1
|
Ding J, Wang X, Yang H, Zhang L, Ying Y, Pi W, Deng G, Zhu Y. IGF2BP3 Triggers STAT3 Pathway by Stabilizing SRC RNA in an m6A-Dependent Manner to Promote Lymphatic Metastasis in LUAD. Cancer Sci 2025; 116:936-950. [PMID: 39805702 PMCID: PMC11967251 DOI: 10.1111/cas.16451] [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: 08/22/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025] Open
Abstract
Lymph node metastasis significantly affects the NSCLC patients' staging, treatment strategy, and prognosis. Studies have shown that IGF2BP3, an oncofetal protein and an m6A reader, overexpresses and correlates to lymph node metastasis and worse overall survival in histopathological studies including NSCLC, but its mechanism needs further study. This study explored IGF2BP3's function and mechanism in LUAD lymphatic metastasis using public databases, a human LUAD tissue microarray, human LUAD cells, and a lymphatic metastasis model in male BALB/c nude mice. Firstly, we proved that IGF2BP3 overexpression was positively correlated to patients' lymph node metastasis and worse overall survival in bioinformatics and a human LUAD tissue microarray analysis. IGF2BP3 was knocked out or overexpressed in human LUAD cell lines. Functionally, IGF2BP3 facilitated NCI-H1299, NCI-H358, and A549 cell growth, migration, invasion, and EMT in vitro, and promoted tumorigenesis, lymphangiogenesis, and lymphatic metastasis of NCI-H1299 cells in BALB/c nude mice. Mechanically, RIP, RNA pull-down assay, MeRIP, mRNA stability assays, rescue experiments, and immunohistochemical assays were conducted. IGF2BP3 was demonstrated to bind to the m6A site of the 3'UTR region of SRC, stabilizing its mRNA and activating the downstream STAT3 signaling pathway and lymphatic growth factors such as VEGF-C, therefore affecting lymphatic metastasis. The cell migration and EMT function of IGF2BP3 were partially rescued by utilizing SRC siRNA or AZD0530, an SRC inhibitor. This study demonstrated that IGF2BP3 promotes lymphatic metastasis in LUAD via activating the m6A-SRC-STAT3-VEGFC signaling axis, indicating that IGF2BP3 is a potential therapeutic target to overcome metastasis in LUAD patients.
Collapse
Affiliation(s)
- Jiapei Ding
- Department of Radiotherapy & OncologyThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health AcademyTaizhou Hospital Affiliated to Wenzhou Medical UniversityTaizhouZhejiangChina
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang ProvinceWenzhou Medical UniversityTaizhouZhejiangChina
| | - Xuequan Wang
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health AcademyTaizhou Hospital Affiliated to Wenzhou Medical UniversityTaizhouZhejiangChina
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang ProvinceWenzhou Medical UniversityTaizhouZhejiangChina
| | - Haihua Yang
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health AcademyTaizhou Hospital Affiliated to Wenzhou Medical UniversityTaizhouZhejiangChina
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang ProvinceWenzhou Medical UniversityTaizhouZhejiangChina
| | - Lele Zhang
- School of Laboratory MedicineNingxia Medical UniversityYinchuanChina
| | - Yongquan Ying
- Department of Thoracic Surgery, Taizhou Hospital of Zhejiang ProvinceWenzhou Medical UniversityTaizhouZhejiangChina
| | - Wenhu Pi
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health AcademyTaizhou Hospital Affiliated to Wenzhou Medical UniversityTaizhouZhejiangChina
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang ProvinceWenzhou Medical UniversityTaizhouZhejiangChina
| | - Guozhong Deng
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health AcademyTaizhou Hospital Affiliated to Wenzhou Medical UniversityTaizhouZhejiangChina
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang ProvinceWenzhou Medical UniversityTaizhouZhejiangChina
| | - Yaqun Zhu
- Department of Radiotherapy & OncologyThe Second Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| |
Collapse
|
2
|
Martina MG, Rubini D, Radi M, Cagno V. Targeting PI4KB and Src/Abl host kinases as broad-spectrum antiviral strategy: Myth or real opportunity? Antiviral Res 2025; 235:106100. [PMID: 39922541 DOI: 10.1016/j.antiviral.2025.106100] [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: 12/16/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Viruses pose a continuous threat to human health. Limited treatment options exist for current viruses, and the risk of infections with newly emerging or re-emerging viruses is increasing. In a pandemic scenario, having a broad-spectrum antiviral to limit viral spread while developing specific antivirals and vaccines is crucial. Targeting host kinases represents a valuable strategy due to the higher barrier to resistance and the broad-spectrum activity it offers. While cells have redundant kinases for the same biological function, viruses rely on specific kinases for their replication cycle, enabling targeted antiviral action with limited toxicity. This review focuses on two extensively studied kinase targets: the lipid kinase phosphatidylinositol 4-kinase IIIβ (PI4KB) and the tyrosine kinase proteins Src and Abl. Compounds active against these targets are reviewed in terms of the viruses they inhibit, their mechanisms of action and their stage of development. While PI4KB inhibitors have reached clinical trials, those targeting Src and Abl remain largely in the preclinical phase. Nevertheless, opportunities exist to improve potency and further understand the specific roles of these kinases in the life cycle of multiple viruses.
Collapse
Affiliation(s)
- Maria Grazia Martina
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy
| | - Daniele Rubini
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy
| | - Marco Radi
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy.
| | - Valeria Cagno
- Institute of Microbiology, University Hospital of Lausanne, University of Lausanne, 1011, Lausanne, Switzerland.
| |
Collapse
|
3
|
Frame G, Huang X, Haas R, Khan KA, Leong HS, Kislinger T, Boutros PC, Downes M, Liu SK. Accelerated growth and local progression of radiorecurrent prostate cancer in an orthotopic bioluminescent mouse model. Sci Rep 2024; 14:31205. [PMID: 39732766 DOI: 10.1038/s41598-024-82546-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
Globally, prostate cancer is the second most common malignancy in males, with over 400 thousand men dying from the disease each year. A common treatment modality for localized prostate cancer is radiotherapy. However, up to half of high-risk patients can relapse with radiorecurrent prostate cancer, the aggressive clinical progression of which remains severely understudied. To address this, we have established an orthotopic mouse model for study that recapitulates the aggressive clinical progression of radiorecurrent prostate cancer. Radiorecurrent DU145 cells which survived conventional fraction (CF) irradiation were orthotopically injected into the prostates of athymic nude mice and monitored with bioluminescent imaging. CF tumours exhibited higher take rates and grew more rapidly than treatment-naïve parental tumours (PAR). Pathohistological analysis revealed extensive seminal vesicle invasion and necrosis in CF tumours, recapitulating the aggressive progression towards locally advanced disease exhibited by radiorecurrent tumours clinically. RNA sequencing of CF and PAR tumours identified ROBO1, CAV1, and CDH1 as candidate targets of radiorecurrent progression associated with biochemical relapse clinically. Together, this study presents a clinically relevant orthotopic model of radiorecurrent prostate cancer progression that will enable discovery of targets for therapeutic intervention to improve outcomes in prostate cancer patients.
Collapse
Affiliation(s)
- Gavin Frame
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Xiaoyong Huang
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Roni Haas
- University of California Los Angeles, Los Angeles, USA
| | - Kabir A Khan
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Hon S Leong
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Thomas Kislinger
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- University Health Network, Toronto, Canada
| | - Paul C Boutros
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- University of California Los Angeles, Los Angeles, USA
| | - Michelle Downes
- Division of Anatomic Pathology, Precision Diagnostics & Therapeutics Program-Laboratory Medicine, Sunnybrook Health Sciences Centre, Toronto, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Stanley K Liu
- Department of Medical Biophysics, University of Toronto, Toronto, Canada.
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada.
- Department of Radiation Oncology, University of Toronto, Toronto, Canada.
| |
Collapse
|
4
|
Ramu A, Ak L, Chinnappan J. Identification of prostate cancer associated genes for diagnosis and prognosis: a modernized in silico approach. Mamm Genome 2024; 35:683-710. [PMID: 39153107 DOI: 10.1007/s00335-024-10060-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024]
Abstract
Prostate cancer (PCa) ranks as the second leading cause of cancer-related deaths in men. Diagnosing PCa relies on molecular markers known as diagnostic biomarkers, while prognostic biomarkers are used to identify key proteins involved in PCa treatments. This study aims to gather PCa-associated genes and assess their potential as either diagnostic or prognostic biomarkers for PCa. A corpus of 152,064 PCa-related data from PubMed, spanning from May 1936 to December 2020, was compiled. Additionally, 4199 genes associated with PCa terms were collected from the National Center of Biotechnology Information (NCBI) database. The PubMed corpus data was extracted using pubmed.mineR to identify PCa-associated genes. Network and pathway analyses were conducted using various tools, such as STRING, DAVID, KEGG, MCODE 2.0, cytoHubba app, CluePedia, and ClueGO app. Significant marker genes were identified using Random Forest, Support Vector Machines, Neural Network algorithms, and the Cox Proportional Hazard model. This study reports 3062 unique PCa-associated genes along with 2518 corresponding unique PMIDs. Diagnostic markers such as IL6, MAPK3, JUN, FOS, ACTB, MYC, and TGFB1 were identified, while prognostic markers like ACTB and HDAC1 were highlighted in PubMed. This suggests that the potential target genes provided by PubMed data outweigh those in the NCBI database.
Collapse
Affiliation(s)
- Akilandeswari Ramu
- Anthropology and Health Informatics Laboratory, Department of Bioinformatics, Bharathiar University, Coimbatore, Tamil Nadu, India.
| | - Lekhashree Ak
- Anthropology and Health Informatics Laboratory, Department of Bioinformatics, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Jayaprakash Chinnappan
- Anthropology and Health Informatics Laboratory, Department of Bioinformatics, Bharathiar University, Coimbatore, Tamil Nadu, India
| |
Collapse
|
5
|
Liu R, Hou W, Li J, Gou X, Gao M, Wang H, Zhang Y, Deng H, Yang X, Zhang W. Co-assembly of cisplatin and dasatinib in hyaluronan nanogel to combat triple negative breast cancer with reduced side effects. Int J Biol Macromol 2024; 269:132074. [PMID: 38705320 DOI: 10.1016/j.ijbiomac.2024.132074] [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: 01/29/2024] [Revised: 04/14/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Treatment for triple negative breast cancer (TNBC) remains a huge challenge due to the lack of targeted therapeutics and tumor heterogenicity. Cisplatin (Cis) have demonstrated favorable therapeutic response in TNBC and thus is used together with various kinase inhibitors to fight the heterogenicity of TNBC. The combination of Cis with SRC inhibitor dasatinib (DAS) has shown encouraging anti-TNBC efficacy although the additive toxicity was commonly observed. To overcome the severe side effects of this Cis involved therapy, here we co-encapsulated Cis and DAS into a self-assembled hyaluronan (HA) nanogel (designated as HA/Cis/DAS (HCD) nanogel) to afford the TNBC targeted delivery by using the 4T1 mouse model. The acquired HCD nanogel was around 181 nm in aqueous solution, demonstrating the pharmacological activities of both Cis and DAS. Taking advantages of HA's targeting capability towards CD44 that is overexpressed on many TNBC cells, the HCD could well maintain the anticancer efficacy of the Cis and DAS combination, significantly increase the maximum tolerated dose and relieve the renal toxicity in vivo. The current HCD nanogel provides a potent strategy to improve the therapeutic outcome of Cis and DAS combination and thus representing a new targeted treatment option for TNBC.
Collapse
Affiliation(s)
- Runmeng Liu
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Wei Hou
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Jiayi Li
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Xiaorong Gou
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Menghan Gao
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Huimin Wang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Yiyi Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Hong Deng
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Xue Yang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China.
| | - Weiqi Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China.
| |
Collapse
|
6
|
Mathkor DM, Mathkor N, Bassfar Z, Bantun F, Slama P, Ahmad F, Haque S. Multirole of the internet of medical things (IoMT) in biomedical systems for managing smart healthcare systems: An overview of current and future innovative trends. J Infect Public Health 2024; 17:559-572. [PMID: 38367570 DOI: 10.1016/j.jiph.2024.01.013] [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: 04/06/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/19/2024] Open
Abstract
Internet of Medical Things (IoMT) is an emerging subset of Internet of Things (IoT), often called as IoT in healthcare, refers to medical devices and applications with internet connectivity, is exponentially gaining researchers' attention due to its wide-ranging applicability in biomedical systems for Smart Healthcare systems. IoMT facilitates remote health biomedical system and plays a crucial role within the healthcare industry to enhance precision, reliability, consistency and productivity of electronic devices used for various healthcare purposes. It comprises a conceptualized architecture for providing information retrieval strategies to extract the data from patient records using sensors for biomedical analysis and diagnostics against manifold diseases to provide cost-effective medical solutions, quick hospital treatments, and personalized healthcare. This article provides a comprehensive overview of IoMT with special emphasis on its current and future trends used in biomedical systems, such as deep learning, machine learning, blockchains, artificial intelligence, radio frequency identification, and industry 5.0.
