1
|
Mitsos P, Anastasiou I, Constantinides C, Deligiannis D, Katafigiotis I, Papakonstantinou A, Tzotzola V, Mitropoulos D, Theocharis S. Clinical Importance of Focal Adhesion Kinase (FAK)-Src and Paxillin Expression in Renal Cell Carcinoma. Cureus 2024; 16:e62706. [PMID: 39036223 PMCID: PMC11259196 DOI: 10.7759/cureus.62706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND AND OBJECTIVE The complex focal adhesion kinase (FAK)/Src and paxillin seem to play a key role in the pathogenesis and progression of cancer. The aim of this study is to evaluate the expression of these proteins in renal cell carcinomas (RCCs), considering the immunoreactive score (IRS), the positivity and the intensity, and to find any association with patients' clinical characteristics, histologic type and other pathological features that imply a possible pathophysiological or prognostic role of FAK/Src and paxillin in RCC. METHODS Patients with RCC who had undergone partial or radical nephrectomy from January 2009 to September 2010 were eligible for this retrospective cross-sectional study. The immunohistochemical expression of FAK, Src and paxillin proteins in formalin-fixed paraffin-embedded tumour tissue was analysed in association with various clinicopathological features. RESULTS Out of ninety patients, 58 had clear cell renal carcinoma, 15 had papillary, 11 had chromophobe and six had unclassified RCC. FAK, Src and paxillin were expressed in 55.6%, 32.2% and 18.9% of all cases, respectively. In univariate analysis, FAK positivity and IRS were more likely in patients with papillary and chromophobe histologic type versus clear cell RCC (p<0.005), Src positivity and IRS presented more frequently in stage T3 versus T1 (p<0.005) and paxillin positivity was more likely in patients with stage T3 versus T2 (p=0.021) and grades 3-4 versus grade 2 (p=0.013). Paxillin-IRS was not associated with any clinicopathological features. The same associations were also reproduced in the multifactorial analysis for the FAK and Src positivity and IRS, while it was found that paxillin positivity and IRS were associated with the female gender (p=0.052, p=0.024), and were higher in grades 3-4 versus grade 2 (p=0.022, p=0.020). CONCLUSIONS Our study suggests that RCC shows immunohistochemical expression of FAK, Src and paxillin proteins, and this expression varies in relation to the histologic type, the stage and the stage/grade/gender, respectively. These findings imply a possible involvement of the FAK/Src signalling pathway in the pathogenesis and progression of cancer in RCC, providing future perspectives for targeted therapies with inhibitors.
Collapse
Affiliation(s)
| | - Ioannis Anastasiou
- First Department of Urology, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, Athens, GRC
| | - Constantinos Constantinides
- First Department of Urology, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, Athens, GRC
| | | | - Ioannis Katafigiotis
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros, The Athens Clinic, Athens, GRC
| | | | - Vasiliki Tzotzola
- Department of Pediatric Hematology-Oncology, Agia Sofia Children's Hospital, Athens, GRC
| | - Dionysios Mitropoulos
- First Department of Urology, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, Athens, GRC
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, GRC
| |
Collapse
|
2
|
Guo Y, Jiang L, Luo S, Hu D, Zhao X, Zhao G, Tang W. Network Analysis and Basic Experiments on the Inhibition of Renal Cancer Proliferation and Migration by Alpinetin through PI3K/AKT/ mTOR Pathway. Curr Mol Med 2024; 24:134-144. [PMID: 37221689 DOI: 10.2174/1566524023666230522145226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 05/25/2023]
Abstract
BACKGROUND Alpinetin, a natural flavonoid, has been shown to have anticancer effects on many tumors. This study investigated the antitumor effect of alpinetin on renal clear cell carcinoma (ccRCC). METHODS Network Pharmacology analysis was carried out on the targets and molecular mechanisms of alpinetin treating ccRCC. The Annexin V PE/7-AAD kit was used to detect apoptosis. Flow cytometry and Cell Counting Kit-8 (CCK-8) were used to detect cell proliferation and cycle. A 24-well transwell chamber and the ibidi scratch insertion performed cell migration analysis. The protein expression of the target molecule was detected by Western blotting. Nude mouse tumorigenesis assays were used to determine the in vivo antitumor effects of alpinetin. RESULTS The network pharmacology revealed that GAPDH, HRAS, SRC, EGFR, and AKT1 are the main targets of alpinetin in treating ccRCC, with the PI3K/AKT signaling pathway being the main pathway of action. We found that alpinetin could significantly inhibit the proliferation and migration of ccRCC cells by inducing apoptosis. In addition, alpinetin also inhibited the cycle progression of ccRCC cells by blocking them in the G1 phase. Furthermore, in vivo and in vitro, alpinetin could inhibit the activation of an important pathway involved in the proliferation and migration of ccRCC cells, namely the PI3K/Akt pathway. CONCLUSION Alpinetin can inhibit the growth of ccRCC cells by inhibiting the activation of the PI3K/Akt pathway and can be a potential anti-cancer drug for ccRCC.
Collapse
Affiliation(s)
- Yu Guo
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Li Jiang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Shengjun Luo
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Daixing Hu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Xin Zhao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Guozhi Zhao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Wei Tang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| |
Collapse
|
3
|
Ortiz MA, Mikhailova T, Li X, Porter BA, Bah A, Kotula L. Src family kinases, adaptor proteins and the actin cytoskeleton in epithelial-to-mesenchymal transition. Cell Commun Signal 2021; 19:67. [PMID: 34193161 PMCID: PMC8247114 DOI: 10.1186/s12964-021-00750-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/14/2021] [Indexed: 12/20/2022] Open
Abstract
Over a century of scientific inquiry since the discovery of v-SRC but still no final judgement on SRC function. However, a significant body of work has defined Src family kinases as key players in tumor progression, invasion and metastasis in human cancer. With the ever-growing evidence supporting the role of epithelial-mesenchymal transition (EMT) in invasion and metastasis, so does our understanding of the role SFKs play in mediating these processes. Here we describe some key mechanisms through which Src family kinases play critical role in epithelial homeostasis and how their function is essential for the propagation of invasive signals. Video abstract.
