1
|
Zheng Y, Yang Y, Zhu W, Liu R, Liu A, Zhang R, Lei W, Huang S, Liu Y, Hu Q. GSK3B inhibition reduced cervical cancer cell proliferation and migration by modulating the PI3K/Akt signaling pathway and epithelial-to-mesenchymal transition. Braz J Med Biol Res 2024; 57:e13796. [PMID: 39166606 PMCID: PMC11338547 DOI: 10.1590/1414-431x2024e13796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024] Open
Abstract
Previous studies show that glycogen synthase kinase 3β (GSK3B) plays an important role in tumorigenesis. However, its role in cervical cancer is unclear. The present study silenced GSK3B with siRNAs and/or chemical inhibitors to determine its role in HeLa cervical cancer cell proliferation and migration as well as in xenograft tumor growth. Cell Counting Kit (CCK)-8 and 5-ethynyl-2'-deoxyuridine (EdU) assays were used to determine cell survival and proliferation. Scratch and Transwell® assays were used to evaluate cell migration. Xenograft tumors were used to evaluate the effect of GSK3B on tumor growth. Transcriptomic sequencing was used to clarify the mechanisms underlying the foregoing processes. Public databases and clinical specimens showed that GSK3B was upregulated in cervical cancer tissues and correlated with poor prognosis. In vitro experiments indicated that GSK3B inhibition reduced cell viability, proliferation, and migration. In vivo experiments demonstrated that GSK3B inhibition slowed xenograft tumor growth. Transcriptomic sequencing revealed that GSK3B inhibition modulated the phosphatidylinositol 3-carboxykinase (PI3K)/protein kinase B (Akt) and extracellular matrix (ECM)-receptor interaction signaling pathways. GSK3B inhibition decreased the protein levels of phosphorylated PI3K and Akt and the levels of mesenchymal markers but increased those of epithelial markers. An activator of the PI3K/Akt signaling pathway counteracted the suppressive effects of GSK3B inhibition on HeLa cell viability and proliferation and on PI3K/Akt signaling. Our data suggested that GSK3B regulated cervical cancer cell proliferation and migration by modulating the PI3K/Akt signaling pathway and epithelial-to-mesenchymal transition (EMT).
Collapse
Affiliation(s)
- Yanhong Zheng
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yang Yang
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Weiyan Zhu
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Ruhao Liu
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Aodong Liu
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Runfeng Zhang
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Weixing Lei
- Luoyuan Center for Disease Control and Prevention, Fuzhou, China
| | - Shifeng Huang
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yongzhu Liu
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Qinglan Hu
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
| |
Collapse
|
2
|
Wani AK, Singh R, Akhtar N, Prakash A, Nepovimova E, Oleksak P, Chrienova Z, Alomar S, Chopra C, Kuca K. Targeted Inhibition of the PI3K/Akt/mTOR Signaling Axis: Potential for Sarcoma Therapy. Mini Rev Med Chem 2024; 24:1496-1520. [PMID: 38265369 DOI: 10.2174/0113895575270904231129062137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 01/25/2024]
Abstract
Sarcoma is a heterogeneous group of malignancies often resistant to conventional chemotherapy and radiation therapy. The phosphatidylinositol-3-kinase/ protein kinase B /mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway has emerged as a critical cancer target due to its central role in regulating key cellular processes such as cell growth, proliferation, survival, and metabolism. Dysregulation of this pathway has been implicated in the development and progression of bone sarcomas (BS) and soft tissue sarcomas (STS). PI3K/Akt/mTOR inhibitors have shown promising preclinical and clinical activity in various cancers. These agents can inhibit the activation of PI3K, Akt, and mTOR, thereby reducing the downstream signaling events that promote tumor growth and survival. In addition, PI3K/Akt/mTOR inhibitors have been shown to enhance the efficacy of other anticancer therapies, such as chemotherapy and radiation therapy. The different types of PI3K/Akt/mTOR inhibitors vary in their specificity, potency, and side effect profiles and may be effective depending on the specific sarcoma type and stage. The molecular targeting of PI3K/Akt/mToR pathway using drugs, phytochemicals, nanomaterials (NMs), and microbe-derived molecules as Pan-PI3K inhibitors, selective PI3K inhibitors, and dual PI3K/mTOR inhibitors have been delineated. While there are still challenges to be addressed, the preclinical and clinical evidence suggests that these inhibitors may significantly improve patient outcomes. Further research is needed to understand the potential of these inhibitors as sarcoma therapeutics and to continue developing more selective and effective agents to meet the clinical needs of sarcoma patients.
Collapse
Affiliation(s)
- Atif Khurshid Wani
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Reena Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Nahid Akhtar
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Ajit Prakash
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Patrik Oleksak
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Zofia Chrienova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Suliman Alomar
- King Saud University, Zoology Department, College of Science, Riyadh, 11451, Saudi Arabia
| | - Chirag Chopra
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Králové, Czechia
| |
Collapse
|
3
|
Cyra M, Schulte M, Berthold R, Heinst L, Jansen EP, Grünewald I, Elges S, Larsson O, Schliemann C, Steinestel K, Hafner S, Simmet T, Wardelmann E, Kailayangiri S, Rossig C, Isfort I, Trautmann M, Hartmann W. SS18-SSX drives CREB activation in synovial sarcoma. Cell Oncol (Dordr) 2022; 45:399-413. [PMID: 35556229 PMCID: PMC9187574 DOI: 10.1007/s13402-022-00673-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2022] [Indexed: 11/28/2022] Open
Abstract
Purpose Synovial sarcoma (SySa) is a rare soft tissue tumor characterized by a reciprocal t(X;18) translocation. The chimeric SS18-SSX fusion protein represents the major driver of the disease, acting as aberrant transcriptional dysregulator. Oncogenic mechanisms whereby SS18-SSX mediates sarcomagenesis are incompletely understood, and strategies to selectively target SySa cells remain elusive. Based on results of Phospho-Kinase screening arrays, we here investigate the functional and therapeutic relevance of the transcription factor CREB in SySa tumorigenesis. Methods Immunohistochemistry of phosphorylated CREB and its downstream targets (Rb, Cyclin D1, PCNA, Bcl-xL and Bcl-2) was performed in a large cohort of SySa. Functional aspects of CREB activity, including SS18-SSX driven circuits involved in CREB activation, were analyzed in vitro employing five SySa cell lines and a mesenchymal stem cell model. CREB mediated transcriptional activity was modulated by RNAi-mediated knockdown and small molecule inhibitors (666-15, KG-501, NASTRp and Ro 31-8220). Anti-proliferative effects of the CREB inhibitor 666-15 were tested in SySa avian chorioallantoic membrane and murine xenograft models in vivo. Results We show that CREB is phosphorylated and activated in SySa, accompanied by downstream target expression. Human mesenchymal stem cells engineered to express SS18-SSX promote CREB expression and phosphorylation. Conversely, RNAi-mediated knockdown of SS18-SSX impairs CREB phosphorylation in SySa cells. Inhibition of CREB activity reduces downstream target expression, accompanied by suppression of SySa cell proliferation and induction of apoptosis invitro and in vivo. Conclusion In conclusion, our data underline an essential role of CREB in SySa tumorigenesis and provides evidence for molecular targeted therapies. Supplementary Information The online version contains supplementary material available at 10.1007/s13402-022-00673-w.
Collapse
Affiliation(s)
- Magdalene Cyra
- Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.,Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Miriam Schulte
- Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.,Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Ruth Berthold
- Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.,Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Lorena Heinst
- Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.,Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Esther-Pia Jansen
- Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.,Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Inga Grünewald
- Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.,Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Sandra Elges
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Olle Larsson
- Departments of Oncology and Pathology, The Karolinska Institute, Stockholm, Sweden
| | - Christoph Schliemann
- Department of Medicine A, Hematology, Oncology and Respiratory Medicine, Münster University Hospital, Münster, Germany
| | - Konrad Steinestel
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.,Institute of Pathology and Molecular Pathology, Bundeswehrkrankenhaus Ulm, Ulm, Germany
| | - Susanne Hafner
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Eva Wardelmann
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Sareetha Kailayangiri
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Claudia Rossig
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Ilka Isfort
- Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.,Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Marcel Trautmann
- Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany. .,Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.
| | - Wolfgang Hartmann
- Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany. .,Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.
| |
Collapse
|
4
|
Lanzi C, Cassinelli G. Combinatorial strategies to potentiate the efficacy of HDAC inhibitors in fusion-positive sarcomas. Biochem Pharmacol 2022; 198:114944. [DOI: 10.1016/j.bcp.2022.114944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/02/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
|
5
|
Feng X, Huang YL, Zhang Z, Wang N, Yao Q, Pang LJ, Li F, Qi Y. The role of SYT-SSX fusion gene in tumorigenesis of synovial sarcoma. Pathol Res Pract 2021; 222:153416. [PMID: 33848939 DOI: 10.1016/j.prp.2021.153416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/09/2021] [Accepted: 03/20/2021] [Indexed: 12/29/2022]
Abstract
Synovial sarcoma (SS) is an aggressive malignancy of an unknown tissue origin that is characterized by biphasic differentiation. A possible basis of the pathogenesis of SS is pathognomonic t(X;18) (p11.2; q11.2) translocation, leading to the formation and expression of the SYT-SSX fusion gene. More than a quarter of the patients die of SS metastasis within 5 years after the diagnosis, but the pathogenic factors are unknown. Therefore, there is an urgent need to explore the pathogenesis, invasion, metastasis, and clinical treatment options for SS, especially molecular-targeted drug therapy. Recent studies have shown that the SYT-SSX fusion gene associated with SS may be regulated by different signaling pathways, microRNAs, and other molecules, which may produce stem cell characteristics or promote epithelial-mesenchymal transition, resulting in SS invasion and metastasis. This review article aims to show the relationship between the SYT-SSX fusion gene and the related pathway molecules as well as other molecules involved from different perspectives, which may provide a deeper and clearer understanding of the SYT-SSX fusion gene function. Therefore, this review may provide a more innovative and broader perspective of the current research, treatment options, and prognosis assessment of SS.
