Innate Immune Cells: A Potential and Promising Cell Population for Treating Osteosarcoma. Front Immunol 2019; 10:1114. [PMID: 31156651 PMCID: PMC6531991 DOI: 10.3389/fimmu.2019.01114] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 05/01/2019] [Indexed: 12/13/2022] Open
Abstract
Advanced, recurrent, or metastasized osteosarcomas remain challenging to cure or even alleviate. Therefore, the development of novel therapeutic strategies is urgently needed. Cancer immunotherapy has greatly improved in recent years, with options including adoptive cellular therapy, vaccination, and checkpoint inhibitors. As such, immunotherapy is becoming a potential strategy for the treatment of osteosarcoma. Innate immunocytes, the first line of defense in the immune system and the bridge to adaptive immunity, are one of the vital effector cell subpopulations in cancer immunotherapy. Innate immune cell-based therapy has shown potent antitumor activity against hematologic malignancies and some solid tumors, including osteosarcoma. Importantly, some immune checkpoints are expressed on both innate and adaptive immune cells, modulating their functions in tumor immunity. Therefore, blocking or activating immune checkpoint-mediated downstream signaling pathways can improve the therapeutic effects of innate immune cell-based therapy. In this review, we summarize the current status and future prospects of innate immune cell-based therapy for the treatment of osteosarcoma, with a focus on the potential synergistic effects of combination therapy involving innate immunotherapy and immune checkpoint inhibitors/oncolytic viruses.
Collapse
Affiliation(s)
- Zenan Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Zhan Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Binghao Li
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Shengdong Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Tao Chen
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Zhaoming Ye
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
Huang Y, Yang W, Zeng H, Hu C, Zhang Y, Ding N, Fan G, Shao L, Kuang B. Droxinostat sensitizes human colon cancer cells to apoptotic cell death via induction of oxidative stress. Cell Mol Biol Lett 2018; 23:34. [PMID: 30065760 PMCID: PMC6064062 DOI: 10.1186/s11658-018-0101-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 07/19/2018] [Indexed: 01/08/2023] Open
Abstract
Upregulation of histone acetylation plays a critical role in the dysregulation of transcription. It alters the structure of chromatin, which leads to the onset of cancer. Histone deacetylase inhibitors may therefore be a promising way to limit cancer progression. In this study, we examined the effects of droxinostat on the growth of HT-29 colon cancer cells. Our results show that droxinostat effectively inhibited cell growth and colony-forming ability by inducing cellular apoptosis and ROS production in HT-29 cells. Notably, the apoptotic inhibitor Z-VAD-FMK significantly decreased the levels of cellular apoptosis and the antioxidant γ-tocotrienol (GT3) significantly decreased ROS production induced by droxinostat treatment. Z-VAD-FMK and GT3 also partially reversed the negative growth effects of droxinstat on HT-29 cells. GT3 treatment decreased cellular apoptosis and increased colony-forming ability upon droxinostat administration. Z-VAD-FMK treatment also partially decreased droxinostat-induced ROS production. Our findings suggest that the effects of droxinostat on colon cancer cells are mediated by the induction of oxidative stress and apoptotic cell death.
