1
|
Wang J, Fang Y, Luo Z, Wang J, Zhao Y. Emerging mRNA Technology for Liver Disease Therapy. ACS NANO 2024; 18:17378-17406. [PMID: 38916747 DOI: 10.1021/acsnano.4c02987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Liver diseases have consistently posed substantial challenges to global health. It is crucial to find innovative methods to effectively prevent and treat these diseases. In recent times, there has been an increasing interest in the use of mRNA formulations that accumulate in liver tissue for the treatment of hepatic diseases. In this review, we start by providing a detailed introduction to the mRNA technology. Afterward, we highlight types of liver diseases, discussing their causes, risks, and common therapeutic strategies. Additionally, we summarize the latest advancements in mRNA technology for the treatment of liver diseases. This includes systems based on hepatocyte growth factor, hepatitis B virus antibody, left-right determination factor 1, human hepatocyte nuclear factor α, interleukin-12, methylmalonyl-coenzyme A mutase, etc. Lastly, we provide an outlook on the potential of mRNA technology for the treatment of liver diseases, while also highlighting the various technical challenges that need to be addressed. Despite these difficulties, mRNA-based therapeutic strategies may change traditional treatment methods, bringing hope to patients with liver diseases.
Collapse
Affiliation(s)
- Ji Wang
- Department of Rheumatology and Immunology, Institute of Translational Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yile Fang
- Department of Rheumatology and Immunology, Institute of Translational Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Zhiqiang Luo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Jinglin Wang
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
2
|
Santoro A, Assenat E, Yau T, Delord JP, Maur M, Knox J, Cattan S, Lee KH, Del Conte G, Springfeld C, Leo E, Xyrafas A, Fairchild L, Mardjuadi F, Chan SL. A phase Ib/II trial of capmatinib plus spartalizumab vs. spartalizumab alone in patients with pretreated hepatocellular carcinoma. JHEP Rep 2024; 6:101021. [PMID: 38617599 PMCID: PMC11009449 DOI: 10.1016/j.jhepr.2024.101021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 12/22/2023] [Accepted: 01/11/2024] [Indexed: 04/16/2024] Open
Abstract
Background & aims This phase Ib/II trial evaluated the safety and efficacy of capmatinib in combination with spartalizumab or spartalizumab alone in patients with advanced hepatocellular carcinoma (HCC). Methods Eligible patients who had progressed or were intolerant to sorafenib received escalating doses of capmatinib 200 mg, 300 mg, and 400 mg twice a day (bid) plus spartalizumab 300 mg every 3 weeks (q3w) in the phase Ib study. Once the recommended phase II dose (RP2D) was determined, the phase II study commenced with randomised 1:1 treatment with either capmatinib + spartalizumab (n = 32) or spartalizumab alone (n = 30). Primary endpoints were safety and tolerability (phase Ib) and investigator-assessed overall response rate per RECIST v1.1 for combination vs. single-agent arms using a Bayesian logistic regression model (phase II). Results In phase Ib, the RP2D for capmatinib in combination with spartalizumab was determined to be 400 mg bid. Dose-limiting toxicity consisting of grade 3 diarrhoea was reported in one patient at the capmatinib 400 mg bid + spartalizumab 300 mg q3w dose level. The primary endpoint in the phase II study was not met. The observed overall response rate in the capmatinib + spartalizumab arm was 9.4% vs. 10% in the spartalizumab arm. The most common any-grade treatment-related adverse events (TRAEs, ≥20%) were nausea (37.5%), asthenia and vomiting (28.1% each), diarrhoea, pyrexia, and decreased appetite (25.0% each) in the combination arm; TRAEs ≥10% were pruritus (23.3%), and rash (10.0%) in the spartalizumab-alone arm. Conclusion Capmatinib at 400 mg bid plus spartalizumab 300 mg q3w was established as the RP2D, with manageable toxicities and no significant safety signals, but the combination did not show superior clinical activity compared with spartalizumab single-agent treatment in patients with advanced HCC who had previously been treated with sorafenib. Impact and implications Simultaneous targeting of MET and programmed cell death protein 1 may provide synergistic clinical benefit in patients with advanced HCC. This is the first trial to report a combination of capmatinib (MET inhibitor) and spartalizumab (programmed cell death protein 1 inhibitor) as second-line treatment after sorafenib for advanced HCC. The combination did not show superior clinical activity compared with spartalizumab single-agent treatment in patients with advanced HCC who had previously been treated with sorafenib. The results indicate that there is a clear need to identify a reliable predictive marker of response for HCC and to identify patients with HCC that would benefit from the combination of checkpoint inhibitor +/- targeted therapy. Clinical trial number NCT02795429.
