1
|
Lai X, Zhang Y, Li M, Yu S, Wang S, Zhang S, Niu H, Chen L, Lan X, Zhang J, Chen S. HGF/c-Met Promotes Breast Cancer Tamoxifen Resistance Through the EZH2/HOTAIR-miR-141/200a Feedback Signaling Pathway. Mol Carcinog 2025; 64:769-783. [PMID: 39853766 DOI: 10.1002/mc.23878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/05/2024] [Accepted: 12/20/2024] [Indexed: 01/26/2025]
Abstract
Tamoxifen is one of the most frequently used endocrine medications for the treatment of estrogen receptor-positive (ER + ) breast cancer (BC). Unfortunately, tamoxifen resistance (TR) brings more challenges to the clinical treatment, and the mechanisms of TR have not yet been fully clarified. HGF/c-Met is closely associated with cancer metastasis, but whether it is involved in TR remains unclear. In our study, we found that the activation of HGF/c-Met was crucial for TR maintenance. Synergistic interaction with HOTAIR and EZH2 accelerated HGF expression by repressing miR-141/200a. Additionally, HGF/c-Met activated NF-κB, forming a positive feedback loop of EZH2/HOTAIR-miR-141/200a-HGF/c-Met-NF-κB. Our findings indicated that HGF/c-Met functioned as an important biomarker for TR, and HGF/c-Met inhibition provided a novel approach to TR treatment.
Collapse
Affiliation(s)
- Xiaofeng Lai
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, China
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (The 900TH Hospital), Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, China
| | - Yuan Zhang
- Department of Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Mengyang Li
- The Faculty of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Shentong Yu
- Department of Pathology, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Shuiliang Wang
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (The 900TH Hospital), Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, China
| | - Shenghang Zhang
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (The 900TH Hospital), Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, China
| | - Huimin Niu
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (The 900TH Hospital), Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, China
| | - Li Chen
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (The 900TH Hospital), Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, China
| | - Xiaopeng Lan
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (The 900TH Hospital), Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, China
| | - Jian Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, China
| | - Suning Chen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
2
|
Jin X, Zhu H, Chen X, Yang Y, Song D. RON receptor tyrosine kinase regulates glycolysis through MAPK/CREB signaling to affect ferroptosis and chemotherapy sensitivity of thyroid cancer cells. Mol Med Rep 2024; 30:234. [PMID: 39422033 PMCID: PMC11529188 DOI: 10.3892/mmr.2024.13359] [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: 01/11/2024] [Accepted: 06/14/2024] [Indexed: 10/19/2024] Open
Abstract
Anaplastic thyroid cancer (ATC) is one of the deadliest and most aggressive human malignancies for which there is currently no effective treatment. Tyrosine kinase receptor RON is highly expressed in various cancer types, including colon, pancreatic and thyroid cancer. However, its underlying role in ATC is not fully understood. The present study investigated the therapeutic potential and molecular mechanism of RON in ATC. RON expression in thyroid cancer cells was detected by western blotting. Glycolysis was assessed by measuring the extracellular acidification rate, glucose uptake, lactate concentration, and expression levels of glucose transporter 1, hexokinase 2 and pyruvate kinase M1/2. In addition, ferroptosis was assessed by detecting the levels of total iron, lipid peroxide and reactive oxygen species, and the expression levels of ferroptosis‑related proteins. Furthermore, mitochondrial function were assessed by JC‑1 staining and detection kits, respectively. The results demonstrated that RON was highly expressed in thyroid cancer cell lines. Furthermore, RON interference inhibited glycolysis, promoted ferroptosis, elevated cell sensitivity to chemotherapy and affected mitochondrial function in thyroid cancer cells. Further experiments demonstrated that RON interference affected the ferroptosis levels in thyroid cancer cells by inhibiting the glycolysis process. Mechanistically, the present results indicated that RON may affect ferroptosis, glycolysis and chemotherapy sensitivity by regulating MAPK/cAMP‑response element binding protein (CREB) signaling in thyroid cancer cells. In conclusion, the present study demonstrated that RON affected ferroptosis, glycolysis and chemotherapy sensitivity in thyroid cancer cells by regulating MAPK/CREB signaling, demonstrating its potential as a therapeutic target in thyroid cancer cells.
Collapse
Affiliation(s)
- Xin Jin
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P.R. China
| | - Haonan Zhu
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P.R. China
| | - Xingyu Chen
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P.R. China
| | - Yining Yang
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P.R. China
| | - Dongliang Song
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|
3
|
de Aguiar ACF, Ferreira NCFDL, Borba MACSM, Filho DDLF, Leitão GM, Mattos LA, Filho JLDL, Martins DBG. TYRO3 and EPHA2 Expression Are Dysregulated in Breast Cancer. Cell Biochem Funct 2024; 42:e4128. [PMID: 39327735 DOI: 10.1002/cbf.4128] [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: 07/09/2024] [Revised: 09/10/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024]
Abstract
Receptor tyrosine kinases (RTKs) are involved in cell growth, motility, and differentiation. Deregulation of RTKs signaling is associated with tumor development and therapy resistance. Potential RTKs like TAM (TYRO3, AXL, MERTK), RON, EPH, and MET have been evaluated in many cancers like lung, prostate, and colorectal, but little is known in breast tumors. In this study, 51 luminal breast cancer tissue and 8 triple negative breast cancer (TNBC) subtypes were evaluated by qPCR for the expression of TAM, RON, EPHA2, and MET genes. Statistical analysis was performed to determine the correlation to clinical data. TYRO3 is related to tumor subtype and stage, patient's age, smoking habits, and obesity. MET expression is correlated to EPHA2 and TAM gene expression. EPHA2 expression is also related to aging and smoking habits. The expression levels of the TAM and EPHA2 genes seem to play an important role in breast cancer, being also influenced by the patient's lifestyle.
Collapse
Affiliation(s)
- Ananda Cristina Fernandes de Aguiar
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
- Molecular Prospection and Bioinformatics Group, Recife, Pernambuco, Brazil
| | | | | | | | - Glauber Moreira Leitão
- Molecular Prospection and Bioinformatics Group, Recife, Pernambuco, Brazil
- Clinical Hospital of Pernambuco-Professor Romero Marques, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | - Luiz Alberto Mattos
- Molecular Prospection and Bioinformatics Group, Recife, Pernambuco, Brazil
- Clinical Hospital of Pernambuco-Professor Romero Marques, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | - José Luiz de Lima Filho
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
- Department of Biochemistry, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | - Danyelly Bruneska Gondim Martins
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
- Molecular Prospection and Bioinformatics Group, Recife, Pernambuco, Brazil
- Department of Biochemistry, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| |
Collapse
|
4
|
Yan S, Ji J, Zhang Z, Imam M, Chen H, Zhang D, Wang J. Targeting the crosstalk between estrogen receptors and membrane growth factor receptors in breast cancer treatment: Advances and opportunities. Biomed Pharmacother 2024; 175:116615. [PMID: 38663101 DOI: 10.1016/j.biopha.2024.116615] [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: 01/10/2024] [Revised: 04/06/2024] [Accepted: 04/17/2024] [Indexed: 06/03/2024] Open
Abstract
Estrogens play a critical role in the initiation and progression of breast cancer. Estrogen receptor (ER)α, ERβ, and G protein-coupled estrogen receptor are the primary receptors for estrogen in breast cancer. These receptors are mainly activated by binding with estrogens. The crosstalk between ERs and membrane growth factor receptors creates additional pathways that amplify the effects of their ligands and promote tumor growth. This crosstalk may cause endocrine therapy resistance in ERα-positive breast cancer. Furthermore, this may explain the resistance to anti-human epidermal growth factor receptor-2 (HER2) treatment in ERα-/HER2-positive breast cancer and chemotherapy resistance in triple-negative breast cancer. Accordingly, it is necessary to understand the complex crosstalk between ERs and growth factor receptors. In this review, we delineate the crosstalk between ERs and membrane growth factor receptors in breast cancer. Moreover, this review highlights the current progress in clinical treatment and discusses how pharmaceuticals target the crosstalk. Lastly, we discuss the current challenges and propose potential solutions regarding the implications of targeting crosstalk via pharmacological inhibition. Overall, the present review provides a landscape of the crosstalk between ERs and membrane growth factor receptors in breast cancer, along with valuable insights for future studies and clinical treatments using a chemotherapy-sparing regimen to improve patient quality of life.
Collapse
Affiliation(s)
- Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110022, China.
| | - Jiale Ji
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Zhijie Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Murshid Imam
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Hong Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Duo Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Jinpeng Wang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| |
Collapse
|
5
|
Wang F, Fu K, Wang Y, Pan C, Wang X, Liu Z, Yang C, Zheng Y, Li X, Lu Y, To KKW, Xia C, Zhang J, Shi Z, Hu Z, Huang M, Fu L. Small-molecule agents for cancer immunotherapy. Acta Pharm Sin B 2024; 14:905-952. [PMID: 38486980 PMCID: PMC10935485 DOI: 10.1016/j.apsb.2023.12.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/28/2023] [Accepted: 12/06/2023] [Indexed: 03/17/2024] Open
Abstract
Cancer immunotherapy, exemplified by the remarkable clinical benefits of the immune checkpoint blockade and chimeric antigen receptor T-cell therapy, is revolutionizing cancer therapy. They induce long-term tumor regression and overall survival benefit in many types of cancer. With the advances in our knowledge about the tumor immune microenvironment, remarkable progress has been made in the development of small-molecule drugs for immunotherapy. Small molecules targeting PRR-associated pathways, immune checkpoints, oncogenic signaling, metabolic pathways, cytokine/chemokine signaling, and immune-related kinases have been extensively investigated. Monotherapy of small-molecule immunotherapeutic drugs and their combinations with other antitumor modalities are under active clinical investigations to overcome immune tolerance and circumvent immune checkpoint inhibitor resistance. Here, we review the latest development of small-molecule agents for cancer immunotherapy by targeting defined pathways and highlighting their progress in recent clinical investigations.
Collapse
Affiliation(s)
- Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Kai Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yujue Wang
- School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing 100084, China
| | - Can Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xueping Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zeyu Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chuan Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ying Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaopeng Li
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yu Lu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Kenneth Kin Wah To
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Chenglai Xia
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan 528000, China
| | - Jianye Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhi Shi
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing 100084, China
| | - Min Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
6
|
Jaradat SK, Ayoub NM, Al Sharie AH, Aldaod JM. Targeting Receptor Tyrosine Kinases as a Novel Strategy for the Treatment of Triple-Negative Breast Cancer. Technol Cancer Res Treat 2024; 23:15330338241234780. [PMID: 38389413 PMCID: PMC10894558 DOI: 10.1177/15330338241234780] [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: 09/10/2023] [Revised: 01/07/2024] [Accepted: 02/05/2024] [Indexed: 02/24/2024] Open
Abstract
Triple-negative breast cancer (TNBC) comprises a group of aggressive and heterogeneous breast carcinoma. Chemotherapy is the mainstay for the treatment of triple-negative tumors. Nevertheless, the success of chemotherapeutic treatments is limited by their toxicity and development of acquired resistance leading to therapeutic failure and tumor relapse. Hence, there is an urgent need to explore novel targeted therapies for TNBC. Receptor tyrosine kinases (RTKs) are a family of transmembrane receptors that are key regulators of intracellular signaling pathways controlling cell proliferation, differentiation, survival, and motility. Aberrant activity and/or expression of several types of RTKs have been strongly connected to tumorigenesis. RTKs are frequently overexpressed and/or deregulated in triple-negative breast tumors and are further associated with tumor progression and reduced survival in patients. Therefore, targeting RTKs could be an appealing therapeutic strategy for the treatment of TNBC. This review summarizes the current evidence regarding the antitumor activity of RTK inhibitors in preclinical models of TNBC. The review also provides insights into the clinical trials evaluating the use of RTK inhibitors for the treatment of patients with TNBC.