Collapse
Affiliation(s)
- Maria A. Ortiz
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, USA
- Department of Urology, SUNY Upstate Medical University, Syracuse, USA
| | - Tatiana Mikhailova
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, USA
| | - Xiang Li
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, USA
- Department of Urology, SUNY Upstate Medical University, Syracuse, USA
| | - Baylee A. Porter
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, USA
- Department of Urology, SUNY Upstate Medical University, Syracuse, USA
| | - Alaji Bah
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, USA
| | - Leszek Kotula
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, USA
- Department of Urology, SUNY Upstate Medical University, Syracuse, USA
| |
Collapse
|
4
|
Gargiuli C, Sepe P, Tessari A, Sheetz T, Colecchia M, de Braud FGM, Procopio G, Sensi M, Verzoni E, Dugo M. Integrative Transcriptomic Analysis Reveals Distinctive Molecular Traits and Novel Subtypes of Collecting Duct Carcinoma. Cancers (Basel) 2021; 13:2903. [PMID: 34200770 PMCID: PMC8230422 DOI: 10.3390/cancers13122903] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
Collecting duct carcinoma (CDC) is a rare and highly aggressive kidney cancer subtype with poor prognosis and no standard treatments. To date, only a few studies have examined the transcriptomic portrait of CDC. Through integration of multiple datasets, we compared CDC to normal tissue, upper-tract urothelial carcinomas, and other renal cancers, including clear cell, papillary, and chromophobe histologies. Association between CDC gene expression signatures and in vitro drug sensitivity data was evaluated using the Cancer Therapeutic Response Portal, Genomics of Drug Sensitivity in Cancer datasets, and connectivity map. We identified a CDC-specific gene signature that predicted in vitro sensitivity to different targeted agents and was associated to worse outcome in clear cell renal cell carcinoma. We showed that CDC are transcriptionally related to the principal cells of the collecting ducts providing evidence that this tumor originates from this normal kidney cell type. Finally, we proved that CDC is a molecularly heterogeneous disease composed of at least two subtypes distinguished by cell signaling, metabolic and immune-related alterations. Our findings elucidate the molecular features of CDC providing novel biological and clinical insights. The identification of distinct CDC subtypes and their transcriptomic traits provides the rationale for patient stratification and alternative therapeutic approaches.
Collapse
Affiliation(s)
- Chiara Gargiuli
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Pierangela Sepe
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (P.S.); (F.G.M.d.B.); (G.P.); (E.V.)
| | - Anna Tessari
- Department of Cancer Biology and Genetics, College of Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.T.); (T.S.)
| | - Tyler Sheetz
- Department of Cancer Biology and Genetics, College of Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.T.); (T.S.)
- Department of Urology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Maurizio Colecchia
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Filippo Guglielmo Maria de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (P.S.); (F.G.M.d.B.); (G.P.); (E.V.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20133 Milan, Italy
| | - Giuseppe Procopio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (P.S.); (F.G.M.d.B.); (G.P.); (E.V.)
| | - Marialuisa Sensi
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Elena Verzoni
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (P.S.); (F.G.M.d.B.); (G.P.); (E.V.)
| | - Matteo Dugo
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| |
Collapse
|
5
|
Kulkarni P, Dasgupta P, Hashimoto Y, Shiina M, Shahryari V, Tabatabai ZL, Yamamura S, Tanaka Y, Saini S, Dahiya R, Majid S. A lncRNA TCL6-miR-155 Interaction Regulates the Src-Akt-EMT Network to Mediate Kidney Cancer Progression and Metastasis. Cancer Res 2021; 81:1500-1512. [PMID: 33500248 DOI: 10.1158/0008-5472.can-20-0832] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/10/2020] [Accepted: 01/20/2021] [Indexed: 11/16/2022]
Abstract
Metastasis is the leading cause of mortality from kidney cancer, and understanding the underlying mechanism of this event will provide better strategies for its management. Here we investigated the biological, functional, and clinical significance of lncTCL6 and its interacting miR-155 in clear cell renal cell carcinoma (ccRCC). We employed a comprehensive approach to investigate the lncTCL6-miR-155-Src/Akt-mediated epithelial-to-mesenchymal transition (EMT) pathway as a novel regulatory mechanism in ccRCC progression. Expression analyses revealed that lncTCL6 is downregulated in ccRCC compared with normal tissues. Overexpression of lncTCL6 in ccRCC cell lines impaired their oncogenic functions, such as cell proliferation and migration/invasion, and induced cell-cycle arrest and apoptosis; conversely, depletion of lncTCL6 rescued these phenotypic effects. Furthermore, lncTCL6 directly interacted with miR-155. Unlike lncTCL6, miR-155 was overexpressed in ccRCC. Stable knockdown of miR-155 phenocopied the effects of lncTCL6 overexpression. Conversely, reconstitution of miR-155 and suppression of lncTCL6 in noncancerous renal cell HK2 induced tumorigenic characteristics. Patients with higher expression of lncTCL6 and lower expression of miR-155 had better survival probability. When overexpressed, lncTCL6 recruited STAU1 and mediated decay of Src mRNA, followed by a marked downregulation of an integrated network of Src target genes involved in migration, invasion, and EMT. However, the interaction between miR-155 and lncTCL6 attenuated the regulatory role of lncTCL6 on Src-mediated EMT. In conclusion, this study is the first report documenting the lncTCL6-miR155-Src/Akt/EMT network as a novel regulatory mechanism in aggressive ccRCC and a promising therapeutic target to inhibit renal cancer. SIGNIFICANCE: This study's investigation of noncoding RNA interactions in renal cell carcinoma identify miRNA-155-lncRNA TCL6-mediated regulation of the Src-Akt-EMT network as a novel mechanism of disease progression and metastasis.
Collapse
Affiliation(s)
- Priyanka Kulkarni
- Department of Urology, Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, California
| | - Pritha Dasgupta
- Department of Urology, Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, California
| | - Yutaka Hashimoto
- Department of Urology, Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, California
| | - Marisa Shiina
- Department of Urology, Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, California
| | - Varahram Shahryari
- Department of Urology, Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, California
| | - Z Laura Tabatabai
- Department of Urology, Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, California
| | - Soichiro Yamamura
- Department of Urology, Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, California
| | - Yuichiro Tanaka
- Department of Urology, Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, California
| | - Sharanjot Saini
- Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Rajvir Dahiya
- Department of Urology, Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, California.
| | - Shahana Majid
- Department of Urology, Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, California.
| |
Collapse
|
6
|
Li Q, Zhang Z, Fan Y, Zhang Q. Epigenetic Alterations in Renal Cell Cancer With TKIs Resistance: From Mechanisms to Clinical Applications. Front Genet 2021; 11:562868. [PMID: 33510766 PMCID: PMC7835797 DOI: 10.3389/fgene.2020.562868] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
The appearance of tyrosine kinase inhibitors (TKIs) has been a major breakthrough in renal cell carcinoma (RCC) therapy. Unfortunately, a portion of patients with TKIs resistance experience disease progression after TKIs therapy. Epigenetic alterations play an important role in the development of TKIs resistance. Current evidence suggests that epigenetic alterations occur frequently in RCC patients with poor response to TKIs therapy, and modulation of them could enhance the cytotoxic effect of antitumor therapy. In this review, we summarize the currently known epigenetic alterations relating to TKIs resistance in RCC, focusing on DNA methylation, non-coding RNAs (ncRNAs), histone modifications, and their interactions with TKIs treatment. In addition, we discuss application of epigenetic alteration analyses in the clinical setting to predict prognosis of patients with TKIs treatment, and the potential use of epigenetics-based therapies to surmount TKIs resistance.