Collapse
Affiliation(s)
- Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele – Milan, Italy
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, Rozzano, Milan, Italy
| | - Eric Assenat
- Hopital Arnaud de Villeneuve Montpellier Cedex 5, Herault, France
| | - Thomas Yau
- Department of Medicine, Queen Mary Hospital, Hong Kong, China
| | | | - Michela Maur
- Oncology Unit, AOU Policlinico Modena and University Study of Modena and Reggio Emilia, Modena, Italy
| | | | | | - Kyung-Hun Lee
- Seoul National University Hospital, Seoul, South Korea
| | - Gianluca Del Conte
- Department of Oncology, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Christoph Springfeld
- Nat. Centrum f. Tumorerkrankungen, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Elisa Leo
- Novartis Pharma AG, Basel, Switzerland
| | | | - Lauren Fairchild
- Oncology Data Science, Novartis Institutes for BioMedical Research, Cambridge, USA
| | - Feby Mardjuadi
- Novartis Institutes for Biomedical Research Co., Ltd., Shanghai, China
| | - Stephen L. Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
3
|
Qin S, Pan H, Blanc JF, Grando V, Lim HY, Chang XY, O'Brate A, Stroh C, Friese-Hamim M, Albers J, Johne A, Faivre S. Activity of Tepotinib in Hepatocellular Carcinoma With High-Level MET Amplification: Preclinical and Clinical Evidence. JCO Precis Oncol 2024; 8:e2300328. [PMID: 38354329 DOI: 10.1200/po.23.00328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/29/2023] [Accepted: 12/11/2023] [Indexed: 02/16/2024] Open
Abstract
PURPOSE MET amplification (METamp) has been reported in 1%-5% of patients with hepatocellular carcinoma (HCC) and may be sensitive to MET inhibition. Tepotinib, a selective MET inhibitor, has shown promising activity in HCC with MET overexpression. We investigated the preclinical and clinical activity of tepotinib in HCC with METamp (MET gene copy number [GCN] ≥5), including high-level METamp (MET GCN ≥10). METHODS Preclinical antitumor activity of tepotinib 100 mg/kg (orally, days 1-5, every 7 days, 3-5 weeks; 3-12 replicates) was evaluated according to METamp status, as determined using the nCounter platform (NanoString), in 37 HCC patient-derived xenografts (PDXs) in immunodeficient mice. Clinical outcomes were evaluated in patients with METamp by fluorescence in situ hybridization who received tepotinib 500 mg (450 mg active moiety) in two phase Ib/II trials in HCC with MET overexpression. RESULTS Across the PDX models, tepotinib induced complete or near-complete tumor regression in the only two models with high-level METamp. Median tumor volume reductions were 100% and 99.8% in models with MET GCN 47.1 and 44.0, respectively. Across the two clinical trials, 15/121 patients had METamp. Disease control was achieved by 11/15 patients with METamp (complete response [CR], n = 1; partial response [PR], n = 4; stable disease [SD], n = 6) and 4/4 with high-level METamp (CR, n = 1; PR, n = 2; SD, n = 1). All three patients with high-level METamp and objective response received treatment for >1 year, including one patient who received first-line tepotinib for >6 years. CONCLUSION High-level METamp may be an oncogenic driver in HCC that is sensitive to MET inhibitors such as tepotinib.
Collapse
Affiliation(s)
- Shukui Qin
- PLA Cancer Center, Nanjing Bayi Hospital, Nanjing, China
- Cancer Center of Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongming Pan
- Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | | | | | - Ho Yeong Lim
- Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Xin Ying Chang
- Global Clinical Development China, Merck Serono Co., Ltd, Beijing, China, an affiliate of Merck KGaA, Darmstadt, Germany
| | - Aurora O'Brate
- The healthcare business of Merck KGaA, Darmstadt, Germany
| | | | | | - Joachim Albers
- The healthcare business of Merck KGaA, Darmstadt, Germany
| | - Andreas Johne
- The healthcare business of Merck KGaA, Darmstadt, Germany
| | | |
Collapse
|
4
|
Qin A, Qin Y, Lee J, Musket A, Ying M, Krenciute G, Marincola FM, Yao ZQ, Musich PR, Xie Q. Tyrosine kinase signaling-independent MET-targeting with CAR-T cells. J Transl Med 2023; 21:682. [PMID: 37779207 PMCID: PMC10544186 DOI: 10.1186/s12967-023-04521-9] [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: 07/28/2023] [Accepted: 09/11/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND Recent progress in cancer immunotherapy encourages the expansion of chimeric antigen receptor (CAR) T cell therapy in solid tumors including hepatocellular carcinoma (HCC). Overexpression of MET receptor tyrosine kinase is common in HCC; however, MET inhibitors are effective only when MET is in an active form, making patient stratification difficult. Specific MET-targeting CAR-T cells hold the promise of targeting HCC with MET overexpression regardless of signaling pathway activity. METHODS MET-specific CARs with CD28ζ or 4-1BBζ as co-stimulation domains were constructed. MET-CAR-T cells derived from healthy subjects (HS) and HCC patients were evaluated for their killing activity and cytokine release against HCC cells with various MET activations in vitro, and for their tumor growth inhibition in orthotopic xenograft models in vivo. RESULTS MET-CAR.CD28ζ and MET-CAR.4-1BBζ T cells derived from both HS and HCC patients specifically killed MET-positive HCC cells. When stimulated with MET-positive HCC cells in vitro, MET-CAR.CD28ζ T cells demonstrated a higher level of cytokine release and expression of programmed cell death protein 1 (PD-1) than MET-CAR.4-1BBζ T cells. When analyzed in vivo, MET-CAR.CD28ζ T cells more effectively inhibited HCC orthotopic tumor growth in mice when compared to MET-CAR.4-1BBζ T cells. CONCLUSION We generated and characterized MET-specific CAR-T cells for targeting HCC with MET overexpression regardless of MET activation. Compared with MET-CAR.4-1BBζ, MET-CAR.CD28ζ T cells showed a higher anti-HCC potency but also a higher level of T cell exhaustion. While MET-CAR.CD28ζ is preferred for further development, overcoming the exhaustion of MET-CAR-T cells is necessary to improve their therapeutic efficacy in vivo.