Collapse
Affiliation(s)
- Xiao Feng
- Department of Pathology, Shihezi University School of Medicine & the First Affiliated Hospital to Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Ya-Lan Huang
- Department of Pathology, Shihezi University School of Medicine & the First Affiliated Hospital to Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China; Department of Pathology Suining Central Hospital, Suining, Sichuan, China
| | - Zhen Zhang
- Department of Pathology, Shihezi University School of Medicine & the First Affiliated Hospital to Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Ning Wang
- Department of Pathology, Shihezi University School of Medicine & the First Affiliated Hospital to Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Qing Yao
- Department of Pathology, Shihezi University School of Medicine & the First Affiliated Hospital to Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Li-Juan Pang
- Department of Pathology, Shihezi University School of Medicine & the First Affiliated Hospital to Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China
| | - Feng Li
- Department of Pathology, Shihezi University School of Medicine & the First Affiliated Hospital to Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China; Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| | - Yan Qi
- Department of Pathology, Shihezi University School of Medicine & the First Affiliated Hospital to Shihezi University School of Medicine, Shihezi, 832002, Xinjiang, China; Department of Pathology, Central People's Hospital of Zhanjiang and Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
6
|
Synovial Sarcoma: A Complex Disease with Multifaceted Signaling and Epigenetic Landscapes. Curr Oncol Rep 2020; 22:124. [PMID: 33025259 DOI: 10.1007/s11912-020-00985-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2020] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW Aside from a characteristic SS18-SSX translocation identified in almost all cases, no genetic anomalies have been reliably isolated yet to drive the pathogenesis of synovial sarcoma. In the following review, we explore the structural units of wild-type SS18 and SSX, particularly as they relate to the transcriptional alterations and cellular pathway changes imposed by SS18-SSX. RECENT FINDINGS Native SS18 and SSX contribute recognizable domains to the SS18-SSX chimeric proteins, which inflict transcriptional and epigenetic changes through selective protein interactions involving the SWI/SNF and Polycomb chromatin remodeling complexes. Multiple oncogenic and developmental pathways become altered, collectively reprogramming the cellular origin of synovial sarcoma and promoting its malignant transformation. Synovial sarcoma is characterized by complex epigenetic and signaling landscapes. Identifying the operational pathways and concomitant genetic changes induced by SS18-SSX fusions could help develop tailored therapeutic strategies to ultimately improve disease control and patient survivorship.
Collapse
|
7
|
Zhang GY, Ma ZJ, Wang L, Sun RF, Jiang XY, Yang XJ, Long B, Ye HL, Zhang SZ, Yu ZY, Shi WG, Jiao ZY. The Role of Shcbp1 in Signaling and Disease. Curr Cancer Drug Targets 2020; 19:854-862. [PMID: 31250756 DOI: 10.2174/1568009619666190620114928] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/19/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022]
Abstract
Src homolog and collagen homolog (Shc) proteins have been identified as adapter proteins associated with cell surface receptors and have been shown to play important roles in signaling and disease. Shcbp1 acts as a Shc SH2-domain binding protein 1 and is involved in the regulation of signaling pathways, such as FGF, NF-κB, MAPK/ERK, PI3K/AKT, TGF-β1/Smad and β -catenin signaling. Shcbp1 participates in T cell development, the regulation of downstream signal transduction pathways, and cytokinesis during mitosis and meiosis. In addition, Shcbp1 has been demonstrated to correlate with Burkitt-like lymphoma, breast cancer, lung cancer, gliomas, synovial sarcoma, human hepatocellular carcinoma and other diseases. Shcbp1 may play an important role in tumorigenesis and progression. Accordingly, recent studies are reviewed herein to discuss and interpret the role of Shcbp1 in normal cell proliferation and differentiation, tumorigenesis and progression, as well as its interactions with proteins.
Collapse
Affiliation(s)
- Geng-Yuan Zhang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhi-Jian Ma
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Long Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Ruo-Fei Sun
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | | | - Xu-Juan Yang
- Lanzhou University Second Hospital, Lanzhou, China
| | - Bo Long
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Hui-Li Ye
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Shu-Ze Zhang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Ze-Yuan Yu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Wen-Gui Shi
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Zuo-Yi Jiao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
8
|
Li YX, Ding SS, Wen WJ, Han L, Wang HQ, Shi HY. Impact of the Activation Status of the Akt/mTOR Signalling Pathway on the Clinical Behaviour of Synovial Sarcoma: Retrospective Analysis of 174 Patients at a Single Institution. Cancer Manag Res 2020; 12:1759-1769. [PMID: 32210617 PMCID: PMC7074818 DOI: 10.2147/cmar.s228578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 02/08/2020] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Phosphoinositide 3-kinase (PI3K) and the downstream Akt/mammalian target of rapamycin (mTOR) pathway are central to the control of cell proliferation and survival. Although abnormal activation of this pathway has been well established in a variety of tumours, limited studies are available on synovial sarcoma. The aim of this study was to investigate the expression of several key proteins of those pathways in synovial sarcomas and to correlate the expression of these proteins with clinicopathologic features and prognosis. PATIENTS AND METHODS A total of 174 patients with synovial sarcomas were recruited for this study. The phosphorylation status of Akt, mTOR, and eukaryotic translation initiation factor 4E binding protein (4E-BP1) was measured by immunohistochemistry assays in formalin-fixed, paraffin-embedded samples. Correlations between the expression levels of these proteins and clinicopathologic features and prognosis were analysed. RESULTS The positive rates of phosphorylated (p)Akt, pmTOR, p4E-BP1, and CyclinD1 were 62.7%, 55.6%, 47.1%, and 52.6%, respectively. The positive results of pmTOR, pAkt, and downstream p4E-BP1 were correlated with each other. The positive pAkt, pmTOR, p4E-BP1, and CyclinD1 results were more highly expressed in head and neck and visceral tumours, and positive p4E-BP1 results were correlated with larger size and larger areas of necrosis. In multivariate analysis of clinicopathologic factors, head and neck and visceral location, large tumour size, larger areas of necrosis and frequent mitosis were confirmed as risk factors for shorter overall survival. Positive pAkt, pmTOR and p4E-BP1 results were correlated significantly with shorter overall survival, and CyclinD1 was not in the univariate analysis. The positive pmTOR, pAkt, p4E-BP1, and CyclinD1 results were significantly poor prognostic factors for overall survival, and only positive p4E-BP1 results were significantly associated with shorter event-free survival in multivariate analysis. CONCLUSION This study demonstrated the high expression of pAkt, pmTOR, and p4E-BP1 associated with aggressive clinical behaviour in synovial sarcomas and provided evidence for prognostic evaluation and targeted therapy.
Collapse
Affiliation(s)
- Ying-Xue Li
- Department of Pathology, Medical School of Chinese People's Liberation Army, Beijing100853, People’s Republic of China
- Department of Pathology, Liaocheng People’s Hospital, Liaocheng252000, Shandong, People’s Republic of China
| | - Shan-Shan Ding
- Department of Pathology, PLA Rocket Force Characteristic Medical Center, Beijing100032, People’s Republic of China
| | - Wen-Juan Wen
- Department of Pathology, Liaocheng People’s Hospital, Liaocheng252000, Shandong, People’s Republic of China
| | - Lin Han
- Department of Pathology, Liaocheng People’s Hospital, Liaocheng252000, Shandong, People’s Republic of China
| | - Hong-Qun Wang
- Department of Pathology, Medical School of Chinese People's Liberation Army, Beijing100853, People’s Republic of China
| | - Huai-Yin Shi
- Department of Pathology, Medical School of Chinese People's Liberation Army, Beijing100853, People’s Republic of China
| |
Collapse
|
9
|
The PTEN Tumor Suppressor Gene in Soft Tissue Sarcoma. Cancers (Basel) 2019; 11:cancers11081169. [PMID: 31416195 PMCID: PMC6721622 DOI: 10.3390/cancers11081169] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/26/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023] Open
Abstract
Soft tissue sarcoma (STS) is a rare malignancy of mesenchymal origin classified into more than 50 different subtypes with distinct clinical and pathologic features. Despite the poor prognosis in the majority of patients, only modest improvements in treatment strategies have been achieved, largely due to the rarity and heterogeneity of these tumors. Therefore, the discovery of new prognostic and predictive biomarkers, together with new therapeutic targets, is of enormous interest. Phosphatase and tensin homolog (PTEN) is a well-known tumor suppressor that commonly loses its function via mutation, deletion, transcriptional silencing, or protein instability, and is frequently downregulated in distinct sarcoma subtypes. The loss of PTEN function has consequent alterations in important pathways implicated in cell proliferation, survival, migration, and genomic stability. PTEN can also interact with other tumor suppressors and oncogenic signaling pathways that have important implications for the pathogenesis in certain STSs. The aim of the present review is to summarize the biological significance of PTEN in STS and its potential role in the development of new therapeutic strategies.
Collapse
|
10
|
Lanzi C, Dal Bo L, Favini E, Tortoreto M, Beretta GL, Arrighetti N, Zaffaroni N, Cassinelli G. Overactive IGF1/Insulin Receptors and NRASQ61R Mutation Drive Mechanisms of Resistance to Pazopanib and Define Rational Combination Strategies to Treat Synovial Sarcoma. Cancers (Basel) 2019; 11:cancers11030408. [PMID: 30909453 PMCID: PMC6468361 DOI: 10.3390/cancers11030408] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/08/2019] [Accepted: 03/18/2019] [Indexed: 12/15/2022] Open
Abstract
Pazopanib is approved for treatment of advanced soft tissue sarcomas, but primary and secondary drug resistance limits its clinical utility. We investigated the molecular mechanisms mediating pazopanib resistance in human synovial sarcoma (SS) models. We found reduced cell sensitivity to pazopanib associated with inefficient inhibition of the two critical signaling nodes, AKT and ERKs, despite strong inhibition of the main drug target, PDGFRα. In the CME-1 cell line, overactivation of IGF1 and Insulin receptors (IGF1R/InsR) sustained AKT activation and pazopanib resistance, which was overcome by a combination treatment with the double IGF1R/InsR inhibitor BMS754807. In the highly pazopanib resistant MoJo cell line, NRASQ61R mutation sustained constitutive ERK activation. Transfection of the NRAS mutant in the pazopanib sensitive SYO-1 cell line increased the drug IC50. MoJo cells treatment with pazopanib in combination with the MEK inhibitor trametinib restored ERK inhibition, synergistically inhibited cell growth, and induced apoptosis. The combination significantly enhanced the antitumor efficacy against MoJo orthotopic xenograft abrogating growth in 38% of mice. These findings identified two different mechanisms of intrinsic pazopanib resistance in SS cells, supporting molecular/immunohistochemical profiling of tumor specimens as a valuable approach to selecting patients who may benefit from rational drug combinations.