Collapse
Affiliation(s)
- Qinhan Li
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Zhenan Zhang
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Yu Fan
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Qian Zhang
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| |
Collapse
|
7
|
Indovina P, Forte IM, Pentimalli F, Giordano A. Targeting SRC Family Kinases in Mesothelioma: Time to Upgrade. Cancers (Basel) 2020; 12:cancers12071866. [PMID: 32664483 PMCID: PMC7408838 DOI: 10.3390/cancers12071866] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/01/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022] Open
Abstract
Malignant mesothelioma (MM) is a deadly tumor mainly caused by exposure to asbestos. Unfortunately, no current treatment is able to change significantly the natural history of the disease, which has a poor prognosis in the majority of patients. The non-receptor tyrosine kinase SRC and other SRC family kinase (SFK) members are frequently hyperactivated in many cancer types, including MM. Several works have indeed suggested that SFKs underlie MM cell proliferation, survival, motility, and invasion, overall affecting multiple oncogenic pathways. Consistently, SFK inhibitors effectively counteracted MM cancerous features at the preclinical level. Dasatinib, a multi-kinase inhibitor targeting SFKs, was also assessed in clinical trials either as second-line treatment for patients with unresectable MM or, more recently, as a neoadjuvant agent in patients with resectable MM. Here, we provide an overview of the molecular mechanisms implicating SFKs in MM progression and discuss possible strategies for a more successful clinical application of SFK inhibitors. Our aim is to stimulate discussion and further consideration of these agents in better designed preclinical and clinical studies to make the most of another class of powerful antitumoral drugs, which too often are lost in translation when applied to MM.
Collapse
Affiliation(s)
- Paola Indovina
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
- Institute for High Performance Computing and Networking, National Research Council of Italy (ICAR-CNR), I-80131 Naples, Italy
- Correspondence: (P.I.); (F.P.)
| | - Iris Maria Forte
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, I-80131 Naples, Italy;
| | - Francesca Pentimalli
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, I-80131 Naples, Italy;
- Correspondence: (P.I.); (F.P.)
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
- Department of Medical Biotechnologies, University of Siena, I-53100 Siena, Italy
| |
Collapse
|
8
|
Li Z, Yu Y, Wang M, Xu H, Han B, Jiang P, Ma H, Li Y, Tian C, Zhou D, Li X, Ye X. Anti-breast Cancer Activity of SPG-56 from Sweet Potato in MCF-7 Bearing Mice in Situ through Promoting Apoptosis and Inhibiting Metastasis. Sci Rep 2019; 9:146. [PMID: 30651572 PMCID: PMC6335419 DOI: 10.1038/s41598-018-29099-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 07/01/2018] [Indexed: 01/19/2023] Open
Abstract
SPG-56 is a newly isolated glycoprotein from sweet potatoes (Zhongshu NO. 1), but its value for suppressing breast cancer progression remains unknown. This study was designed to investigate the potential anti-cancer effects of SPG-56, which consists of 2.9% sugar and 97.1% protein. The effects of SPG-56 on the proliferation and apoptosis of breast cancer cells were determined using CCK-8 and Hoechst 33342 assays and flow cytometry, after staining with Annexin V and PI respectively. The activities of SPG-56 against breast cancer were examined using female BALB/c nude mice orthotopically implanted with human breast carcinoma cells of the types MCF-7 and 4T1-Luc. The cellular experiments showed that SPG-56 inhibited proliferation and promoted apoptosis of MCF-7 cells dose- and time-dependently. Oral administration of SPG-56 significantly suppressed the development of MCF-7 tumor cells (P < 0.01) as compared with an untreated group. The serum tumor markers CEA, CA125 and CA153 in a 240 mg/kg/d SPG-56 decreased by 54.8%, 91.8%, and 90.3%, respectively. The experiments further demonstrated that SPG-56 inhibited the metastasis of breast cancer in MCF-7 and 4T1-bearing mice by altering the expression of MMP2, MMP9, VEGF, Occludin and Claudin. It is concluded that SPG-56 may have potential as a novel anti-tumor candidate for breast cancer.
Collapse
Affiliation(s)
- Zhaoxing Li
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China.,McLean Hospital, Harvard Medical School, Belmont, 02478, MA, USA
| | - Yang Yu
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China
| | - Meimei Wang
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China
| | - Heshan Xu
- Chongqing Engineering Research Centre for Sweet Potato, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Bing Han
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China
| | - Pu Jiang
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China
| | - Hang Ma
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China
| | - Yuanfeng Li
- Chongqing Engineering Research Centre for Sweet Potato, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Cheng Tian
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China
| | - Deqi Zhou
- Oncology Department, Chongqing Beibei District Hospital of Traditional Chinese Medicine, Chongqing, 400700, China
| | - Xuegang Li
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China.
| | - Xiaoli Ye
- Chongqing Engineering Research Centre for Sweet Potato, School of Life Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
9
|
Roelants C, Giacosa S, Pillet C, Bussat R, Champelovier P, Bastien O, Guyon L, Arnoux V, Cochet C, Filhol O. Combined inhibition of PI3K and Src kinases demonstrates synergistic therapeutic efficacy in clear-cell renal carcinoma. Oncotarget 2018; 9:30066-30078. [PMID: 30046388 PMCID: PMC6059021 DOI: 10.18632/oncotarget.25700] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 06/12/2018] [Indexed: 12/13/2022] Open
Abstract
Potent inhibitors of PI3K (GDC-0941) and Src (Saracatinib) exhibit as individual agents, excellent oral anticancer activity in preclinical models and have entered phase II clinical trials in various cancers. We found that PI3K and Src kinases are dysregulated in clear cell renal carcinomas (ccRCCs), an aggressive disease without effective targeted therapies. In this study we addressed this challenge by testing GDC-0941 and Saracatinib as either single agents or in combination in ccRCC cell lines, as well as in mouse and PDX models. Our findings demonstrate that combined inhibition of PI3K and Src impedes cell growth and invasion and induces cell death of renal carcinoma cells providing preclinical evidence for a pairwise combination of these anticancer drugs as a rational strategy to improve renal cancer treatment.