Collapse
Affiliation(s)
- Ying Huang
- 1Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006 China.,2Medical School of Nanchang University, 461 Bayi Road, Nanchang, 330006 Jiangxi China
| | - Wuping Yang
- 1Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006 China.,2Medical School of Nanchang University, 461 Bayi Road, Nanchang, 330006 Jiangxi China
| | - Huihong Zeng
- 2Medical School of Nanchang University, 461 Bayi Road, Nanchang, 330006 Jiangxi China
| | - Chuan Hu
- 2Medical School of Nanchang University, 461 Bayi Road, Nanchang, 330006 Jiangxi China
| | - Yaqiong Zhang
- 2Medical School of Nanchang University, 461 Bayi Road, Nanchang, 330006 Jiangxi China
| | - Nanhua Ding
- 2Medical School of Nanchang University, 461 Bayi Road, Nanchang, 330006 Jiangxi China
| | - Guangqin Fan
- 1Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006 China.,3School of Public Health, Nanchang University, Nanchang, 330006 China
| | - Lijian Shao
- 1Jiangxi provincial key laboratory of preventive medicine, Nanchang University, Nanchang, 330006 China.,3School of Public Health, Nanchang University, Nanchang, 330006 China
| | - Bohai Kuang
- 2Medical School of Nanchang University, 461 Bayi Road, Nanchang, 330006 Jiangxi China
| |
Collapse
|
11
|
Expression patterns of class I histone deacetylases in osteosarcoma: a novel prognostic marker with potential therapeutic implications. Mod Pathol 2018; 31:264-274. [PMID: 28984297 PMCID: PMC5811636 DOI: 10.1038/modpathol.2017.125] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/18/2017] [Accepted: 08/20/2017] [Indexed: 12/28/2022]
Abstract
Epigenetic aberrations are recognized as having pivotal roles in cancer etiology and progression. Histone deacetylases are among the most studied epigenetic modulators in various cancer types. The expression levels of class I histone deacetylase isoforms 1, 2, and 3 in patient-derived primary osteosarcoma cells (6 cases) was investigated, comparing them to normal bone graft-derived osteoblasts (6 cases) using the immunoblotting technique. Expression profiles of histone deacetylases in high-grade osteosarcoma tissue of 89 patients were examined and their association with clinicopathologic parameters and the patient survival was evaluated. Histone deacetylases were immunohistochemically stained on formalin-fixed paraffin-embedded biopsied tissue. Primary osteosarcoma cells expressed higher levels of histone deacetylase 1 and histone deacetylase 2, but lower levels of histone deacetylase 3 compared to benign osteoblasts. Overall, 82, 99, and 93% of 89 osteosarcomas showed nuclear expression of the histone deacetylase isoforms 1, 2, and 3, respectively. Low levels of histone deacetylase 1 were significantly associated with a high Enneking stage (P=0.014) and the presence of initial metastasis (P=0.040), while low levels of histone deacetylase 3 were significantly correlated with age >15 years (P=0.026). Univariate survival analysis found significantly shorter survival in the patients with a high Enneking stage (P<0.001), axial location (P=0.009), presence of initial metastasis (P<0.001), low-histone deacetylase 1 expression (P=0.038), and low-all-histone deacetylases expression (P=0.016). Multivariate survival analysis showed that only axial location (P=0.011) and low-all-histone deacetylases expression (P=0.039) were independent prognostic factors. In subgroup analysis of stage IIB patients (n=45), only axial location and low-all-histone deacetylases expression were associated with shorter survival in both univariate and multivariate analysis (axial location, P=0.008 and 0.010; low-all-HDACs, P=0.013 and 0.038, respectively). Low levels of all-histone deacetylases expression were significantly associated with advanced disease status and short survival. These findings may be a guide to future use of histone deacetylase inhibitors in osteosarcoma patients.
Collapse
|
12
|
Yang Y, Huang G, Zhou Z, Fewell JG, Kleinerman ES. miR-20a Regulates FAS Expression in Osteosarcoma Cells by Modulating FAS Promoter Activity and Can be Therapeutically Targeted to Inhibit Lung Metastases. Mol Cancer Ther 2017; 17:130-139. [PMID: 29079708 DOI: 10.1158/1535-7163.mct-17-0042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 06/02/2017] [Accepted: 10/05/2017] [Indexed: 11/16/2022]
Abstract
The metastatic potential of osteosarcoma cells is inversely correlated to cell surface FAS expression. Downregulation of FAS allows osteosarcoma cells to escape FAS ligand-mediated apoptosis when they enter a FAS ligand-positive microenvironment such as the lung. We have previously demonstrated that miR-20a, encoded by the miR-17-92 cluster, downregulates FAS expression in osteosarcoma. We further demonstrated an inverse correlation between FAS expression and miR-20a expression. However, the mechanism of FAS regulation by miR-20a was still unclear. The purpose of the current study was to evaluate the mechanism of FAS regulation by miR-20a in vitro and test the effect of targeting miR-20a in vivo We investigated whether miR-20a's downregulation of FAS was mediated by binding to the 3'-untranslated region (3'-UTR) of FAS mRNA with the consequent induction of mRNA degradation or translational suppression. We identified and mutated two miR-20a binding sites on the FAS mRNA 3'-UTR. Using luciferase reporter assays, we demonstrated that miR-20a did not bind to either the wild-type or mutated FAS 3'-UTR. In contrast, overexpression of miR-20a resulted in downregulation of FAS promoter activity. Similarly, the inhibition of miR-20a increased FAS promoter activity. The critical region identified on the FAS promoter was between -240 bp and -150 bp. Delivery of anti-miR-20a in vivo using nanoparticles in mice with established osteosarcoma lung metastases resulted in upregulation of FAS and tumor growth inhibition. Taken together, our data suggest that miR-20a regulates FAS expression through the modulation of the FAS promoter and that targeting miR-20a using anti-miR-20a has therapeutic potential. Mol Cancer Ther; 17(1); 130-9. ©2017 AACR.