Collapse
Affiliation(s)
- Darin Mansor Mathkor
- Research and Scientific Studies Unit, Department of Nursing, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Noof Mathkor
- Department of Pathology, Ministry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
| | - Zaid Bassfar
- Department of Information Technology, Faculty of Computers and Information Technology, University of Tabuk, Tabuk, Saudi Arabia
| | - Farkad Bantun
- Department of Microbiology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Petr Slama
- Laboratory of Animal Immunology and Biotechnology, Department of Animal Morphology, Physiology and Genetics, Mendel University in Brno, 61300 Brno, Czech Republic
| | - Faraz Ahmad
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Shafiul Haque
- Research and Scientific Studies Unit, Department of Nursing, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon; Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| |
Collapse
|
7
|
Magrath JW, Flinchum DA, Hartono AB, Sampath SS, O'Grady TM, Baddoo M, Haoyang L, Xu X, Flemington EK, Lee SB. Transcriptomic analysis identifies B-lymphocyte kinase as a therapeutic target for desmoplastic small round cell tumor cancer stem cell-like cells. Oncogenesis 2024; 13:2. [PMID: 38177125 PMCID: PMC10767073 DOI: 10.1038/s41389-023-00504-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/28/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024] Open
Abstract
Desmoplastic small round cell tumor (DSRCT) is an aggressive pediatric cancer caused by the EWSR1-WT1 fusion oncoprotein. The tumor is refractory to treatment with a 5-year survival rate of only 15-25%, necessitating the development of novel therapeutics, especially those able to target chemoresistant subpopulations. Novel in vitro cancer stem cell-like (CSC-like) culture conditions increase the expression of stemness markers (SOX2, NANOG) and reduce DSRCT cell line susceptibility to chemotherapy while maintaining the ability of DSRCT cells to form xenografts. To gain insights into this chemoresistant model, RNA-seq was performed to elucidate transcriptional alterations between DSRCT cells grown in CSC-like spheres and normal 2-dimensional adherent state. Commonly upregulated and downregulated genes were identified and utilized in pathway analysis revealing upregulation of pathways related to chromatin assembly and disassembly and downregulation of pathways including cell junction assembly and extracellular matrix organization. Alterations in chromatin assembly suggest a role for epigenetics in the DSRCT CSC-like state, which was further investigated with ATAC-seq, identifying over 10,000 differentially accessible peaks, including 4444 sphere accessible peaks and 6,120 adherent accessible peaks. Accessible regions were associated with higher gene expression, including increased accessibility of the CSC marker SOX2 in CSC-like culture conditions. These analyses were further utilized to identify potential CSC therapeutic targets, leading to the identification of B-lymphocyte kinase (BLK) as a CSC-enriched, EWSR1-WT1-regulated, druggable target. BLK inhibition and knockdown reduced CSC-like properties, including abrogation of tumorsphere formation and stemness marker expression. Importantly, BLK knockdown reduced DSRCT CSC-like cell chemoresistance, making its inhibition a promising target for future combination therapy.
Collapse
Affiliation(s)
- Justin W Magrath
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Dane A Flinchum
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Alifiani B Hartono
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
- Department of Molecular & Medical Pharmacology, University of California Los Angeles, 630 Charles E Young Dr. S., Los Angeles, CA, 90095, USA
| | - Shruthi Sanjitha Sampath
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Tina M O'Grady
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Melody Baddoo
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Liang Haoyang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Xiaojiang Xu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Erik K Flemington
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Sean B Lee
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA.
| |
Collapse
|
8
|
Miller KA, Degan S, Wang Y, Cohen J, Ku SY, Goodrich DW, Gelman IH. PTEN-regulated PI3K-p110 and AKT isoform plasticity controls metastatic prostate cancer progression. Oncogene 2024; 43:22-34. [PMID: 37875657 PMCID: PMC10766561 DOI: 10.1038/s41388-023-02875-4] [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: 05/11/2023] [Revised: 10/04/2023] [Accepted: 10/13/2023] [Indexed: 10/26/2023]
Abstract
PTEN loss, one of the most frequent mutations in prostate cancer (PC), is presumed to drive disease progression through AKT activation. However, two transgenic PC models with Akt activation plus Rb loss exhibited different metastatic development: Pten/RbPE:-/- mice produced systemic metastatic adenocarcinomas with high AKT2 activation, whereas RbPE:-/- mice deficient for the Src-scaffolding protein, Akap12, induced high-grade prostatic intraepithelial neoplasias and indolent lymph node dissemination, correlating with upregulated phosphotyrosyl PI3K-p85α. Using PC cells isogenic for PTEN, we show that PTEN-deficiency correlated with dependence on both p110β and AKT2 for in vitro and in vivo parameters of metastatic growth or motility, and with downregulation of SMAD4, a known PC metastasis suppressor. In contrast, PTEN expression, which dampened these oncogenic behaviors, correlated with greater dependence on p110α plus AKT1. Our data suggest that metastatic PC aggressiveness is controlled by specific PI3K/AKT isoform combinations influenced by divergent Src activation or PTEN-loss pathways.
Collapse
Affiliation(s)
- Karina A Miller
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14209, USA
- American Society of Human Genetics, Rockville, MD, 20852, USA
| | - Seamus Degan
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14209, USA
| | - Yanqing Wang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14209, USA
| | - Joseph Cohen
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14209, USA
- Sequence, Inc., Morrisville, NC, USA
| | - Sheng Yu Ku
- Dana Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - David W Goodrich
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14209, USA
| | - Irwin H Gelman
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14209, USA.
| |
Collapse
|
9
|
Zamora I, Freeman MR, Encío IJ, Rotinen M. Targeting Key Players of Neuroendocrine Differentiation in Prostate Cancer. Int J Mol Sci 2023; 24:13673. [PMID: 37761978 PMCID: PMC10531052 DOI: 10.3390/ijms241813673] [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: 08/17/2023] [Revised: 09/02/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Neuroendocrine prostate cancer (NEPC) is a highly aggressive subtype of prostate cancer (PC) that commonly emerges through a transdifferentiation process from prostate adenocarcinoma and evades conventional therapies. Extensive molecular research has revealed factors that drive lineage plasticity, uncovering novel therapeutic targets to be explored. A diverse array of targeting agents is currently under evaluation in pre-clinical and clinical studies with promising results in suppressing or reversing the neuroendocrine phenotype and inhibiting tumor growth and metastasis. This new knowledge has the potential to contribute to the development of novel therapeutic approaches that may enhance the clinical management and prognosis of this lethal disease. In the present review, we discuss molecular players involved in the neuroendocrine phenotype, and we explore therapeutic strategies that are currently under investigation for NEPC.
Collapse
Affiliation(s)
- Irene Zamora
- Department of Health Science, Public University of Navarre, 31008 Pamplona, Spain
| | - Michael R. Freeman
- Departments of Urology and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ignacio J. Encío
- Department of Health Science, Public University of Navarre, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Navarre Institute for Health Research, 31008 Pamplona, Spain
| | - Mirja Rotinen
- Department of Health Science, Public University of Navarre, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Navarre Institute for Health Research, 31008 Pamplona, Spain
| |
Collapse
|
10
|
Ye C, Gosser C, Runyon ED, Zha J, Cai J, Beharry Z, Bowes Rickman C, Klingeborn M, Liu Y, Xie J, Cai H. Src family kinases engage differential pathways for encapsulation into extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e96. [PMID: 37588411 PMCID: PMC10426749 DOI: 10.1002/jex2.96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 05/26/2023] [Indexed: 08/18/2023]
Abstract
Extracellular vesicles (EVs) are heterogeneous biological nanoparticles secreted by all cell types. Identifying the proteins preferentially encapsulated in secreted EVs will help understand their heterogeneity. Src family kinases including Src and Fyn are a group of tyrosine kinases with fatty acylation modifications and/or multiple lysine residues (contributing charge interaction) at their N-terminus. Here, we demonstrate that Src and Fyn kinases were preferentially encapsulated in EVs and fatty acylation including myristoylation and palmitoylation facilitated their encapsulation. Genetic loss or pharmacological inhibition of myristoylation suppressed Src and/or Fyn kinase levels in EVs. Similarly, loss of palmitoylation reduced Fyn levels in EVs. Additionally, mutation of lysine at sites 5, 7, and 9 of Src kinase also inhibited the encapsulation of myristoylated Src into EVs. Knockdown of TSG101, which is a protein involved in the endosomal sorting complexes required for transport (ESCRT) protein complex mediated EVs biogenesis and led to a reduction of Src levels in EVs. In contrast, filipin III treatment, which disturbed the lipid raft structure, reduced Fyn kinase levels, but not Src kinase levels in EVs. Finally, elevated levels of Src protein were detected in the serum EVs of host mice carrying constitutively active Src-mediated prostate tumors in vivo. Collectively, the data suggest that different EVs biogenesis pathways exist and can regulate the encapsulation of specific proteins into EVs. This study provides an understanding of the EVs heterogeneity created by different EVs biogenesis pathways.
Collapse
Affiliation(s)
- Chenming Ye
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of Georgia AthensAthensGeorgiaUSA
| | - Cade Gosser
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of Georgia AthensAthensGeorgiaUSA
| | - Ethan Daniel Runyon
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of Georgia AthensAthensGeorgiaUSA
| | - Junyi Zha
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of Georgia AthensAthensGeorgiaUSA
| | - Jingwen Cai
- Department of Cellular Biology and AnatomyAugusta UniversityAugustaGeorgiaUSA
| | - Zanna Beharry
- Department of Chemical and Physical SciencesUniversity of Virgin IslandsUSA
| | - Catherine Bowes Rickman
- Department of OphthalmologyDuke UniversityDurhamNorth CarolinaUSA
- Department of Cell BiologyDuke UniversityDurhamNorth CarolinaUSA
| | | | - Yutao Liu
- Department of Cellular Biology and AnatomyAugusta UniversityAugustaGeorgiaUSA
| | - Jin Xie
- Department of ChemistryUniversity of Georgia AthensAthensGeorgiaUSA
| | - Houjian Cai
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of Georgia AthensAthensGeorgiaUSA
| |
Collapse
|
11
|
Miller K, Degan S, Wang Y, Cohen J, Ku SY, Goodrich D, Gelman I. PTEN regulated PI3K-p110 and AKT isoform plasticity controls metastatic prostate cancer progression. RESEARCH SQUARE 2023:rs.3.rs-2924750. [PMID: 37292818 PMCID: PMC10246239 DOI: 10.21203/rs.3.rs-2924750/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
PTEN loss, one of the most frequent mutations in prostate cancer (PC), is presumed to drive disease progression through AKT activation. However, two transgenic PC models with Akt activation plus Rb loss exhibited different metastasis development: Pten/RbPE:-/- mice produced systemic metastatic adenocarcinomas with high AKT2 activation, whereas RbPE:-/- mice deficient for the Src-scaffolding protein, Akap12, induced high-grade prostatic intraepithelial neoplasias and indolent lymph node disseminations, correlating with upregulated phosphotyrosyl PI3K-p85α. Using PC cells isogenic for PTEN, we show that PTEN-deficiency correlated with dependence on both p110β and AKT2 for in vitro and in vivo parameters of metastatic growth or motility, and with downregulation of SMAD4, a known PC metastasis suppressor. In contrast, PTEN expression, which dampened these oncogenic behaviors, correlated with greater dependence on p110α plus AKT1. Our data suggest that metastatic PC aggressiveness is controlled by specific PI3K/AKT isoform combinations influenced by divergent Src activation or PTEN-loss pathways.