Collapse
Affiliation(s)
- Caroline Roelants
- Université Grenoble-Alpes, Inserm U1036, CEA, BIG-BCI, Grenoble, France.,Inovarion, Paris, France
| | - Sofia Giacosa
- Université Grenoble-Alpes, Inserm U1036, CEA, BIG-BCI, Grenoble, France
| | - Catherine Pillet
- Université Grenoble-Alpes, Inserm U1038, CEA, BIG-BGE, Grenoble, France
| | - Rémi Bussat
- Université Grenoble-Alpes, Inserm U1036, CEA, BIG-BCI, Grenoble, France
| | | | - Olivier Bastien
- Université Grenoble-Alpes, CNRS-CEA-INRA, Laboratoire de Physiologie Cellulaire et Végétale, Grenoble, France
| | - Laurent Guyon
- Université Grenoble-Alpes, Inserm U1036, CEA, BIG-BCI, Grenoble, France
| | - Valentin Arnoux
- Centre Hospitalier Université Grenoble-Alpes, CS 10217, Grenoble, France
| | - Claude Cochet
- Université Grenoble-Alpes, Inserm U1036, CEA, BIG-BCI, Grenoble, France
| | - Odile Filhol
- Université Grenoble-Alpes, Inserm U1036, CEA, BIG-BCI, Grenoble, France
| |
Collapse
|
10
|
Association of Expression Levels or Activation Status of STAT3 with Treatment Outcomes of Sunitinib in Patients with Renal Cell Carcinoma. Target Oncol 2018; 13:371-378. [DOI: 10.1007/s11523-018-0563-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
11
|
Adelaiye-Ogala R, Damayanti NP, Orillion AR, Arisa S, Chintala S, Titus MA, Kao C, Pili R. Therapeutic Targeting of Sunitinib-Induced AR Phosphorylation in Renal Cell Carcinoma. Cancer Res 2018; 78:2886-2896. [PMID: 29572225 DOI: 10.1158/0008-5472.can-17-3386] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/27/2018] [Accepted: 03/20/2018] [Indexed: 12/11/2022]
Abstract
Androgen receptor (AR) plays a crucial role in the development and progression of prostate cancer. AR expression has also been reported in other solid tumors, including renal cell carcinoma (RCC), but its biological role here remains unclear. Through integrative analysis of a reverse phase protein array, we discovered increased expression of AR in an RCC patient-derived xenograft model of acquired resistance to the receptor tyrosine kinase inhibitor (RTKi) sunitinib. AR expression was increased in RCC cell lines with either acquired or intrinsic sunitinib resistance in vitro An AR signaling gene array profiler indicated elevated levels of AR target genes in sunitinib-resistant cells. Sunitinib-induced AR transcriptional activity was associated with increased phosphorylation of serine 81 (pS81) on AR. Additionally, AR overexpression resulted in acquired sunitinib resistance and the AR antagonist enzalutamide-induced AR degradation and attenuated AR downstream activity in sunitinib-resistant cells, also indicated by decreased secretion of human kallikrein 2. Enzalutamide-induced AR degradation was rescued by either proteasome inhibition or by knockdown of the AR ubiquitin ligase speckle-type POZ protein (SPOP). In vivo treatment with enzalutamide and sunitinib demonstrated that this combination efficiently induced tumor regression in a RCC model following acquired sunitinib resistance. Overall, our results suggest the potential role of AR as a target for therapeutic interventions, in combination with RTKi, to overcome drug resistance in RCC.Significance: These findings highlight the therapeutic potential of targeting the androgen receptor to overcome RCC resistance to receptor tyrosine kinase inhibitors. Cancer Res; 78(11); 2886-96. ©2018 AACR.
Collapse
Affiliation(s)
- Remi Adelaiye-Ogala
- Genitourinary Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana.,Department of Cancer Pathology and Prevention, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - Nur P Damayanti
- Genitourinary Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Ashley R Orillion
- Genitourinary Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana.,Department of Cellular and Molecular Biology, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - Sreevani Arisa
- Genitourinary Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Sreenivasulu Chintala
- Genitourinary Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Mark A Titus
- Department of Genitourinary Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chinghai Kao
- Department of Urology, Indiana University, Indianapolis, Indiana
| | - Roberto Pili
- Genitourinary Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana. .,Department of Urology, Indiana University, Indianapolis, Indiana
| |
Collapse
|
12
|
Antcin-H Isolated from Antrodia cinnamomea Inhibits Renal Cancer Cell Invasion Partly through Inactivation of FAK-ERK-C/EBP- β/c-Fos-MMP-7 Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:5052870. [PMID: 29234409 PMCID: PMC5688354 DOI: 10.1155/2017/5052870] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/29/2017] [Accepted: 10/09/2017] [Indexed: 12/19/2022]
Abstract
Antcin-H, a natural triterpene, is purified from a famous anticancer medicinal mushroom, Antrodia cinnamomea, in Taiwan. This study showed that antcin-H inhibited the growth of human renal carcinoma 786-0 cells; the IC50 value (for 48 h) was 170 μM. Besides, the migration and invasion of 786-0 cells were suppressed by antcin-H under noncytotoxic concentrations (<100 μM); these events were accompanied by inhibition of FAK and Src kinase activities, decrease of paxillin phosphorylation, impairment of lamellipodium formation, and upregulation of TIMPs and downregulation of MMPs, especially MMP-7 expression. Luciferase reporter assay showed that antcin-H repressed the MMP-7 promoter activity, in parallel to inhibiting c-Fos/AP-1 and C/EBP-β transactivation abilities. Moreover, antcin-H suppressed the activity of ERK1/2 and decreased the binding ability of C/EBP-β and c-Fos on the upstream/enhancer region of MMP-7 promoter. Overall, this study demonstrated that the anti-invasive effect of antcin-H in human renal carcinoma 786-0 cells might be at least in part by abrogating focal adhesion complex and lamellipodium formation through inhibiting the Src/FAK-paxillin signaling pathways and decreasing MMP-7 expression through suppressing the ERK1/2-AP-1/c-Fos and C/EBP-β signaling axis. Our findings provide the evidence that antcin-H may be an active component existing in A. cinnamomea with anticancer effect.
Collapse
|
13
|
Sato H, Uzu M, Kashiba T, Suzuki R, Fujiwara T, Okuzawa H, Ueno K. Sodium butyrate enhances the growth inhibitory effect of sunitinib in human renal cell carcinoma cells. Oncol Lett 2017; 14:937-943. [PMID: 28693255 DOI: 10.3892/ol.2017.6217] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 03/14/2017] [Indexed: 12/12/2022] Open
Abstract
Sunitinib (SU) is a small molecule that inhibits the receptor tyrosine kinase (RTK) signaling pathway, and has been clinically used to treat advanced renal cell carcinoma (RCC). However, SU is not always effective as RCC is a highly chemoresistant type of cancer. One of the factors that confer chemoresistance to RCC is a hypoxic condition. Lack of oxygen activates hypoxia-inducible factor (HIF) protein, which is followed by the upregulation of growth factors, including vascular endothelial growth factor and activation of the RTK signaling pathway. In this context, histone deacetylase inhibitors (HDACIs) are considered prominent combined agents for SU as they downregulate the expression of HIFs. Therefore, the present study aimed to investigate the effectiveness of combined treatment with SU and sodium butyrate (NaBu), an HDACI. Long-term exposure to these agents exerted a stronger growth inhibitory effect in RCC cell lines compared with single treatment groups. Furthermore, combined treatment suppressed HIF-2α protein, which was induced under hypoxic conditions. In addition, this combination sustained the activity of the RTK signaling pathway to the level of intact cells, although a single treatment with SU or NaBu was demonstrated to increase this activity. Overall, it is suggested that the combination of SU and NaBu is effective for overcoming drug resistance in RCC.
Collapse
Affiliation(s)
- Hiromi Sato
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chuo-ku, Chiba 260-8675, Japan
| | - Miaki Uzu
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chuo-ku, Chiba 260-8675, Japan
| | - Tatsuro Kashiba
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chuo-ku, Chiba 260-8675, Japan
| | - Rina Suzuki
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chuo-ku, Chiba 260-8675, Japan
| | - Takuya Fujiwara
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chuo-ku, Chiba 260-8675, Japan
| | - Hiroko Okuzawa
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chuo-ku, Chiba 260-8675, Japan
| | - Koichi Ueno
- Center of Preventive Medical Science, Chiba University, Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
14
|
Vallard A, Trone JC, Langrand-Escure J, Espenel S, Guy JB, Rancoule C, Xia Y, El Meddeb Hamrouni A, Ben Mrad M, Magné N. The world of targeted therapies in kidney cancers: pitfalls, tips and tricks. Onco Targets Ther 2017; 10:1375-1380. [PMID: 28424553 PMCID: PMC5344426 DOI: 10.2147/ott.s127919] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In the past few years, metastatic renal cell carcinoma prognosis was improved by the development of molecular targeted therapies (TTs). At the metastatic stage, the tolerance to treatment is a major concern, not only because of the challenge of the efficacy/toxicity ratio improvement but also because of the importance of an optimal adherence to oral treatments. The present case series relates the issues of dealing with uncommon and sometimes never described side effects of sunitinib and sorafenib. The first case report deals with grade 3 vomiting during hemodialysis with concurrent administration of sunitinib. The second case is an iterative gout attack induced by sunitinib. The third case presents a grade 3 scalp dysesthesia with sorafenib. The fourth case includes an astonishing efficacy of metronomic (ie, low doses during a long period of time) bevacizumab in monotherapy. Multidisciplinary management and systematic reporting of unexpected efficacies and toxicities are needed to better understand TTs real therapeutic index. Although TTs revolutionized metastatic renal cell cancer prognosis, they also brought about previously unknown side effects. Identification and management of these off-target effects may be tricky, and therefore, comedication must be wisely chosen. As the physiopathology of these side effects is still unclear, multidisciplinary management and systematic reporting of toxicities are essential.