Collapse
Affiliation(s)
- Cinzia Lanzi
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| | - Laura Dal Bo
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| | - Enrica Favini
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| | - Monica Tortoreto
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| | - Giovanni Luca Beretta
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| | - Noemi Arrighetti
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| | - Nadia Zaffaroni
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| | - Giuliana Cassinelli
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| |
Collapse
|
11
|
Isfort I, Cyra M, Elges S, Kailayangiri S, Altvater B, Rossig C, Steinestel K, Grünewald I, Huss S, Eßeling E, Mikesch JH, Hafner S, Simmet T, Wozniak A, Schöffski P, Larsson O, Wardelmann E, Trautmann M, Hartmann W. SS18-SSX–Dependent YAP/TAZ Signaling in Synovial Sarcoma. Clin Cancer Res 2019; 25:3718-3731. [DOI: 10.1158/1078-0432.ccr-17-3553] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/02/2018] [Accepted: 02/21/2019] [Indexed: 11/16/2022]
|
12
|
Trautmann M, Cyra M, Isfort I, Jeiler B, Krüger A, Grünewald I, Steinestel K, Altvater B, Rossig C, Hafner S, Simmet T, Becker J, Åman P, Wardelmann E, Huss S, Hartmann W. Phosphatidylinositol-3-kinase (PI3K)/Akt Signaling is Functionally Essential in Myxoid Liposarcoma. Mol Cancer Ther 2019; 18:834-844. [PMID: 30787173 DOI: 10.1158/1535-7163.mct-18-0763] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/13/2018] [Accepted: 01/28/2019] [Indexed: 11/16/2022]
Abstract
Myxoid liposarcoma (MLS) is an aggressive soft-tissue tumor characterized by a specific reciprocal t(12;16) translocation resulting in expression of the chimeric FUS-DDIT3 fusion protein, an oncogenic transcription factor. Similar to other translocation-associated sarcomas, MLS is characterized by a low frequency of somatic mutations, albeit a subset of MLS has previously been shown to be associated with activating PIK3CA mutations. This study was performed to assess the prevalence of PI3K/Akt signaling alterations in MLS and the potential of PI3K-directed therapeutic concepts. In a large cohort of MLS, key components of the PI3K/Akt signaling cascade were evaluated by next generation seqeuncing (NGS), fluorescence in situ hybridization (FISH), and immunohistochemistry (IHC). In three MLS cell lines, PI3K activity was inhibited by RNAi and the small-molecule PI3K inhibitor BKM120 (buparlisib) in vitro An MLS cell line-based avian chorioallantoic membrane model was applied for in vivo confirmation. In total, 26.8% of MLS cases displayed activating alterations in PI3K/Akt signaling components, with PIK3CA gain-of-function mutations representing the most prevalent finding (14.2%). IHC suggested PI3K/Akt activation in a far larger subgroup of MLS, implying alternative mechanisms of pathway activation. PI3K-directed therapeutic interference showed that MLS cell proliferation and viability significantly depended on PI3K-mediated signals in vitro and in vivo Our preclinical study underlines the elementary role of PI3K/Akt signals in MLS tumorigenesis and provides a molecularly based rationale for a PI3K-targeted therapeutic approach which may be particularly effective in the subgroup of tumors carrying activating genetic alterations in PI3K/Akt signaling components.
Collapse
Affiliation(s)
- Marcel Trautmann
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany. .,Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Magdalene Cyra
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.,Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Ilka Isfort
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.,Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Birte Jeiler
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.,Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Arne Krüger
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.,Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Inga Grünewald
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.,Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Konrad Steinestel
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany.,Institute of Pathology and Molecular Pathology, Bundeswehrkrankenhaus Ulm, Ulm, Germany
| | - Bianca Altvater
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Claudia Rossig
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany.,Cells in Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Susanne Hafner
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Jessica Becker
- Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Pierre Åman
- Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Eva Wardelmann
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Sebastian Huss
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Wolfgang Hartmann
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany. .,Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| |
Collapse
|
13
|
Pollock RE, Payne JE, Rogers AD, Smith SM, Iwenofu OH, Valerio IL, Zomerlei TA, Howard JH, Dornbos D, Galgano MA, Goulart C, Mendel E, Miller ED, Xu-Welliver M, Martin DD, Haglund KE, Bupathi M, Chen JL, Yeager ND. Multidisciplinary sarcoma care. Curr Probl Surg 2018; 55:517-580. [PMID: 30526918 DOI: 10.1067/j.cpsurg.2018.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Raphael E Pollock
- The Ohio State University Comprehensive Cancer Center, Columbus, OH.
| | - Jason E Payne
- The Ohio State University Wexner Medical Center, Columbus, OH
| | - Alan D Rogers
- The Ohio State University Wexner Medical Center, Columbus, OH
| | - Stephen M Smith
- The Ohio State University Wexner Medical Center, Columbus, OH
| | - O Hans Iwenofu
- Department of Pathology & Laboratory Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Ian L Valerio
- The Ohio State University Wexner Medical Center, Columbus, OH
| | | | | | - David Dornbos
- The Ohio State University Wexner Medical Center, Columbus, OH
| | | | | | - Ehud Mendel
- The Ohio State University Wexner Medical Center, Columbus, OH
| | - Eric D Miller
- The Ohio State University Wexner Medical Center, Columbus, OH
| | | | | | - Karl E Haglund
- The Ohio State University Wexner Medical Center, Columbus, OH
| | | | - James L Chen
- The Ohio State University Wexner Medical Center, Columbus, OH
| | - Nicholas D Yeager
- Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH
| |
Collapse
|
14
|
Namatame N, Tamaki N, Yoshizawa Y, Okamura M, Nishimura Y, Yamazaki K, Tanaka M, Nakamura T, Semba K, Yamori T, Yaguchi SI, Dan S. Antitumor profile of the PI3K inhibitor ZSTK474 in human sarcoma cell lines. Oncotarget 2018; 9:35141-35161. [PMID: 30416685 PMCID: PMC6205545 DOI: 10.18632/oncotarget.26216] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 09/26/2018] [Indexed: 11/25/2022] Open
Abstract
Treatment of patients with advanced sarcoma remains challenging due to lack of effective medicine, with the development of novel drugs being of keen interest. A pan-PI3K inhibitor, ZSTK474, has been evaluated in clinical trials against a range of advanced solid tumors, with clinical benefit shown in sarcoma patients. In the present study, we developed a panel of 14 human sarcoma cell lines and investigated the antitumor effect of 24 anticancer agents including ZSTK474, other PI3K inhibitors, and those clinically used for sarcoma treatment. ZSTK474 exhibited a similar antiproliferative profile to other PI3K inhibitors but was clearly different from the other drugs examined. Indeed, ZSTK474 inhibited PI3K-downstream pathways, in parallel to growth inhibition, in all cell lines examined, showing proof-of-concept of PI3K inhibition. In addition, ZSTK474 induced apoptosis selectively in Ewing's sarcoma (RD-ES and A673), alveolar rhabdomyosarcoma (SJCRH30) and synovial sarcoma (SYO-1, Aska-SS and Yamato-SS) cell lines, all of which harbor chromosomal translocation and resulting oncogenic fusion genes, EWSR1-FLI1, PAX3-FOXO1 and SS18-SSX, respectively. Finally, animal experiments confirmed the antitumor activity of ZSTK474 in vivo, with superior efficacy observed in translocation-positive cells. These results suggest that ZSTK474 could be a promising drug candidate for treating sarcomas, especially those harboring chromosomal translocation.
Collapse
Affiliation(s)
- Nachi Namatame
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.,R&D Center, Zenyaku Kogyo Co. Ltd, Tokyo, Japan
| | - Naomi Tamaki
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yuya Yoshizawa
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Mutsumi Okamura
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yumiko Nishimura
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kanami Yamazaki
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Miwa Tanaka
- Division of Carcinogenesis, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takuro Nakamura
- Division of Carcinogenesis, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kentaro Semba
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Takao Yamori
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.,Present address: Center for Product Evaluation, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Shin-Ichi Yaguchi
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.,R&D Center, Zenyaku Kogyo Co. Ltd, Tokyo, Japan
| | - Shingo Dan
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
15
|
Peng C, Zhao H, Chen W, Song Y, Wang X, Li J, Qiao Y, Wu D, Ma S, Wang X, Gao C. Identification of SHCBP1 as a novel downstream target gene of SS18-SSX1 and its functional analysis in progression of synovial sarcoma. Oncotarget 2018; 7:66822-66834. [PMID: 27572315 PMCID: PMC5341840 DOI: 10.18632/oncotarget.11651] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 08/21/2016] [Indexed: 01/10/2023] Open
Abstract
The SS18-SSX1 fusion gene has been shown to play important roles in the development of synovial sarcoma (SS), but the underlying molecular mechanisms and its downstream target genes are still not clear. Here SHC SH2-domain binding protein 1 (SHCBP1) was identified and validated to be a novel downstream target gene of SS18-SSX1 by using microarray assay, quantitative real-time (qPCR) and western blot. Expression of SHCBP1 was firstly confirmed in SS cell line and SS tissues. The effects of SHCBP1 overexpression or knockdown on SS cell proliferation and tumorigenicity were then studied by cell proliferation, DNA replication, colony formation, flow cytometric assays, and its in vivo tumorigenesis was determined in the nude mice. Meanwhile, the related signaling pathways of SHCBP1 were also examined in SS cells. The results indicated that SHCBP1 was significantly increased in SS cells and SS tissues compared with adjacent noncancerous tissues. The expression of SHCBP1 was demonstrated to be positively correlated with the SS18-SSX1 level. Overexpression and ablation of SHCBP1 promoted and inhibited, respectively, the proliferation and tumorigenicity of SS cells in vitro. SHCBP1 knockdown also significantly inhibited SS cell growth in nude mice, and lowered the MAPK/ERK and PI3K/AKT/mTOR signaling pathways and cyclin D1 expression. Our findings disclose that SHCBP1 is a novel downstream target gene of SS18-SSX1, and demonstrate that the oncogene SS18-SSX1 promotes tumorigenesis by increasing the expression of SHCBP1, which normally acts as a tumor promoting factor.