Collapse
Affiliation(s)
- Sara K. Jaradat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Ahmed H. Al Sharie
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Julia M. Aldaod
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| |
Collapse
|
7
|
Hunt BG, Davis JC, Fox LH, Vicente-Muñoz S, Lester C, Wells SI, Waltz SE. RON-augmented cholesterol biosynthesis in breast cancer metastatic progression and recurrence. Oncogene 2023; 42:1716-1727. [PMID: 37029299 PMCID: PMC10205688 DOI: 10.1038/s41388-023-02688-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/09/2023]
Abstract
Recurrence remains a significant clinical barrier to improving breast cancer patient outcomes. The RON receptor is a predictor of metastatic progression and recurrence in breast cancers of all subtypes. RON directed therapies are in development, but preclinical data directly testing the impact of RON inhibition on metastatic progression/recurrence are lacking, and mechanisms to exert this function remain unclear. Herein, we modeled breast cancer recurrence using implantation of RON-overexpressing murine breast cancer cells. Recurrent growth was examined after tumor resection via in vivo imaging and ex vivo culture of circulating tumor cells from whole blood samples from tumor bearing mice. In vitro functional assessment of was performed using mammosphere formation assays. Transcriptomic pathway enrichment identified glycolysis and cholesterol biosynthesis pathways, transcription factor targets, and signaling pathways enriched in RON-overexpressing breast cancer cells. BMS777607, a RON inhibitor, abrogated CTC colony formation tumor cells and tumor recurrence. RON promoted mammosphere formation through upregulated cholesterol production that utilizes glycolysis-derived substrates. In mouse models with RON overexpression, statin-mediated inhibition of cholesterol biosynthesis impeded metastatic progression and recurrence but does not affect the primary tumor. RON upregulates glycolysis and cholesterol biosynthesis gene expression by two pathways: MAPK-dependent c-Myc expression and β-catenin -dependent SREBP2 expression.
Collapse
Affiliation(s)
- Brian G Hunt
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA
| | - James C Davis
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA
| | - Levi H Fox
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA
| | - Sara Vicente-Muñoz
- Division of Pathology, NMR-Metabolomics Core, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229-3026, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229-3026, USA
| | - Carissa Lester
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA
| | - Susanne I Wells
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229-3026, USA
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Susan E Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA.
- Research Service, Cincinnati Veterans Affairs Hospital Medical Center, Cincinnati, OH, 45220, USA.
| |
Collapse
|
8
|
Hunt BG, Fox LH, Davis JC, Jones A, Lu Z, Waltz SE. An Introduction and Overview of RON Receptor Tyrosine Kinase Signaling. Genes (Basel) 2023; 14:517. [PMID: 36833444 PMCID: PMC9956929 DOI: 10.3390/genes14020517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/07/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
RON is a receptor tyrosine kinase (RTK) of the MET receptor family that is canonically involved in mediating growth and inflammatory signaling. RON is expressed at low levels in a variety of tissues, but its overexpression and activation have been associated with malignancies in multiple tissue types and worse patient outcomes. RON and its ligand HGFL demonstrate cross-talk with other growth receptors and, consequentially, positions RON at the intersection of numerous tumorigenic signaling programs. For this reason, RON is an attractive therapeutic target in cancer research. A better understanding of homeostatic and oncogenic RON activity serves to enhance clinical insights in treating RON-expressing cancers.
Collapse
Affiliation(s)
- Brian G. Hunt
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Levi H. Fox
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - James C. Davis
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Angelle Jones
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Zhixin Lu
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Susan E. Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
- Research Service, Cincinnati Veterans Affairs Hospital Medical Center, Cincinnati, OH 45220, USA
| |
Collapse
|
9
|
Ruiz-Torres SJ, Bourn JR, Benight NM, Hunt BG, Lester C, Waltz SE. Macrophage-mediated RON signaling supports breast cancer growth and progression through modulation of IL-35. Oncogene 2022; 41:321-333. [PMID: 34743208 PMCID: PMC8758553 DOI: 10.1038/s41388-021-02091-y] [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: 03/18/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 01/20/2023]
Abstract
Tumor associated macrophages (TAMs) play a major role in regulating mammary tumor growth and in directing the responses of tumor infiltrating leukocytes in the microenvironment. However, macrophage-specific mechanisms regulating the interactions of macrophages with tumor cells and other leukocytes that support tumor progression have not been extensively studied. In this study, we show that the activation of the RON receptor tyrosine kinase signaling pathway specifically in macrophages supports breast cancer growth and metastasis. Using clinically relevant murine models of breast cancer, we demonstrate that loss of macrophage RON expression results in decreases in mammary tumor cell proliferation, survival, cancer stem cell self-renewal, and metastasis. Macrophage RON signaling modulates these phenotypes via direct effects on the tumor proper and indirectly by regulating leukocyte recruitment including macrophages, T-cells, and B-cells in the mammary tumor microenvironment. We further show that macrophage RON expression regulates the macrophage secretome including IL-35 and other immunosuppressive factors. Overall, our studies implicate activation of RON signaling in macrophages as a key player in supporting a thriving mammary pro-tumor microenvironment through novel mechanisms including the augmentation of tumor cell properties through IL-35.
Collapse
Affiliation(s)
- Sasha J. Ruiz-Torres
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Jennifer R. Bourn
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Nancy M. Benight
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Brian G. Hunt
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Carissa Lester
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Susan E. Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA,Research Service, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH 45267, USA,Address correspondence to: Susan E. Waltz, PhD, Department of Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, 3125 Eden Ave, Cincinnati, OH 45267-0521, Tel: 513.558.8675,
| |
Collapse
|
10
|
Prognostic role of the recepteur d'origine nantais (RON) expression in primary high-grade osteosarcoma. J Orthop Sci 2021; 26:1100-1106. [PMID: 32972820 DOI: 10.1016/j.jos.2020.08.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/11/2020] [Accepted: 08/05/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND Osteosarcoma is a common primary malignant bone tumor susceptible to distant metastasis. The clinical outcome for patients remains poor due to the resistance to chemotherapy and lacking effective therapeutic targets. Recepteur d'origine nantais (RON), a transmembrane protein of the c-MET proto-oncogene family, has been reported to contribute to the malignant progression and bone metastasis in several tumors. The present study aimed to explore the prognostic significance of RON in primary high-grade osteosarcoma. METHODS Immunohistochemistry (IHC) and western blotting (WB) were used to investigate the protein expression of RON in 80 surgically resected specimens (50 high-grade osteosarcoma specimens and 30 non-neoplastic bone tissues) and 6 cell lines. The χ2 test or independent-sample Student's t-test was used to assess the significance of RON difference between osteosarcoma and non-neoplastic bone tissues. The χ2 test and Fisher's exact test were used to analyze the association of RON with the clinicopathological features of osteosarcoma patients. Kaplan-Meier method and Cox proportional hazards model were used to assess the significance of RON for the survival of osteosarcoma patients. RESULTS The results of IHC and WB observed significant overexpression of RON in osteosarcoma specimens (P < 0.001) and osteosarcoma cell lines. Moreover, immunohistochemical high expression of RON was associated with a poor response to chemotherapy (P = 0.032) as well as worse progression-free (P = 0.003) and overall (P < 0.001) survival of osteosarcoma patients. Multivariate analysis revealed that high expression of RON was independently associated with reduced progression-free (P = 0.027, HR = 2.31) and overall survival (P = 0.004, HR = 5.06) time of osteosarcoma patients. CONCLUSIONS The present study demonstrated that high expression of RON held independent value for unfavorable survival in primary high-grade osteosarcoma. Its potential role as a therapeutic target for osteosarcoma treatment deserves further research.
Collapse
|
11
|
Singh D, Dheer D, Samykutty A, Shankar R. Antibody drug conjugates in gastrointestinal cancer: From lab to clinical development. J Control Release 2021; 340:1-34. [PMID: 34673122 DOI: 10.1016/j.jconrel.2021.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022]
Abstract
The antibody-drug conjugates (ADCs) are one the fastest growing biotherapeutics in oncology and are still in their infancy in gastrointestinal (GI) cancer for clinical applications to improve patient survival. The ADC based approach is developed with tumor specific antigen, antibody carrying cytotoxic agents to precisely target and deliver chemotherapeutics at the tumor site. To date, 11 ADCs have been approved by US-FDA, and more than 80 are in the clinical development phase for different oncological indications. However, The ADCs based therapies in GI cancers are still far from having high-efficient clinical outcomes. The limited success of these ADCs and lessons learned from the past are now being used to develop a newer generation of ADC against GI cancers. In this review, we did a comprehensive assessment of the key components of ADCs, including tumor marker, antibody, cytotoxic payload, and linkage strategy, with a focus on technical improvement and some future trends in the pipeline for clinical translation. The various preclinical and clinical ADCs used in gastrointestinal malignancies, their target, composition and bioconjugation, along with preclinical and clinical outcomes, are discussed. The emphasis is also given to new generation ADCs employing novel mAb, payload, linker, and bioconjugation methods are also included.
Collapse
Affiliation(s)
- Davinder Singh
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Divya Dheer
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Abhilash Samykutty
- Stephenson Comprehensive Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA.
| | - Ravi Shankar
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
12
|
Bourn JR, Ruiz-Torres SJ, Hunt BG, Benight NM, Waltz SE. Tumor cell intrinsic RON signaling suppresses innate immune responses in breast cancer through inhibition of IRAK4 signaling. Cancer Lett 2021; 503:75-90. [PMID: 33508385 PMCID: PMC7981256 DOI: 10.1016/j.canlet.2021.01.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/11/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022]
Abstract
Increasing evidence suggests that cancer cells require both alterations in intrinsic cellular processes and the tumor microenvironment for tumor establishment, growth, and progression to metastatic disease. Despite this, knowledge of tumor-cell intrinsic molecular mechanisms controlling both tumor cell processes as well as the tumor microenvironment is limited. In this study, we provide evidence demonstrating the novel role of RON signaling in regulating breast cancer initiation, progression, and metastasis through modulation of tumor cell intrinsic processes and the tumor microenvironment. Using clinically relevant models of breast cancer, we show that RON signaling in the mammary epithelial tumor cells promotes tumor cell survival and proliferation as well as an immunopermissive microenvironment associated with decreased M1 macrophage, natural killer (NK) cell, and CD8+ T cell recruitment. Moreover, we demonstrate that RON signaling supports these phenotypes through novel mechanisms involving suppression of IRAK4 signaling and inhibition of type I Interferons. Our studies indicate that activation of RON signaling within breast cancer cells promotes tumor cell intrinsic growth and immune evasion which support breast cancer progression and highlight the role of targeting RON signaling as a potential therapeutic strategy against breast cancer.