Collapse
Affiliation(s)
- Yuanzheng Yang
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gangxiong Huang
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Immunotherapy Institute, Fujian Medical University, University Town, Fuzhou, China
| | - Zhichao Zhou
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Eugenie S Kleinerman
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
13
|
Tang F, Choy E, Tu C, Hornicek F, Duan Z. Therapeutic applications of histone deacetylase inhibitors in sarcoma. Cancer Treat Rev 2017; 59:33-45. [PMID: 28732326 DOI: 10.1016/j.ctrv.2017.06.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 02/05/2023]
Abstract
Sarcomas are a rare group of malignant tumors originating from mesenchymal stem cells. Surgery, radiation and chemotherapy are currently the only standard treatments for sarcoma. However, their response rates to chemotherapy are quite low. Toxic side effects and multi-drug chemoresistance make treatment even more challenging. Therefore, better drugs to treat sarcomas are needed. Histone deacetylase inhibitors (HDAC inhibitors, HDACi, HDIs) are epigenetic modifying agents that can inhibit sarcoma growth in vitro and in vivo through a variety of pathways, including inducing tumor cell apoptosis, causing cell cycle arrest, impairing tumor invasion and preventing metastasis. Importantly, preclinical studies have revealed that HDIs can not only sensitize sarcomas to chemotherapy and radiotherapy, but also increase treatment responses when combined with other chemotherapeutic drugs. Several phase I and II clinical trials have been conducted to assess the efficacy of HDIs either as monotherapy or in combination with standard chemotherapeutic agents or targeted therapeutic drugs for sarcomas. Combination regimen for sarcomas appear to be more promising than monotherapy when using HDIs. This review summarizes our current understanding and therapeutic applications of HDIs in sarcomas.
Collapse
Affiliation(s)
- Fan Tang
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, USA; Department of Orthopedics, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, Sichuan 610041, China
| | - Edwin Choy
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, USA
| | - Chongqi Tu
- Department of Orthopedics, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, Sichuan 610041, China
| | - Francis Hornicek
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, USA
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, USA.
| |
Collapse
|
14
|
Yang J, Nishihara R, Zhang X, Ogino S, Qian ZR. Energy sensing pathways: Bridging type 2 diabetes and colorectal cancer? J Diabetes Complications 2017; 31:1228-1236. [PMID: 28465145 PMCID: PMC5501176 DOI: 10.1016/j.jdiacomp.2017.04.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 04/04/2017] [Accepted: 04/10/2017] [Indexed: 12/14/2022]
Abstract
The recently rapid increase of obesity and type 2 diabetes mellitus has caused great burden to our society. A positive association between type 2 diabetes and risk of colorectal cancer has been reported by increasing epidemiological studies. The molecular mechanism of this connection remains elusive. However, type 2 diabetes may result in abnormal carbohydrate and lipid metabolism, high levels of circulating insulin, insulin growth factor-1, and adipocytokines, as well as chronic inflammation. All these factors could lead to the alteration of energy sensing pathways such as the AMP activated kinase (PRKA), mechanistic (mammalian) target of rapamycin (mTOR), SIRT1, and autophagy signaling pathways. The resulted impaired SIRT1 and autophagy signaling pathway could increase the risk of gene mutation and cancer genesis by decreasing genetic stability and DNA mismatch repair. The dysregulated mTOR and PRKA pathway could remodel cell metabolism during the growth and metastasis of cancer in order for the cancer cell to survive the unfavorable microenvironment such as hypoxia and low blood supply. Moreover, these pathways may be coupling metabolic and epigenetic alterations that are central to oncogenic transformation. Further researches including molecular pathologic epidemiologic studies are warranted to better address the precise links between these two important diseases.