Collapse
|
12
|
Nepali PR, Kyprianou N. Anoikis in phenotypic reprogramming of the prostate tumor microenvironment. Front Endocrinol (Lausanne) 2023; 14:1160267. [PMID: 37091854 PMCID: PMC10113530 DOI: 10.3389/fendo.2023.1160267] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/09/2023] [Indexed: 04/25/2023] Open
Abstract
Prostate cancer is one of the most common malignancies in males wherein 1 in 8 men are diagnosed with this disease in their lifetime. The urgency to find novel therapeutic interventions is associated with high treatment resistance and mortality rates associated with castration-resistant prostate cancer. Anoikis is an apoptotic phenomenon for normal epithelial or endothelial cells that have lost their attachment to the extracellular matrix (ECM). Tumor cells that lose their connection to the ECM can die via apoptosis or survive via anoikis resistance and thus escaping to distant organs for metastatic progression. This review discusses the recent advances made in our understanding of the signaling effectors of anoikis in prostate cancer and the approaches to translate these mechanistic insights into therapeutic benefits for reducing lethal disease outcomes (by overcoming anoikis resistance). The prostate tumor microenvironment is a highly dynamic landscape wherein the balance between androgen signaling, cell lineage changes, epithelial-mesenchymal transition (EMT), extracellular matrix interactions, actin cytoskeleton remodeling as well as metabolic changes, confer anoikis resistance and metastatic spread. Thus, these mechanisms also offer unique molecular treatment signatures, exploitation of which can prime prostate tumors to anoikis induction with a high translational significance.
Collapse
Affiliation(s)
- Prerna R. Nepali
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Natasha Kyprianou
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pathology and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
13
|
Tarfeen N, Nisa KU, Ali S, Yatoo AM, Shah AM, Sabba A, Maqbool R, Ahmad MB. Utility of proteomics and phosphoproteomics in the tailored medication of cancer. Proteomics 2023. [DOI: 10.1016/b978-0-323-95072-5.00006-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
14
|
Myeloid-derived suppressor cells in head and neck squamous cell carcinoma. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 375:33-92. [PMID: 36967154 DOI: 10.1016/bs.ircmb.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs), which originated from hematopoietic stem cells, are heterogeneous population of cells that have different differentiation patterns and widely presented in tumor microenvironment. For tumor research, myeloid suppressor cells have received extensive attention since their discovery due to their specific immunosuppressive properties, and the mechanisms of immunosuppression and therapeutic approaches for MDSCs have been investigated in a variety of different types of malignancies. To improve the efficacy of treatment for head and neck squamous cell carcinoma (HNSCC), a disease with a high occurrence, immunotherapy has gradually emerged in after traditional surgery and subsequent radiotherapy and chemotherapy, and has made some progress. In this review, we introduced the mechanisms on the development, differentiation, and elimination of MDSCs and provided a detailed overview of the mechanisms behind the immunosuppressive properties of MDSCs. We summarized the recent researches on MDSCs in HNSCC, especially for targeting-MDSCs therapy and combination with other types of therapy such as immune checkpoint blockade (ICB). Furthermore, we looked at drug delivery patterns and collected the current diverse drug delivery systems for the improvement that contributed to therapy against MDSCs in HNSCC. Most importantly, we made possible outlooks for the future research priorities, which provide a basis for further study on the clinical significance and therapeutic value of MDSCs in HNSCC.
Collapse
|
15
|
Johnson RP, Ratnacaram CK, Kumar L, Jose J. Combinatorial approaches of nanotherapeutics for inflammatory pathway targeted therapy of prostate cancer. Drug Resist Updat 2022; 64:100865. [PMID: 36099796 DOI: 10.1016/j.drup.2022.100865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 12/24/2022]
Abstract
Prostate cancer (PC) is the most prevalent male urogenital cancer worldwide. PC patients presenting an advanced or metastatic cancer succumb to the disease, even after therapeutic interventions including radiotherapy, surgery, androgen deprivation therapy (ADT), and chemotherapy. One of the hallmarks of PC is evading immune surveillance and chronic inflammation, which is a major challenge towards designing effective therapeutic formulations against PC. Chronic inflammation in PC is often characterized by tumor microenvironment alterations, epithelial-mesenchymal transition and extracellular matrix modifications. The inflammatory events are modulated by reactive nitrogen and oxygen species, inflammatory cytokines and chemokines. Major signaling pathways in PC includes androgen receptor, PI3K and NF-κB pathways and targeting these inter-linked pathways poses a major therapeutic challenge. Notably, many conventional treatments are clinically unsuccessful, due to lack of targetability and poor bioavailability of the therapeutics, untoward toxicity and multidrug resistance. The past decade witnessed an advancement of nanotechnology as an excellent therapeutic paradigm for PC therapy. Modern nanovectorization strategies such as stimuli-responsive and active PC targeting carriers offer controlled release patterns and superior anti-cancer effects. The current review initially describes the classification, inflammatory triggers and major inflammatory pathways of PC, various PC treatment strategies and their limitations. Subsequently, recent advancement in combinatorial nanotherapeutic approaches, which target PC inflammatory pathways, and the mechanism of action are discussed. Besides, the current clinical status and prospects of PC homing nanovectorization, and major challenges to be addressed towards the advancement PC therapy are also addressed.
Collapse
Affiliation(s)
- Renjith P Johnson
- Polymer Nanobiomaterial Research Laboratory, Nanoscience and Microfluidics Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Chandrahas Koumar Ratnacaram
- Cell Signaling and Cancer Biology Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Lalit Kumar
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka 576 104, India
| | - Jobin Jose
- NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangalore 575018, India.
| |
Collapse
|
16
|
Moreira-Silva F, Henrique R, Jerónimo C. From Therapy Resistance to Targeted Therapies in Prostate Cancer. Front Oncol 2022; 12:877379. [PMID: 35686097 PMCID: PMC9170957 DOI: 10.3389/fonc.2022.877379] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is the second most common malignancy among men worldwide. Although early-stage disease is curable, advanced stage PCa is mostly incurable and eventually becomes resistant to standard therapeutic options. Different genetic and epigenetic alterations are associated with the development of therapy resistant PCa, with specific players being particularly involved in this process. Therefore, identification and targeting of these molecules with selective inhibitors might result in anti-tumoral effects. Herein, we describe the mechanisms underlying therapy resistance in PCa, focusing on the most relevant molecules, aiming to enlighten the current state of targeted therapies in PCa. We suggest that selective drug targeting, either alone or in combination with standard treatment options, might improve therapeutic sensitivity of resistant PCa. Moreover, an individualized analysis of tumor biology in each PCa patient might improve treatment selection and therapeutic response, enabling better disease management.
Collapse
Affiliation(s)
- Filipa Moreira-Silva
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (He-alth Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (He-alth Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences of the University of Porto (ICBAS-UP), Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (He-alth Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), Porto, Portugal.,Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences of the University of Porto (ICBAS-UP), Porto, Portugal
| |
Collapse
|
17
|
Computational modeling identifies multitargeted kinase inhibitors as effective therapies for metastatic, castration-resistant prostate cancer. Proc Natl Acad Sci U S A 2021; 118:2103623118. [PMID: 34593636 PMCID: PMC8501846 DOI: 10.1073/pnas.2103623118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2021] [Indexed: 01/02/2023] Open
Abstract
Metastatic, castration-resistant prostate cancer (mCRPC) is an advanced prostate cancer with limited therapeutic options and poor patient outcomes. To investigate whether multitargeted kinase inhibitors (KIs) represent an opportunity for mCRPC drug development, we applied machine learning–based functional screening and identified two KIs, PP121 and SC-1, which demonstrated strong suppression of CRPC growth in vitro and in vivo. Furthermore, we show the marked ability of these KIs to improve on standard-of-care chemotherapy in both tumor response and survival, suggesting that combining multitargeted KIs with chemotherapy represents a promising avenue for mCRPC treatment. Overall, our findings demonstrate the application of a multidisciplinary strategy that blends bench science with machine-learning approaches for rapidly identifying KIs that result in desired phenotypic effects. Castration-resistant prostate cancer (CRPC) is an advanced subtype of prostate cancer with limited therapeutic options. Here, we applied a systems-based modeling approach called kinome regularization (KiR) to identify multitargeted kinase inhibitors (KIs) that abrogate CRPC growth. Two predicted KIs, PP121 and SC-1, suppressed CRPC growth in two-dimensional in vitro experiments and in vivo subcutaneous xenografts. An ex vivo bone mimetic environment and in vivo tibia xenografts revealed resistance to these KIs in bone. Combining PP121 or SC-1 with docetaxel, standard-of-care chemotherapy for late-stage CRPC, significantly reduced tibia tumor growth in vivo, decreased growth factor signaling, and vastly extended overall survival, compared to either docetaxel monotherapy. These results highlight the utility of computational modeling in forming physiologically relevant predictions and provide evidence for the role of multitargeted KIs as chemosensitizers for late-stage, metastatic CRPC.
Collapse
|
18
|
Ranjithkumar R, Saravanan K, Balaji B, Hima S, Sreeja S, Timane SR, Ram Pravin Kumar M, Kabilan S, Ramanathan M. Novel daidzein molecules exhibited anti-prostate cancer activity through nuclear receptor ERβ modulation, in vitro and in vivo studies. J Chemother 2021; 33:582-594. [PMID: 34060437 DOI: 10.1080/1120009x.2021.1924935] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Eight novel ERβ selective daidzein analogues (NCE1-8) were synthesized and their anti-cancer activity was evaluated by in vitro and in vivo methods. Cytotoxicity study, Receptor binding studies, Luciferase assay, cMYC & Cyclin D1 expression and Caspase 3, 8 & 9 activities were measured to ascertain the anticancer activity and mechanism. Uterotropic, anti-androgenic and anti-tumour activities were performed in rodents. The results revealed that NCEs produced anti-prostate cancer activity in DU145, LNCaP and PC3 cell lines and 50% more active than genistein. NCEs was significantly down-regulated cMYC & Cyclin D1 genes and elevated caspase 3 & 9 levels and did not show any difference in uterotropic, anti-androgenic activities. The tumour weight was also reduced. The NCE 1 and 2 have shown ERβ selectivity in receptor binding studies. Daidzein with methyl substitution at R or R1 position exhibited more ERβ selectivity and could be considered as lead molecules for anti-prostate cancer activity.
Collapse
Affiliation(s)
- R Ranjithkumar
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, Tamil Nadu, India
| | - K Saravanan
- Drug Discovery Lab, Department of Chemistry, Annamalai University, Annamalai Nagar, Tamil Nadu, India
| | - B Balaji
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, Tamil Nadu, India
| | - S Hima
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - S Sreeja
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - S R Timane
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, Tamil Nadu, India
| | - M Ram Pravin Kumar
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, Tamil Nadu, India
| | - S Kabilan
- Drug Discovery Lab, Department of Chemistry, Annamalai University, Annamalai Nagar, Tamil Nadu, India
| | - M Ramanathan
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, Tamil Nadu, India
| |
Collapse
|
19
|
Tong D. Unravelling the molecular mechanisms of prostate cancer evolution from genotype to phenotype. Crit Rev Oncol Hematol 2021; 163:103370. [PMID: 34051300 DOI: 10.1016/j.critrevonc.2021.103370] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PC) is the most frequently diagnosed cancer and the second leading cause of cancer-related death in men in the Western society. Unfortunately, although the vast majority of patients are initially responsive to androgen-deprivation therapy (ADT), most cases eventually develop from hormone-sensitive prostate cancer (HSPC) to castration-resistant prostate cancer (CRPC). The main reason is PC heterogeneity and evolution during therapy. PC evolution is a continuously progressive process with combination of genomic alterations including canonical AR, TMPRSS2-ERG fusion, SPOP/FOXA1, TP53/RB1/PTEN, BRCA2. Meanwhile, signaling pathways including PI3K, WNT/β-catenin, SRC, IL-6/STAT3 are activated, to promote epithelial mesenchymal transition (EMT), cancer stem cell (CSC)-like features/stemness and neuroendocrine differentiation (NED) of PC. These improve our understanding of the genotype-phenotype relationships. The identification of canonical genetic alterations and signaling pathway activation in PC has shed more insight into genetic background, molecular subtype and disease landscape of PC evolution, resulting in a more flexible role of individual therapies targeting diverse genotype and phenotype presentation.