Collapse
Affiliation(s)
- Changliang Peng
- Department of Orthopaedics, Shandong University Second Hospital, Jinan, China
| | - Hui Zhao
- Department of Orthopaedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Wei Chen
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yan Song
- Nephrology Research Institute, Shandong University Second Hospital, Jinan, China
| | - Xiaoying Wang
- Department of Pathology, Shandong University Second Hospital, Jinan, China
| | - Ji Li
- Department of Orthopaedics, Shandong University Second Hospital, Jinan, China
| | - Yong Qiao
- Department of Orthopaedics, Shandong University Second Hospital, Jinan, China
| | - Dongjin Wu
- Department of Orthopaedics, Shandong University Second Hospital, Jinan, China
| | - Shengzhong Ma
- Department of Orthopaedics, Shandong University Second Hospital, Jinan, China
| | - Xiuwen Wang
- Department of Orthopaedics, Shandong University Second Hospital, Jinan, China
| | - Chunzheng Gao
- Department of Orthopaedics, Shandong University Second Hospital, Jinan, China
| |
Collapse
|
16
|
Cassinelli G, Dal Bo L, Favini E, Cominetti D, Pozzi S, Tortoreto M, De Cesare M, Lecis D, Scanziani E, Minoli L, Naggi A, Vlodavsky I, Zaffaroni N, Lanzi C. Supersulfated low-molecular weight heparin synergizes with IGF1R/IR inhibitor to suppress synovial sarcoma growth and metastases. Cancer Lett 2017; 415:187-197. [PMID: 29225052 DOI: 10.1016/j.canlet.2017.12.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 01/26/2023]
Abstract
Synovial sarcoma (SS) is an aggressive tumor with propensity for lung metastases which significantly impact patients' prognosis. New therapeutic approaches are needed to improve treatment outcome. Targeting the heparanase/heparan sulfate proteoglycan system by heparin derivatives which act as heparanase inhibitors/heparan sulfate mimetics is emerging as a therapeutic approach that can sensitize the tumor response to chemotherapy. We investigated the therapeutic potential of a supersulfated low molecular weight heparin (ssLMWH) in preclinical models of SS. ssLMWH showed a potent anti-heparanase activity, dose-dependently inhibited SS colony growth and cell invasion, and downregulated the activation of receptor tyrosine kinases including IGF1R and IR. The combination of ssLMWH and the IGF1R/IR inhibitor BMS754807 synergistically inhibited proliferation of cells exhibiting IGF1R hyperactivation, also abrogating cell motility and promoting apoptosis in association with PI3K/AKT pathway inhibition. The drug combination strongly enhanced the antitumor effect against the CME-1 model, as compared to single agent treatment, abrogating orthotopic tumor growth and significantly repressing spontaneous lung metastatic dissemination in treated mice. These findings provide a strong preclinical rationale for developing drug regimens combining heparanase inhibitors/HS mimetics with IGF1R antagonists for treatment of metastatic SS.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Apoptosis/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Drug Synergism
- Glucuronidase/antagonists & inhibitors
- Glucuronidase/metabolism
- Heparin, Low-Molecular-Weight/administration & dosage
- Heparin, Low-Molecular-Weight/metabolism
- Heparin, Low-Molecular-Weight/pharmacology
- Humans
- Mice, SCID
- Neoplasm Metastasis
- Pyrazoles/administration & dosage
- Pyrazoles/pharmacology
- Receptor, IGF Type 1
- Receptors, Somatomedin/antagonists & inhibitors
- Receptors, Somatomedin/metabolism
- Sarcoma, Synovial/drug therapy
- Sarcoma, Synovial/metabolism
- Sarcoma, Synovial/pathology
- Sulfates
- Triazines/administration & dosage
- Triazines/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Giuliana Cassinelli
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| | - Laura Dal Bo
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Enrica Favini
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Denis Cominetti
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Sabina Pozzi
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Monica Tortoreto
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Michelandrea De Cesare
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Daniele Lecis
- Research Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Eugenio Scanziani
- Department of Veterinary Medicine, Università Degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; Mouse and Animal Pathology Laboratory, Fondazione Filarete, Viale Ortles 22/4, 20139 Milan, Italy
| | - Lucia Minoli
- Department of Veterinary Medicine, Università Degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; Mouse and Animal Pathology Laboratory, Fondazione Filarete, Viale Ortles 22/4, 20139 Milan, Italy
| | - Annamaria Naggi
- G. Ronzoni Institute for Chemical and Biochemical Research, Via G. Colombo 81, 20133 Milan, Italy
| | - Israel Vlodavsky
- Cancer and Vascular Biology Research Center, Bruce Rappaport Faculty of Medicine, Technion, P.O. Box 9649, Haifa 31096, Israel
| | - Nadia Zaffaroni
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Cinzia Lanzi
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| |
Collapse
|
17
|
Martinez-Font E, Felipe-Abrio I, Calabuig-Fariñas S, Ramos R, Terrasa J, Vögler O, Alemany R, Martín-Broto J, Obrador-Hevia A. Disruption of TCF/β-Catenin Binding Impairs Wnt Signaling and Induces Apoptosis in Soft Tissue Sarcoma Cells. Mol Cancer Ther 2017; 16:1166-1176. [DOI: 10.1158/1535-7163.mct-16-0585] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/13/2016] [Accepted: 03/02/2017] [Indexed: 12/29/2022]
Abstract
Abstract
Soft tissue sarcomas (STS) are malignant tumors of mesenchymal origin and represent around 1% of adult cancers, being a very heterogeneous group of tumors with more than 50 different subtypes. The Wnt signaling pathway is involved in the development and in the regulation, self-renewal, and differentiation of mesenchymal stem cells, and plays a role in sarcomagenesis. In this study, we have tested pharmacologic inhibition of Wnt signaling mediated by disruption of TCF/β-catenin binding and AXIN stabilization, being the first strategy more efficient in reducing cell viability and downstream effects. We have shown that disruption of TCF/β-catenin binding with PKF118-310 produces in vitro antitumor activity in a panel of prevalent representative STS cell lines and primary cultures. At the molecular level, PKF118-310 treatment reduced β-catenin nuclear localization, reporter activity, and target genes, resulting in an increase in apoptosis. Importantly, combination of PKF118-310 with doxorubicin resulted in enhanced reduction of cell viability, suggesting that Wnt inhibition could be a new combination regime in these patients. Our findings support the usefulness of Wnt inhibitors as new therapeutic strategies for the prevalent STS. Mol Cancer Ther; 16(6); 1166–76. ©2017 AACR.
Collapse
Affiliation(s)
- Esther Martinez-Font
- 1Group of Advanced Therapies and Biomarkers in Clinical Oncology, Institut d'Investigació Sanitària de Palma (IdISPa), Palma de Mallorca, Spain
| | - Irene Felipe-Abrio
- 2Group of Molecular Oncology and New Therapies, Oncohematology and Genetics Department, Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain
| | - Silvia Calabuig-Fariñas
- 3Molecular Oncology Laboratory, Fundación de Investigación, Hospital General Universitario de Valencia, Valencia, Spain
- 4Department of Pathology, Universitat de Valencia, Valencia, Spain
| | - Rafael Ramos
- 5Department of Pathology, Hospital Universitari Son Espases, Palma de Mallorca, Spain
| | - Josefa Terrasa
- 1Group of Advanced Therapies and Biomarkers in Clinical Oncology, Institut d'Investigació Sanitària de Palma (IdISPa), Palma de Mallorca, Spain
- 6Department of Oncology, Hospital Universitari Son Espases, Palma de Mallorca, Spain
| | - Oliver Vögler
- 1Group of Advanced Therapies and Biomarkers in Clinical Oncology, Institut d'Investigació Sanitària de Palma (IdISPa), Palma de Mallorca, Spain
- 7Group of Clinical and Translational Research, Department of Biology, Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), University of the Balearic Islands, Spain
| | - Regina Alemany
- 1Group of Advanced Therapies and Biomarkers in Clinical Oncology, Institut d'Investigació Sanitària de Palma (IdISPa), Palma de Mallorca, Spain
- 7Group of Clinical and Translational Research, Department of Biology, Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), University of the Balearic Islands, Spain
| | - Javier Martín-Broto
- 2Group of Molecular Oncology and New Therapies, Oncohematology and Genetics Department, Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain
- 8Department of Oncology, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Antònia Obrador-Hevia
- 1Group of Advanced Therapies and Biomarkers in Clinical Oncology, Institut d'Investigació Sanitària de Palma (IdISPa), Palma de Mallorca, Spain
- 6Department of Oncology, Hospital Universitari Son Espases, Palma de Mallorca, Spain
| |
Collapse
|
18
|
Yamada S, Imura Y, Nakai T, Nakai S, Yasuda N, Kaneko K, Outani H, Takenaka S, Hamada K, Myoui A, Araki N, Ueda T, Itoh K, Yoshikawa H, Naka N. Therapeutic potential of TAS-115 via c-MET and PDGFRα signal inhibition for synovial sarcoma. BMC Cancer 2017; 17:334. [PMID: 28511645 PMCID: PMC5434537 DOI: 10.1186/s12885-017-3324-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 05/04/2017] [Indexed: 01/16/2023] Open
Abstract
Background The prognosis of synovial sarcoma (SS), an aggressive soft tissue sarcoma, remains poor. We previously reported that c-MET or platelet-derived growth factor receptor α (PDGFRα) signalling pathway is related to SS progression based upon the findings of phospho-receptor tyrosine kinase (RTK) arrays. TAS-115 is a novel c-MET/ vascular endothelial growth factor receptor-targeting tyrosine kinase inhibitor that has been shown to inhibit multiple RTKs. Here we aimed to investigate the therapeutic potential of TAS-115 against SS. Methods We first evaluated which signalling pathway was relevant to the viability of three human SS cell lines: Yamato-SS, SYO-1 and HS-SY-II. Next, we assessed the anticancer activity and mechanism of action of TAS-115 in these SS cell lines. Finally, we compared the ability of TAS-115 to inhibit c-MET and PDGFRα phosphorylation with that of pazopanib. Results We classified the SS cell lines as c-MET-dependent or PDGFRα-dependent based upon the differences in the signalling pathway relevant for growth and/or survival. We also found that c-MET and PDGFRα were the primary activators of both phosphatidylinositol 3-kinase/AKT and mitogen-activated protein kinase pathways in c-MET-dependent and PDGFRα-dependent SS cells, respectively. TAS-115 treatment blocked the phosphorylation of PDGFRα as well as that of c-MET and their downstream effectors, leading to marked growth inhibition in both types of SS cell lines in in vitro and in vivo studies. Furthermore, PDGFRα phosphorylation, on at least four representative autophosphorylation sites, was impeded by TAS-115 equivalently to pazopanib. Conclusions These experimental results have demonstrated the significance of c-MET and PDGFRα signalling for growth and/or survival of SS tumours. TAS-115 monotherapy may benefit SS patients whose tumours are dependent upon either c-MET or PDGFRα signalling by functioning as a multiple tyrosine kinase inhibitor to suppress c-MET as well as PDGFRα pathways. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3324-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shutaro Yamada
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshinori Imura
- Musculoskeletal Oncology Service, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka, 541-8567, Japan
| | - Takaaki Nakai
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Sho Nakai
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Naohiro Yasuda
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Keiko Kaneko
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hidetatsu Outani
- Musculoskeletal Oncology Service, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka, 541-8567, Japan
| | - Satoshi Takenaka
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kenichiro Hamada
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Akira Myoui
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Nobuhito Araki
- Musculoskeletal Oncology Service, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka, 541-8567, Japan
| | - Takafumi Ueda
- Department of Orthopaedic Surgery, Osaka National Hospital, 2-1-14 Hoenzaka, Chuo-ku, Osaka, 540-0006, Japan
| | - Kazuyuki Itoh
- Research Institute, Nozaki Tokushukai, 2-10-50 Tanigawa, Daitou, Osaka, 574-0074, Japan
| | - Hideki Yoshikawa
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Norifumi Naka
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
19
|
El Beaino M, Araujo DM, Lazar AJ, Lin PP. Synovial Sarcoma: Advances in Diagnosis and Treatment Identification of New Biologic Targets to Improve Multimodal Therapy. Ann Surg Oncol 2017; 24:2145-2154. [PMID: 28397189 DOI: 10.1245/s10434-017-5855-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Indexed: 12/14/2022]
Abstract
Synovial sarcoma is a translocation-associated soft-tissue malignancy that frequently affects adolescents and young adults. It is driven by one of the fusion oncoproteins SS18-SSX1, SS18-SSX2, or rarely, SS18-SSX4. Prognosis of patients with recurrent or metastatic disease is generally poor, and newer therapeutic strategies are needed. In this review, we present recent discoveries in the pathogenesis, diagnosis, and treatment of synovial sarcoma. We discuss potential therapeutic strategies to improve clinical outcomes in this disease.