Collapse
Affiliation(s)
- Jennifer R Bourn
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA
| | - Sasha J Ruiz-Torres
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA
| | - Brian G Hunt
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA
| | - Nancy M Benight
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA
| | - Susan E Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA; Research Service, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH, 45267, USA.
| |
Collapse
|
13
|
Combined crizotinib and endocrine drugs inhibit proliferation, migration, and colony formation of breast cancer cells via downregulation of MET and estrogen receptor. Med Oncol 2021; 38:8. [DOI: 10.1007/s12032-021-01458-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/01/2021] [Indexed: 02/07/2023]
|
14
|
RON signalling promotes therapeutic resistance in ESR1 mutant breast cancer. Br J Cancer 2020; 124:191-206. [PMID: 33257837 PMCID: PMC7782501 DOI: 10.1038/s41416-020-01174-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/16/2022] Open
Abstract
Background Oestrogen Receptor 1 (ESR1) mutations are frequently acquired in oestrogen receptor (ER)-positive metastatic breast cancer (MBC) patients who were treated with aromatase inhibitors (AI) in the metastatic setting. Acquired ESR1 mutations are associated with poor prognosis and there is a lack of effective therapies that selectively target these cancers. Methods We performed a proteomic kinome analysis in ESR1 Y537S mutant cells to identify hyperactivated kinases in ESR1 mutant cells. We validated Recepteur d’Origine Nantais (RON) and PI3K hyperactivity through phospho-immunoblot analysis, organoid growth assays, and in an in vivo patient-derived xenograft (PDX) metastatic model. Results We demonstrated that RON was hyperactivated in ESR1 mutant models, and in acquired palbociclib-resistant (PalbR) models. RON and insulin-like growth factor 1 receptor (IGF-1R) interacted as shown through pharmacological and genetic inhibition and were regulated by the mutant ER as demonstrated by reduced phospho-protein expression with endocrine therapies (ET). We show that ET in combination with a RON inhibitor (RONi) decreased ex vivo organoid growth of ESR1 mutant models, and as a monotherapy in PalbR models, demonstrating its therapeutic efficacy. Significantly, ET in combination with the RONi reduced metastasis of an ESR1 Y537S mutant PDX model. Conclusions Our results demonstrate that RON/PI3K pathway inhibition may be an effective treatment strategy in ESR1 mutant and PalbR MBC patients. Clinically our data predict that ET resistance mechanisms can also contribute to CDK4/6 inhibitor resistance. ![]()
Collapse
|
15
|
Antibody-drug conjugates targeting RON receptor tyrosine kinase as a novel strategy for treatment of triple-negative breast cancer. Drug Discov Today 2020; 25:1160-1173. [PMID: 32479905 DOI: 10.1016/j.drudis.2020.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/05/2020] [Accepted: 05/18/2020] [Indexed: 12/17/2022]
Abstract
Treatment of triple-negative breast cancer (TNBC) is a challenge to oncologists. Currently, the lack of effective therapy has fostered a major effort to discover new targets and therapeutics to combat this disease. The recepteur d'origine nantais (RON) receptor has been implicated in the pathogenesis of TNBC. Clinical studies have revealed that aberrant RON expression is crucial in regulating TNBC malignant phenotypes. Increased RON expression also has prognostic value for breast cancer progress. These features provide the rationale to target RON for TNBC treatment. In this review, we discuss the importance of RON in TNBC tumorigenesis and the development of anti-RON antibody-drug conjugates (ADCs) for clinical application. The findings from preclinical studies lay the foundation for clinical trials of this novel biotherapeutic for TNBC therapy.
Collapse
|
16
|
MST1R (RON) expression is a novel prognostic biomarker for metastatic progression in breast cancer patients. Breast Cancer Res Treat 2020; 181:529-540. [PMID: 32342233 DOI: 10.1007/s10549-020-05653-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/17/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE This study evaluates the prognostic significance of MST1R (RON) expression in breast cancer with respect to disease progression, long-term survival, subtype, and association with conventional prognostic factors. METHODS The approach includes interrogation of survival and tumor staging with paired MST1R RNA expression from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets. Protein expression evaluation was performed using immunohistochemistry (IHC) staining of MST1R on breast cancer tissue samples from the Cancer Diagnosis Program Breast Cancer Progression tissue microarray and locally obtained breast tumor tissue samples analyzed with paired survival, metastasis, and subtype. RESULTS Data from TCGA (n = 774) show poorer relapse-free survival (RFS) in patients with high MST1R expression (P = 0.32) and no difference in MST1R expression based on tumor stage (P = 0.77) or nodal status (P = 0.94). Patients in the GEO-derived Kaplan-Meier Plotter microarray dataset demonstrate the association of MST1R and poorer overall survival (n = 1402, P = 0.018) and RFS in patients receiving chemotherapy (n = 798, P = 0.041). Patients with high MST1R expression display worse overall survival (P = 0.01) and receiver operator characteristic (ROC) analysis demonstrate the predictive capacity of increased MST1R with early death (P = 0.0017) in IHC-stained samples. Paired IHC-stained breast tumor samples from the primary versus metastatic site show MST1R expression is associated with metastatic progression (P = 0.032), and ROC analysis supports the predictive capacity of MST1R in metastatic progression (P = 0.031). No associations of MST1R with estrogen receptor (ER), progesterone receptor (PR), both ER and PR, HER2 positivity, or triple-negativity were found (P = 0.386, P = 0.766, P = 0.746, P = 0.457, P = 0.947, respectively). CONCLUSIONS MST1R expression has prognostic value in breast cancer with respect to survival and metastatic progression. MST1R expression is not associated with tumor stage, nodal status, or subtype.
Collapse
|
17
|
Hu CY, Xu XM, Hong B, Wu ZG, Qian Y, Weng TH, Liu YZ, Tang TM, Wang MH, Yao HP. Aberrant RON and MET Co-overexpression as Novel Prognostic Biomarkers of Shortened Patient Survival and Therapeutic Targets of Tyrosine Kinase Inhibitors in Pancreatic Cancer. Front Oncol 2019; 9:1377. [PMID: 31867280 PMCID: PMC6906148 DOI: 10.3389/fonc.2019.01377] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 11/22/2019] [Indexed: 12/14/2022] Open
Abstract
RON (recepteur d'origine nantais) and MET (hepatocyte growth factor receptor) are tyrosine kinase receptors. Various cancers have aberrant RON and MET expression and activation, which contribute to cancer cell proliferation, invasiveness, and metastasis. Here, we explored RON and MET expression in pancreatic cancer and their relationship with overall survival (OS) time, and evaluated their significance as therapeutic targets of tyrosine kinase inhibitors in pancreatic cancer. We enrolled 227 patients with pancreatic cancer in the study. RON and MET expression was analyzed by immunohistochemical staining. Four human pancreatic cancer cell lines expressing variable levels of RON or MET and four MET superfamily inhibitors (BMS777607, PHA665752, INCB28060, Tivantinib) were used. The effect of the four tyrosine kinase inhibitors on cell viability, migration, and apoptosis were determined using cell viability, scratch wound healing, and Caspase-Glo 3/7 assays. Cellular signaling was analyzed by immunoprecipitation and western blotting. The therapeutic efficacy of the tyrosine kinase inhibitors was determined with mouse xenograft pancreatic cancer models in vivo. There was wide aberrant RON and MET expression in the cancer tissues. In 227 pancreatic cancer samples, 33% had RON overexpression, 41% had MET overexpression, and 15.4% had RON and MET co-overexpression. RON and MET expression were highly correlated. RON and MET expression levels were significantly related to OS. Patients with RON and MET co-overexpression had poorer OS. BMS777607 and PHA665752 inhibited pancreatic cancer cell viability and migration, and promoted apoptosis by inhibiting RON and MET phosphorylation and further inhibiting the downstream signaling pathways in vitro. They also inhibited tumor growth and further inhibited phosphorylated (phosphor)-RON and phospho-MET expression in the mouse xenograft models in vivo effectively. INCB28060, which inhibits the MET signaling pathway alone, was not effective. RON and MET can be important indicators of prognosis in pancreatic cancer. Tyrosine kinase inhibitors targeting RON and MET in pancreatic cancer are a novel and potential approach for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Chen-Yu Hu
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiang-Ming Xu
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bo Hong
- Department of Pathology, The Second Affiliated Hospital, Hangzhou, China
| | - Zhi-Gang Wu
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yun Qian
- Department of Clinical Laboratory, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian-Hao Weng
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi-Zhi Liu
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao-Ming Tang
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming-Hai Wang
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Biology Research Center, Amarillo, TX, United States.,Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX, United States
| | - Hang-Ping Yao
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
18
|
Suthe SR, Yao HP, Weng TH, Hu CY, Feng L, Wu ZG, Wang MH. RON Receptor Tyrosine Kinase as a Therapeutic Target for Eradication of Triple-Negative Breast Cancer: Efficacy of Anti-RON ADC Zt/g4-MMAE. Mol Cancer Ther 2018; 17:2654-2664. [PMID: 30275241 DOI: 10.1158/1535-7163.mct-18-0252] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/21/2018] [Accepted: 09/26/2018] [Indexed: 12/21/2022]
Abstract
Triple-negative breast cancer (TNBC) is a highly diverse group of malignant neoplasia with poor outcome. Currently, the lack of effective therapy has fostered a major effort to discover new targets to treat this malignant cancer. Here we identified the RON receptor tyrosine kinase as a therapeutic target for potential TNBC treatment. We analyzed RON expression in 168 primary TNBC samples via tissue microarray using anti-RON IHC staining and demonstrated that RON was widely expressed in 76.8% TNBC samples with overexpression in 76 cases (45.2%). These results provide the molecular basis to target RON for TNBC therapy. To this end, anti-RON monoclonal antibody Zt/g4-drug monomethyl auristatin E conjugate (Zt/g4-MMAE) was developed with a drug to antibody ratio of 3.29 and tested in a panel of TNBC cell lines with different phenotypes. In vitro, Zt/g4-MMAE rapidly induced RON internalization, resulted in cell-cycle arrest followed by massive cell death. The calculated IC50 values ranged from 0.06 to 3.46 μg/mL dependent on individual TNBC cell lines tested. Zt/g4-MMAE also effectively killed TNBC stem-like cells with RON+/CD44+/CD24- phenotypes and RON-negative TNBC cells through the bystander effect. In vivo, Zt/g4-MMAE at 10 mg/kg in a Q12 × 2 regimen completely eradicated TNBC xenografts without the regrowth of xenograft tumors. In conclusion, increased RON expression is a pathogenic feature in primary TNBC samples. Zt/g4-MMAE is highly effective in eradicating TNBC xenografts in preclinical models. These findings lay the foundation for using anti-RON Zt/g4-MMAE in clinical trials as a novel strategy for TNBC treatment.