Collapse
Affiliation(s)
- Dali Tong
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, PR China.
| |
Collapse
|
20
|
Spetsieris N, Boukovala M, Weldon JA, Tsikkinis A, Hoang A, Aparicio A, Tu SM, Araujo JC, Zurita AJ, Corn PG, Pagliaro L, Kim J, Wang J, Subudhi SK, Tannir NM, Logothetis CJ, Troncoso P, Wang X, Wen S, Efstathiou E. A Phase 2 Trial of Abiraterone Followed by Randomization to Addition of Dasatinib or Sunitinib in Men With Metastatic Castration-Resistant Prostate Cancer. Clin Genitourin Cancer 2021; 19:22-31.e5. [PMID: 32675015 PMCID: PMC10014037 DOI: 10.1016/j.clgc.2020.05.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/19/2020] [Accepted: 05/19/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Resistance to novel androgen signaling inhibition and metastatic castration-resistant prostate cancer (mCRPC) progression is likely dependent on tumor microenvironment interactions. The Src pathway and neoangiogenesis have been implicated in prostate cancer progression. We studied the effect of adding the targeted agents dasatinib and sunitinib to abiraterone acetate (AA) in men with mCRPC. PATIENTS AND METHODS In this open-label randomized phase 2 study, mCRPC patients received AA. At resistance to AA, they were randomized 1:1 to combination with dasatinib or sunitinib. At second progression, patients crossed over. The primary end point was time to treatment failure (TTF), defined as time to progression or death. Secondary end points included overall survival and safety. RESULTS From March 2011 to February 2015, a total of 179 patients were enrolled and 132 subsequently randomized. Median TTF was 5.7 months in the dasatinib group and 5.5 months in the sunitinib group. There was no difference between the two groups in terms of TTF (hazard ratio, 0.85; 95% confidence interval, 0.59-1.22). Median overall survival from study entry was 26.3 months in the dasatinib group and 27.7 months in the sunitinib group (hazard ratio, 1.02; 95% confidence interval, 0.71-1.47). Grade 3 or higher adverse events related to study medication were more frequent with sunitinib (n = 44, 46%) compared to dasatinib (n = 26, 24%). At data cutoff, 7 patients were experiencing a continuous response to AA, with a median duration of treatment of 5.7 years. CONCLUSION There is no difference in overall survival and TTF between dasatinib and sunitinib combined with abiraterone in the treatment of patients with bone mCRPC.
Collapse
Affiliation(s)
- Nicholas Spetsieris
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Myrto Boukovala
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Justin A Weldon
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Alexandros Tsikkinis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anh Hoang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ana Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Shi-Ming Tu
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - John C Araujo
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Amado J Zurita
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Paul G Corn
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lance Pagliaro
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jeri Kim
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jennifer Wang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sumit K Subudhi
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Patricia Troncoso
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xuemei Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sijin Wen
- Department of Biostatistics, West Virginia University School of Public Health, Morgantown, WV
| | - Eleni Efstathiou
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
21
|
Moro L, Simoneschi D, Kurz E, Arbini AA, Jang S, Guaragnella N, Giannattasio S, Wang W, Chen YA, Pires G, Dang A, Hernandez E, Kapur P, Mishra A, Tsirigos A, Miller G, Hsieh JT, Pagano M. Epigenetic silencing of the ubiquitin ligase subunit FBXL7 impairs c-SRC degradation and promotes epithelial-to-mesenchymal transition and metastasis. Nat Cell Biol 2020; 22:1130-1142. [PMID: 32839549 PMCID: PMC7484425 DOI: 10.1038/s41556-020-0560-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/14/2020] [Indexed: 12/12/2022]
Abstract
Epigenetic plasticity is a pivotal factor that drives metastasis. Here, we show that the promoter of the gene that encodes the ubiquitin ligase subunit FBXL7 is hypermethylated in advanced prostate and pancreatic cancers, correlating with decreased FBXL7 mRNA and protein levels. Low FBXL7 mRNA levels are predictive of poor survival in patients with pancreatic and prostatic cancers. FBXL7 mediates the ubiquitylation and proteasomal degradation of active c-SRC after its phosphorylation at Ser 104. The DNA-demethylating agent decitabine recovers FBXL7 expression and limits epithelial-to-mesenchymal transition and cell invasion in a c-SRC-dependent manner. In vivo, FBXL7-depleted cancer cells form tumours with a high metastatic burden. Silencing of c-SRC or treatment with the c-SRC inhibitor dasatinib together with FBXL7 depletion prevents metastases. Furthermore, decitabine reduces metastases derived from prostate and pancreatic cancer cells in a FBXL7-dependent manner. Collectively, this research implicates FBXL7 as a metastasis-suppressor gene and suggests therapeutic strategies to counteract metastatic dissemination of pancreatic and prostatic cancer cells.
Collapse
Affiliation(s)
- Loredana Moro
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA.
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA.
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy.
| | - Daniele Simoneschi
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Emma Kurz
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Arnaldo A Arbini
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Shaowen Jang
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Nicoletta Guaragnella
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "A. Moro", Bari, Italy
| | - Sergio Giannattasio
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy
| | - Wei Wang
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA
- Department of Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Yu-An Chen
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Geoffrey Pires
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, USA
| | - Andrew Dang
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elizabeth Hernandez
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ankita Mishra
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA
- Department of Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Aristotelis Tsirigos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - George Miller
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA
- Department of Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michele Pagano
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA.
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA.
- Howard Hughes Medical Institute, New York, NY, USA.
| |
Collapse
|
22
|
Lorusso G, Rüegg C, Kuonen F. Targeting the Extra-Cellular Matrix-Tumor Cell Crosstalk for Anti-Cancer Therapy: Emerging Alternatives to Integrin Inhibitors. Front Oncol 2020; 10:1231. [PMID: 32793493 PMCID: PMC7387567 DOI: 10.3389/fonc.2020.01231] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) is a complex network composed of a multitude of different macromolecules. ECM components typically provide a supportive structure to the tissue and engender positional information and crosstalk with neighboring cells in a dynamic reciprocal manner, thereby regulating tissue development and homeostasis. During tumor progression, tumor cells commonly modify and hijack the surrounding ECM to sustain anchorage-dependent growth and survival, guide migration, store pro-tumorigenic cell-derived molecules and present them to enhance receptor activation. Thereby, ECM potentially supports tumor progression at various steps from initiation, to local growth, invasion, and systemic dissemination and ECM-tumor cells interactions have long been considered promising targets for cancer therapy. Integrins represent key surface receptors for the tumor cell to sense and interact with the ECM. Yet, attempts to therapeutically impinge on these interactions using integrin inhibitors have failed to deliver anticipated results, and integrin inhibitors are still missing in the emerging arsenal of drugs for targeted therapies. This paradox situation should urge the field to reconsider the role of integrins in cancer and their targeting, but also to envisage alternative strategies. Here, we review the therapeutic targets implicated in tumor cell adhesion to the ECM, whose inhibitors are currently in clinical trials and may offer alternatives to integrin inhibition.
Collapse
Affiliation(s)
- Girieca Lorusso
- Experimental and Translational Oncology, Department of Oncology Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Curzio Rüegg
- Experimental and Translational Oncology, Department of Oncology Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - François Kuonen
- Department of Dermatology and Venereology, Hôpital de Beaumont, Lausanne University Hospital Center, Lausanne, Switzerland
| |
Collapse
|
23
|
Small Ones to Fight a Big Problem-Intervention of Cancer Metastasis by Small Molecules. Cancers (Basel) 2020; 12:cancers12061454. [PMID: 32503267 PMCID: PMC7352875 DOI: 10.3390/cancers12061454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
Metastasis represents the most lethal attribute of cancer and critically limits successful therapies in many tumor entities. The clinical need is defined by the fact that all cancer patients, who have or who will develop distant metastasis, will experience shorter survival. Thus, the ultimate goal in cancer therapy is the restriction of solid cancer metastasis by novel molecularly targeted small molecule based therapies. Biomarkers identifying cancer patients at high risk for metastasis and simultaneously acting as key drivers for metastasis are extremely desired. Clinical interventions targeting these key molecules will result in high efficiency in metastasis intervention. In result of this, personalized tailored interventions for restriction and prevention of cancer progression and metastasis will improve patient survival. This review defines crucial biological steps of the metastatic cascade, such as cell dissemination, migration and invasion as well as the action of metastasis suppressors. Targeting these biological steps with tailored therapeutic strategies of intervention or even prevention of metastasis using a wide range of small molecules will be discussed.
Collapse
|
24
|
Src Family Kinases as Therapeutic Targets in Advanced Solid Tumors: What We Have Learned so Far. Cancers (Basel) 2020; 12:cancers12061448. [PMID: 32498343 PMCID: PMC7352436 DOI: 10.3390/cancers12061448] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 12/17/2022] Open
Abstract
Src is the prototypal member of Src Family tyrosine Kinases (SFKs), a large non-receptor kinase class that controls multiple signaling pathways in animal cells. SFKs activation is necessary for the mitogenic signal from many growth factors, but also for the acquisition of migratory and invasive phenotype. Indeed, oncogenic activation of SFKs has been demonstrated to play an important role in solid cancers; promoting tumor growth and formation of distant metastases. Several drugs targeting SFKs have been developed and tested in preclinical models and many of them have successfully reached clinical use in hematologic cancers. Although in solid tumors SFKs inhibitors have consistently confirmed their ability in blocking cancer cell progression in several experimental models; their utilization in clinical trials has unveiled unexpected complications against an effective utilization in patients. In this review, we summarize basic molecular mechanisms involving SFKs in cancer spreading and metastasization; and discuss preclinical and clinical data highlighting the main challenges for their future application as therapeutic targets in solid cancer progression
Collapse
|
25
|
Liu YL, Horning AM, Lieberman B, Kim M, Lin CK, Hung CN, Chou CW, Wang CM, Lin CL, Kirma NB, Liss MA, Vasisht R, Perillo EP, Blocher K, Horng H, Taverna JA, Ruan J, Yankeelov TE, Dunn AK, Huang THM, Yeh HC, Chen CL. Spatial EGFR Dynamics and Metastatic Phenotypes Modulated by Upregulated EphB2 and Src Pathways in Advanced Prostate Cancer. Cancers (Basel) 2019; 11:cancers11121910. [PMID: 31805710 PMCID: PMC6966510 DOI: 10.3390/cancers11121910] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/23/2019] [Accepted: 11/26/2019] [Indexed: 12/21/2022] Open
Abstract
Advanced prostate cancer is a very heterogeneous disease reflecting in diverse regulations of oncogenic signaling pathways. Aberrant spatial dynamics of epidermal growth factor receptor (EGFR) promote their dimerization and clustering, leading to constitutive activation in oncogenesis. The EphB2 and Src signaling pathways are associated with the reorganization of the cytoskeleton leading to malignancy, but their roles in regulating EGFR dynamics and activation are scarcely reported. Using single-particle tracking techniques, we found that highly phosphorylated EGFR in the advanced prostate cancer cell line, PC3, was associated with higher EGFR diffusivity, as compared with LNCaP and less aggressive DU145. The increased EGFR activation and biophysical dynamics were consistent with high proliferation, migration, and invasion. After performing single-cell RNA-seq on prostate cancer cell lines and circulating tumor cells from patients, we identified that upregulated gene expression in the EphB2 and Src pathways are associated with advanced malignancy. After dasatinib treatment or siRNA knockdowns of EphB2 or Src, the PC3 cells exhibited significantly lower EGFR dynamics, cell motility, and invasion. Partial inhibitory effects were also found in DU145 cells. The upregulation of parts of the EphB2 and Src pathways also predicts poor prognosis in the prostate cancer patient cohort of The Cancer Genome Atlas. Our results provide evidence that overexpression of the EphB2 and Src signaling pathways regulate EGFR dynamics and cellular aggressiveness in some advanced prostate cancer cells.