Collapse
Affiliation(s)
- Juhong Yang
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215; 211 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormone and Development (Ministry of Health), Metabolic Disease Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China.
| | - Reiko Nishihara
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215; Division of MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, 75 Francis Street, Boston, MA 02115; Department of Epidemiology, Harvard School of Public Health, 677 Huntington Ave., Boston, MA 02115
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, 75 Francis Street, Boston, MA 02115
| | - Shuji Ogino
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215; Division of MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, 75 Francis Street, Boston, MA 02115; Department of Epidemiology, Harvard School of Public Health, 677 Huntington Ave., Boston, MA 02115
| | - Zhi Rong Qian
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215.
| |
Collapse
|
15
|
Huang YC, Huang FI, Mehndiratta S, Lai SC, Liou JP, Yang CR. Anticancer activity of MPT0G157, a derivative of indolylbenzenesulfonamide, inhibits tumor growth and angiogenesis. Oncotarget 2016; 6:18590-601. [PMID: 26087180 PMCID: PMC4621912 DOI: 10.18632/oncotarget.4068] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/15/2015] [Indexed: 11/30/2022] Open
Abstract
Histone deacetylases (HDACs) display multifaceted functions by coordinating the interaction of signal pathways with chromatin structure remodeling and the activation of non-histone proteins; these epigenetic regulations play an important role during malignancy progression. HDAC inhibition shows promise as a new strategy for cancer therapy; three HDAC inhibitors have been approved. We previously reported that N-hydroxy-3-{4-[2-(2-methyl-1H-indol-3-yl)-ethylsulfamoyl]-phenyl}-acrylamide (MPT0G157), a novel indole-3-ethylsulfamoylphenylacrylamide compound, demonstrated potent HDAC inhibition and anti-inflammatory effects. In this study, we evaluated its anti-cancer activity in vitro and in vivo. MPT0G157 treatment significantly inhibited different tumor growth at submicromolar concentration and was particularly potent in human colorectal cancer (HCT116) cells. Apoptosis and inhibited HDACs activity induced by MPT0G157 was more potent than that by the marketed drugs PXD101 (Belinostat) and SAHA (Vorinostat). In an in vivo model, MPT0G157 markedly inhibited HCT116 xenograft tumor volume and reduced matrigel-induced angiogenesis. The anti-angiogenetic effect of MPT0G157 was found to increase the hyperacetylation of heat shock protein 90 (Hsp90) and promote hypoxia-inducible factor-1α (HIF-1α) degradation followed by down-regulation of vascular endothelial growth factor (VEGF) expression. Our results demonstrate that MPT0G157 has potential as a new drug candidate for cancer therapy.
Collapse
Affiliation(s)
- Yen-Chia Huang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Fang-I Huang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Samir Mehndiratta
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Ssu-Chia Lai
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chia-Ron Yang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
16
|
Alfranca A, Martinez-Cruzado L, Tornin J, Abarrategi A, Amaral T, de Alava E, Menendez P, Garcia-Castro J, Rodriguez R. Bone microenvironment signals in osteosarcoma development. Cell Mol Life Sci 2015; 72:3097-113. [PMID: 25935149 PMCID: PMC11113487 DOI: 10.1007/s00018-015-1918-y] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 04/24/2015] [Accepted: 04/27/2015] [Indexed: 02/06/2023]
Abstract
The bone is a complex connective tissue composed of many different cell types such as osteoblasts, osteoclasts, chondrocytes, mesenchymal stem/progenitor cells, hematopoietic cells and endothelial cells, among others. The interaction between them is finely balanced through the processes of bone formation and bone remodeling, which regulates the production and biological activity of many soluble factors and extracellular matrix components needed to maintain the bone homeostasis in terms of cell proliferation, differentiation and apoptosis. Osteosarcoma (OS) emerges in this complex environment as a result of poorly defined oncogenic events arising in osteogenic lineage precursors. Increasing evidence supports that similar to normal development, the bone microenvironment (BME) underlies OS initiation and progression. Here, we recapitulate the physiological processes that regulate bone homeostasis and review the current knowledge about how OS cells and BME communicate and interact, describing how these interactions affect OS cell growth, metastasis, cancer stem cell fate and therapy outcome.