Collapse
Affiliation(s)
- Sreedhar Reddy Suthe
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Biology Research Center.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Hang-Ping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian-Hao Weng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen-Yu Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liang Feng
- Cancer Biology Research Center.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Zhi-Gang Wu
- Zhejiang Provincial Key Laboratory for Precision Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancers, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming-Hai Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Cancer Biology Research Center.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas.,Zhejiang Provincial Key Laboratory for Precision Diagnosis and Treatment of Hepatobiliary and Pancreatic Cancers, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
19
|
Liu L, Shen W, Zhu Z, Lin J, Fang Q, Ruan Y, Zhao H. Combined inhibition of EGFR and c-ABL suppresses the growth of fulvestrant-resistant breast cancer cells through miR-375-autophagy axis. Biochem Biophys Res Commun 2018. [PMID: 29522716 DOI: 10.1016/j.bbrc.2018.03.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Fulvestrant is the FDA-approved "pure anti-estrogen" agent for malignant breast cancer therapy. But endocrine resistance causes drug failure. A new approach is desired for fulvestrant-resistant breast cancer (FRBC) therapy. This study aims to find an effective approach to inhibit FRBC for patients with advanced breast cancer. MTT assay was first performed to detect the effect of inhibitors of c-ABL (imatinib) and EGFR (lapatinib) on FRBC cells. Microarray analysis was carried out to identify microRNA which is significantly changed between parental and FRBC cells. The related mechanisms were analyzed by qRT-PCR, MTT, AO staining and western blotting. Dual treatment significantly inhibited cell growth of FRBC and upregulated microRNA-375 (miR-375). Overexpression of miR-375 inhibited growth of FRBC cells, reduced autophagy, and decreased expression of ATG7 and LC3-II. Dual treatment elevated expression of miR-375 more than any single one of these two inhibitors. Overexpression of miR-375 increased cell growth inhibition induced by dual treatment, and the effect was attenuated when miR-375 was inhibited. In conclusion, we identified that combined inhibition of EGFR and c-ABL can suppress the growth of FRBC cells and elucidated a mechanism within FRBC cells involving regulation of miR-375 and autophagy. Dual treatment may be useful for inhibiting fulvestrant-resistant breast cancer.
Collapse
Affiliation(s)
- Lunming Liu
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, China
| | - Weifeng Shen
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, China
| | - Zhihui Zhu
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, China
| | - Jinxiong Lin
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, China
| | - Qingxia Fang
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, China
| | - Yeping Ruan
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, China.
| | - Huajun Zhao
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, China.
| |
Collapse
|
20
|
Zhang Y, Xia M, Jin K, Wang S, Wei H, Fan C, Wu Y, Li X, Li X, Li G, Zeng Z, Xiong W. Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities. Mol Cancer 2018; 17:45. [PMID: 29455668 PMCID: PMC5817860 DOI: 10.1186/s12943-018-0796-y] [Citation(s) in RCA: 380] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 02/01/2018] [Indexed: 12/15/2022] Open
Abstract
c-Met is a receptor tyrosine kinase belonging to the MET (MNNG HOS transforming gene) family, and is expressed on the surfaces of various cells. Hepatocyte growth factor (HGF) is the ligand for this receptor. The binding of HGF to c-Met initiates a series of intracellular signals that mediate embryogenesis and wound healing in normal cells. However, in cancer cells, aberrant HGF/c-Met axis activation, which is closely related to c-Met gene mutations, overexpression, and amplification, promotes tumor development and progression by stimulating the PI3K/AKT, Ras/MAPK, JAK/STAT, SRC, Wnt/β-catenin, and other signaling pathways. Thus, c-Met and its associated signaling pathways are clinically important therapeutic targets. In this review, we elaborate on the molecular structure of c-Met and HGF and the mechanism through which their interaction activates the PI3K/AKT, Ras/MAPK, and Wnt signaling pathways. We also summarize the connection between c-Met and RON and EGFR, which are also receptor tyrosine kinases. Finally, we introduce the current therapeutic drugs that target c-Met in primary tumors, and their use in clinical research.
Collapse
Affiliation(s)
- Yazhuo Zhang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mengfang Xia
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ke Jin
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Shufei Wang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Hang Wei
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Chunmei Fan
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yingfen Wu
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Wei Xiong
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
21
|
Bhatnagar S, Soni A, Kaushik S, Rikhi M, Santhoshkumar TR, Jayaram B. Nonsteroidal estrogen receptor isoform-selective biphenyls. Chem Biol Drug Des 2017; 91:620-630. [PMID: 29052968 DOI: 10.1111/cbdd.13126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/18/2017] [Accepted: 10/04/2017] [Indexed: 12/17/2022]
Abstract
Estrogen receptor (ER) has been a therapeutic target to treat ER-positive breast cancer, most notably by agents known as selective estrogen receptor modulators (SERMs). However, resistance and severe adverse effects of known drugs gave impetus to the search for newer agents with better therapeutic profile. ERα and ERβ are two isoforms sharing 56% identity and having different physiological functions and expressions in various tissues. Only two residues differ in the active sites of the two isoforms motivating us to design isoform-selective ligands. Guided by computational docking and molecular dynamics simulations, we have designed, synthesized, and tested, substituted biphenyl-2,6-diethanones and their derivatives as potential agents targeting ERα. Four of the molecules synthesized exhibited preferential cytotoxicity in ERα+ cell line (MCF-7) compared to ERβ+ cell line (MDA-MB-231). Molecular dynamics (MD) in combination with molecular mechanics-generalized Born surface area (MM-GBSA) methods could account for binding selectivity. Further cotreatment and E-screen studies with known ER ligands-estradiol (E2 ) and tamoxifen (Tam)-indicated isoform-selective anti-estrogenicity in ERα+ cell line which might be ER-mediated. ERα siRNA silencing experiments further confirmed the ER selective nature of ligands.
Collapse
Affiliation(s)
- Seema Bhatnagar
- Amity Institute of Biotechnology, Amity University, Noida, India
| | - Anjali Soni
- Department of Chemistry and Supercomputing Facility for Bioinformatics and Computational Biology, Indian Institute of Technology, Hauz Khas, New Delhi, India
| | - Swati Kaushik
- Amity Institute of Biotechnology, Amity University, Noida, India.,Cancer Research Programme Lab1, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, Kerala, India
| | - Megha Rikhi
- Amity Institute of Biotechnology, Amity University, Noida, India
| | | | - Bhyravabhotla Jayaram
- Department of Chemistry and Supercomputing Facility for Bioinformatics and Computational Biology, Indian Institute of Technology, Hauz Khas, New Delhi, India.,Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi, India
| |
Collapse
|
22
|
Yang SY, Nguyen TT, Ung TT, Jung YD. Role of Recepteur D'origine Nantais on Gastric Cancer Development and Progression. Chonnam Med J 2017; 53:178-186. [PMID: 29026705 PMCID: PMC5636756 DOI: 10.4068/cmj.2017.53.3.178] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/13/2017] [Accepted: 07/17/2017] [Indexed: 01/12/2023] Open
Abstract
Recepteur d'origine nantais (RON) is a receptor tyrosine kinase belonging to the subfamily of which c-MET is the prototype. Large epidemiologic studies have confirmed the strong association between RON and gastric cancer development. Constitutive activation of RON signaling directly correlates with tumorigenic phenotypes of gastric cancer and a poor survival rate in advanced gastric cancer patients. In this review, we focus on recent evidence of the aberrant expression and activation of RON in gastric cancer tumors and provide insights into the mechanism of RON signaling associated with gastric cancer progression and metastasis. Current therapeutics against RON in gastric cancer are summarized.
Collapse
Affiliation(s)
- Sung Yeul Yang
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea
| | - Thi Thinh Nguyen
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea
| | - Trong Thuan Ung
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea
| | - Young Do Jung
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
23
|
Zarei O, Benvenuti S, Ustun-Alkan F, Hamzeh-Mivehroud M, Dastmalchi S. Identification of a RON tyrosine kinase receptor binding peptide using phage display technique and computational modeling of its binding mode. J Mol Model 2017; 23:267. [DOI: 10.1007/s00894-017-3437-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/01/2017] [Indexed: 12/18/2022]
|
24
|
Ruiz-Torres SJ, Benight NM, Karns RA, Lower EE, Guan JL, Waltz SE. HGFL-mediated RON signaling supports breast cancer stem cell phenotypes via activation of non-canonical β-catenin signaling. Oncotarget 2017; 8:58918-58933. [PMID: 28938607 PMCID: PMC5601703 DOI: 10.18632/oncotarget.19441] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/11/2017] [Indexed: 12/16/2022] Open
Abstract
Breast cancer stem cells (BCSCs), which drive tumor progression, recurrence, and metastasis, are considered a major challenge for breast cancer treatments, thus the discovery of novel pathways regulating BCSC maintenance remains essential to develop new strategies to effectively target this population and combat disease mortality. The HGFL-RON signaling is overexpressed in human breast cancers and is associated with increased breast cancer progression, metastasis, and poor prognosis. Here, we report that overexpression of RON/MST1R and HGFL/MST1 in cell lines and primary tumors increases BCSC self-renewal, numbers, and tumorigenic potential after syngeneic transplantation. Transcriptome analyses also reveal that the HGFL-RON signaling pathway regulates additional BCSC functions and supports an immunosuppressive microenvironment to stimulate tumor formation and progression. Moreover, we show that genetic and chemical downregulation of HGFL-RON signaling disrupts BCSC phenotypes and tumor growth by suppressing the RON-mediated phosphorylation/activation of β-CATENIN/CTNNB1 and its effector NF-κB/RELA. These studies indicate that HGFL-RON signaling regulates BCSC phenotypes and the tumor microenvironment to drive tumorigenesis and present HGFL/RON as novel therapeutic targets to effectively eradicate BCSCs in patients.
Collapse
Affiliation(s)
- Sasha J Ruiz-Torres
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Nancy M Benight
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Rebekah A Karns
- Division of Bioinformatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Elyse E Lower
- Department of Internal Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Susan E Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.,Research Service, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH 45267, USA
| |
Collapse
|
25
|
Wang J, Li L, Liu S, Zhao Y, Wang L, Du G. FOXC1 promotes melanoma by activating MST1R/PI3K/AKT. Oncotarget 2016; 7:84375-84387. [PMID: 27533251 PMCID: PMC5356666 DOI: 10.18632/oncotarget.11224] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/19/2016] [Indexed: 12/27/2022] Open
Abstract
FOXC1 is a member of Forkhead box family transcription factors. We showed that FOXC1 level was increased in melanoma cells and tissues and correlated with hypomethylation of the FOXC1 gene. Overexpression of FOXC1 promoted proliferation, migration, invasion, colony formation and growth in 3D Matrigel of melanoma cells. FOXC1 increased MST1R and activated the PI3K/AKT pathway. Also, FOXC1 expression was associated with disease progression and poor prognosis of melanoma. We suggest that FOXC1 is a potential prognostic biomarker for treating melanoma and predicting outcome of patients.
Collapse
Affiliation(s)
- Jinhua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Department of Molecular Oncology, John Wayne Cancer Institute (JWCI) at Providence Saint John's Health Center, Santa Monica 90404, CA, USA
| | - Li Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Shiwei Liu
- Department of Endocrinology, Shanxi DAYI Hospital, Shanxi Medical University, Taiyuan, Shanxi 030002, China
| | - Ying Zhao
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Lin Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
26
|
Zarei O, Benvenuti S, Ustun-Alkan F, Hamzeh-Mivehroud M, Dastmalchi S. Strategies of targeting the extracellular domain of RON tyrosine kinase receptor for cancer therapy and drug delivery. J Cancer Res Clin Oncol 2016; 142:2429-2446. [PMID: 27503093 DOI: 10.1007/s00432-016-2214-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 08/01/2016] [Indexed: 01/22/2023]
Abstract
PURPOSE Cancer is one of the most important life-threatening diseases in the world. The current efforts to combat cancer are being focused on molecular-targeted therapies. The main purpose of such approaches is based on targeting cancer cell-specific molecules to minimize toxicity for the normal cells. RON (Recepteur d'Origine Nantais) tyrosine kinase receptor is one of the promising targets in cancer-targeted therapy and drug delivery. METHODS In this review, we will summarize the available agents against extracellular domain of RON with potential antitumor activities. RESULTS The presented antibodies and antibody drug conjugates against RON in this review showed wide spectrum of in vitro and in vivo antitumor activities promising the hope for them entering the clinical trials. CONCLUSION Due to critical role of extracellular domain of RON in receptor activation, the development of therapeutic agents against this region could lead to fruitful outcome in cancer therapy.