Collapse
Affiliation(s)
- Yen-Liang Liu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan;
- Department of Biomedical Engineering, University of Texas at Austin, 107 W. Dean Keeton, BME Building, Austin, TX 78712, USA; (M.K.); (R.V.); (E.P.P.); (K.B.); (T.E.Y.); (A.K.D.)
| | - Aaron M. Horning
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center, 8210 Floyd Curl Drive, Mail code: 8257, San Antonio, TX 78229, USA; (A.M.H.); (C.-K.L.); (C.-N.H.); (C.-W.C.); (C.-M.W.); (C.-L.L.); (N.B.K.); (T.H.-M.H.)
| | - Brandon Lieberman
- Department of Biology, Trinity University, San Antonio, TX 78212, USA;
| | - Mirae Kim
- Department of Biomedical Engineering, University of Texas at Austin, 107 W. Dean Keeton, BME Building, Austin, TX 78712, USA; (M.K.); (R.V.); (E.P.P.); (K.B.); (T.E.Y.); (A.K.D.)
| | - Che-Kuang Lin
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center, 8210 Floyd Curl Drive, Mail code: 8257, San Antonio, TX 78229, USA; (A.M.H.); (C.-K.L.); (C.-N.H.); (C.-W.C.); (C.-M.W.); (C.-L.L.); (N.B.K.); (T.H.-M.H.)
| | - Chia-Nung Hung
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center, 8210 Floyd Curl Drive, Mail code: 8257, San Antonio, TX 78229, USA; (A.M.H.); (C.-K.L.); (C.-N.H.); (C.-W.C.); (C.-M.W.); (C.-L.L.); (N.B.K.); (T.H.-M.H.)
| | - Chih-Wei Chou
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center, 8210 Floyd Curl Drive, Mail code: 8257, San Antonio, TX 78229, USA; (A.M.H.); (C.-K.L.); (C.-N.H.); (C.-W.C.); (C.-M.W.); (C.-L.L.); (N.B.K.); (T.H.-M.H.)
| | - Chiou-Miin Wang
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center, 8210 Floyd Curl Drive, Mail code: 8257, San Antonio, TX 78229, USA; (A.M.H.); (C.-K.L.); (C.-N.H.); (C.-W.C.); (C.-M.W.); (C.-L.L.); (N.B.K.); (T.H.-M.H.)
| | - Chun-Lin Lin
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center, 8210 Floyd Curl Drive, Mail code: 8257, San Antonio, TX 78229, USA; (A.M.H.); (C.-K.L.); (C.-N.H.); (C.-W.C.); (C.-M.W.); (C.-L.L.); (N.B.K.); (T.H.-M.H.)
| | - Nameer B. Kirma
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center, 8210 Floyd Curl Drive, Mail code: 8257, San Antonio, TX 78229, USA; (A.M.H.); (C.-K.L.); (C.-N.H.); (C.-W.C.); (C.-M.W.); (C.-L.L.); (N.B.K.); (T.H.-M.H.)
| | - Michael A. Liss
- Department of Urology, University of Texas Health Science Center, San Antonio, TX 78229, USA;
| | - Rohan Vasisht
- Department of Biomedical Engineering, University of Texas at Austin, 107 W. Dean Keeton, BME Building, Austin, TX 78712, USA; (M.K.); (R.V.); (E.P.P.); (K.B.); (T.E.Y.); (A.K.D.)
| | - Evan P. Perillo
- Department of Biomedical Engineering, University of Texas at Austin, 107 W. Dean Keeton, BME Building, Austin, TX 78712, USA; (M.K.); (R.V.); (E.P.P.); (K.B.); (T.E.Y.); (A.K.D.)
| | - Katherine Blocher
- Department of Biomedical Engineering, University of Texas at Austin, 107 W. Dean Keeton, BME Building, Austin, TX 78712, USA; (M.K.); (R.V.); (E.P.P.); (K.B.); (T.E.Y.); (A.K.D.)
| | - Hannah Horng
- Department of Bioengineering, the University of Maryland, College Park, MD 20742, USA;
| | - Josephine A. Taverna
- Department of Medicine, Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX 78229, USA;
| | - Jianhua Ruan
- Department of Computer Science, University of Texas at San Antonio, San Antonio, TX 78249, USA;
| | - Thomas E. Yankeelov
- Department of Biomedical Engineering, University of Texas at Austin, 107 W. Dean Keeton, BME Building, Austin, TX 78712, USA; (M.K.); (R.V.); (E.P.P.); (K.B.); (T.E.Y.); (A.K.D.)
- Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX 78712, USA
- Department of Diagnostic Medicine, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA
- Livestrong Cancer Institutes, University of Texas at Austin, Austin, TX 78712, USA
| | - Andrew K. Dunn
- Department of Biomedical Engineering, University of Texas at Austin, 107 W. Dean Keeton, BME Building, Austin, TX 78712, USA; (M.K.); (R.V.); (E.P.P.); (K.B.); (T.E.Y.); (A.K.D.)
| | - Tim H.-M. Huang
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center, 8210 Floyd Curl Drive, Mail code: 8257, San Antonio, TX 78229, USA; (A.M.H.); (C.-K.L.); (C.-N.H.); (C.-W.C.); (C.-M.W.); (C.-L.L.); (N.B.K.); (T.H.-M.H.)
| | - Hsin-Chih Yeh
- Department of Biomedical Engineering, University of Texas at Austin, 107 W. Dean Keeton, BME Building, Austin, TX 78712, USA; (M.K.); (R.V.); (E.P.P.); (K.B.); (T.E.Y.); (A.K.D.)
- Texas Materials Institute, University of Texas at Austin, Austin, TX 78712, USA
- Correspondence: (H.-C.Y.); (C.-L.C.); Tel.: +1-512-471-7931 (H.-C.Y.); +1-210-562-4143 (C.-L.C.); Fax: +1-512-471-0616 (H.-C.Y.); +1-210-562-4161 (C.-L.C.)
| | - Chun-Liang Chen
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center, 8210 Floyd Curl Drive, Mail code: 8257, San Antonio, TX 78229, USA; (A.M.H.); (C.-K.L.); (C.-N.H.); (C.-W.C.); (C.-M.W.); (C.-L.L.); (N.B.K.); (T.H.-M.H.)
- Correspondence: (H.-C.Y.); (C.-L.C.); Tel.: +1-512-471-7931 (H.-C.Y.); +1-210-562-4143 (C.-L.C.); Fax: +1-512-471-0616 (H.-C.Y.); +1-210-562-4161 (C.-L.C.)
| |
Collapse
|
26
|
Parkin A, Man J, Timpson P, Pajic M. Targeting the complexity of Src signalling in the tumour microenvironment of pancreatic cancer: from mechanism to therapy. FEBS J 2019; 286:3510-3539. [PMID: 31330086 PMCID: PMC6771888 DOI: 10.1111/febs.15011] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 05/26/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer, a disease with extremely poor prognosis, has been notoriously resistant to virtually all forms of treatment. The dynamic crosstalk that occurs between tumour cells and the surrounding stroma, frequently mediated by intricate Src/FAK signalling, is increasingly recognised as a key player in pancreatic tumourigenesis, disease progression and therapeutic resistance. These important cues are fundamental for defining the invasive potential of pancreatic tumours, and several components of the Src and downstream effector signalling have been proposed as potent anticancer therapeutic targets. Consequently, numerous agents that block this complex network are being extensively investigated as potential antiinvasive and antimetastatic therapeutic agents for this disease. In this review, we will discuss the latest evidence of Src signalling in PDAC progression, fibrotic response and resistance to therapy. We will examine future opportunities for the development and implementation of more effective combination regimens, targeting key components of the oncogenic Src signalling axis, and in the context of a precision medicine-guided approach.
Collapse
Affiliation(s)
- Ashleigh Parkin
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
| | - Jennifer Man
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
| | - Paul Timpson
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
- Faculty of MedicineSt Vincent's Clinical SchoolUniversity of NSWSydneyAustralia
| | - Marina Pajic
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
- Faculty of MedicineSt Vincent's Clinical SchoolUniversity of NSWSydneyAustralia
| |
Collapse
|
27
|
3D-QSAR, molecular docking, and new compound design of pyrimidine derivatives as Src small molecule inhibitors. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02370-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
28
|
Miao W, Yuan J, Li L, Wang Y. Parallel-Reaction-Monitoring-Based Proteome-Wide Profiling of Differential Kinase Protein Expression during Prostate Cancer Metastasis in Vitro. Anal Chem 2019; 91:9893-9900. [PMID: 31241916 DOI: 10.1021/acs.analchem.9b01561] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Prostate cancer is the most common type of cancer in men, and kinases are heavily pursued as drug targets for anticancer therapy. In this study, we applied our recently reported parallel-reaction-monitoring (PRM)-based targeted proteomic method to examine the reprogramming of the human kinome associated with bone metastasis of prostate cancer in vitro. The method displayed superior sensitivity over the shotgun-proteomic approach, and it facilitated the quantification of the relative expression of 276 kinase proteins in a pair of bone metastatic prostate cancer cells. Among the differentially expressed kinases, mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) stimulates the migration and invasion of cultured prostate cancer cells, partially by modulating the activity of secreted matrix metalloproteinases 9 (MMP-9). We also found that the upregulation of MAP4K4 in metastatic prostate cancer cells is driven by the MYC proto-oncogene. Cumulatively, we identify MAP4K4 as a potential promoter for prostate cancer metastasis in vitro.
Collapse
|
29
|
Targeting the Hepatocyte Growth Factor and c-Met Signaling Axis in Bone Metastases. Int J Mol Sci 2019; 20:ijms20020384. [PMID: 30658428 PMCID: PMC6359064 DOI: 10.3390/ijms20020384] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 12/22/2022] Open
Abstract
Bone metastasis is the terminal stage disease of prostate, breast, renal, and lung cancers, and currently no therapeutic approach effectively cures or prevents its progression to bone metastasis. One of the hurdles to the development of new drugs for bone metastasis is the complexity and heterogeneity of the cellular components in the metastatic bone microenvironment. For example, bone cells, including osteoblasts, osteoclasts, and osteocytes, and the bone marrow cells of diverse hematopoietic lineages interact with each other via numerous cytokines and receptors. c-Met tyrosine kinase receptor and its sole ligand hepatocyte growth factor (HGF) are enriched in the bone microenvironment, and their expression correlates with the progression of bone metastasis. However, no drugs or antibodies targeting the c-Met/HGF signaling axis are currently available in bone metastatic patients. This significant discrepancy should be overcome by further investigation of the roles and regulation of c-Met and HGF in the metastatic bone microenvironment. This review paper summarizes the key findings of c-Met and HGF in the development of novel therapeutic approaches for bone metastasis.
Collapse
|
30
|
Singla N, Ghandour RA, Raj GV. Investigational luteinizing hormone releasing hormone (LHRH) agonists and other hormonal agents in early stage clinical trials for prostate cancer. Expert Opin Investig Drugs 2019; 28:249-259. [PMID: 30649971 DOI: 10.1080/13543784.2019.1570130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The treatment and management of prostate cancer continues to evolve; newer classes of agents and combination therapies are being developed and some are being investigated in early phase clinical trials. AREAS COVERED We discuss investigational hormonal agents for the treatment of prostate cancer and focus primarily on luteinizing hormone releasing hormone (LHRH) agonists in early stage trials. We look at agents that target the hormonal axis, including anti-androgens, gonadotropins, estrogenic agents and progestogenic agents and other non-hormonal agents often used in combination with LHRH agonists. We review these candidates in the specific clinical niche in which they might find utility. EXPERT OPINION Of all candidate compounds being evaluated in clinical trials, very few will receive FDA approval. Few, if any of the investigational agents discussed here will be used routinely in clinical practice for treating prostate cancer. Recognizing the reasons for the failure of agents to advance to later stage trials is important. Furthermore, a thorough understanding of the mechanisms underlying prostate cancer pathogenesis, including various points in the HGPA and parallel pathways, will help identify potentially actionable targets.
Collapse
Affiliation(s)
- Nirmish Singla
- a Department of Urology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Rashed A Ghandour
- a Department of Urology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Ganesh V Raj
- a Department of Urology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| |
Collapse
|
31
|
Luo D, Zhan S, Xia W, Huang L, Ge W, Wang T. Proteomics study of serum exosomes from papillary thyroid cancer patients. Endocr Relat Cancer 2018; 25:879-891. [PMID: 29895528 DOI: 10.1530/erc-17-0547] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 06/11/2018] [Indexed: 01/11/2023]
Abstract
Lymph node metastasis (LNM) in papillary thyroid cancer (PTC) is related to increased risk of recurrence and poor prognosis. Tumour exosomes have been shown to be associated with metastasis of cancer cells. Therefore, we aim to identify the characteristics and biological functions of serum exosomes in lymph node metastases of PTC. We compared proteome profiles of serum-purified exosomes (SPEs) from PTC patients with LNM, PTC patients without LNM, and healthy donors, using a combination of liquid chromatography-tandem mass spectroscopy analyses and tandem mass tag label quantitation analysis. We identified 1569 proteins by two or more unique peptides. Compared with the SPEs of PTC patients without LNM, we found 697 differentially expressed proteins in the SPEs of PTC patients with LNM. Our results revealed overexpression of specific proteins with well-established links to cancer cell metastasis, such as SRC, TLN1, ITGB2 and CAPNS1. Consistent with mass spectrum results, we performed Western blot to detect the expression of these proteins in individual sample. Biological pathway analyses showed that integrin signalling was aberrantly activated in the SPEs of PTC patients with LNM compared to those without LNM. Our study reveals that SPEs of PTC patients with lymph node metastases promote BHT101 thyroid cancer cell invasiveness, but have no apparent influence on cell migration. In the serum exosomes of PTC patients with LNM, integrin-associated proteins are obviously upregulated. These proteomic findings will contribute to elucidation of the pathophysiological functions of tumour-derived exosomes.