Collapse
Affiliation(s)
- Arantzazu Alfranca
- Unidad de Biotecnología Celular, Área de Genética Humana, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Lucia Martinez-Cruzado
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| | - Juan Tornin
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| | - Ander Abarrategi
- Unidad de Biotecnología Celular, Área de Genética Humana, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, UK
| | - Teresa Amaral
- Molecular Pathology Program, Institute of Biomedical Research of Salamanca-Centro de Investigación del Cáncer, Centro de Investigación del Cáncer (IBSAL-CIC), Salamanca, Spain
- Department of Pathology and Biobank, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS), CSIC-Universidad de Sevilla, Seville, Spain
| | - Enrique de Alava
- Molecular Pathology Program, Institute of Biomedical Research of Salamanca-Centro de Investigación del Cáncer, Centro de Investigación del Cáncer (IBSAL-CIC), Salamanca, Spain
- Department of Pathology and Biobank, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS), CSIC-Universidad de Sevilla, Seville, Spain
| | - Pablo Menendez
- Cell Therapy Program, School of Medicine, Josep Carreras Leukemia Research Institute, University of Barcelona, Barcelona, Spain
- Instituciò Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| | - Javier Garcia-Castro
- Unidad de Biotecnología Celular, Área de Genética Humana, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Rene Rodriguez
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| |
Collapse
|
17
|
Abstract
Osteosarcoma is the most common primary malignancy of bone, typically presenting in the first or second decade of life. Unfortunately, clinical outcomes for osteosarcoma patients have not substantially improved in over 30 years. This stagnation in therapeutic advances is perhaps explained by the genetic, epigenetic, and biological complexities of this rare tumor. In this review we provide a general background on the biology of osteosarcoma and the clinical status quo. We go on to enumerate the genetic and epigenetic defects identified in osteosarcoma. Finally, we discuss ongoing large-scale studies in the field and potential new therapies that are currently under investigation.
Collapse
Affiliation(s)
- James J. Morrow
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Chand Khanna
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
18
|
Abstract
Osteosarcoma (OS) is the most common primary malignant tumor of bone and the third most common cancer in childhood and adolescence. Nowadays, early diagnosis, drug resistance and recurrence of the disease represent the major challenges in OS treatment. Post-genomics, and in particular proteomic technologies, offer an invaluable opportunity to address the level of biological complexity expressed by OS. Although the main goal of OS oncoproteomics is focused on diagnostic and prognostic biomarker discovery, in this review we describe and discuss global protein profiling approaches to other aspects of OS biology and pathophysiology, or to investigate the mechanism of action of chemotherapeutics. In addition, we present proteomic analyses carried out on OS cell lines as in vitro models for studying osteoblastic cell biology and the attractive opportunity offered by proteomics of OS cancer stem cells.
Collapse
Affiliation(s)
- Giulia Bernardini
- Dipartimento di Biotecnologie, Chimica e Farmacia, via Fiorentina 1, Università degli Studi di Siena, 53100 Siena, Italy
| | | | | | | |
Collapse
|
19
|
Rao H, Ma LX, Xu TT, Li J, Deng ZY, Fan YW, Li HY. Lipid rafts and Fas/FasL pathway may involve in elaidic acid-induced apoptosis of human umbilical vein endothelial cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:798-807. [PMID: 24364735 DOI: 10.1021/jf404834e] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Our previous study showed that trans-fatty acids can cause apoptosis of endothelial cells through the caspase pathway and the mitochondrial pathway. The objective of this study was to explore how trans-fatty acids activate the caspase pathway, whether there exist specific receptors induced apoptosis by comparing normal cells and non-rafts cells treated with elaidic acid (9t18:1) and oleic acid (9c18:1), respectively. Compared to normal cells treated with 9t18:1, the cell viability increased by 13% and the number of apoptotic cells decreased by 3% in non-rafts cells treated with 9t18:1 (p < 0.05), and the expression levels of pro-apoptotic proteins such as caspase-3, -8, -9, Bax, and Bid decreased, and expression of antiapoptotic protein Bcl-2 increased (p < 0.05). In addition, Fas/FasL expression in cell membrane decreased significantly (p < 0.05). In conclusion, the lipid rafts and Fas/FasL pathway may involve in 9t18:1-induced apoptosis of human umbilical vein endothelial cells.