Collapse
Affiliation(s)
- Marc El Beaino
- Department of Orthopaedic Oncology - Unit 1448, MD Anderson Cancer Center, Houston, TX, USA
| | - Dejka M Araujo
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander J Lazar
- Department of Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick P Lin
- Department of Orthopaedic Oncology - Unit 1448, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
20
|
Barrott JJ, Kafchinski LA, Jin H, Potter JW, Kannan SD, Kennedy R, Mosbruger T, Wang WL, Tsai JW, Araujo DM, Liu T, Capecchi MR, Lazar AJ, Jones KB. Modeling synovial sarcoma metastasis in the mouse: PI3'-lipid signaling and inflammation. J Exp Med 2016; 213:2989-3005. [PMID: 27956588 PMCID: PMC5154942 DOI: 10.1084/jem.20160817] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/09/2016] [Accepted: 10/17/2016] [Indexed: 12/25/2022] Open
Abstract
Solid tumor metastasis is a complex biology, impinged upon by a variety of dysregulated signaling pathways. PI3'-lipid signaling has been associated with metastasis and inflammation in many cancers, but the relationship between tumor cell-intrinsic PI3'-lipid signaling and inflammatory cell recruitment has remained enigmatic. Elevated PI3'-lipid signaling associates with progression of synovial sarcoma, a deadly soft tissue malignancy initiated by a t(X;18) chromosomal translocation that generates an SS18-SSX fusion oncoprotein. Here, we show in genetically engineered mouse models of locally induced expression of SS18-SSX1 or SS18-SSX2 that Pten silencing dramatically accelerated and enhanced sarcomagenesis without compromising synovial sarcoma characteristics. PTEN deficiency increased tumor angiogenesis, promoted inflammatory gene expression, and enabled highly penetrant spontaneous pulmonary metastasis. PTEN-deficient sarcomas revealed infiltrating myeloid-derived hematopoietic cells, particularly macrophages and neutrophils, recruited via PI3'-lipid-induced CSF1 expression in tumor cells. Moreover, in a large panel of human synovial sarcomas, enhanced PI3'-lipid signaling also correlated with increased inflammatory cell recruitment and CSF1R signal transduction in both macrophages and endothelial cells. Thus, both in the mouse model and in human synovial sarcomas, PI3'-lipid signaling drives CSF1 expression and associates with increased infiltration of the monocyte/macrophage lineage as well as neutrophils.
Collapse
Affiliation(s)
- Jared J Barrott
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - Lisa A Kafchinski
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112
| | - Huifeng Jin
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - Jared W Potter
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - Sarmishta D Kannan
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - Robert Kennedy
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - Tim Mosbruger
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112.,Department of Bioinformatics, University of Utah, Salt Lake City, UT 84112
| | - Wei-Lien Wang
- Departments of Pathology and Translational Molecular Pathology, M.D. Anderson Cancer Center, Houston, TX 77030
| | - Jen-Wei Tsai
- Departments of Pathology and Translational Molecular Pathology, M.D. Anderson Cancer Center, Houston, TX 77030
| | - Dejka M Araujo
- Sarcoma Medical Oncology, M.D. Anderson Cancer Center, Houston, TX 77030
| | - Ting Liu
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112.,Department of Pathology, University of Utah, Salt Lake City, UT 84112
| | - Mario R Capecchi
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112
| | - Alexander J Lazar
- Departments of Pathology and Translational Molecular Pathology, M.D. Anderson Cancer Center, Houston, TX 77030
| | - Kevin B Jones
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112 .,Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| |
Collapse
|
21
|
Next generation sequencing in synovial sarcoma reveals novel gene mutations. Oncotarget 2016; 6:34680-90. [PMID: 26415226 PMCID: PMC4741482 DOI: 10.18632/oncotarget.5786] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 08/31/2015] [Indexed: 01/05/2023] Open
Abstract
Over 95% of all synovial sarcomas (SS) share a unique translocation, t(X;18), however, they show heterogeneous clinical behavior. We analyzed multiple SS to reveal additional genetic alterations besides the translocation. Twenty-six SS from 22 patients were sequenced for 409 cancer-related genes using the Comprehensive Cancer Panel (Life Technologies, USA) on an Ion Torrent platform. The detected variants were verified by Sanger sequencing and compared to matched normal DNAs. Copy number variation was assessed in six tumors using the Oncoscan array (Affymetrix, USA). In total, eight somatic mutations were detected in eight samples. These mutations have not been reported previously in SS. Two of these, in KRAS and CCND1, represent known oncogenic mutations in other malignancies. Additional mutations were detected in RNF213, SEPT9, KDR, CSMD3, MLH1 and ERBB4. DNA alterations occurred more often in adult tumors. A distinctive loss of 6q was found in a metastatic lesion progressing under pazopanib, but not in the responding lesion. Our results emphasize t(X;18) as a single initiating event in SS and as the main oncogenic driver. Our results also show the occurrence of additional genetic events, mutations or chromosomal aberrations, occurring more frequently in SS with an onset in adults.
Collapse
|
22
|
Vlenterie M, Hillebrandt-Roeffen MHS, Schaars EWM, Flucke UE, Fleuren EDG, Navis AC, Leenders WPJ, Versleijen-Jonkers YMH, van der Graaf WTA. Targeting Cyclin-Dependent Kinases in Synovial Sarcoma: Palbociclib as a Potential Treatment for Synovial Sarcoma Patients. Ann Surg Oncol 2016; 23:2745-52. [PMID: 27334220 PMCID: PMC4972869 DOI: 10.1245/s10434-016-5341-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Indexed: 11/18/2022]
Abstract
Background In synovial sarcomas alterations in the cyclin D1-CDK4/6-Rb axis have been described. Also, β-catenin, a cyclin D1 regulator, is often overexpressed. Additionally, studies have shown that the t(X;18) translocation influences tumor behavior partly through cyclin D1 activation. We investigated how alterations in the cyclin D1-CDK4/6-Rb axis impact prognosis and studied effects of targeting this axis with the CDK4/6 inhibitor palbociclib. Methods Synovial sarcoma samples (n = 43) were immunohistochemically stained for β-catenin, cyclin D1, p16, p21, p27, Rb, and phospho-Rb. Fluorescent in situ hybridization (FISH) was performed to detect CCND1 amplification or translocation. In 4 synovial sarcoma cell lines sensitivity to palbociclib was investigated using cell viability assays, and effects on the sensitive cell lines were evaluated on protein level and by cell cycle arrest. Results Expression of nuclear phospho-Rb and nuclear β-catenin in the patient samples was associated with poor survival. FISH showed a sporadic translocation of CCND1 in a subset of tumors. An 8-fold CCND1 amplification was found in 1 cell line, but not in the patient samples investigated. Palbociclib effectively inhibited Rb-phosphorylation in 3 cell lines, resulting in an induction of a G1 arrest and proliferation block. Conclusions In this series nuclear phospho-Rb and nuclear β-catenin expression were negative prognostic factors. In vitro data suggest that palbociclib may be a potential treatment for a subset of synovial sarcoma patients. Whether this effect can be enhanced by combination treatment deserves further preclinical investigations. Electronic supplementary material The online version of this article (doi:10.1245/s10434-016-5341-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Myrella Vlenterie
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | | | - Esther W M Schaars
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Uta E Flucke
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Emmy D G Fleuren
- The Institute of Cancer Research London and the Royal Marsden NHS Foundation Trust, London, UK
| | - Anna C Navis
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - William P J Leenders
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Winette T A van der Graaf
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.,The Institute of Cancer Research London and the Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
23
|
Villar VH, Vögler O, Barceló F, Martín-Broto J, Martínez-Serra J, Ruiz-Gutiérrez V, Alemany R. Down-Regulation of AKT Signalling by Ursolic Acid Induces Intrinsic Apoptosis and Sensitization to Doxorubicin in Soft Tissue Sarcoma. PLoS One 2016; 11:e0155946. [PMID: 27219337 PMCID: PMC4878803 DOI: 10.1371/journal.pone.0155946] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 05/06/2016] [Indexed: 01/11/2023] Open
Abstract
Several important biological activities have been attributed to the pentacyclic triterpene ursolic acid (UA), being its antitumoral effect extensively studied in human adenocarcinomas. In this work, we focused on the efficacy and molecular mechanisms involved in the antitumoral effects of UA, as single agent or combined with doxorubicin (DXR), in human soft tissue sarcoma cells. UA (5-50 μM) strongly inhibited (up to 80%) the viability of STS cells at 24 h and its proliferation in soft agar, with higher concentrations increasing apoptotic death up to 30%. UA treatment (6-9 h) strongly blocked the survival AKT/GSK3β/β-catenin signalling pathway, which led to a concomitant reduction of the anti-apoptotic proteins c-Myc and p21, altogether resulting in the activation of intrinsic apoptosis. Interestingly, UA at low concentrations (10-15 μM) enhanced the antitumoral effects of DXR by up to 2-fold, while in parallel inhibiting DXR-induced AKT activation and p21 expression, two proteins implicated in antitumoral drug resistance and cell survival. In conclusion, UA is able to induce intrinsic apoptosis in human STS cells and also to sensitize these cells to DXR by blocking the AKT signalling pathway. Therefore, UA may have beneficial effects, if used as nutraceutical adjuvant during standard chemotherapy treatment of STS.