Collapse
Affiliation(s)
- Omid Zarei
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Silvia Benvenuti
- Molecular Therapeutics and Exploratory Research Laboratory, Candiolo Cancer Institute-FPO-IRCCS, Candiolo, Turin, Italy
| | - Fulya Ustun-Alkan
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Istanbul University, Istanbul, Turkey
| | - Maryam Hamzeh-Mivehroud
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
27
|
Pires-Luís AS, Vieira-Coimbra M, Ferreira MJ, Ramalho-Carvalho J, Costa-Pinheiro P, Antunes L, Dias PC, Lobo F, Oliveira J, Graça I, Henrique R, Jerónimo C. Prognostic significance of MST1R dysregulation in renal cell tumors. Am J Cancer Res 2016; 6:1799-1811. [PMID: 27648366 PMCID: PMC5004080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 05/29/2016] [Indexed: 06/06/2023] Open
Abstract
Macrophage stimulating 1 receptor (MST1R) is a C-MET proto-oncogene family receptor tyrosine kinase. Promoter methylation patterns determine transcription of MST1R variants as hypermethylation of a region upstream of transcription start site (TSS) is associated with lack of MST1R long transcript (MST1R long) and expression of a short transcript with oncogenic potential. Thus, we aimed to investigate MST1R variant transcript regulation in renal cell tumors (RCT) and assess their prognostic potential. We found, in a series of 120 RCT comprising the four main subtypes (clear cell, papillary and chromophobe renal cell carcinoma, and oncocytoma), that higher methylation levels close to TSS were associated with total MST1R expression levels (MST1R total) in primary tumors (p=0.049) and renal cancer cell lines. After demethylating treatment, MST1R long/MST1R total ratio increased, as expected, in two renal cell carcinoma cell lines tested. However, in primary tumors with hypermethylation upstream of TSS, a decrease in MST1R long/MST1R total ratio was not detected, although higher expression ratio of nuclear factor-κB was apparent. Furthermore, survival analysis demonstrated that MST1R long/MST1R total ratio was independently associated with shorter disease-specific and disease-free survival, whereas MST1R total expression associated with shorter disease-specific survival. In conclusion, although promoter methylation patterns seem to determine MST1R global transcription regulation in renal cell carcinoma, other mechanisms might contribute to deregulate MST1R variant expression in RCT. Nevertheless, MST1R total expression and MST1R long/MST1R total ratio modulate the biological and clinical aggressiveness of renal cell carcinoma, as depicted by its prognostic significance, a finding that requires validation in a larger independent series.
Collapse
Affiliation(s)
- Ana S Pires-Luís
- Cancer Biology and Epigenetics Group, Research Center of The Portuguese Oncology Institute of PortoPorto, Portugal
- Department of Pathology - Portuguese Oncology Institute of PortoPorto, Portugal
| | - Márcia Vieira-Coimbra
- Cancer Biology and Epigenetics Group, Research Center of The Portuguese Oncology Institute of PortoPorto, Portugal
- Department of Pathology - Portuguese Oncology Institute of PortoPorto, Portugal
| | - Maria João Ferreira
- Cancer Biology and Epigenetics Group, Research Center of The Portuguese Oncology Institute of PortoPorto, Portugal
| | - João Ramalho-Carvalho
- Cancer Biology and Epigenetics Group, Research Center of The Portuguese Oncology Institute of PortoPorto, Portugal
| | - Pedro Costa-Pinheiro
- Cancer Biology and Epigenetics Group, Research Center of The Portuguese Oncology Institute of PortoPorto, Portugal
| | - Luís Antunes
- Department of Epidemiology - Portuguese Oncology Institute of PortoPorto, Portugal
| | - Paula C Dias
- Department of Pathology - Portuguese Oncology Institute of PortoPorto, Portugal
| | - Francisco Lobo
- Department of Urology - Portuguese Oncology Institute of PortoPorto, Portugal
| | - Jorge Oliveira
- Department of Urology - Portuguese Oncology Institute of PortoPorto, Portugal
| | - Inês Graça
- Cancer Biology and Epigenetics Group, Research Center of The Portuguese Oncology Institute of PortoPorto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center of The Portuguese Oncology Institute of PortoPorto, Portugal
- Department of Pathology - Portuguese Oncology Institute of PortoPorto, Portugal
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of PortoPorto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of The Portuguese Oncology Institute of PortoPorto, Portugal
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of PortoPorto, Portugal
| |
Collapse
|
28
|
Ha JR, Siegel PM, Ursini-Siegel J. The Tyrosine Kinome Dictates Breast Cancer Heterogeneity and Therapeutic Responsiveness. J Cell Biochem 2016; 117:1971-90. [PMID: 27392311 DOI: 10.1002/jcb.25561] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 03/24/2016] [Indexed: 12/13/2022]
Abstract
Phospho-tyrosine signaling networks control numerous biological processes including cellular differentiation, cell growth and survival, motility, and invasion. Aberrant regulation of the tyrosine kinome is a hallmark of malignancy and influences all stages of breast cancer progression, from initiation to the development of metastatic disease. The success of specific tyrosine kinase inhibitors strongly validates the clinical relevance of tyrosine phosphorylation networks in breast cancer pathology. However, a significant degree of redundancy exists within the tyrosine kinome. Numerous receptor and cytoplasmic tyrosine kinases converge on a core set of signaling regulators, including adaptor proteins and tyrosine phosphatases, to amplify pro-tumorigenic signal transduction pathways. Mutational activation, amplification, or overexpression of one or more components of the tyrosine kinome represents key contributing events responsible for the tumor heterogeneity that is observed in breast cancers. It is this molecular heterogeneity that has become the most significant barrier to durable clinical responses due to the development of therapeutic resistance. This review focuses on recent literature that supports a prominent role for specific components of the tyrosine kinome in the emergence of unique breast cancer subtypes and in shaping breast cancer plasticity, sensitivity to targeted therapies, and the eventual emergence of acquired resistance. J. Cell. Biochem. 117: 1971-1990, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jacqueline R Ha
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Peter M Siegel
- Department of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Oncology, McGill University, Montreal, Quebec, Canada.,Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.,Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Josie Ursini-Siegel
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Oncology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
29
|
Mereiter S, Balmaña M, Gomes J, Magalhães A, Reis CA. Glycomic Approaches for the Discovery of Targets in Gastrointestinal Cancer. Front Oncol 2016; 6:55. [PMID: 27014630 PMCID: PMC4783390 DOI: 10.3389/fonc.2016.00055] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 02/24/2016] [Indexed: 12/22/2022] Open
Abstract
Gastrointestinal (GI) cancer is the most common group of malignancies and many of its types are among the most deadly. Various glycoconjugates have been used in clinical practice as serum biomarker for several GI tumors, however, with limited diagnose application. Despite the good accessibility by endoscopy of many GI organs, the lack of reliable serum biomarkers often leads to late diagnosis of malignancy and consequently low 5-year survival rates. Recent advances in analytical techniques have provided novel glycoproteomic and glycomic data and generated functional information and putative biomarker targets in oncology. Glycosylation alterations have been demonstrated in a series of glycoconjugates (glycoproteins, proteoglycans, and glycosphingolipids) that are involved in cancer cell adhesion, signaling, invasion, and metastasis formation. In this review, we present an overview on the major glycosylation alterations in GI cancer and the current serological biomarkers used in the clinical oncology setting. We further describe recent glycomic studies in GI cancer, namely gastric, colorectal, and pancreatic cancer. Moreover, we discuss the role of glycosylation as a modulator of the function of several key players in cancer cell biology. Finally, we address several state-of-the-art techniques currently applied in this field, such as glycomic and glycoproteomic analyses, the application of glycoengineered cell line models, microarray and proximity ligation assay, and imaging mass spectrometry, and provide an outlook to future perspectives and clinical applications.
Collapse
Affiliation(s)
- Stefan Mereiter
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Meritxell Balmaña
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona , Girona , Spain
| | - Joana Gomes
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal; Medical Faculty, University of Porto, Porto, Portugal
| |
Collapse
|
30
|
Zhong ZF, Qiang WA, Wang CM, Tan W, Wang YT. Furanodiene enhances the anti-cancer effects of doxorubicin on ERα-negative breast cancer cells in vitro. Eur J Pharmacol 2016; 774:10-9. [DOI: 10.1016/j.ejphar.2015.11.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/18/2015] [Accepted: 11/18/2015] [Indexed: 12/26/2022]
|
31
|
Abstract
The majority of metastatic breast cancers cannot be cured and present a major public health problem worldwide. Approximately 70% of breast cancers express the estrogen receptor, and endocrine-based therapies have significantly improved patient outcomes. However, the development of endocrine resistance is extremely common. Understanding the molecular pathways that regulate the hormone sensitivity of breast cancer cells is important to improving the efficacy of endocrine therapy. It is becoming clearer that the PI3K-AKT-forkhead box O (FOXO) signaling axis is a key player in the hormone-independent growth of many breast cancers. Constitutive PI3K-AKT pathway activation, a driver of breast cancer growth, causes down-regulation of FOXO tumor suppressor functions. This review will summarize what is currently known about the role of FOXOs in endocrine-resistance mechanisms. It will also suggest potential therapeutic strategies for the restoration of normal FOXO transcriptional activity.
Collapse
Affiliation(s)
- M Bullock
- Hormones and Cancer GroupCancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, Pacific Highway Saint Leonards, Sydney, New South Wales 2065, Australia
| |
Collapse
|
32
|
Chang K, Karnad A, Zhao S, Freeman JW. Roles of c-Met and RON kinases in tumor progression and their potential as therapeutic targets. Oncotarget 2016; 6:3507-18. [PMID: 25784650 PMCID: PMC4414132 DOI: 10.18632/oncotarget.3420] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 01/21/2015] [Indexed: 02/06/2023] Open
Abstract
c-Met and receptor originated from nantes (RON) are structurally related transmembrane phosphotyrosine kinase receptors. c-Met and RON show increased expression or activity in a variety of tumors leading to tumor progression and may play a role in acquired resistance to therapy. Although often co-expressed, the distinct functional roles of c-Met and RON are not fully understood. c-Met and RON form both activated homodimers and heterodimers with themselves and other families of phosphotyrosine kinase receptors. Inhibitors for c-Met and RON including small molecular weigh kinase inhibitors and neutralizing antibodies are in pre-clinical investigation and clinical trials. Several of the tyrosine kinase inhibitors have activity against both c-Met and RON kinases whereas the antibodies generally are target specific. As with many targeted agents used to treat solid tumors, it is likely that c-Met/RON inhibitors will have greater benefit when used in combination with chemotherapy or other targeted agents. A careful analysis of c-Met/RON expression or activity and a better elucidation of how they influence cell signaling will be useful in predicting which tumors respond best to these inhibitors as well as determining which agents can be used with these inhibitors for combined therapy.