Collapse
Affiliation(s)
- Dan Luo
- National Key Laboratory of Medical Molecular Biology & Department of ImmunologyInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Shaohua Zhan
- National Key Laboratory of Medical Molecular Biology & Department of ImmunologyInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenchao Xia
- National Key Laboratory of Medical Molecular Biology & Department of ImmunologyInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Liang Huang
- Department of Blood TransfusionTongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Ge
- National Key Laboratory of Medical Molecular Biology & Department of ImmunologyInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Tianxiao Wang
- Department of Head and Neck SurgeryKey Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
32
|
Tan J, Liu Y, Maimaiti Y, Wang C, Yan Y, Zhou J, Ruan S, Huang T. Combination of SIRT1 and Src overexpression suggests poor prognosis in luminal breast cancer. Onco Targets Ther 2018; 11:2051-2061. [PMID: 29695913 PMCID: PMC5905521 DOI: 10.2147/ott.s162503] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Objectives 1) Analyze the correlation of SIRT1 and Src with human breast cancer (BC) prognosis; 2) explore the roles of SIRT1 and Src in BC cell proliferation, tumor invasion, and metastasis; and 3) analyze the correlation and interaction between SIRT1 and Src. Materials and methods 1) Tissue microarray was used to analyze the expression of SIRT1 and Src in human BC tissues and the correlation between protein expression and cancer prognosis; 2) CCK8 assay was used to determine the influence of SIRT1 and Src inhibitors on BC cell proliferation; 3) Transwell migration assay and wound healing assay were used to determine the effect of SIRT1 and Src inhibitors on BC cell migration and invasion; and 4) Western blotting was used to analyze the correlation and interaction between SIRT1 and Src. Results 1) Combination of SIRT1 and/or Src positivity is a prognosis factor in BC, especially in luminal type; 2) MCF-7 cell proliferation is suppressed by SIRT1 inhibitor Ex527, and cell migration and invasion were inhibited by Src inhibitor bosutinib; 3) combined with Ex527, bosutinib has a significantly increased effect on MCF-7 cell migration suppression; and 4) there is a positive association between SIRT1 and Src both in BC tissues and in MCF-7 cells. Conclusion 1) SIRT1 and Src overexpression are both correlated with poor prognosis in human BC; 2) SIRT1 + Src (SIRT1 and/or Src positivity) is a fine prognosis model for luminal-type BC; 3) SIRT1 is a copromotor of Src in BC migration and invasion, but not in cell proliferation; and 4) our results suggest a potential interaction or a common regulation pathway between SIRT1 and Src expression and activity.
Collapse
Affiliation(s)
- Jie Tan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuyin Liu
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yusufu Maimaiti
- Department of General Surgery, Research Institute of Minimally Invasive, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Changwen Wang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Yan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zhou
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengnan Ruan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
33
|
Ramroop JR, Stein MN, Drake JM. Impact of Phosphoproteomics in the Era of Precision Medicine for Prostate Cancer. Front Oncol 2018; 8:28. [PMID: 29503809 PMCID: PMC5820335 DOI: 10.3389/fonc.2018.00028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/29/2018] [Indexed: 11/13/2022] Open
Abstract
Prostate cancer is the most common malignancy in men in the United States. While androgen deprivation therapy results in tumor responses initially, there is relapse and progression to metastatic castration-resistant prostate cancer. Currently, all prostate cancer patients receive essentially the same treatment, and there is a need for clinically applicable technologies to provide predictive biomarkers toward personalized therapies. Genomic analyses of tumors are used for clinical applications, but with a paucity of obvious driver mutations in metastatic castration-resistant prostate cancer, other applications, such as phosphoproteomics, may complement this approach. Immunohistochemistry and reverse phase protein arrays are limited by the availability of reliable antibodies and evaluates a preselected number of targets. Mass spectrometry-based phosphoproteomics has been used to profile tumors consisting of thousands of phosphopeptides from individual patients after surgical resection or at autopsy. However, this approach is time consuming, and while a large number of candidate phosphopeptides are obtained for evaluation, limitations are reduced reproducibility, sensitivity, and precision. Targeted mass spectrometry can help eliminate these limitations and is more cost effective and less time consuming making it a practical platform for future clinical testing. In this review, we discuss the use of phosphoproteomics in prostate cancer and other clinical cancer tissues for target identification, hypothesis testing, and possible patient stratification. We highlight the majority of studies that have used phosphoproteomics in prostate cancer tissues and cell lines and propose ways forward to apply this approach in basic and clinical research. Overall, the implementation of phosphoproteomics via targeted mass spectrometry has tremendous potential to aid in the development of more rational, personalized therapies that will result in increased survival and quality of life enhancement in patients suffering from metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Johnny R. Ramroop
- Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Mark N. Stein
- Developmental Therapeutics/Phase I Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
- Department of Medicine, Division of Medical Oncology and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Justin M. Drake
- Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
- Department of Medicine, Division of Medical Oncology and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| |
Collapse
|
34
|
Begemann D, Anastos H, Kyprianou N. Cell death under epithelial-mesenchymal transition control in prostate cancer therapeutic response. Int J Urol 2018; 25:318-326. [DOI: 10.1111/iju.13505] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 11/05/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Diane Begemann
- Department of Urology; University of Kentucky College of Medicine; Lexington Kentucky USA
- Department of Toxicology and Cancer Biology; University of Kentucky College of Medicine; Lexington Kentucky USA
| | - Harry Anastos
- Department of Urology; University of Kentucky College of Medicine; Lexington Kentucky USA
| | - Natasha Kyprianou
- Department of Urology; University of Kentucky College of Medicine; Lexington Kentucky USA
- Department of Toxicology and Cancer Biology; University of Kentucky College of Medicine; Lexington Kentucky USA
- Department of Molecular Biochemistry; University of Kentucky College of Medicine; Lexington Kentucky USA
| |
Collapse
|
35
|
Abstract
Dasatinib is an oral available short-acting inhibitor of multiple tyrosine kinases. It was designed to inhibit ABL and SRC, but also has activity in multiple other kinases, including c-KIT, PDGFR-α, PDGFR-β, and ephrin receptor kinases. Dasatinib is a very potent inhibitor of BCR-ABL and an effective treatment for the BCR-ABL-driven diseases chronic myeloid leukemia (CML) and Philadelphia-chromosome-positive acute lymphoblastic leukemia (Ph+ ALL), characterized by the constitutively active tyrosine kinase, BCR-ABL. Dasatinib is approved for the treatment of CML (all phases) including children and for the treatment of Ph+ ALL, resistant or intolerant to prior imatinib treatment. Randomized trials in CML comparing dasatinib with imatinib show that first-line dasatinib causes significantly deeper and faster molecular remissions. In accelerated and blastic phase CML, as well as in Ph+ ALL, dasatinib frequently induces complete hematologic and cytogenetic remissions even in imatinib pretreated patients. Remissions however are often short. Dasatinib is administered independent of food intake as a once-daily dose of 100 mg in chronic phase CML and 140 mg in Ph+ ALL or blastic phase. Side effects of dasatinib are frequent but mostly moderate and manageable and include cytopenias and pleural effusions. The review presents the preclinical and clinical activity of dasatinib with a focus on clinical studies in CML.
Collapse
Affiliation(s)
- Markus Lindauer
- Klinik für Innere Medizin III, Klinikum am Gesundbrunnen, Am Gesundbrunnen 20-24, 74078, Heilbronn, Germany.
| | - Andreas Hochhaus
- Abteilung Hämatologie/Onkologie, Klinik für Innere Medizin II, Universitätsklinikum Jena, Erlanger Allee 101, 07740, Jena, Germany
| |
Collapse
|
36
|
Kong X, Wu W, Yuan Y, Pandey V, Wu Z, Lu X, Zhang W, Chen Y, Wu M, Zhang M, Li G, Tan S, Qian P, Perry JK, Lobie PE, Zhu T. Human growth hormone and human prolactin function as autocrine/paracrine promoters of progression of hepatocellular carcinoma. Oncotarget 2017; 7:29465-79. [PMID: 27102295 PMCID: PMC5045410 DOI: 10.18632/oncotarget.8781] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 03/24/2016] [Indexed: 11/25/2022] Open
Abstract
The death rates of hepatocellular carcinoma (HCC) are extremely high due to the paucity of therapeutic options. Animal models and anecdotal clinical evidence indicate a potential role of hGH and hPRL in HCC. However, the prognostic relevance and the functional role of tumor expression of these hormones in human HCC are not defined. Herein, we analyzed the mRNA and protein expression of hGH and hPRL in histopathological samples of non-neoplastic liver and HCC by in situ hybridization, PCR and immunohistochemistry techniques. Increased mRNA and protein expression of both hormones was observed in HCC compared with non-neoplastic liver tissues. hGH expression was significantly associated with tumor size and tumor grade. No significant association was observed between the expression of hPRL and any histopathological features. Amplification of both hGH and hPRL genes in HCC was observed when compared to non-neoplastic tissue. Expression of both hGH and hPRL was associated with worse relapse-free and overall survival in HCC patients. In vitro and in vivo functional assays performed with HCC cell lines demonstrated that autocrine expression of hGH or hPRL in HCC cells increased STAT3 activation, oncogenicity and tumor growth while functional antagonism with hGH-G120R significantly reduced these parameters. Hence, tumor expression of hGH/hPRL is associated with a worse survival outcome for patients with HCC and hGH/hPRL function as autocrine/paracrine promoters of HCC progression.
Collapse
Affiliation(s)
- Xiangjun Kong
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Wenyong Wu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yan Yuan
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Vijay Pandey
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore
| | - Zhengsheng Wu
- Department of Pathology, Anhui Medical University, Hefei, Anhui, China
| | - Xuefei Lu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Weijie Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Yijun Chen
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore
| | - Mingming Wu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Min Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Gaopeng Li
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Sheng Tan
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Pengxu Qian
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| | - Jo K Perry
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Peter E Lobie
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore.,National University Cancer Institute of Singapore, National University Health System, Singapore
| | - Tao Zhu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China
| |
Collapse
|
37
|
Cao Z, Livas T, Kyprianou N. Anoikis and EMT: Lethal "Liaisons" during Cancer Progression. Crit Rev Oncog 2017; 21:155-168. [PMID: 27915969 DOI: 10.1615/critrevoncog.2016016955] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Anoikis is a unique mode of apoptotic cell death that occurs consequentially to insufficient cell-matrix interactions. Resistance to anoikis is a critical contributor to tumor invasion and metastasis. The phenomenon is regulated by integrins, which upon engagement with components of the extracellular matrix (ECM) form adhesion complexes and the actin cytoskeleton drives the formation of cell protrusions used to adhere to ECM, directing cell migration. The epithelial-mesenchymal transition (EMT) confers stem cell properties and leads to acquisition of a migratory and invasive phenotype by causing adherens junction breakdown and circumventing anoikis in the tumor microenvironment. The investigation of drug discovery platforms for apoptosis-driven therapeutics identified several novel agents with antitumor action via reversing resistance to anoikis, inhibiting survival pathways and impacting the EMT landscape in human cancer. In this review, we discuss current evidence on the contribution of the anoikis phenomenon functionally linked to EMT to cancer metastasis and the therapeutic value of antitumor drugs that selectively reverse anoikis resistance and/or EMT to impair tumor progression toward the development/optimization of apoptosis-driven therapeutic targeting of metastatic disease.