Collapse
Affiliation(s)
- Alexis Vallard
- Department of Radiation Oncology, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Jane-Chloé Trone
- Department of Radiation Oncology, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Julien Langrand-Escure
- Department of Radiation Oncology, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Sophie Espenel
- Department of Radiation Oncology, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Jean-Baptiste Guy
- Department of Radiation Oncology, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Chloé Rancoule
- Department of Radiation Oncology, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Yaoxiong Xia
- Department of Radiation Oncology, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Anis El Meddeb Hamrouni
- Department of Radiation Oncology, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Majed Ben Mrad
- Department of Radiation Oncology, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| | - Nicolas Magné
- Department of Radiation Oncology, Lucien Neuwirth Cancer Institute, Saint-Priest en Jarez, France
| |
Collapse
|
15
|
van der Mijn JC, Broxterman HJ, Knol JC, Piersma SR, De Haas RR, Dekker H, Pham TV, Van Beusechem VW, Halmos B, Mier JW, Jiménez CR, Verheul HMW. Sunitinib activates Axl signaling in renal cell cancer. Int J Cancer 2016; 138:3002-10. [PMID: 26815723 DOI: 10.1002/ijc.30022] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 12/01/2015] [Accepted: 01/14/2016] [Indexed: 12/14/2022]
Abstract
Mass spectrometry-based phosphoproteomics provides a unique unbiased approach to evaluate signaling network in cancer cells. The tyrosine kinase inhibitor sunitinib is registered as treatment for patients with renal cell cancer (RCC). We investigated the effect of sunitinib on tyrosine phosphorylation in RCC tumor cells to get more insight in its mechanism of action and thereby to find potential leads for combination treatment strategies. Sunitinib inhibitory concentrations of proliferation (IC50) of 786-O, 769-p and A498 RCC cells were determined by MTT-assays. Global tyrosine phosphorylation was measured by LC-MS/MS after immunoprecipitation with the antiphosphotyrosine antibody p-TYR-100. Phosphoproteomic profiling of 786-O cells yielded 1519 phosphopeptides, corresponding to 675 unique proteins including 57 different phosphorylated protein kinases. Compared to control, incubation with sunitinib at its IC50 of 2 µM resulted in downregulation of 86 phosphopeptides including CDK5, DYRK3, DYRK4, G6PD, PKM and LDH-A, while 94 phosphopeptides including Axl, FAK, EPHA2 and p38α were upregulated. Axl- (y702), FAK- (y576) and p38α (y182) upregulation was confirmed by Western Blot in 786-O and A498 cells. Subsequent proliferation assays revealed that inhibition of Axl with a small molecule inhibitor (R428) sensitized 786-O RCC cells and immortalized endothelial cells to sunitinib up to 3 fold. In conclusion, incubation with sunitinib of RCC cells causes significant upregulation of multiple phosphopeptides including Axl. Simultaneous inhibition of Axl improves the antitumor activity of sunitinib. We envision that evaluation of phosphoproteomic changes by TKI treatment enables identification of new targets for combination treatment strategies.
Collapse
Affiliation(s)
- Johannes C van der Mijn
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands.,Department of Hematology/Oncology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Henk J Broxterman
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Jaco C Knol
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Sander R Piersma
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Richard R De Haas
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Henk Dekker
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Thang V Pham
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Victor W Van Beusechem
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Balazs Halmos
- Department of Hematology/Oncology, Columbia University Medical Center, New York, NY
| | - James W Mier
- Department of Hematology/Oncology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Connie R Jiménez
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Powles T, Brown J, Larkin J, Jones R, Ralph C, Hawkins R, Chowdhury S, Boleti E, Bhal A, Fife K, Webb A, Crabb S, Geldart T, Hill R, Dunlop J, Hall PE, McLaren D, Ackerman C, Beltran L, Nathan P. A randomized, double-blind phase II study evaluating cediranib versus cediranib and saracatinib in patients with relapsed metastatic clear-cell renal cancer (COSAK). Ann Oncol 2016; 27:880-6. [PMID: 26802156 DOI: 10.1093/annonc/mdw014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 01/06/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Preclinical work suggests SRC proteins have a role in the development of resistance to vascular endothelial growth factor (VEGF) targeted therapy in metastatic clear-cell renal cancer (mRCC). This hypothesis was tested in this trial using the SRC inhibitor saracatinib and the VEGF inhibitor cediranib. PATIENTS AND METHODS Patients with disease progression after ≥1 VEGF-targeted therapy were eligible to participate in this double-blind, randomized (1:1) phase II study. The study compared the combination cediranib 30 mg once daily (o.d.) and saracatinib 175 mg o.d. (CS) (n = 69) or cediranib 45 mg o.d. and placebo o.d. (C) (n = 69). Archived tissue was used for biomarker analysis [SRC, focal adhesion kinase (FAK), von Hippel-Lindau, protein tyrosine phosphatase 1b and hypoxia-inducible factor 2α : n = 86]. The primary end point was progression-free survival (PFS) by RECIST v1.1. RESULTS Between 2010 and 2012, 138 patients were randomized across 16 UK sites. The characteristics of the two groups were well balanced. Partial responses were seen in 13.0% for C and 14.5% for CS (P > 0.05). There was no significant difference in PFS [5.4 months (3.6-7.3 months) for C and 3.9 (2.4-5.3 months) for CS; hazard ratio (HR) 1.18 (0.94-1.48)] or overall survival (OS) [14.2 months (11.2-16.8 months) for C and 10.0 (6.7-13.2 months) for CS; HR 1.28 (1.00-1.63)]. There was no significant difference in the frequency of key adverse events, dose reductions or drug discontinuations. None of the biomarkers were prognostic for PFS or OS. FAK overexpression correlated with an OS benefit [HR 2.29 (1.09-4.82), P > 0.05], but not PFS, for CS. CONCLUSIONS Saracatinib did not increase the efficacy of a VEGF-targeted therapy (cediranib) in this setting. Biomarker analysis did not identify consistent predictive biomarkers. CLINICALTRIALSGOV NCT00942877.