Collapse
Affiliation(s)
- Victor Hugo Villar
- Group of Clinical and Translational Research, Department of Biology, Institut Universitari d’Investigacions en Ciències de la Salut (IUNICS), University of the Balearic Islands, Palma de Mallorca, Spain
| | - Oliver Vögler
- Group of Clinical and Translational Research, Department of Biology, Institut Universitari d’Investigacions en Ciències de la Salut (IUNICS), University of the Balearic Islands, Palma de Mallorca, Spain
| | - Francisca Barceló
- Group of Clinical and Translational Research, Department of Biology, Institut Universitari d’Investigacions en Ciències de la Salut (IUNICS), University of the Balearic Islands, Palma de Mallorca, Spain
| | - Javier Martín-Broto
- Group of Clinical and Translational Research, Department of Biology, Institut Universitari d’Investigacions en Ciències de la Salut (IUNICS), University of the Balearic Islands, Palma de Mallorca, Spain
- Department of Oncology, University Hospital Virgen del Rocío and Biomedicine Institute of Sevilla (IBIS), Sevilla, Spain
| | - Jordi Martínez-Serra
- Group of Clinical and Translational Research, Department of Biology, Institut Universitari d’Investigacions en Ciències de la Salut (IUNICS), University of the Balearic Islands, Palma de Mallorca, Spain
- Department of Hematology, University Hospital Son Espases, Palma de Mallorca, Spain
| | - Valentina Ruiz-Gutiérrez
- Instituto de la Grasa, Consejo Superior de Investigaciones Científicas (CSIC), Sevilla, Spain
- CIBER:CB06/03 Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Instituto de Salud, Carlos III (ISCIII), Spain
| | - Regina Alemany
- Group of Clinical and Translational Research, Department of Biology, Institut Universitari d’Investigacions en Ciències de la Salut (IUNICS), University of the Balearic Islands, Palma de Mallorca, Spain
- CIBER:CB06/03 Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Instituto de Salud, Carlos III (ISCIII), Spain
| |
Collapse
|
24
|
Ferrari A, Chiaravalli S, Casanova M, Gasparini P, Corradini N, Orbach D. Considering chemotherapy in synovial sarcoma. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1076723] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
25
|
Nielsen TO, Poulin NM, Ladanyi M. Synovial sarcoma: recent discoveries as a roadmap to new avenues for therapy. Cancer Discov 2015; 5:124-34. [PMID: 25614489 DOI: 10.1158/2159-8290.cd-14-1246] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
UNLABELLED Oncogenesis in synovial sarcoma is driven by the chromosomal translocation t(X,18; p11,q11), which generates an in-frame fusion of the SWI/SNF subunit SS18 to the C-terminal repression domains of SSX1 or SSX2. Proteomic studies have identified an integral role of SS18-SSX in the SWI/SNF complex, and provide new evidence for mistargeting of polycomb repression in synovial sarcoma. Two recent in vivo studies are highlighted, providing additional support for the importance of WNT signaling in synovial sarcoma: One used a conditional mouse model in which knockout of β-catenin prevents tumor formation, and the other used a small-molecule inhibitor of β-catenin in xenograft models. SIGNIFICANCE Synovial sarcoma appears to arise from still poorly characterized immature mesenchymal progenitor cells through the action of its primary oncogenic driver, the SS18-SSX fusion gene, which encodes a multifaceted disruptor of epigenetic control. The effects of SS18-SSX on polycomb-mediated gene repression and SWI/SNF chromatin remodeling have recently come into focus and may offer new insights into the basic function of these processes. A central role for deregulation of WNT-β-catenin signaling in synovial sarcoma has also been strengthened by recent in vivo studies. These new insights into the the biology of synovial sarcoma are guiding novel preclinical and clinical studies in this aggressive cancer.
Collapse
Affiliation(s)
- Torsten O Nielsen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Neal M Poulin
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Marc Ladanyi
- Department of Pathology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
26
|
Morgan SS, Cranmer LD. Vorinostat synergizes with ridaforolimus and abrogates the ridaforolimus-induced activation of AKT in synovial sarcoma cells. BMC Res Notes 2014; 7:812. [PMID: 25406429 PMCID: PMC4247709 DOI: 10.1186/1756-0500-7-812] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/31/2014] [Indexed: 11/13/2022] Open
Abstract
Background Curative treatments for patients with metastatic synovial sarcoma (SS) do not exist, and such patients have a poor prognosis. We explored combinations of molecularly-targeted and cytotoxic agents to identify synergistic treatment combinations in SS cells. Methods Two SS cell lines (HS-SY-II and SYO-I) were treated with single agents or combinations of molecularly targeted therapies (HDAC inhibitor, vorinostat; mTOR inhibitor, ridaforolimus) and cytotoxic agents. After 72 hours, cell viability was measured using the MTS cell proliferation assay. Combination Indices (CI) were calculated to determine whether each combination was synergistic, additive, or antagonistic. Western Blot analysis assessed alterations in total and phospho-AKT protein levels in response to drug treatment. Results We determined the single-agent IC50 for ridaforolimus, vorinostat, doxorubicin, and melphalan in HS-SY-II and SYO-I. Synergism was apparent in cells co-treated with ridaforolimus and vorinostat: CI was 0.28 and 0.63 in HS-SY-II and SYO-I, respectively. Ridaforolimus/doxorubicin and ridaforolimus/melphalan exhibited synergism in both cell lines. An additive effect was observed with combination of vorinostat/doxorubicin in both cell lines. Vorinostat/melphalan was synergistic in HS-SY-II and additive in SYO-I. Western blot analysis demonstrated that ridaforolimus increased pAKT-ser473 levels; this effect was abrogated by vorinostat co-treatment. Conclusions The combination of ridaforolimus and vorinostat demonstrates in vitro synergism in SS. Addition of vorinostat abrogated ridaforolimus-induced AKT activation. Since AKT activation is a possible mechanism of resistance to mTOR inhibitors, adding vorinostat (or another HDAC inhibitor) may be a route to circumvent AKT-mediated resistance to mTOR inhibitors.
Collapse
Affiliation(s)
| | - Lee D Cranmer
- The University of Arizona Cancer Center, 1515 N, Campbell Avenue, Tucson, AZ 85724-5024, USA.
| |
Collapse
|
27
|
Yasui H, Naka N, Imura Y, Outani H, Kaneko K, Hamada KI, Sasagawa S, Araki N, Ueda T, Itoh K, Myoui A, Yoshikawa H. Tailored therapeutic strategies for synovial sarcoma: Receptor tyrosine kinase pathway analyses predict sensitivity to the mTOR inhibitor RAD001. Cancer Lett 2014; 347:114-22. [DOI: 10.1016/j.canlet.2014.01.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 01/15/2014] [Accepted: 01/27/2014] [Indexed: 01/03/2023]
|
28
|
Palmerini E, Paioli A, Ferrari S. Emerging therapeutic targets for synovial sarcoma. Expert Rev Anticancer Ther 2014; 14:791-806. [DOI: 10.1586/14737140.2014.901155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
29
|
Takahashi Y, Kohashi K, Yamada Y, Endo M, Setsu N, Ishii T, Yamamoto H, Iwamoto Y, Oda Y. Activation of the Akt/mammalian target of rapamycin pathway in myxofibrosarcomas. Hum Pathol 2014; 45:984-93. [PMID: 24746202 DOI: 10.1016/j.humpath.2013.12.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 12/17/2013] [Accepted: 12/18/2013] [Indexed: 10/25/2022]
Abstract
The Akt/mammalian target of rapamycin (mTOR) pathway plays important roles in modulating cellular function in response to extracellular signals such as growth factors and cytokines. The Akt/mTOR signaling pathway is activated in certain kinds of sarcomas. Myxofibrosarcoma is a soft tissue sarcoma, characterized by abundant myxoid stroma and frequent local recurrence. Here, we conducted a large-scale examination of the clinicopathological and activation statuses of the Akt/mTOR pathways in myxofibrosarcoma. The phosphorylation status of Akt, mTOR, S6 ribosomal protein, and the eukaryotic translation initiation factor 4E-binding protein, and mitogen-activated protein kinase were assessed by immunohistochemistry in 101 formalin-fixed, paraffin-embedded samples, including 68 primary tumors in myxofibrosarcoma. Immunohistochemical expressions were confirmed by Western blotting with 20 frozen samples, which were paired with normal tissue samples. PIK3CA and AKT1 gene mutations were also analyzed using 12 primary tumor frozen samples. Immunohistochemically, phosphorylations of Akt, mTOR, S6 ribosomal protein, 4E-binding protein, and mitogen-activated protein kinase 1/2 were observed in 64.7%, 45.6%, 42.6%, 63.2%, and 64.7% of samples. Phosphorylated Akt/mTOR pathway proteins were correlated with one another and were also correlated with the phosphorylation of these proteins in the concordant recurrent tumors. Immunoblotting showed a high degree of phosphorylation in tumor samples, compared with that in normal tissue samples. Activation of the Akt/mTOR pathway was correlated with histologic grade and tumor progression. Mutational analysis failed to reveal any PIK3CA or AKT1 mutations around the hot spots. Activation of the Akt/mTOR pathway was associated with histologic malignancy and tumor progression in primary and recurrent myxofibrosarcoma.