Collapse
Affiliation(s)
- Katherine Chang
- Department of Medicine, Division of Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy and Research Center, Experimental and Developmental Therapeutics Program, San Antonio, TX, USA
| | - Anand Karnad
- Department of Medicine, Division of Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy and Research Center, Experimental and Developmental Therapeutics Program, San Antonio, TX, USA
| | - Shujie Zhao
- Department of Medicine, Division of Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - James W Freeman
- Department of Medicine, Division of Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy and Research Center, Experimental and Developmental Therapeutics Program, San Antonio, TX, USA.,Research and Development, Audie Murphy Veterans Administration Hospital, San Antonio, TX, USA
| |
Collapse
|
33
|
Mereiter S, Magalhães A, Adamczyk B, Jin C, Almeida A, Drici L, Ibáñez-Vea M, Gomes C, Ferreira JA, Afonso LP, Santos LL, Larsen MR, Kolarich D, Karlsson NG, Reis CA. Glycomic analysis of gastric carcinoma cells discloses glycans as modulators of RON receptor tyrosine kinase activation in cancer. Biochim Biophys Acta Gen Subj 2015; 1860:1795-808. [PMID: 26721331 DOI: 10.1016/j.bbagen.2015.12.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 01/16/2023]
Abstract
BACKGROUND Terminal α2-3 and α2-6 sialylation of glycans precludes further chain elongation, leading to the biosynthesis of cancer relevant epitopes such as sialyl-Lewis X (SLe(X)). SLe(X) overexpression is associated with tumor aggressive phenotype and patients' poor prognosis. METHODS MKN45 gastric carcinoma cells transfected with the sialyltransferase ST3GAL4 were established as a model overexpressing sialylated terminal glycans. We have evaluated at the structural level the glycome and the sialoproteome of this gastric cancer cell line applying liquid chromatography and mass spectrometry. We further validated an identified target expression by proximity ligation assay in gastric tumors. RESULTS Our results showed that ST3GAL4 overexpression leads to several glycosylation alterations, including reduced O-glycan extension and decreased bisected and increased branched N-glycans. A shift from α2-6 towards α2-3 linked sialylated N-glycans was also observed. Sialoproteomic analysis further identified 47 proteins with significantly increased sialylated N-glycans. These included integrins, insulin receptor, carcinoembryonic antigens and RON receptor tyrosine kinase, which are proteins known to be key players in malignancy. Further analysis of RON confirmed its modification with SLe(X) and the concomitant activation. SLe(X) and RON co-expression was validated in gastric tumors. CONCLUSION The overexpression of ST3GAL4 interferes with the overall glycophenotype of cancer cells affecting a multitude of key proteins involved in malignancy. Aberrant glycosylation of the RON receptor was shown as an alternative mechanism of oncogenic activation. GENERAL SIGNIFICANCE This study provides novel targets and points to an integrative tumor glycomic/proteomic-profiling for gastric cancer patients' stratification. This article is part of a Special Issue entitled "Glycans in personalised medicine" Guest Editor: Professor Gordan Lauc.
Collapse
Affiliation(s)
- Stefan Mereiter
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar - ICBAS, University of Porto, Portugal
| | - Ana Magalhães
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal
| | - Barbara Adamczyk
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Chunsheng Jin
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Andreia Almeida
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany; Free University Berlin, Berlin, Germany
| | - Lylia Drici
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Maria Ibáñez-Vea
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Catarina Gomes
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal
| | - José A Ferreira
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal; Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology of Porto, Portugal
| | - Luis P Afonso
- Department of Pathology, Portuguese Institute of Oncology of Porto, Portugal
| | - Lúcio L Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology of Porto, Portugal; Department of Surgical Oncology, Portuguese Institute of Oncology of Porto, Portugal
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Daniel Kolarich
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Niclas G Karlsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Celso A Reis
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar - ICBAS, University of Porto, Portugal; Medical Faculty, University of Porto, Portugal.
| |
Collapse
|
34
|
Nardone A, De Angelis C, Trivedi MV, Osborne CK, Schiff R. The changing role of ER in endocrine resistance. Breast 2015; 24 Suppl 2:S60-6. [PMID: 26271713 DOI: 10.1016/j.breast.2015.07.015] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Estrogen receptor (ER) is expressed in approximately 70% of newly diagnosed breast tumors. Although endocrine therapy targeting ER is highly effective, intrinsic or acquired resistance is common, significantly jeopardizing treatment outcomes and minimizing overall survival. Even in the presence of endocrine resistance, a continued role of ER signaling is suggested by several lines of clinical and preclinical evidence. Indeed, inhibition or down-regulation of ER reduces tumor growth in preclinical models of acquired endocrine resistance, and many patients with recurrent ER+ breast tumors progressing on one type of ER-targeted treatment still benefit from sequential endocrine treatments that target ER by a different mechanism. New insights into the nature and biology of ER have revealed several mechanisms sustaining altered ER signaling in endocrine-resistant tumors, including deregulated growth factor receptor signaling that results in ligand-independent ER activation, unbalanced ER co-regulator activity, and genomic alterations involving the ER gene ESR1. Therefore, biopsies of recurrent lesions are needed to assess the changes in epi/genomics and signaling landscape of ER and associated pathways in order to tailor therapies to effectively overcome endocrine resistance. In addition, more completely abolishing the levels and activity of ER and its co-activators, in combination with selected signal transduction inhibitors or agents blocking the upstream or downstream targets of the ER pathway, may provide a better therapeutic strategy in combating endocrine resistance.
Collapse
Affiliation(s)
- Agostina Nardone
- Lester and Sue Smith Breast Center, Baylor College of Medicine, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, USA; Department of Medicine, Baylor College of Medicine, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, USA
| | - Carmine De Angelis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, USA; Department of Medicine, Baylor College of Medicine, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, USA; Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Italy
| | - Meghana V Trivedi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, USA; Department of Medicine, Baylor College of Medicine, USA; Department of Pharmacy Practice and Translational Research, University of Houston, College of Pharmacy, Houston, TX 77030, USA
| | - C Kent Osborne
- Lester and Sue Smith Breast Center, Baylor College of Medicine, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, USA; Department of Medicine, Baylor College of Medicine, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, USA
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Baylor College of Medicine, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, USA; Department of Medicine, Baylor College of Medicine, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, USA.
| |
Collapse
|
35
|
Batth IS, Yun H, Kumar AP. Recepteur d'origine nantais (RON), more than a kinase: Role in castrate-resistant prostate cancer. Mol Carcinog 2015; 54:937-46. [PMID: 26152593 DOI: 10.1002/mc.22354] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/20/2015] [Accepted: 05/28/2015] [Indexed: 12/30/2022]
Abstract
Prostate cancer (PCA) is the second leading cause of cancer-related deaths in men in the United States. It is natural for a hormone-driven malignancy such as prostate cancer that androgen deprivation therapy (ADT) would be the preferred treatment for clinical disease management. However, after initial treatment response a vast majority of patients develop metastatic castrate-resistant prostate cancer (CRPC), which is fatal. While great headway has been made to understand the possible mechanisms that drive castrate-resistant disease, a bonafide cure remains elusive. Reactivation of androgen receptor (AR) signaling partly contributes to the emergence of CRPC. Here we briefly examine some of the known mechanisms of AR reactivation including intratumoral synthesis of androgens, modulation of AR coregulators, and AR variants with constitutive activity as well as activation of receptor tyrosine kinases. We primarily focus on the emerging dual function of the receptor tyrosine kinase (recepteur d'origine nantais; RON) as a traditional tyrosine kinase and transcription factor. We further discuss activation of RON as an alternate mechanism in the development of CRPC and available therapeutic approaches for clinical management of CRPC by combined inhibition of RON and AR.
Collapse
Affiliation(s)
- Izhar Singh Batth
- Department of Urology, University of Texas Health Science Center, San Antonio, Texas
| | - Huiyoung Yun
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, Texas
| | - Addankl P Kumar
- Department of Urology, University of Texas Health Science Center, San Antonio, Texas.,Department of Pharmacology, University of Texas Health Science Center, San Antonio, Texas.,Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas.,Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, Texas.,South Texas Veterans Health Care System, San Antonio, Texas
| |
Collapse
|
36
|
Zhang C, Zhong Q, Zhang Q, Zheng S, Miele L, Wang G. Boronic prodrug of endoxifen as an effective hormone therapy for breast cancer. Breast Cancer Res Treat 2015; 152:283-91. [PMID: 26071758 PMCID: PMC4524496 DOI: 10.1007/s10549-015-3461-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/05/2015] [Indexed: 11/25/2022]
Abstract
As a prodrug, tamoxifen is activated by the P450 enzyme CYP2D6 that is responsible for converting it to the active metabolites, 4-hydroxytamoxifen and endoxifen. Patients with genetic polymorphisms of CYP2D6 may not receive the full benefit of tamoxifen therapy. There is increasing evidence that poor metabolizer patients have lower plasma concentrations of endoxifen and suffer worse disease outcome, although some clinical studies reported no correlation between CYP2D6 polymorphism and tamoxifen therapy outcome. Endoxifen is currently undergoing clinical trials as a potentially improved and more potent SERM (Selective Estrogen Receptor Modulator) for endocrine therapy that is independent of CYP2D6 status in patients. However, direct administration of endoxifen may present the problem of low bioavailability due to its rapid first-pass metabolism via O-glucuronidation. We have designed and synthesized ZB483, a boronic prodrug of endoxifen suitable for oral administration with greatly enhanced bioavailability by increasing the concentration of endoxifen in mouse blood. Our study demonstrated that ZB483 potently inhibited growth of ER+ breast cancer cells in vitro and was efficiently converted to endoxifen in cell culture media by oxidative deboronation. This metabolic conversion is equally efficient in vivo as indicated in the pharmacokinetic study in mice. Moreover, when administered at the same dose, oral ZB483 afforded a 30- to 40-fold higher plasma level of endoxifen in mice than oral administration of endoxifen. The significantly enhanced bioavailability of endoxifen conferred by the boronic prodrug was further validated in an in vivo efficacy study. ZB483 was demonstrated to be more efficacious than endoxifen in inhibiting xenograft tumor growth in mice at equal dosage but more so at lower dosage. Together, these preclinical studies demonstrate that ZB483 is a promising endocrine therapy agent with markedly enhanced bioavailability in systemic circulation and superior efficacy compared to endoxifen.