Collapse
Affiliation(s)
- T Powles
- Department of Medical Oncology, The Royal Free NHS Foundation Trust, London Barts Cancer Institute, CRUK Experiment Cancer Medicine Centre, London
| | - J Brown
- Department of Medical Oncology, University of Sheffield, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield Department of Medical Oncology, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, West Yorkshire
| | - J Larkin
- Department of Medical Oncology, The Royal Marsden Hospital, London
| | - R Jones
- The Beatson Cancer Centre, University of Glasgow, Glasgow
| | - C Ralph
- Department of Medical Oncology, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, West Yorkshire
| | - R Hawkins
- Department of Medical Oncology, The Christie Hospital, Manchester
| | - S Chowdhury
- Department of Medical Oncology, Guys and St Thomas' NHS Foundation Trust, London
| | - E Boleti
- Department of Medical Oncology, The Royal Free NHS Foundation Trust, London
| | - A Bhal
- Department of Oncology, University Hospital Bristol NHS Foundation trust, Bristol
| | - K Fife
- Addenbrooke's Cancer Centre, University of Cambridge, Cambridge
| | - A Webb
- Department of Medical Oncology, West Sussex Cancer Centre, Brighton
| | - S Crabb
- Cancer Sciences Unit, Southampton NHS Foundation Trust, Southampton
| | - T Geldart
- Department of Medical Oncology, Royal Bournemouth Hospital, Bournemouth
| | - R Hill
- Scottish Clinical Trials Research Unit (SCTRU), Partners in CaCTUS, Edinburgh
| | - J Dunlop
- Scottish Clinical Trials Research Unit (SCTRU), Partners in CaCTUS, Edinburgh
| | - P E Hall
- Barts Cancer Institute, CRUK Experiment Cancer Medicine Centre, London
| | - D McLaren
- Edinburgh Cancer Centre, Western General Hospital, Edinburgh
| | - C Ackerman
- Barts Cancer Institute, CRUK Experiment Cancer Medicine Centre, London
| | - L Beltran
- Barts Cancer Institute, CRUK Experiment Cancer Medicine Centre, London
| | - P Nathan
- Department of Medical Oncology, Mount Vernon Cancer Centre, Northwood, UK
| |
Collapse
|
17
|
Poh AR, O'Donoghue RJ, Ernst M. Hematopoietic cell kinase (HCK) as a therapeutic target in immune and cancer cells. Oncotarget 2015; 6:15752-71. [PMID: 26087188 PMCID: PMC4599235 DOI: 10.18632/oncotarget.4199] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/29/2015] [Indexed: 12/21/2022] Open
Abstract
The hematopoietic cell kinase (HCK) is a member of the SRC family of cytoplasmic tyrosine kinases (SFKs), and is expressed in cells of the myeloid and B-lymphocyte cell lineages. Excessive HCK activation is associated with several types of leukemia and enhances cell proliferation and survival by physical association with oncogenic fusion proteins, and with functional interactions with receptor tyrosine kinases. Elevated HCK activity is also observed in many solid malignancies, including breast and colon cancer, and correlates with decreased patient survival rates. HCK enhances the secretion of growth factors and pro-inflammatory cytokines from myeloid cells, and promotes macrophage polarization towards a wound healing and tumor-promoting alternatively activated phenotype. Within tumor associated macrophages, HCK stimulates the formation of podosomes that facilitate extracellular matrix degradation, which enhance immune and epithelial cell invasion. By virtue of functional cooperation between HCK and bona fide oncogenic tyrosine kinases, excessive HCK activation can also reduce drug efficacy and contribute to chemo-resistance, while genetic ablation of HCK results in minimal physiological consequences in healthy mice. Given its known crystal structure, HCK therefore provides an attractive therapeutic target to both, directly inhibit the growth of cancer cells, and indirectly curb the source of tumor-promoting changes in the tumor microenvironment.
Collapse
Affiliation(s)
- Ashleigh R. Poh
- The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Victoria, Australia
| | - Robert J.J. O'Donoghue
- The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Victoria, Australia
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Victoria, Australia
| | - Matthias Ernst
- The Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Victoria, Australia
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Victoria, Australia
| |
Collapse
|
18
|
Chu Q, Han N, Yuan X, Nie X, Wu H, Chen Y, Guo M, Yu S, Wu K. DACH1 inhibits cyclin D1 expression, cellular proliferation and tumor growth of renal cancer cells. J Hematol Oncol 2014; 7:73. [PMID: 25322986 PMCID: PMC4203876 DOI: 10.1186/s13045-014-0073-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/22/2014] [Indexed: 12/15/2022] Open
Abstract
Background Renal cell carcinoma (RCC) is a complex with diverse biological characteristics and distinct molecular signature. New target therapies to molecules that drive RCC initiation and progression have achieved promising responses in some patients, but the total effective rate is still far from satisfaction. Dachshund (DACH1) network is a key signaling pathway for kidney development and has recently been identified as a tumor suppressor in several cancer types. However, its role in renal cell carcinoma has not been fully investigated. Methods Immunohistochemical staining for DACH1, PCNA and cyclin D1 was performed on human renal tissue microaraays and correlation with clinic-pathological characteristics was analyzed. In vitro proliferation, apoptosis and in vivo tumor growth were evaluated on human renal cancer cell lines with decitabine treatment or ectopic expression of DACH1. Downstream targets and potential molecular mechanism were investigated through western blot, immunoprecipitation and reporter gene assays. Results Expression of DACH1 was significantly decreased in human renal carcinoma tissue. DACH1 protein abundance was inversely correlated with the expression of PCNA and cyclin D1, tumor grade, and TNM stage. Restoration of DACH1 function in renal clear cell cancer cells inhibited in vitro cellular proliferation, S phase progression, clone formation, and in vivo tumor growth. In mechanism, DACH1 repressed cyclin D1 transcription through association with AP-1 protein. Conclusion Our results indicated that DACH1 was a novel molecular marker of RCC and it attributed to the malignant behavior of renal cancer cells. Re-activation of DACH1 may represent a potential therapeutic strategy.
Collapse
Affiliation(s)
- Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430030, China.
| | - Na Han
- Department of Oncology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430030, China.
| | - Xun Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430030, China.
| | - Xin Nie
- Department of Oncology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430030, China.
| | - Hua Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430030, China.
| | - Yu Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430030, China.
| | - Mingzhou Guo
- Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing, 100853, China.
| | - Shiying Yu
- Department of Oncology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430030, China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430030, China.