Collapse
Affiliation(s)
- Zheng Cao
- Department of Urology, Molecular Biochemistry, Pathology, Toxicology & Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, U.S.A
| | - Theodore Livas
- Department of Urology, Molecular Biochemistry, Pathology, Toxicology & Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, U.S.A
| | - Natasha Kyprianou
- Department of Urology, Molecular Biochemistry, Pathology, Toxicology & Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, U.S.A
| |
Collapse
|
38
|
Resistance to dasatinib is associated with the activation of Akt in oral squamous cell carcinoma. TRANSLATIONAL RESEARCH IN ORAL ONCOLOGY 2017. [DOI: 10.1177/2057178x17702920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
39
|
Zhang K, Myllymäki SM, Gao P, Devarajan R, Kytölä V, Nykter M, Wei GH, Manninen A. Oncogenic K-Ras upregulates ITGA6 expression via FOSL1 to induce anoikis resistance and synergizes with αV-Class integrins to promote EMT. Oncogene 2017; 36:5681-5694. [PMID: 28604746 PMCID: PMC5658677 DOI: 10.1038/onc.2017.177] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 04/11/2017] [Accepted: 05/04/2017] [Indexed: 12/17/2022]
Abstract
In many cancer types, integrin-mediated signaling regulates proliferation, survival and invasion of tumorigenic cells. However, it is still unclear how integrins crosstalk with oncogenes to regulate tumorigenesis and metastasis. Here we show that oncogenic K-RasV12 upregulates α6-integrin expression in Madin–Darby canine kidney (MDCK) cells via activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK)/Fos-related antigen 1-signaling cascade. Activated α6-integrins promoted metastatic capacity and anoikis resistance, and led to perturbed growth of MDCK cysts. Transcriptomic analysis of K-RasV12-transformed MDCK cells also revealed robust downregulation of αV-class integrins. Re-expression of αV-integrin in K-RasV12-transformed MDCK cells synergistically upregulated the expression of Zinc finger E-box-binding homeobox 1 and Twist-related protein 1 and triggered epithelial-mesenchymal transition leading to induced cell motility and invasion. These results delineate the signaling cascades connecting oncogenic K-RasV12 with α6- and αV-integrin functions to modulate cancer cell survival and tumorigenesis, and reveal new possible strategies to target highly oncogenic K-RasV12 mutants.
Collapse
Affiliation(s)
- K Zhang
- Biocenter Oulu, Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - S-M Myllymäki
- Biocenter Oulu, Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - P Gao
- Biocenter Oulu, Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - R Devarajan
- Biocenter Oulu, Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - V Kytölä
- Prostate Cancer Research Center, Institute of Biomedical Technology and BioMediTech, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - M Nykter
- Prostate Cancer Research Center, Institute of Biomedical Technology and BioMediTech, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - G-H Wei
- Biocenter Oulu, Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - A Manninen
- Biocenter Oulu, Centre of Excellence in Cell-Extracellular Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
40
|
Castoria G, Auricchio F, Migliaccio A. Extranuclear partners of androgen receptor: at the crossroads of proliferation, migration, and neuritogenesis. FASEB J 2016; 31:1289-1300. [PMID: 28031322 DOI: 10.1096/fj.201601047r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/19/2016] [Indexed: 01/11/2023]
Abstract
In this review, we focus on the role played by the protein partners of ligand-activated extranuclear androgen receptor (AR) in the final effects of hormone action, such as proliferation, migration, and neuritogenesis. The choice of AR partner, at least in part, depends on cell type. Androgen-activated receptor directly associates with cytoplasmic Src tyrosine kinase in epithelial cells, whereas in mesenchymal and neuronal cells, it prevalently interacts with filamin A. In the former, proliferation represents the final hormonal outcome, whereas in the latter, either migration or neuritogenesis, respectively, occurs. Furthermore, AR partner filamin A is replaced with Src when mesenchymal cells are stimulated with very low androgen concentrations. Consequently, the migratory effect is replaced by mitogenesis. Use of peptides that prevent receptor/partner assembly abolishes the effects that are dependent on their association and offers new therapeutic approaches to AR-related diseases. Perturbation of migration is often associated with metastatic spreading in cancer. In turn, cell cycle aberration causes tumors to grow faster, whereas toxic signaling triggers neurodegenerative events in the CNS. Here, we provide examples of new tools that interfere in rapid androgen effects, including migration, proliferation, and neuronal differentiation, together with their potential therapeutic applications in AR-dependent diseases-mainly prostate cancer and neurodegenerative disorders.-Castoria, G., Auricchio, F., Migliaccio, A. Extranuclear partners of androgen receptor: at the crossroads of proliferation, migration, and neuritogenesis.
Collapse
Affiliation(s)
- Gabriella Castoria
- Department of Biochemistry, Biophysics, and General Pathology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Ferdinando Auricchio
- Department of Biochemistry, Biophysics, and General Pathology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Antimo Migliaccio
- Department of Biochemistry, Biophysics, and General Pathology, University of Campania "Luigi Vanvitelli," Naples, Italy
| |
Collapse
|
41
|
Zhang M, Zhang W, Wu Z, Liu S, Sun L, Zhong Y, Zhang X, Kong X, Qian P, Zhang H, Lobie PE, Zhu T. Artemin is hypoxia responsive and promotes oncogenicity and increased tumor initiating capacity in hepatocellular carcinoma. Oncotarget 2016; 7:3267-82. [PMID: 26675549 PMCID: PMC4823105 DOI: 10.18632/oncotarget.6572] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/21/2015] [Indexed: 12/28/2022] Open
Abstract
Hypoxia has been reported to regulate the cancer stem cell (CSC) population yet the underlying mechanism is poorly characterized. Herein, we show that Artemin (ARTN), a member of the glial cell derived neurotrophic factor family of ligands, is a hypoxia-responsive factor and is essential for hypoxia-induced CSC expansion in hepatocellular carcinoma (HCC). Clinically, elevated expression of ARTN in HCC was associated with larger tumor size, faster relapse and shorter survival. In vitro, HCC cells with forced expression of ARTN exhibited reduced apoptosis, increased proliferation, epithelial-mesenchymal transition (EMT) and enhanced motility. Additionally, ARTN dramatically increased xenograft tumor size and metastasis in vivo. Moreover, ARTN also enhanced tumorsphere formation and the tumor initiating capacity of HCC cells, consequent to expansion of the CD133+ CSC population. ARTN transcription was directly activated by hypoxia-induced factor-1α (HIF-1α) and hypoxia induced ARTN promoted EMT and increased the CSC population via AKT signaling. We herein identify a novel HIF-1α/ARTN axis promoting CSC-like behavior in hypoxic environments which implicates ARTN as a valuable therapeutic target for HCC.
Collapse
Affiliation(s)
- Min Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Weijie Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Zhengsheng Wu
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Shumin Liu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Linchong Sun
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Yanghao Zhong
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Xiao Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Xiangjun Kong
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Pengxu Qian
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Huafeng Zhang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Peter E Lobie
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore
| | - Tao Zhu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| |
Collapse
|
42
|
Mao L, Deng WW, Yu GT, Bu LL, Liu JF, Ma SR, Wu L, Kulkarni AB, Zhang WF, Sun ZJ. Inhibition of SRC family kinases reduces myeloid-derived suppressor cells in head and neck cancer. Int J Cancer 2016; 140:1173-1185. [PMID: 27798955 DOI: 10.1002/ijc.30493] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/11/2016] [Accepted: 10/24/2016] [Indexed: 12/30/2022]
Abstract
SRC family kinases (SFKs), a group of nonreceptor tyrosine kinases, modulate multiple cellular functions, such as cell proliferation, differentiation and metabolism. SFKs display aberrant activity in progressive stages of human cancers. However, the precise role of SFKs in the head and neck squamous cell carcinoma (HNSCC) signaling network is far from clear. In this study, we found that the inhibition of SFKs activity by dasatinib effectively reduced the tumor size and population of MDSCs in the HNSCC mouse model. Molecular analysis indicates that phosphorylation of LYN, rather than SRC, was inhibited by dasatinib treatment. Next, we analyzed LYN expression by immunostaining and found that it was overexpressed in the human HNSCC specimens. Moreover, LYN expression in stromal cells positively correlated with myeloid-derived suppressor cells (MDSCs) makers CD11b and CD33 in human HNSCC. The dual positive expression of LYN in epithelial and stromal cells (EPI+ SRT+ ) was associated with unfavorable overall survival of HNSCC patients. These findings indicate that SFKs may be a potential target for an effective immunotherapy of HNSCC by decreasing MDSCs and moreover, LYN will have an impact on such therapeutic strategy.
Collapse
Affiliation(s)
- Liang Mao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei-Wei Deng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guang-Tao Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lin-Lin Bu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jian-Feng Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Si-Rui Ma
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lei Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ashok B Kulkarni
- Functional Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Wen-Feng Zhang
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
43
|
SRC family kinase FYN promotes the neuroendocrine phenotype and visceral metastasis in advanced prostate cancer. Oncotarget 2016; 6:44072-83. [PMID: 26624980 PMCID: PMC4792542 DOI: 10.18632/oncotarget.6398] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 11/14/2015] [Indexed: 01/08/2023] Open
Abstract
FYN is a SRC family kinase (SFK) that has been shown to be up-regulated in human prostate cancer (PCa) tissues and cell lines. In this study, we observed that FYN is strongly up-regulated in human neuroendocrine PCa (NEPC) tissues and xenografts, as well as cells derived from a NEPC transgenic mouse model. In silico analysis of FYN expression in prostate cancer cell line databases revealed an association with the expression of neuroendocrine (NE) markers such as CHGA, CD44, CD56, and SYP. The loss of FYN abrogated the invasion of PC3 and ARCaPM cells in response to MET receptor ligand HGF. FYN also contributed to the metastatic potential of NEPC cells in two mouse models of visceral metastasis with two different cell lines (PC3 and TRAMPC2-RANKL). The activation of MET appeared to regulate neuroendocrine (NE) features as evidenced by increased expression of NE markers in PC3 cells with HGF. Importantly, the overexpression of FYN protein in DU145 cells was directly correlated with the increase of CHGA. Thus, our data demonstrated that the neuroendocrine differentiation that occurs in PCa cells is, at least in part, regulated by FYN kinase. Understanding the role of FYN in the regulation of NE markers will provide further support for ongoing clinical trials of SFK and MET inhibitors in castration-resistant PCa patients.
Collapse
|
44
|
Tampellini M, La Salvia A, Scagliotti GV. Novel investigational therapies for treating biliary tract carcinoma. Expert Opin Investig Drugs 2016; 25:1423-1436. [PMID: 27771967 DOI: 10.1080/13543784.2016.1252330] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Cholangiocarcinoma (CCA) is an epithelial cell malignancy arising from bile ducts and/or peribiliary glands. Even though it is considered as a rare neoplasm, its incidence is raising, particularly in developed countries. Prognosis is generally poor with few patients who present the inclusion criteria for surgery (the mainstay treatment for this tumour). Several genetic alterations potentially driving tumour progression have been described, representing a possible target for new compounds. Areas covered: A clinical trial search in Clinicaltrials.gov encompassing a literature search in PubMed and ASCO/ESMO Websites was undertaken in March 2016. Expert opinion: Notwithstanding a large number of drug tested, results are still disappointing. The main reasons could be the low number of patients enrolled in trials, and the lack of a patient selection based on the biological profile of the tumours. Potential active drugs could have been discharged simply because beneficial in a particular subgroup of patients and not in un unselected population. The future direction of the research should consider biomarker evaluation in order to describe the genetic alteration/s that drive tumour progression and aggressiveness and the mechanisms of drug resistance. Finally, it will be of great interest to consider the results of immunotherapy whenever available.