Collapse
Affiliation(s)
- Changde Zhang
- RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA, 70125, USA
| | | | | | | | | | | |
Collapse
|
37
|
Yan S, Jiao X, Zou H, Li K. Prognostic significance of c-Met in breast cancer: a meta-analysis of 6010 cases. Diagn Pathol 2015; 10:62. [PMID: 26047809 PMCID: PMC4458003 DOI: 10.1186/s13000-015-0296-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 05/12/2015] [Indexed: 12/21/2022] Open
Abstract
Background The prognostic value of c-Met in breast cancer remains controversial. A meta-analysis of the impact of c-Met in breast cancer was performed by searching published data. Methods Published studies analyzing overall survival (OS) or relapse free survival (RFS) according to c-Met expression were searched. The principal outcome measures were hazard ratios (HRs) for RFS or OS according to c-Met expression. Combined HRs were calculated using fixed- or random- effects models according to the heterogeneity. Results Twenty-one studies involving 6,010 patients met our selection criteria. The impact of c-Met on RFS and OS was investigated in 12 and 17 studies, respectively. The meta-analysis results showed that c-Met overexpression significantly predicted poor RFS and OS in unselected breast cancer. Subgroup analysis indicated that c-Met overexpression was correlated with poor RFS and OS in Western patients, but was not associated with RFS or OS in Asian patients. C-Met was associated with poor OS in lymph node negative breast cancer and with poor RFS in hormone-receptor positive and triple negative breast cancer, but was not associated with prognosis in human epidermal growth factor receptor (HER)-2 positive breast cancer. Conclusions C-Met overexpression is an adverse prognostic marker in breast cancer, except among Asian and HER-2 positive patients. Virtual slides The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/1869780799156041
Collapse
Affiliation(s)
- Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| | - Xin Jiao
- Department of Respiratory Medicine, Shenyang Chest Hospital, Shenyang, 110044, China.
| | - Huawei Zou
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| | - Kai Li
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| |
Collapse
|
38
|
Vasiliauskas J, Nashu MA, Pathrose P, Starnes SL, Waltz SE. Hepatocyte growth factor-like protein is required for prostate tumor growth in the TRAMP mouse model. Oncotarget 2015; 5:5547-58. [PMID: 24980820 PMCID: PMC4170603 DOI: 10.18632/oncotarget.2139] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The Ron receptor is deregulated in a variety of cancers. Hepatocyte growth factor-like protein (HGFL) is the ligand for Ron and is constitutively secreted from hepatocytes into the circulation. While a few recent reports have emerged analyzing ectopic HGFL overexpression in cancer cells, no studies have examined the effect of host-produced HGFL in tumorigenesis. To examine HGFL function in prostate cancer, the TRAMP mouse model, which is predisposed to develop prostate tumors, was utilized. Prostate tumors from TRAMP mice exhibit elevated levels of HGFL, which correlated with upregulation in human prostate cancer. To directly implicate HGFL in prostate tumorigenesis, TRAMP mice deficient in HGFL (HGFL-/-TRAMP+) were generated. HGFL-/- TRAMP+ mice developed significantly smaller prostate tumors compared to controls. Analysis of HGFL-/- tumors revealed reduced tumor vascularization. No differences in cancer cell proliferation were detected between HGFL-/- TRAMP+ and HGFL+/+ TRAMP+ mice. However, a significant increase in cancer cell death was detected in HGFL-/- TRAMP+ prostates which correlated with decreased pro-survival targets. In vitro analysis demonstrated robust STAT3 activation resulting in Bcl2-dependent survival following treatment of prostate cancer cells with HGFL. These data document a novel function for endogenous HGFL in prostate cancer by imparting a critical survival signal to tumor cells.
Collapse
Affiliation(s)
- Juozas Vasiliauskas
- Departments of Cancer Biology , Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio
| | - Madison A Nashu
- Departments of Cancer Biology , Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio
| | - Peterson Pathrose
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio
| | - Sandra L Starnes
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio
| | - Susan E Waltz
- Departments of Cancer Biology , Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio. Research Service, Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio
| |
Collapse
|
39
|
Privette Vinnedge LM, Benight NM, Wagh PK, Pease NA, Nashu MA, Serrano-Lopez J, Adams AK, Cancelas JA, Waltz SE, Wells SI. The DEK oncogene promotes cellular proliferation through paracrine Wnt signaling in Ron receptor-positive breast cancers. Oncogene 2015; 34:2325-36. [PMID: 24954505 PMCID: PMC4275425 DOI: 10.1038/onc.2014.173] [Citation(s) in RCA: 52] [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: 11/26/2013] [Revised: 04/18/2014] [Accepted: 05/09/2014] [Indexed: 12/12/2022]
Abstract
Disease progression and recurrence are major barriers to survival for breast cancer patients. Understanding the etiology of recurrent or metastatic breast cancer and underlying mechanisms is critical for the development of new treatments and improved survival. Here, we report that two commonly overexpressed breast cancer oncogenes, Ron (Recepteur d'Origine Nantaise) and DEK, cooperate to promote advanced disease through multipronged effects on β-catenin signaling. The Ron receptor is commonly activated in breast cancers, and Ron overexpression in human disease stimulates β-catenin nuclear translocation and is an independent predictor of metastatic dissemination. Dek is a chromatin-associated oncogene whose expression has been linked to cancer through multiple mechanisms, including β-catenin activity. We demonstrate here that Dek is a downstream target of Ron receptor activation in murine and human models. The absence of Dek in the MMTV-Ron mouse model led to a significant delay in tumor development, characterized by decreased cell proliferation, diminished metastasis and fewer cells expressing mammary cancer stem cell markers. Dek complementation of cell lines established from this model was sufficient to promote cellular growth and invasion. Mechanistically, Dek expression stimulated the production and secretion of Wnt ligands to sustain an autocrine/paracrine canonical β-catenin signaling loop. Finally, we show that Dek overexpression promotes tumorigenic phenotypes in immortalized human mammary epithelial MCF10A cells and, in the context of Ron receptor activation, correlates with disease recurrence and metastasis in patients. Overall, our studies demonstrate that DEK overexpression, due in part to Ron receptor activation, drives breast cancer progression through the induction of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
| | - Nancy M. Benight
- Department of Cancer Biology, University of Cincinnati College of Medicine Cincinnati, Cincinnati, OH
| | - Purnima K. Wagh
- Department of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Nicholas A. Pease
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Madison A. Nashu
- Department of Cancer Biology, University of Cincinnati College of Medicine Cincinnati, Cincinnati, OH
| | - Juana Serrano-Lopez
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- IMIBIC/UCO/University Hospital Reina Sofia, Cordoba, Spain
| | - Allie K. Adams
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Jose A. Cancelas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Susan E. Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine Cincinnati, Cincinnati, OH
- Department of Research, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH 45220
| | - Susanne I. Wells
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
40
|
Bruce MC, McAllister D, Murphy LC. The kinome associated with estrogen receptor-positive status in human breast cancer. Endocr Relat Cancer 2014; 21:R357-70. [PMID: 25056177 DOI: 10.1530/erc-14-0232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Estrogen receptor alpha (ERα) regulates and is regulated by kinases involved in several functions associated with the hallmarks of cancer. The following literature review strongly suggests that distinct kinomes exist for ERα-positive and -negative human breast cancers. Importantly, consistent with the known heterogeneity of ERα-positive cancers, different subgroups exist, which can be defined by different kinome signatures, which in turn are correlated with clinical outcome. Strong evidence supports the interplay of kinase networks, suggesting that targeting a single node may not be sufficient to inhibit the network. Therefore, identifying the important hubs/nodes associated with each clinically relevant kinome in ER+ tumors could offer the ability to implement the best therapy options at diagnosis, either endocrine therapy alone or together with other targeted therapies, for improved overall outcome.
Collapse
Affiliation(s)
- M Christine Bruce
- Department of Biochemistry and Medical GeneticsManitoba Institute of Cell Biology, University of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9
| | - Danielle McAllister
- Department of Biochemistry and Medical GeneticsManitoba Institute of Cell Biology, University of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9
| | - Leigh C Murphy
- Department of Biochemistry and Medical GeneticsManitoba Institute of Cell Biology, University of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9
| |
Collapse
|
41
|
Evaluation of stromal HGF immunoreactivity as a biomarker for melanoma response to RAF inhibitors. Mod Pathol 2014; 27:1193-202. [PMID: 24434899 PMCID: PMC4107197 DOI: 10.1038/modpathol.2013.226] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 10/12/2013] [Accepted: 10/13/2013] [Indexed: 12/13/2022]
Abstract
Of more than 150 000 published studies evaluating new biomarkers, fewer than 100 biomarkers have been implemented for patient care. One reason for this is lack of rigorous testing by the medical community to validate claims for biomarker clinical relevance, and potential reluctance to publish negative results when confirmation is not obtained. Here we sought to determine the utility and reproducibility of immunohistochemical detection of hepatocyte growth factor (HGF) in melanoma tissue, an approach of potential assistance in defining patients with innate resistance to BRAF inhibitor therapy. To this end, a published and a revised method that retained sensitivity but with greater specificity for HGF detection, were evaluated in cells known to endogenously express HGF, and in models where HGF is upregulated via cytokine induction and via overexpression by gene transfection. Consequent patient evaluation in collaboration with the Melanoma Institute Australia of a cohort of 41 melanoma specimens with extensive clinical annotation failed to validate HGF immunohistochemistry as a predictor of response to BRAF inhibitors. Targeted therapies for advanced melanoma and other cancers show great promise, and rigorous validation studies are thus indicated for approaches that seek to personalize such therapies to maximize therapeutic efficacy.
Collapse
|
42
|
Gurusamy D, Ruiz-Torres SJ, Johnson AL, Smith DA, Waltz SE. Hepatocyte growth factor-like protein is a positive regulator of early mammary gland ductal morphogenesis. Mech Dev 2014; 133:11-22. [PMID: 25049204 DOI: 10.1016/j.mod.2014.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 05/15/2014] [Accepted: 07/08/2014] [Indexed: 12/15/2022]
Abstract
The Ron receptor tyrosine kinase regulates multiple cellular processes and is important during mammary gland development and tumor progression. Hepatocyte growth factor-like protein [HGFL] is the only known ligand for the Ron receptor and recent studies have identified major roles for HGFL during breast cancer metastasis. Understanding the functional importance HGFL during mammary gland development will provide significant insights onto its contribution during tumor development and metastasis. In this study, we assessed the role of HGFL during postnatal mammary gland development using mice that were either proficient [HGFL +/+] or deficient [HGFL-/-] for HGFL. Postnatal ductal morphology and stromal cell associations were analyzed at multiple time points through puberty until adulthood. HGFL deficiency resulted in several mammary gland developmental defects including smaller terminal end buds [TEBs], significantly fewer TEBs, and delayed ductal outgrowth during early puberty. Additionally, HGFL deficient animals exhibited significantly altered TEB epithelial cell turnover with decreased proliferation and increased apoptosis coupled with decreased TEB diameter. Macrophage recruitment to the TEBs was also significantly decreased in the HGFL-/- mice compared to controls. Moreover, the levels of STAT3 mRNA as well as the phosphorylation status of this protein were lower in the HGFL-/- mammary glands compared to controls. Taken together, our data provide the first evidence for HGFL as a positive regulator of mammary gland ductal morphogenesis by controlling overall epithelial cell turnover, macrophage recruitment, and STAT3 activation in the developing mammary gland. With a function in early mammary gland development, HGFL represents a potential target for the development of novel breast cancer therapies.