| |
Collapse
|
19
|
Yamamoto K, Mizumoto A, Nishimura K, Uda A, Mukai A, Yamashita K, Kume M, Makimoto H, Bito T, Nishigori C, Nakagawa T, Hirano T, Hirai M. Association of toxicity of sorafenib and sunitinib for human keratinocytes with inhibition of signal transduction and activator of transcription 3 (STAT3). PLoS One 2014; 9:e102110. [PMID: 25013907 PMCID: PMC4094497 DOI: 10.1371/journal.pone.0102110] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 06/15/2014] [Indexed: 11/30/2022] Open
Abstract
Hand–foot skin reaction is a most common multi-kinase inhibitor-related adverse event. This study aimed to examine whether the toxicity of sorafenib and sunitinib for human keratinocytes was associated with inhibiting signal transduction and activator of transcription 3 (STAT3). We studied whether STAT3 activity affects sorafenib- and sunitinib-induced cell growth inhibition in HaCaT cells by WST-8 assay. Stattic enhanced the cell-growth inhibitory and apoptotic effects of sorafenib and sunitinib. HaCaT cells transfected with constitutively-active STAT3 (STAT3C) were resistant to the sorafenib- and sunitinib-induced cell growth inhibition. STAT3 activity decreased after short-term treatment with sorafenib and sunitinib in a dose-dependent manner and recovered after long-term treatment with sorafenib and sunitinib at low doses. Moreover, the expression of survivin and bcl-2 decreased after treatment with sorafenib and sunitinib was concomitant with variations in STAT3 activity. Sorafenib-induced STAT3 inhibition was mediated by regulation via MAPK pathways in HaCaT cells, while sunitinib-induced STAT3 inhibition was not. Thus, STAT3 activation mediating apoptosis suppressors may be a key factor in sorafenib and sunitinib-induced keratinocyte cytotoxicity.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Department of Pharmacy, Kobe University Hospital, Kobe, Japan
- * E-mail:
| | - Atsushi Mizumoto
- Division of Pharmacokinetics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kohji Nishimura
- Division of Pharmacokinetics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Atsushi Uda
- Department of Pharmacy, Kobe University Hospital, Kobe, Japan
| | - Akira Mukai
- Department of Pharmacy, Kobe University Hospital, Kobe, Japan
| | | | - Manabu Kume
- Department of Pharmacy, Kobe University Hospital, Kobe, Japan
| | - Hiroo Makimoto
- Department of Pharmacy, Kobe University Hospital, Kobe, Japan
| | - Toshinori Bito
- Division of Dermatology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Chikako Nishigori
- Division of Dermatology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tsutomu Nakagawa
- Department of Pharmacy, Kobe University Hospital, Kobe, Japan
- Division of Pharmacokinetics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takeshi Hirano
- Department of Pharmacy, Kobe University Hospital, Kobe, Japan
- Division of Pharmacokinetics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Midori Hirai
- Department of Pharmacy, Kobe University Hospital, Kobe, Japan
- Division of Pharmacokinetics, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
20
|
Shanmugam MK, Rajendran P, Li F, Kim C, Sikka S, Siveen KS, Kumar AP, Ahn KS, Sethi G. Abrogation of STAT3 signaling cascade by zerumbone inhibits proliferation and induces apoptosis in renal cell carcinoma xenograft mouse model. Mol Carcinog 2014; 54:971-85. [PMID: 24797723 DOI: 10.1002/mc.22166] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 03/19/2014] [Accepted: 03/31/2014] [Indexed: 11/10/2022]
Abstract
Persistent activation of signal transducer and activator of transcription 3 (STAT3) is one of the characteristic features of renal cell carcinoma (RCC) and often linked to its deregulated proliferation, survival, and angiogenesis. In the present report, we investigated whether zerumbone, a sesquiterpene, exerts its anticancer effect through modulation of STAT3 activation pathway. The pharmacological effect of zerumbone on STAT3 activation, associated protein kinases and phosphatase, and apoptosis was investigated using both RCC cell lines and xenograft mouse model. We observed that zerumbone suppressed STAT3 activation in a dose- and time-dependent manner in RCC cells. The suppression was mediated through the inhibition of activation of upstream kinases c-Src, Janus-activated kinase 1, and Janus-activated kinase 2. Pervanadate treatment reversed zerumbone-induced downregulation of STAT3, suggesting the involvement of a tyrosine phosphatase. Indeed, we found that zerumbone induced the expression of tyrosine phosphatase SHP-1 that correlated with its ability to inhibit STAT3 activation. Interestingly, deletion of SHP-1 gene by siRNA abolished the ability of zerumbone to inhibit STAT3 activation. The inhibition of STAT3 activation by zerumbone also caused the suppression of the gene products involved in proliferation, survival, and angiogenesis. Finally, when administered i.p., zerumbone inhibited STAT3 activation in tumor tissues and the growth of human RCC xenograft tumors in athymic nu/nu mice without any side effects. Overall, our results suggest for the first time that zerumbone is a novel blocker of STAT3 signaling cascade and thus has an enormous potential for the treatment of RCC and other solid tumors.
Collapse
Affiliation(s)
- Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Peramaiyan Rajendran
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Feng Li
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chulwon Kim
- Department of Oriental Pathology, College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Sakshi Sikka
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Cancer Science Institute of Singapore, Centre for Translational Medicine, Singapore, Singapore
| | - Kodappully Sivaraman Siveen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Cancer Science Institute of Singapore, Centre for Translational Medicine, Singapore, Singapore.,Faculty of Health Sciences, School of Biomedical Sciences, Curtin University, Western Australia, Australia.,Department of Biological Sciences, University of North Texas, Denton, Texas
| | - Kwang Seok Ahn
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Cancer Science Institute of Singapore, Centre for Translational Medicine, Singapore, Singapore
| |
Collapse
|
21
|
Megison ML, Gillory LA, Stewart JE, Nabers HC, Mrozcek-Musulman E, Beierle EA. FAK inhibition abrogates the malignant phenotype in aggressive pediatric renal tumors. Mol Cancer Res 2014; 12:514-26. [PMID: 24464916 DOI: 10.1158/1541-7786.mcr-13-0505] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
UNLABELLED Despite the tremendous advances in the treatment of childhood kidney tumors, there remain subsets of pediatric renal tumors that continue to pose a therapeutic challenge, mainly malignant rhabdoid kidney tumors and nonosseous renal Ewing sarcoma. Children with advanced, metastatic, or relapsed disease have a poor disease-free survival rate. Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase that is important in many facets of tumor development and progression. FAK has been found in other pediatric solid tumors and in adult renal cellular carcinoma, leading to the hypothesis that FAK contributes to pediatric kidney tumors and would affect cellular survival. In the current study, FAK was present and phosphorylated in pediatric kidney tumor specimens. Moreover, the effects of FAK inhibition upon G401 and SK-NEP-1 cell lines were examined using a number of parallel approaches to block FAK, including RNA interference and small-molecule FAK inhibitors. FAK inhibition resulted in decreased cellular survival, invasion and migration, and increased apoptosis. Furthermore, small-molecule inhibition of FAK led to decreased SK-NEP-1 xenograft growth in vivo. These data deepen the knowledge of the tumorigenic process in pediatric renal tumors, and provide desperately needed therapeutic strategies and targets for these rare, but difficult to treat, malignancies. IMPLICATIONS This study provides a fundamental understanding of tumorigenesis in difficult to treat renal tumors and provides an impetus for new avenues of research and potential for novel, targeted therapies.