Collapse
Affiliation(s)
- Yusuke Takahashi
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yuichi Yamada
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Makoto Endo
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Nokitaka Setsu
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takeaki Ishii
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yukihide Iwamoto
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
30
|
Levard A, Tassy L, Cassier PA. Emerging Therapies for Soft-Tissue Sarcomas. Hematol Oncol Clin North Am 2013; 27:1063-78. [DOI: 10.1016/j.hoc.2013.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
31
|
Zhu WL, Wang SJ, Liu MM, Shi HS, Zhang RX, Liu JF, Ding ZB, Lu L. Glycine site N-methyl-D-aspartate receptor antagonist 7-CTKA produces rapid antidepressant-like effects in male rats. J Psychiatry Neurosci 2013; 38:306-16. [PMID: 23611177 PMCID: PMC3756114 DOI: 10.1503/jpn.120228] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Glutamate N-methyl-D-aspartate (NMDA) receptor antagonists exert fast-acting antidepressant effects, providing a promising way to develop a new classification of antidepressant that targets the glutamatergic system. In the present study, we examined the potential antidepressant action of 7-chlorokynurenic acid (7-CTKA), a glycine recognition site NMDA receptor antagonist, in a series of behavioural models of depression and determined the molecular mechanisms that underlie the behavioural actions of 7-CTKA. METHODS We administered the forced swim test, novelty-suppressed feeding test, learned helplessness paradigm and chronic mild stress (CMS) paradigm in male rats to evaluate the possible rapid antidepressant-like actions of 7-CTKA. In addition, we assessed phospho-glycogen synthase kinase-3β (p-GSK3β) level, mammalian target of rapamycin (mTOR) function, and postsynaptic protein expression in the medial prefrontal cortex (mPFC) and hippocampus. RESULTS Acute 7-CTKA administration produced rapid antidepressant-like actions in several behavioural tests. It increased p-GSK3β, enhanced mTOR function and increased postsynaptic protein levels in the mPFC. Activation of GSK3β by LY294002 completely blocked the antidepressant-like effects of 7-CTKA. Moreover, 7-CTKA did not produce rewarding properties or abuse potential. LIMITATIONS It is possible that 7-CTKA modulates glutamatergic transmission, thereby causing enduring alterations of GSK3β and mTOR signalling, although we did not provide direct evidence to support this possibility. Thus, the therapeutic involvement of synaptic adaptions engaged by 7-CTKA requires further study. CONCLUSION Our findings demonstrate that acute 7-CTKA administration produced rapid antidepressant-like effects, indicating that the behavioural response to 7-CTKA is mediated by GSK3β and mTOR signalling function in the mPFC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lin Lu
- Correspondence to: L. Lu, National Institute on Drug Dependence, Peking University, 38 Xue Yuan Rd., Hai Dian District, Beijing 100191, China;
| |
Collapse
|
32
|
Setsu N, Kohashi K, Fushimi F, Endo M, Yamamoto H, Takahashi Y, Yamada Y, Ishii T, Yokoyama K, Iwamoto Y, Oda Y. Prognostic impact of the activation status of the Akt/mTOR pathway in synovial sarcoma. Cancer 2013; 119:3504-13. [DOI: 10.1002/cncr.28255] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 05/08/2013] [Accepted: 05/28/2013] [Indexed: 01/24/2023]
Affiliation(s)
- Nokitaka Setsu
- Department of Anatomic Pathology; Graduate School of Medical Science, Kyushu University; Fukuoka Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology; Graduate School of Medical Science, Kyushu University; Fukuoka Japan
| | - Fumiyoshi Fushimi
- Department of Pathology; National Kyushu Cancer Center; Fukuoka Japan
| | - Makoto Endo
- Department of Anatomic Pathology; Graduate School of Medical Science, Kyushu University; Fukuoka Japan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology; Graduate School of Medical Science, Kyushu University; Fukuoka Japan
| | - Yusuke Takahashi
- Department of Anatomic Pathology; Graduate School of Medical Science, Kyushu University; Fukuoka Japan
| | - Yuichi Yamada
- Department of Anatomic Pathology; Graduate School of Medical Science, Kyushu University; Fukuoka Japan
| | - Takeaki Ishii
- Department of Anatomic Pathology; Graduate School of Medical Science, Kyushu University; Fukuoka Japan
| | | | - Yukihide Iwamoto
- Department of Orthopedic Surgery; Graduate School of Medical Science, Kyushu University; Fukuoka Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology; Graduate School of Medical Science, Kyushu University; Fukuoka Japan
| |
Collapse
|
33
|
Bekarev M, Elsinger EC, Villanueva-Siles E, Borzykowski RM, Geller DS. Synovial sarcoma of the foot. J Foot Ankle Surg 2013; 52:513-7. [PMID: 23632071 DOI: 10.1053/j.jfas.2013.03.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Indexed: 02/03/2023]
Abstract
We report the case of a 75-year-old male who underwent lung lobectomy for presumed lung cancer. Thereafter, he presented with a painful mass between the third and fourth metatarsal heads in the foot that was assumed to be Morton's neuroma. After extensive oncologic evaluation, the foot mass was diagnosed as a synovial sarcoma. In retrospect, his lung lesion was understood to be metastatic disease.
Collapse
|
34
|
Michels S, Trautmann M, Sievers E, Kindler D, Huss S, Renner M, Friedrichs N, Kirfel J, Steiner S, Endl E, Wurst P, Heukamp L, Penzel R, Larsson O, Kawai A, Tanaka S, Sonobe H, Schirmacher P, Mechtersheimer G, Wardelmann E, Büttner R, Hartmann W. SRC signaling is crucial in the growth of synovial sarcoma cells. Cancer Res 2013; 73:2518-28. [PMID: 23580575 DOI: 10.1158/0008-5472.can-12-3023] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Synovial sarcoma is a soft-tissue malignancy characterized by a reciprocal t(X;18) translocation encoding a chimeric transcriptional modifier. Several receptor tyrosine kinases have been found activated in synovial sarcoma; however, no convincing therapeutic concept has emerged from these findings. On the basis of the results of phosphokinase screening arrays, we here investigate the functional and therapeutic relevance of the SRC kinase in synovial sarcoma. Immunohistochemistry of phosphorylated SRC and its regulators CSK and PTP1B (PTPN1) was conducted in 30 synovial sarcomas. Functional aspects of SRC, including dependence of SRC activation on the SS18/SSX fusion proteins, were analyzed in vitro. Eventually, synovial sarcoma xenografts were treated with the SRC inhibitor dasatinib in vivo. Activated phospho (p)-(Tyr416)-SRC was detected in the majority of tumors; dysregulation of CSK or PTP1B was excluded as the reason for the activation of the kinase. Expression of the SS18/SSX fusion proteins in T-REx-293 cells was associated with increased p-(Tyr416)-SRC levels, linked with an induction of the insulin-like growth factor pathway. Treatment of synovial sarcoma cells with dasatinib led to apoptosis and inhibition of cellular proliferation, associated with reduced phosphorylation of FAK (PTK2), STAT3, IGF-IR, and AKT. Concurrent exposure of cells to dasatinib and chemotherapeutic agents resulted in additive effects. Cellular migration and invasion were dependent on signals transmitted by SRC involving regulation of the Rho GTPases Rac and RhoA. Treatment of nude mice with SYO-1 xenografts with dasatinib significantly inhibited tumor growth in vivo. In summary, SRC is of crucial biologic importance and represents a promising therapeutic target in synovial sarcoma.
Collapse
Affiliation(s)
- Sebastian Michels
- Department of Pathology, University Hospital Cologne, Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zheng J, Hu JD, Chen YY, Chen BY, Huang Y, Zheng ZH, Liu TB. Baicalin induces apoptosis in leukemia HL-60/ADR cells via possible down-regulation of the PI3K/Akt signaling pathway. Asian Pac J Cancer Prev 2013; 13:1119-24. [PMID: 22799292 DOI: 10.7314/apjcp.2012.13.4.1119] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The effect and possible mechanism of traditional Chinese medicine, baicalin, on the PI3K/ Akt signaling pathway in drug-resistant human myeloid leukemia HL-60/ADR cells have been investigated in this current study. METHODS HL-60/ADR cells were treated by 20, 40, 80 μmol/L baicalin followed by cell cycle analysis at 24h. The mRNA expression level of the apoptosis related gene, Bcl-2 and bad, were measured by RT-PCR on cells treated with 80 μmol/L baicalin at 12, 24 and 48hr. Western blot was performed to detect the changes in the expression of the proteins related to HL-60/ADR cell apoptosis and the signaling pathway before and after baicalin treatment, including Bcl-2, PARP, Bad, Caspase 3, Akt, p-Akt, NF-κB, p-NF-κB, mTOR and p-mTOR. RESULTS Sub-G1 peak of HL-60/ADR cells appeared 24 h after 20 μmol/L baicalin treatment, and the ratio increased as baicalin concentration increased. Cell cycle analysis showed 44.9% G0/G1 phase cells 24 h after baicalin treatment compared to 39.6% in the control group. Cells treated with 80 μmol/L baicalin displayed a trend in decreasing of Bcl-2 mRNA expression over time. Expression level of the Bcl-2 and PARP proteins decreased significantly while that of the PARP, Caspase-3, and Bad proteins gradually increased. No significant difference in Akt expression was observed between treated and the control groups. However, the expression levels of p-Akt, NF-κB, p-NF-κB, mTOR and p-mTOR decreased significantly in a time-dependent manner. CONCLUSIONS We conclude that baicalin may induce HL-60/ADR cell apoptosis through the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Jing Zheng
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | | | | | | | | | | | | |
Collapse
|
36
|
Hosaka S, Horiuchi K, Yoda M, Nakayama R, Tohmonda T, Susa M, Nakamura M, Chiba K, Toyama Y, Morioka H. A novel multi-kinase inhibitor pazopanib suppresses growth of synovial sarcoma cells through inhibition of the PI3K-AKT pathway. J Orthop Res 2012; 30:1493-8. [PMID: 22359392 DOI: 10.1002/jor.22091] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Accepted: 02/02/2012] [Indexed: 02/04/2023]
Abstract
Synovial sarcoma is an aggressive soft tissue sarcoma with only a modest response to conventional cytotoxic agents. In the present study, we evaluated the potential antitumor effects of a novel anti-angiogenesis agent, pazopanib, against synovial sarcoma cells. We found that pazopanib directly inhibited the growth of synovial sarcoma cells by inducing G1 arrest. Multiplex analyses revealed that the PI3K-AKT pathway was highly suppressed in pazopanib-sensitive synovial sarcoma cells. Furthermore, administration of pazopanib highly suppressed the tumor growth in a xenograft model. Taken together, these results suggest pazopanib as a possible agent against synovial sarcoma and may warrant further clinical studies.