Collapse
Affiliation(s)
- M Tampellini
- a Department of Oncology, AOU San Luigi di Orbassano , University of Turin , Torino , Italy
| | - A La Salvia
- a Department of Oncology, AOU San Luigi di Orbassano , University of Turin , Torino , Italy
| | - G V Scagliotti
- a Department of Oncology, AOU San Luigi di Orbassano , University of Turin , Torino , Italy
| |
Collapse
|
45
|
The molecular effect of metastasis suppressors on Src signaling and tumorigenesis: new therapeutic targets. Oncotarget 2016; 6:35522-41. [PMID: 26431493 PMCID: PMC4742122 DOI: 10.18632/oncotarget.5849] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 08/15/2015] [Indexed: 02/07/2023] Open
Abstract
A major problem for cancer patients is the metastasis of cancer cells from the primary tumor. This involves: (1) migration through the basement membrane; (2) dissemination via the circulatory system; and (3) invasion into a secondary site. Metastasis suppressors, by definition, inhibit metastasis at any step of the metastatic cascade. Notably, Src is a non-receptor, cytoplasmic, tyrosine kinase, which becomes aberrantly activated in many cancer-types following stimulation of plasma membrane receptors (e.g., receptor tyrosine kinases and integrins). There is evidence of a prominent role of Src in tumor progression-related events such as the epithelial–mesenchymal transition (EMT) and the development of metastasis. However, the precise molecular interactions of Src with metastasis suppressors remain unclear. Herein, we review known metastasis suppressors and summarize recent advances in understanding the mechanisms of how these proteins inhibit metastasis through modulation of Src. Particular emphasis is bestowed on the potent metastasis suppressor, N-myc downstream regulated gene 1 (NDRG1) and its interactions with the Src signaling cascade. Recent studies demonstrated a novel mechanism through which NDRG1 plays a significant role in regulating cancer cell migration by inhibiting Src activity. Moreover, we discuss the rationale for targeting metastasis suppressor genes as a sound therapeutic modality, and we review several examples from the literature where such strategies show promise. Collectively, this review summarizes the essential interactions of metastasis suppressors with Src and their effects on progression of cancer metastasis. Moreover, interesting unresolved issues regarding these proteins as well as their potential as therapeutic targets are also discussed.
Collapse
|
46
|
Yi F, Tan XL, Yan X, Liu HB. In silico profiling for secondary metabolites from Lepidium meyenii (maca) by the pharmacophore and ligand-shape-based joint approach. Chin Med 2016; 11:42. [PMID: 27708692 PMCID: PMC5037646 DOI: 10.1186/s13020-016-0112-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 09/19/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Lepidium meyenii Walpers (maca) is an herb known as a traditional nutritional supplement and widely used in Peru, North America, and Europe to enhance human fertility and treat osteoporosis. The secondary metabolites of maca, namely, maca alkaloids, macaenes, and macamides, are bioactive compounds, but their targets are undefined. METHODS The pharmacophore-based PharmaDB targets database screening joint the ligand shape similarity-based WEGA validation approach is proposed to predict the targets of these unique constituents and was performed using Discovery Studio 4.5 and PharmaDB. A compounds-targets-diseases network was established using Cytoscape 3.2. These suitable targets and their genes were calculated and analyzed using ingenuity pathway analysis and GeneMANIA. RESULTS Certain targets were identified in osteoporosis (8 targets), prostate cancer (9 targets), and kidney diseases (11 targets). This was the first study to identify the targets of these bioactive compounds in maca for cardiovascular diseases (29 targets). The compound with the most targets (46) was an amide alkaloid (MA-24). CONCLUSION In silico target fishing identified maca's traditional effects on treatment and prevention of osteoporosis, prostate cancer, and kidney diseases, and its potential function of treating cardiovascular diseases, as the most important of this herb's possible activities.
Collapse
Affiliation(s)
- Fan Yi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, 151 Malianwa N, Haidian District, Beijing, 100193 China ; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, 100193 China
| | - Xiao-Lei Tan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, 151 Malianwa N, Haidian District, Beijing, 100193 China ; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, 100193 China
| | - Xin Yan
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Circle at University City, Guangzhou, 510006 China
| | - Hai-Bo Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, 151 Malianwa N, Haidian District, Beijing, 100193 China ; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, 100193 China
| |
Collapse
|
47
|
Shahryari V, Nip H, Saini S, Dar AA, Yamamura S, Mitsui Y, Colden M, Bucay N, Tabatabai LZ, Greene K, Deng G, Tanaka Y, Dahiya R, Majid S. Pre-clinical Orthotopic Murine Model of Human Prostate Cancer. J Vis Exp 2016. [PMID: 27684100 DOI: 10.3791/54125] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
To study the multifaceted biology of prostate cancer, pre-clinical in vivo models offer a range of options to uncover critical biological information about this disease. The human orthotopic prostate cancer xenograft mouse model provides a useful alternative approach for understanding the specific interactions between genetically and molecularly altered tumor cells, their organ microenvironment, and for evaluation of efficacy of therapeutic regimens. This is a well characterized model designed to study the molecular events of primary tumor development and it recapitulates the early events in the metastatic cascade prior to embolism and entry of tumor cells into the circulation. Thus it allows elucidation of molecular mechanisms underlying the initial phase of metastatic disease. In addition, this model can annotate drug targets of clinical relevance and is a valuable tool to study prostate cancer progression. In this manuscript we describe a detailed procedure to establish a human orthotopic prostate cancer xenograft mouse model.
Collapse
Affiliation(s)
| | - Hannah Nip
- Department of Urology, VA Medical Center and UCSF
| | | | - Altaf A Dar
- California Pacific Medical Center Research Institute
| | | | - Yozo Mitsui
- Department of Urology, VA Medical Center and UCSF
| | | | - Nathan Bucay
- Department of Urology, VA Medical Center and UCSF
| | | | | | - Guoren Deng
- Department of Urology, VA Medical Center and UCSF
| | | | | | | |
Collapse
|
48
|
Zarif JC, Miranti CK. The importance of non-nuclear AR signaling in prostate cancer progression and therapeutic resistance. Cell Signal 2016; 28:348-356. [PMID: 26829214 PMCID: PMC4788534 DOI: 10.1016/j.cellsig.2016.01.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 01/28/2016] [Indexed: 01/22/2023]
Abstract
The androgen receptor (AR) remains the major oncogenic driver of prostate cancer, as evidenced by the efficacy of androgen deprivation therapy (ADT) in naïve patients, and the continued effectiveness of second generation ADTs in castration resistant disease. However, current ADTs are limited to interfering with AR ligand binding, either through suppression of androgen production or the use of competitive antagonists. Recent studies demonstrate 1) the expression of constitutively active AR splice variants that no longer depend on androgen, and 2) the ability of AR to signal in the cytoplasm independently of its transcriptional activity (non-genomic); thus highlighting the need to consider other ways to target AR. Herein, we review canonical AR signaling, but focus on AR non-genomic signaling, some of its downstream targets and how these effectors contribute to prostate cancer cell behavior. The goals of this review are to 1) re-highlight the continued importance of AR in prostate cancer as the primary driver, 2) discuss the limitations in continuing to use ligand binding as the sole targeting mechanism, 3) discuss the implications of AR non-genomic signaling in cancer progression and therapeutic resistance, and 4) address the need to consider non-genomic AR signaling mechanisms and pathways as a viable targeting strategy in combination with current therapies.
Collapse
Affiliation(s)
- Jelani C Zarif
- The James Buchanan Brady Urological Institute at The Johns Hopkins University School of Medicine Baltimore, MD 21287, United States
| | - Cindy K Miranti
- Lab of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, United States.
| |
Collapse
|
49
|
Chatterji T, Varkaris AS, Parikh NU, Song JH, Cheng CJ, Schweppe RE, Alexander S, Davis JW, Troncoso P, Friedl P, Kuang J, Lin SH, Gallick GE. Yes-mediated phosphorylation of focal adhesion kinase at tyrosine 861 increases metastatic potential of prostate cancer cells. Oncotarget 2016; 6:10175-94. [PMID: 25868388 PMCID: PMC4496348 DOI: 10.18632/oncotarget.3391] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/16/2015] [Indexed: 01/15/2023] Open
Abstract
To study the role of FAK signaling complexes in promoting metastatic properties of prostate cancer (PCa) cells, we selected stable, highly migratory variants, termed PC3 Mig-3 and DU145 Mig-3, from two well-characterized PCa cell lines, PC3 and DU145. These variants were not only increased migration and invasion in vitro, but were also more metastatic to lymph nodes following intraprostatic injection into nude mice. Both PC3 Mig-3 and DU145 Mig-3 were specifically increased in phosphorylation of FAK Y861. We therefore examined potential alterations in Src family kinases responsible for FAK phosphorylation and determined only Yes expression was increased. Overexpression of Yes in PC3 parental cells and src-/-fyn-/-yes-/- fibroblasts selectively increased FAK Y861 phosphorylation, and increased migration. Knockdown of Yes in PC3 Mig-3 cells decreased migration and decreased lymph node metastasis following orthotopic implantation of into nude mice. In human specimens, Yes expression was increased in lymph node metastases relative to paired primary tumors from the same patient, and increased pFAK Y861 expression in lymph node metastases correlated with poor prognosis. These results demonstrate a unique role for Yes in phosphorylation of FAK and in promoting PCa metastasis. Therefore, phosphorylated FAK Y861 and increased Yes expression may be predictive markers for PCa metastasis.
Collapse
Affiliation(s)
- Tanushree Chatterji
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Programs in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, TX, USA
| | - Andreas S Varkaris
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nila U Parikh
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jian H Song
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chien-Jui Cheng
- Department of Pathology, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pathology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Rebecca E Schweppe
- Division of Endocrinology, Metabolism, and Diabetes, and Department of Pathology, University of Colorado Anschutz Medical Campus, University of Colorado Cancer Center, Aurora, CO, USA
| | - Stephanie Alexander
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Cell Biology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - John W Davis
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patricia Troncoso
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Peter Friedl
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Division of Endocrinology, Metabolism, and Diabetes, and Department of Pathology, University of Colorado Anschutz Medical Campus, University of Colorado Cancer Center, Aurora, CO, USA
| | - Jian Kuang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sue-Hwa Lin
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Programs in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, TX, USA.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gary E Gallick
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Programs in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, TX, USA
| |
Collapse
|
50
|
Liu W, Yue F, Zheng M, Merlot A, Bae DH, Huang M, Lane D, Jansson P, Lui GYL, Richardson V, Sahni S, Kalinowski D, Kovacevic Z, Richardson DR. The proto-oncogene c-Src and its downstream signaling pathways are inhibited by the metastasis suppressor, NDRG1. Oncotarget 2016; 6:8851-74. [PMID: 25860930 PMCID: PMC4496188 DOI: 10.18632/oncotarget.3316] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 02/08/2015] [Indexed: 11/25/2022] Open
Abstract
N-myc downstream regulated gene-1 (NDRG1) is a potent metastasis suppressor that plays a key role in regulating signaling pathways involved in mediating cancer cell invasion and migration, including those derived from prostate, colon, etc. However, the mechanisms and molecular targets through which NDRG1 reduces cancer cell invasion and migration, leading to inhibition of cancer metastasis, are not fully elucidated. In this investigation, using NDRG1 over-expression models in three tumor cell-types (namely, DU145, PC3MM and HT29) and also NDRG1 silencing in DU145 and HT29 cells, we reveal that NDRG1 decreases phosphorylation of a key proto-oncogene, cellular Src (c-Src), at a well-characterized activating site (Tyr416). NDRG1-mediated down-regulation of EGFR expression and activation were responsible for the decreased phosphorylation of c-Src (Tyr416). Indeed, NDRG1 prevented recruitment of c-Src to EGFR and c-Src activation. Moreover, NDRG1 suppressed Rac1 activity by modulating phosphorylation of a c-Src downstream effector, p130Cas, and its association with CrkII, which acts as a "molecular switch" to activate Rac1. NDRG1 also affected another signaling molecule involved in modulating Rac1 signaling, c-Abl, which then inhibited CrkII phosphorylation. Silencing NDRG1 increased cell migration relative to the control and inhibition of c-Src signaling using siRNA, or a pharmacological inhibitor (SU6656), prevented this increase. Hence, the role of NDRG1 in decreasing cell migration is, in part, due to its inhibition of c-Src activation. In addition, novel pharmacological agents, which induce NDRG1 expression and are currently under development as anti-metastatic agents, markedly increase NDRG1 and decrease c-Src activation. This study leads to important insights into the mechanism involved in inhibiting metastasis by NDRG1 and how to target these pathways with novel therapeutics.
Collapse
Affiliation(s)
- Wensheng Liu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R.China.,Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Fei Yue
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R.China
| | - Minhua Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R.China
| | - Angelica Merlot
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Dong-Hun Bae
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Michael Huang
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Darius Lane
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Patric Jansson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Goldie Yuan Lam Lui
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Vera Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Sumit Sahni
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Danuta Kalinowski
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Zaklina Kovacevic
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|