Collapse
Affiliation(s)
- Michael L Megison
- University of Alabama at Birmingham, 1600 7th Avenue South, Lowder, Room 300, Birmingham, AL 35233.
| | | | | | | | | | | |
Collapse
|
22
|
Phosphorylation of paxillin confers cisplatin resistance in non-small cell lung cancer via activating ERK-mediated Bcl-2 expression. Oncogene 2013; 33:4385-95. [DOI: 10.1038/onc.2013.389] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 07/05/2013] [Accepted: 08/02/2013] [Indexed: 12/21/2022]
|
23
|
STAT3 and HIF1α cooperatively activate HIF1 target genes in MDA-MB-231 and RCC4 cells. Oncogene 2013; 33:1670-9. [PMID: 23604114 DOI: 10.1038/onc.2013.115] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 01/19/2013] [Accepted: 02/13/2013] [Indexed: 12/26/2022]
Abstract
Solid tumors often exhibit simultaneously inflammatory and hypoxic microenvironments. The 'signal transducer and activator of transcription-3' (STAT3)-mediated inflammatory response and the hypoxia-inducible factor (HIF)-mediated hypoxia response have been independently shown to promote tumorigenesis through the activation of HIF or STAT3 target genes and to be indicative of a poor prognosis in a variety of tumors. We report here for the first time that STAT3 is involved in the HIF1, but not HIF2-mediated hypoxic transcriptional response. We show that inhibiting STAT3 activity in MDA-MB-231 and RCC4 cells by a STAT3 inhibitor or STAT3 small interfering RNA significantly reduces the levels of HIF1, but not HIF2 target genes in spite of normal levels of hypoxia-inducible transcription factor 1α (HIF1α) and HIF2α protein. Mechanistically, STAT3 activates HIF1 target genes by binding to HIF1 target gene promoters, interacting with HIF1α protein and recruiting coactivators CREB binding protein (CBP) and p300, and RNA polymerase II (Pol II) to form enhanceosome complexes that contain HIF1α, STAT3, CBP, p300 and RNA Pol II on HIF1 target gene promoters. Functionally, the effect of STAT3 knockdown on proliferation, motility and clonogenic survival of tumor cells in vitro is phenocopied by HIF1α knockdown in hypoxic cells, whereas STAT3 knockdown in normoxic cells also reduces cell proliferation, motility and clonogenic survival. This indicates that STAT3 works with HIF1 to activate HIF1 target genes and to drive HIF1-depedent tumorigenesis under hypoxic conditions, but also has HIF-independent activity in normoxic and hypoxic cells. Identifying the role of STAT3 in the hypoxia response provides further data supporting the effectiveness of STAT3 inhibitors in solid tumor treatment owing to their usefulness in inhibiting both the STAT3 and HIF1 pro-tumorigenic signaling pathways in some cancer types.
Collapse
|
24
|
Tan X, He S, Han Y, Yu Y, Xiao J, Xu D, Wang G, Du Y, Chang W, Yin J, Su T, Hou J, Cao G. Establishment and characterization of clear cell renal cell carcinoma cell lines with different metastatic potential from Chinese patients. Cancer Cell Int 2013; 13:20. [PMID: 23442546 PMCID: PMC3599881 DOI: 10.1186/1475-2867-13-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 12/19/2012] [Indexed: 11/23/2022] Open
Abstract
Abstracts
Collapse
Affiliation(s)
- Xiaojie Tan
- Department of Epidemiology, Second Military Medical University, 800 Xiangyin Rd, Shanghai 200433, China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Cavalloni G, Peraldo-Neia C, Sarotto I, Gammaitoni L, Migliardi G, Soster M, Marchiò S, Aglietta M, Leone F. Antitumor activity of Src inhibitor saracatinib (AZD-0530) in preclinical models of biliary tract carcinomas. Mol Cancer Ther 2012; 11:1528-38. [PMID: 22452946 DOI: 10.1158/1535-7163.mct-11-1020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Biliary tract carcinoma (BTC) has a poor prognosis due to limited treatment options. There is, therefore, an urgent need to identify new targets and to design innovative therapeutic approaches. Among potential candidate molecules, we evaluated the nonreceptor tyrosine kinase Src, observing promising antitumor effects of its small-molecule inhibitor saracatinib in BTC preclinical models. The presence of an active Src protein was investigated by immunohistochemistry in 19 surgical samples from patients with BTC. Upon saracatinib treatment, the phosphorylation of Src and of its downstream transducers was evaluated in the BTC cell lines TFK-1, EGI-1, HuH28, and TGBC1-TKB. The effect of saracatinib on proliferation and migration was analyzed in these same cell lines, and its antitumor activity was essayed in EGI-1 mouse xenografts. Saracatinib-modulated transcriptome was profiled in EGI-1 cells and in tumor samples of the xenograft model. Src was activated in about 80% of the human BTC samples. In cultured BTC cell lines, low-dose saracatinib counteracted the activation of Src and of its downstream effectors, increased the fraction of cells in G(0)-G(1) phase, and inhibited cell migration. At high concentrations (median dose from 2.26-6.99 μmol/L), saracatinib was also capable of inhibiting BTC cell proliferation. In vivo, saracatinib treatment resulted in delayed tumor growth, associated with an impaired vascular network. Here, we provide a demonstration that the targeted inhibition of Src kinase by saracatinib is of therapeutic benefit in preclinical models of BTC. We propose our results as a basis for the design of saracatinib-based clinical applications.
Collapse
Affiliation(s)
- Giuliana Cavalloni
- Department of Medical Oncology, University of Turin Medical School, Turin, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zhang S, Yu D. Targeting Src family kinases in anti-cancer therapies: turning promise into triumph. Trends Pharmacol Sci 2011; 33:122-8. [PMID: 22153719 DOI: 10.1016/j.tips.2011.11.002] [Citation(s) in RCA: 226] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 11/07/2011] [Accepted: 11/08/2011] [Indexed: 01/11/2023]
Abstract
Src is a non-receptor tyrosine kinase that is deregulated in many types of cancer. Decades of research have revealed the crucial role of Src in many aspects of tumor development, including proliferation, survival, adhesion, migration, invasion and, most importantly, metastasis, in multiple tumor types. Despite extensive preclinical evidence that warrants targeting Src as a promising therapeutic approach for cancer, Src inhibitor(s) showed only minimal therapeutic activity in various types of solid tumors when used as a single agent in recent early-phase clinical trials. In this review, we highlight the most recent advances from preclinical studies and clinical trials that shed light on potential clinical use of Src inhibitor-containing combinatorial regimens in overcoming resistance to current anticancer therapies and in preventing metastatic recurrence.
Collapse
Affiliation(s)
- Siyuan Zhang
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | | |
Collapse
|
27
|
Zhang S, Yu D. Targeting Src family kinases in anti-cancer therapies: turning promise into triumph. Trends Pharmacol Sci 2011. [PMID: 22153719 DOI: 10.1016/j.tips.2011.11.002s0165-6147(11)00208-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Src is a non-receptor tyrosine kinase that is deregulated in many types of cancer. Decades of research have revealed the crucial role of Src in many aspects of tumor development, including proliferation, survival, adhesion, migration, invasion and, most importantly, metastasis, in multiple tumor types. Despite extensive preclinical evidence that warrants targeting Src as a promising therapeutic approach for cancer, Src inhibitor(s) showed only minimal therapeutic activity in various types of solid tumors when used as a single agent in recent early-phase clinical trials. In this review, we highlight the most recent advances from preclinical studies and clinical trials that shed light on potential clinical use of Src inhibitor-containing combinatorial regimens in overcoming resistance to current anticancer therapies and in preventing metastatic recurrence.
Collapse
Affiliation(s)
- Siyuan Zhang
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | | |
Collapse
|