Collapse
Affiliation(s)
- Huan Rao
- State Key Laboratory of Food Science and Technology, Institute for Advanced Study, Nanchang University , Nanchang, Jiangxi 330047, P.R. China
| | | | | | | | | | | | | |
Collapse
|
20
|
Tarek N, Lee DA. Natural Killer Cells for Osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 804:341-53. [DOI: 10.1007/978-3-319-04843-7_19] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
21
|
Participation of the Fas/FasL signaling pathway and the lung microenvironment in the development of osteosarcoma lung metastases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 804:203-17. [PMID: 24924176 DOI: 10.1007/978-3-319-04843-7_11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The lungs are the most common site for the metastatic spread of osteosarcoma. Success in using chemotherapy to improve overall survival has reached a plateau. Understanding the biologic properties that permit osteosarcoma cells to grow in the lungs may allow the identification of novel therapeutic approaches-the goal being to alter the tumor cells' expression of cell surface proteins so that there is no longer compatibility with the metastatic niche. We have demonstrated that the Fas Ligand positive (FasL(+)) lung microenvironment eliminates Fas(+) osteosarcoma cells that metastasize to the lungs. Indeed, osteosarcoma lung metastases from patients are Fas(-), similar to what we found in several different mouse models. The Fas(+) cells are cleared from the lungs through apoptosis induced by the Fas signaling pathway following interaction of Fas on the tumor cell surface with the lung FasL. Blocking the Fas signaling pathway interferes with this process, allowing the Fas(+) cells to grow in the lungs. Our investigations show that Fas expression in osteosarcoma cells is regulated epigenetically by the micro-RNA miR-20a, encoded by the miR-17-92 cluster. Our studies support the feasibility of finding agents that can re-induce Fas expression as a novel therapeutic approach to treat osteosarcoma patients with lung metastases. We have identified two such agents, the histone deacetylase inhibitor entinostat and the chemotherapeutic agent gemcitabine (GCB). Aerosol GCB and oral entinostat induce the upregulation of Fas and the regression of established osteosarcoma lung metastases. Aerosol GCB was not effective in the FasL-deficient gld mouse confirming that the lung microenvironment was central to the success of this therapy. Our studies establish the critical role of the lung microenvironment in the metastatic process of osteosarcoma to the lungs and suggest an alternative focus for therapy, that is, incorporating the lung microenvironment as part of the treatment strategy against established osteosarcoma disease in the lungs.
Collapse
|
22
|
Sigalotti L, Fratta E, Coral S, Maio M. Epigenetic drugs as immunomodulators for combination therapies in solid tumors. Pharmacol Ther 2013; 142:339-50. [PMID: 24384533 DOI: 10.1016/j.pharmthera.2013.12.015] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 12/10/2013] [Indexed: 12/14/2022]
Abstract
Continuously improving knowledge of the fine mechanisms regulating cross-talk between immune cells, and of their multi-faceted interactions with cancer cells, has prompted the development of several novel immunotherapeutic strategies for cancer treatment. Among these, modulation of the host's immune system by targeting immunological synapses has shown notable clinical efficacy in different tumor types. Despite this, objective clinical responses and, more importantly, long-term survival are achieved only by a fraction of patients; therefore, identification of the mechanism(s) responsible for the differential effectiveness of immune checkpoint blockade in specific patient populations is an area of intense investigation. Neoplastic cells can activate multiple mechanisms to escape from immune control; among these, epigenetic reprogramming is emerging as a key player. Selected tumor-associated antigens, Human Leukocyte Antigens, and accessory/co-stimulatory molecules required for efficient recognition of neoplastic cells by the immune system have been shown to be epigenetically silenced or down-regulated in cancer. Consistent with the inherent reversibility of epigenetic silencing, "epigenetic" drugs, such as inhibitors of DNA methyltransferases and of histone deacetylases, can restore the functional expression of these down-regulated molecules, thus improving the recognition of cancer cells by both the innate and adaptive immune responses. This review focuses on the immunomodulatory activity of epigenetic drugs and on their proposed clinical use in novel combined chemo-immunotherapeutic regimens for the treatment of solid tumors.