Collapse
Affiliation(s)
- Devikala Gurusamy
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Sasha J Ruiz-Torres
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Abby L Johnson
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Dana A Smith
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Susan E Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA; Research Service, Cincinnati Veterans Hospital Medical Center, Cincinnati, OH 45267-0521, USA.
| |
Collapse
|
43
|
Wang Q, Quan H, Zhao J, Xie C, Wang L, Lou L. RON confers lapatinib resistance in HER2-positive breast cancer cells. Cancer Lett 2013; 340:43-50. [DOI: 10.1016/j.canlet.2013.06.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 06/04/2013] [Accepted: 06/20/2013] [Indexed: 12/26/2022]
|
44
|
Wang MH, Zhang R, Zhou YQ, Yao HP. Pathogenesis of RON receptor tyrosine kinase in cancer cells: activation mechanism, functional crosstalk, and signaling addiction. J Biomed Res 2013; 27:345-56. [PMID: 24086167 PMCID: PMC3783819 DOI: 10.7555/jbr.27.20130038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 05/27/2013] [Indexed: 12/15/2022] Open
Abstract
The RON receptor tyrosine kinase, a member of the MET proto-oncogene family, is a pathogenic factor implicated in tumor malignancy. Specifically, aberrations in RON signaling result in increased cancer cell growth, survival, invasion, angiogenesis, and drug resistance. Biochemical events such as ligand binding, receptor overexpression, generation of structure-defected variants, and point mutations in the kinase domain contribute to RON signaling activation. Recently, functional crosstalk between RON and signaling proteins such as MET and EFGR has emerged as an additional mechanism for RON activation, which is critical for tumorigenic development. The RON signaling crosstalk acts either as a regulatory feedback loop that strengthens or enhances tumorigenic phenotype of cancer cells or serves as a signaling compensatory pathway providing a growth/survival advantage for cancer cells to escape targeted therapy. Moreover, viral oncoproteins derived from Friend leukemia or Epstein-Barr viruses interact with RON to drive viral oncogenesis. In cancer cells, RON signaling is integrated into cellular signaling network essential for cancer cell growth and survival. These activities provide the molecular basis of targeting RON for cancer treatment. In this review, we will discuss recent data that uncover the mechanisms of RON activation in cancer cells, review evidence of RON signaling crosstalk relevant to cancer malignancy, and emphasize the significance of the RON signaling addiction by cancer cells for tumor therapy. Understanding aberrant RON signaling will not only provide insight into the mechanisms of tumor pathogenesis, but also lead to the development of novel strategies for molecularly targeted cancer treatment.
Collapse
Affiliation(s)
- Ming-Hai Wang
- Cancer Biology Research Center, ; Department of Biomedical Sciences, and
| | | | | | | |
Collapse
|
45
|
Overcoming intratumor heterogeneity of polygenic cancer drug resistance with improved biomarker integration. Neoplasia 2013; 14:1278-89. [PMID: 23308059 DOI: 10.1593/neo.122096] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 12/11/2012] [Accepted: 12/11/2012] [Indexed: 12/14/2022] Open
Abstract
Improvements in technology and resources are helping to advance our understanding of cancer-initiating events as well as factors involved with tumor progression, adaptation, and evasion of therapy. Tumors are well known to contain diverse cell populations and intratumor heterogeneity affords neoplasms with a diverse set of biologic characteristics that can be used to evolve and adapt. Intratumor heterogeneity has emerged as a major hindrance to improving cancer patient care. Polygenic cancer drug resistance necessitates reconsidering drug designs to include polypharmacology in pursuit of novel combinatorial agents having multitarget activity to overcome the diverse and compensatory signaling pathways in which cancer cells use to survive and evade therapy. Advances will require integration of different biomarkers such as genomics and imaging to provide for more adequate elucidation of the spatially varying location, type, and extent of diverse intratumor signaling molecules to provide for a rationale-based personalized cancer medicine strategy.
Collapse
|
46
|
Abstract
Since the discovery of MSP (macrophage-stimulating protein; also known as MST1 and hepatocyte growth factor-like (HGFL)) as the ligand for the receptor tyrosine kinase RON (also known as MST1R) in the early 1990s, the roles of this signalling axis in cancer pathogenesis has been extensively studied in various model systems. Both in vitro and in vivo evidence has revealed that MSP-RON signalling is important for the invasive growth of different types of cancers. Currently, small-molecule inhibitors and antibodies blocking RON signalling are under investigation. Substantial responses have been achieved in human tumour xenograft models, laying the foundation for clinical validation. In this Review, we discuss recent advances that demonstrate the importance of MSP-RON signalling in cancer and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Hang-Ping Yao
- Viral Oncogenesis Section in State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P. R. China
| | | | | | | |
Collapse
|
47
|
Ron tyrosine kinase receptor synergises with EGFR to confer adverse features in head and neck squamous cell carcinoma. Br J Cancer 2013; 109:482-92. [PMID: 23799848 PMCID: PMC3721396 DOI: 10.1038/bjc.2013.321] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/30/2013] [Accepted: 06/04/2013] [Indexed: 02/04/2023] Open
Abstract
Background: Although EGFR inhibitors have shown some success in the treatment of head and neck squamous cell carcinomas (HNSCCs), the results are not dramatic. Additional molecular targets are urgently needed. We previously showed that the loss of Ron receptor activity significantly slowed squamous tumour growth and progression in a murine model. Based on these data, we hypothesised that Ron expression confers an aggressive phenotype in HNSCCs. Methods: We prospectively collected and evaluated 154 snap-frozen, primary HNSCCs for Ron and EGFR expression/phosphorylation. Biomarker correlation with clinical, pathological and outcome data was performed. The biological responses of HNSCC cell lines to Ron knockdown, its activation and the biochemical interaction between Ron and EGFR were examined. Results: We discovered that 64.3% (99 out of 154) HNSCCs expressed Ron. The carcinomas expressed exclusively mature functional Ron, whereas the adjacent nonmalignant epithelium expressed predominantly nonfunctional Ron precursor. There was no significant association between Ron and sex, tumour differentiation, perineural/vascular invasion or staging. However, patients with Ron+HNSCC were significantly older and more likely to have oropharyngeal tumours. Ron+HNSCC also had significantly higher EGFR expression and correlated strongly with phosphorylated EGFR (pEGFR). Newly diagnosed HNSCC with either Ron/pEGFR or both had lower disease-free survival than those without Ron and pEGFR. Knocking down Ron in SCC9 cells significantly blunted their migratory response to not only the Ron ligand, MSP, but also EGF. Stimulation of Ron in SCC9 cells significantly augmented the growth effect of EGF; the synergistic effect of both growth factors in SCC9 cells was dependent on Ron expression. Activated Ron also interacted with and transactivated EGFR. Conclusion: Ron synergises with EGFR to confer certain adverse features in HNSCCs.
Collapse
|
48
|
Kim SH, Sehrawat A, Singh SV. Dietary chemopreventative benzyl isothiocyanate inhibits breast cancer stem cells in vitro and in vivo. Cancer Prev Res (Phila) 2013; 6:782-90. [PMID: 23661606 DOI: 10.1158/1940-6207.capr-13-0100] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A small subset of mammary tumor-initiating cells (also known as breast cancer stem cells; bCSC), characterized by expression of different markers [CD44(high)/CD24(low)/epithelial-specific antigen (ESA)+], aldehyde dehydrogenase-1 (ALDH1) activity, and ability to form mammospheres under ultra-low attachment culture conditions, are suspected to evade conventional therapies leading to disease recurrence. Elimination of both therapy-sensitive epithelial tumor cells and therapy-resistant bCSC is therefore necessary for prevention of breast cancer. We have shown previously that a nontoxic small-molecule constituent of edible cruciferous vegetables (benzyl isothiocyanate; BITC) inhibits mammary cancer development in mouse mammary tumor virus-neu (MMTV-neu) transgenic mice by causing epithelial tumor cell apoptosis. The present study shows efficacy of BITC against bCSC in vitro and in vivo. Mammosphere formation frequency and CD44(high)/CD24(low)/ESA+ and/or ALDH1+ populations in cultured MCF-7 (estrogen receptor-positive) and SUM159 (triple-negative) human breast cancer cells were decreased significantly in the presence of plasma achievable concentrations of BITC. BITC administration in the diet (3 μmol BITC/g diet for 29 weeks) resulted in a marked decrease in bCSCs in the MMTV-neu mice tumors in vivo. Overexpression of full-length Ron as well as its truncated form (sfRon), but not urokinase-type plasminogen activator receptor, conferred near complete protection against BITC-mediated inhibition of bCSCs in MCF-7 cells. The BITC treatment downregulated protein levels of Ron and sfRon in cultured breast cancer cells and in tumor xenografts. Ron overexpression resulted in upregulation of bCSC-associated genes Oct-4, SOX-2, and Nanog. In conclusion, the present study indicates that BITC treatment eliminates bCSCs in vitro and in vivo.
Collapse
Affiliation(s)
- Su-Hyeong Kim
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
49
|
Zhao H, Chen MS, Lo YH, Waltz SE, Wang J, Ho PC, Vasiliauskas J, Plattner R, Wang YL, Wang SC. The Ron receptor tyrosine kinase activates c-Abl to promote cell proliferation through tyrosine phosphorylation of PCNA in breast cancer. Oncogene 2013; 33:1429-37. [PMID: 23542172 DOI: 10.1038/onc.2013.84] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 12/12/2012] [Accepted: 01/11/2013] [Indexed: 12/14/2022]
Abstract
Multiple growth pathways lead to enhanced proliferation in malignant cells. However, how the core machinery of DNA replication is regulated by growth signaling remains largely unclear. The sliding clamp proliferating cell nuclear antigen (PCNA) is an indispensable component of the DNA machinery responsible for replicating the genome and maintaining genomic integrity. We previously reported that epidermal growth factor receptor (EGFR) triggered tyrosine 211 (Y211) phosphorylation of PCNA, which in turn stabilized PCNA on chromatin to promote cell proliferation. Here we show that the phosphorylation can also be catalyzed by the non-receptor tyrosine kinase c-Abl. We further demonstrate that, in the absence of EGFR, signaling to PCNA can be attained through the activation of the Ron receptor tyrosine kinase and the downstream non-receptor tyrosine kinase c-Abl. We show that Ron and c-Abl form a complex, and that activation of Ron by its ligand, hepatocyte growth factor-like protein (HGFL), stimulates c-Abl kinase activity, which in turn directly phosphorylates PCNA at Y211 and leads to an increased level of chromatin-associated PCNA. Correspondingly, HGFL-induced Ron activation resulted in Y211 phosphorylation of PCNA while silencing of c-Abl blocked this effect. We show that c-Abl and Y211 phosphorylation of PCNA is an important axis downstream of Ron, which is required for cell proliferation. Treatment with a specific peptide that inhibits Y211 phosphorylation of PCNA or with the c-Abl pharmacological inhibitor imatinib suppressed HGFL-induced cell proliferation. Our findings identify the pathway of Ron-c-Abl-PCNA as a mechanism of oncogene-induced cell proliferation, with potentially important implications for development of combination therapy of breast cancer.
Collapse
Affiliation(s)
- H Zhao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - M-S Chen
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Y-H Lo
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - S E Waltz
- 1] Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA [2] Cincinnati Veterans Affairs Medical Center, Cincinnati, OH, USA
| | - J Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - P-C Ho
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - J Vasiliauskas
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - R Plattner
- Department of Molecular and Biomedical Pharmacology, University of Kentucky School of Medicine, Lexington, KY, USA
| | - Y-L Wang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - S-C Wang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
50
|
Kapoor S. Ron and its expanding role in tumor carcinogenesis. APMIS 2013; 121:898-9. [PMID: 23294341 DOI: 10.1111/apm.12045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|