Collapse
Affiliation(s)
- Seiichi Hosaka
- Department of Orthopedic Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Synovial sarcomas are regarded as chemosensitive tumors compared to other types of soft tissue sarcomas, however, prognosis for advanced refractory disease remains poor. In light of the vascular nature of sarcomas, current molecularly targeted therapies aim at an antiangiogenic approach to management of this disease. RECENT FINDINGS Recent studies with oral vascular endothelial growth factor receptor (VEGFR) inhibitors such as sunitinib, sorafenib and cediranib have shown disease stabilization in patients with advanced synovial sarcoma. Forty-nine percent of patients with synovial sarcoma on the phase II trial of pazopanib had no evidence of disease progression at 12 weeks. SUMMARY The overall impact of chemotherapy on survival has been minimal in advanced soft tissue sarcomas. Anti-VEGF therapies have resulted in improved outcomes for patients with various solid tumors, and have shown preliminary evidence of activity in synovial sarcomas. Combinations of anti-VEGF therapies with agents targeting other pathways dysregulated in sarcomas have the potential to improve the outcome of this difficult-to-treat disease.
Collapse
Affiliation(s)
- Khanh Do
- Medical Oncology Branch, Center for Cancer Research, Bethesda, Maryland, USA
| | - James H. Doroshow
- Medical Oncology Branch, Center for Cancer Research, Bethesda, Maryland, USA
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland, USA
| | - Shivaani Kummar
- Medical Oncology Branch, Center for Cancer Research, Bethesda, Maryland, USA
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
38
|
von dem Knesebeck A, Felsberg J, Waha A, Hartmann W, Scheffler B, Glas M, Hammes J, Mikeska T, Yan PS, Endl E, Simon M, Reifenberger G, Pietsch T, Waha A. RANK (TNFRSF11A) is epigenetically inactivated and induces apoptosis in gliomas. Neoplasia 2012; 14:526-34. [PMID: 22787434 PMCID: PMC3394195 DOI: 10.1596/neo.12360] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 04/27/2012] [Accepted: 04/30/2012] [Indexed: 12/13/2022]
Abstract
Alterations of DNA methylation play an important role in gliomas. In a genome-wide screen, we identified a CpG-rich fragment within the 5' region of the tumor necrosis factor receptor superfamily, member 11A gene (TNFRSF11A) that showed de novo methylation in gliomas. TNFRSF11A, also known as receptor activator of NF-κB (RANK), activates several signaling pathways, such as NF-κB, JNK, ERK, p38α, and Akt/PKB. Using pyrosequencing, we detected RANK/TNFRSF11A promoter methylation in 8 (57.1%) of 14 diffuse astrocytomas, 17 (77.3%) of 22 anaplastic astrocytomas, 101 (84.2%) of 120 glioblastomas, 6 (100%) of 6 glioma cell lines, and 7 (100%) of 7 glioma stem cell-enriched glioblastoma primary cultures but not in four normal white matter tissue samples. Treatment of glioma cell lines with the demethylating agent 5-aza-2'-deoxycytidine significantly reduced the methylation level and resulted in increased RANK/TNFRSF11A mRNA expression. Overexpression of RANK/TNFRSF11A in glioblastoma cell lines leads to a significant reduction in focus formation and elevated apoptotic activity after flow cytometric analysis. Reporter assay studies of transfected glioma cells supported these results by showing the activation of signaling pathways associated with regulation of apoptosis. We conclude that RANK/TNFRSF11A is a novel and frequent target for de novo methylation in gliomas, which affects apoptotic activity and focus formation thereby contributing to the molecular pathogenesis of gliomas.
Collapse
Affiliation(s)
| | - Jörg Felsberg
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Anke Waha
- Department of Neuropathology, University of Bonn, Bonn, Germany
| | | | - Björn Scheffler
- Institute of Reconstructive Neurobiology, University of Bonn, Bonn, Germany
| | - Martin Glas
- Institute of Reconstructive Neurobiology, University of Bonn, Bonn, Germany
- Clinical Neurooncology Unit, Department of Neurology, University of Bonn, Bonn, Germany
| | - Jennifer Hammes
- Department of Neuropathology, University of Bonn, Bonn, Germany
| | - Thomas Mikeska
- Department of Neuropathology, University of Bonn, Bonn, Germany
| | - Pearlly S Yan
- Division of Human Cancer Genetics, Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, Ohio State University, Columbus, OH
| | - Elmar Endl
- Institute of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany
| | - Matthias Simon
- Department of Neurosurgery, University of Bonn, Bonn, Germany
| | - Guido Reifenberger
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Torsten Pietsch
- Department of Neuropathology, University of Bonn, Bonn, Germany
| | - Andreas Waha
- Department of Neuropathology, University of Bonn, Bonn, Germany
| |
Collapse
|
39
|
Kimura T, Wang L, Tabu K, Nishihara H, Mashita Y, Kikuchi N, Tanino M, Hiraga H, Tanaka S. CD133 Negatively Regulates Tumorigenicity via AKT Pathway in Synovial Sarcoma. Cancer Invest 2012; 30:390-7. [DOI: 10.3109/07357907.2012.672607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Taichi Kimura
- 1Department of Pathology, Laboratory of Cancer Research, Hokkaido University Graduate School of Medicine,
N15, W7, Sapporo, Japan,
| | - Lei Wang
- 1Department of Pathology, Laboratory of Cancer Research, Hokkaido University Graduate School of Medicine,
N15, W7, Sapporo, Japan,
| | - Kouichi Tabu
- 2Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University,
1-5-45 Yushima, Bunkyo-ku, Tokyo, Japan,
| | - Hiroshi Nishihara
- 3Department of Translational Pathology, Hokkaido University Graduate School of Medicine,
N15, W7, Sapporo, Japan,
| | - Yuji Mashita
- 1Department of Pathology, Laboratory of Cancer Research, Hokkaido University Graduate School of Medicine,
N15, W7, Sapporo, Japan,
| | - Naoyuki Kikuchi
- 1Department of Pathology, Laboratory of Cancer Research, Hokkaido University Graduate School of Medicine,
N15, W7, Sapporo, Japan,
| | - Mishie Tanino
- 1Department of Pathology, Laboratory of Cancer Research, Hokkaido University Graduate School of Medicine,
N15, W7, Sapporo, Japan,
| | - Hiroaki Hiraga
- 4Division of Orthopedics, National Hospital Organization Hokkaido Cancer Center,
Sapporo, Japan
| | - Shinya Tanaka
- 1Department of Pathology, Laboratory of Cancer Research, Hokkaido University Graduate School of Medicine,
N15, W7, Sapporo, Japan,
- 3Department of Translational Pathology, Hokkaido University Graduate School of Medicine,
N15, W7, Sapporo, Japan,
| |
Collapse
|
40
|
Ray A, Huh WW. Current State-of-the-Art Systemic Therapy for Pediatric Soft Tissue Sarcomas. Curr Oncol Rep 2012; 14:311-9. [DOI: 10.1007/s11912-012-0243-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
41
|
Downstream and intermediate interactions of synovial sarcoma-associated fusion oncoproteins and their implication for targeted therapy. Sarcoma 2012; 2012:249219. [PMID: 22550415 PMCID: PMC3329658 DOI: 10.1155/2012/249219] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 01/09/2012] [Indexed: 12/14/2022] Open
Abstract
Synovial sarcoma (SS), an aggressive type of soft tissue tumor, occurs mostly in adolescents and young adults. The origin and molecular mechanism of the development of SS remain only partially known. Over 90% of SS cases are characterized by the t(X;18)(p11.2;q11.2) translocation, which results mainly in the formation of
SS18-SSX1 or SS18-SSX2 fusion genes. In recent years, several reports describing direct and indirect interactions of SS18-SSX1/SSX2 oncoproteins have been published. These reports suggest that the fusion proteins particularly affect the cell growth, cell proliferation, TP53 pathway, and chromatin remodeling mechanisms, contributing to SS oncogenesis. Additional research efforts are required to fully explore the protein-protein interactions of SS18-SSX oncoproteins and the pathways that are regulated by these partnerships for the development of effective targeted therapy.
Collapse
|
42
|
Wardelmann E, Chemnitz JM, Wendtner CM. Targeted Therapy of Soft Tissue Sarcomas. ACTA ACUST UNITED AC 2012; 35 Suppl 1:21-7. [DOI: 10.1159/000334958] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
43
|
Vincenzi B, Napolitano A, D'Onofrio L, Frezza AM, Silletta M, Venditti O, Santini D, Tonini G. Targeted therapy in sarcomas: mammalian target of rapamycin inhibitors from bench to bedside. Expert Opin Investig Drugs 2011; 20:1685-705. [DOI: 10.1517/13543784.2011.628984] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Bruno Vincenzi
- University Campus Bio-Medico, Medical Oncology, via Alvaro del Portillo, 200, Rome, Italy
| | - Andrea Napolitano
- University Campus Bio-Medico, Medical Oncology, via Alvaro del Portillo, 200, Rome, Italy
| | - Loretta D'Onofrio
- University Campus Bio-Medico, Medical Oncology, via Alvaro del Portillo, 200, Rome, Italy
| | - Anna Maria Frezza
- University Campus Biomedico, Via Emilio Longoni 69, 155, Rome, Italy
| | - Marianna Silletta
- University Campus Bio-Medico, Medical Oncology, via Alvaro del Portillo, 200, Rome, Italy
| | - Olga Venditti
- University Campus Bio-Medico, Medical Oncology, via Alvaro del Portillo, 200, Rome, Italy
| | - Daniele Santini
- University Campus Biomedico, Via Emilio Longoni 69, 155, Rome, Italy
| | - Giuseppe Tonini
- University Campus Biomedico, Via Emilio Longoni 69, 155, Rome, Italy
| |
Collapse
|