Collapse
Affiliation(s)
- Luca Sigalotti
- Cancer Bioimmunotherapy Unit, Centro di Riferimento Oncologico Aviano, National Cancer Institute, Aviano, Italy
| | - Elisabetta Fratta
- Cancer Bioimmunotherapy Unit, Centro di Riferimento Oncologico Aviano, National Cancer Institute, Aviano, Italy
| | - Sandra Coral
- Medical Oncology and Immunotherapy, Department of Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Michele Maio
- Medical Oncology and Immunotherapy, Department of Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy.
| |
Collapse
|
23
|
The narrow-spectrum HDAC inhibitor entinostat enhances NKG2D expression without NK cell toxicity, leading to enhanced recognition of cancer cells. Pharm Res 2013; 32:779-92. [PMID: 24203492 DOI: 10.1007/s11095-013-1231-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 10/14/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE Natural killer (NK) cell cytotoxicity correlates with the ligation of activating receptors (e.g., NKG2D) by their ligands (e.g., MHC class I-related chains [MIC] A and B) on target cells. Histone deacetylase inhibitors (HDACi) at high concentrations inhibit tumor growth and can increase NKG2D ligand expression on tumor targets, but are widely regarded as toxic to NK cells. METHODS We investigated the mechanism of entinostat, a benzamide-derivative narrow-spectrum HDACi, in augmenting the cytotoxicity of NK cells against human colon carcinoma and sarcoma by assessing gene and protein expression, histone acetylation, and cytotoxicity in in vitro and murine models. RESULTS We observed that entinostat dose- and time-dependent increase in MIC expression in tumor targets and NKG2D in primary human NK cells, both correlating with increased acetylated histone 3 (AcH3) binding to associated promoters. Entinostat pretreatment of colon carcinoma and sarcoma cells, NK cells, or both led to enhanced overall cytotoxicity in vitro, which was reversed by NKG2D blockade, and inhibited growth of tumor xenografts. Lastly, we showed decreased expression of MICA and ULBP2 transcription in primary human osteosarcoma. CONCLUSIONS Entinostat enhances NK cell killing of cancer cells through upregulation of both NKG2D and its ligands, suggesting an attractive approach for augmenting NK cell immunotherapy of solid tumors such as colon carcinoma and sarcomas.
Collapse
|
24
|
Rao-Bindal K, Rao CK, Yu L, Kleinerman ES. Expression of c-FLIP in pulmonary metastases in osteosarcoma patients and human xenografts. Pediatr Blood Cancer 2013; 60:575-9. [PMID: 23255321 PMCID: PMC3883385 DOI: 10.1002/pbc.24412] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 11/01/2012] [Indexed: 01/09/2023]
Abstract
OBJECTIVE(S) We have previously shown that Fas expression inversely correlates with the metastatic potential of osteosarcoma (OS) to the lung. FasL is constitutively expressed in the lung microenvironment and eliminates Fas(+) OS cells leaving Fas(-) cells to form metastases. Absence of FasL in the lung epithelium or blocking the Fas-signaling pathway interfered with this clearance mechanism allowing Fas(+) cells to remain and form lung metastases. We also demonstrated that while the majority of patient OS lung metastases were Fas(-), 10-20% of the lesions contained Fas(+) cells, suggesting that these cells were not sensitive to FasL-induced apoptosis. The expression of c-FLIP, an inhibitor of the Fas pathway, has been associated with tumor development, progression, and resistance to chemotherapy. We therefore evaluated the expression of c-FLIP in OS patient tumor specimens and human xenograft lung metastases. METHODS OS patient tissues, which included both primary and metastatic lesions, were evaluated for the expression of c-FLIP. In addition, tumors from human OS xenografts were examined for c-FLIP expression. RESULTS c-FLIP expression was significantly higher in the lung metastases than in the primary tumors. CONCLUSION(S) c-FLIP may play an important role in the metastatic potential of OS to the lung. Inhibition of c-FLIP may be a future therapeutic target.
Collapse
Affiliation(s)
- Krithi Rao-Bindal
- Division of Pediatrics, The University of Texas, MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|