Collapse
Affiliation(s)
- Anna Qin
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Yuan Qin
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Joseph Lee
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Anna Musket
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Mingyao Ying
- Department of Neurology, Hugo W. Moser Research Institute at Kennedy Krieger, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Giedre Krenciute
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | | | - Zhi Q Yao
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Phillip R Musich
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Qian Xie
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.
| |
Collapse
|
5
|
Affo S, Filliol A, Gores GJ, Schwabe RF. Fibroblasts in liver cancer: functions and therapeutic translation. Lancet Gastroenterol Hepatol 2023; 8:748-759. [PMID: 37385282 DOI: 10.1016/s2468-1253(23)00111-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 07/01/2023]
Abstract
Accumulation of fibroblasts in the premalignant or malignant liver is a characteristic feature of liver cancer, but has not been therapeutically leveraged despite evidence for pathophysiologically relevant roles in tumour growth. Hepatocellular carcinoma is a largely non-desmoplastic tumour, in which fibroblasts accumulate predominantly in the pre-neoplastic fibrotic liver and regulate the risk for hepatocellular carcinoma development through a balance of tumour-suppressive and tumour-promoting mediators. By contrast, cholangiocarcinoma is desmoplastic, with cancer-associated fibroblasts contributing to tumour growth. Accordingly, restoring the balance from tumour-promoting to tumour-suppressive fibroblasts and mediators might represent a strategy for hepatocellular carcinoma prevention, whereas in cholangiocarcinoma, fibroblasts and their mediators could be leveraged for tumour treatment. Importantly, fibroblast mediators regulating hepatocellular carcinoma development might exert opposite effects on cholangiocarcinoma growth. This Review translates the improved understanding of tumour-specific, location-specific, and stage-specific roles of fibroblasts and their mediators in liver cancer into novel and rational therapeutic concepts.
Collapse
Affiliation(s)
- Silvia Affo
- Department of Liver, Digestive System, and Metabolism, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Aveline Filliol
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
6
|
Patil VS, Harish DR, Sampat GH, Roy S, Jalalpure SS, Khanal P, Gujarathi SS, Hegde HV. System Biology Investigation Revealed Lipopolysaccharide and Alcohol-Induced Hepatocellular Carcinoma Resembled Hepatitis B Virus Immunobiology and Pathogenesis. Int J Mol Sci 2023; 24:11146. [PMID: 37446321 DOI: 10.3390/ijms241311146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 07/15/2023] Open
Abstract
Hepatitis B infection caused by the hepatitis B virus is a life-threatening cause of liver fibrosis, cirrhosis, and hepatocellular carcinoma. Researchers have produced multiple in vivo models for hepatitis B virus (HBV) and, currently, there are no specific laboratory animal models available to study HBV pathogenesis or immune response; nonetheless, their limitations prevent them from being used to study HBV pathogenesis, immune response, or therapeutic methods because HBV can only infect humans and chimpanzees. The current study is the first of its kind to identify a suitable chemically induced liver cirrhosis/HCC model that parallels HBV pathophysiology. Initially, data from the peer-reviewed literature and the GeneCards database were compiled to identify the genes that HBV and seven drugs (acetaminophen, isoniazid, alcohol, D-galactosamine, lipopolysaccharide, thioacetamide, and rifampicin) regulate. Functional enrichment analysis was performed in the STRING server. The network HBV/Chemical, genes, and pathways were constructed by Cytoscape 3.6.1. About 1546 genes were modulated by HBV, of which 25.2% and 17.6% of the genes were common for alcohol and lipopolysaccharide-induced hepatitis. In accordance with the enrichment analysis, HBV activates the signaling pathways for apoptosis, cell cycle, PI3K-Akt, TNF, JAK-STAT, MAPK, chemokines, NF-kappa B, and TGF-beta. In addition, alcohol and lipopolysaccharide significantly activated these pathways more than other chemicals, with higher gene counts and lower FDR scores. In conclusion, alcohol-induced hepatitis could be a suitable model to study chronic HBV infection and lipopolysaccharide-induced hepatitis for an acute inflammatory response to HBV.
Collapse
Affiliation(s)
- Vishal S Patil
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi 590010, India
| | - Darasaguppe R Harish
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi 590010, India
| | - Ganesh H Sampat
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi 590010, India
| | - Subarna Roy
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi 590010, India
| | - Sunil S Jalalpure
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi 590010, India
| | - Pukar Khanal
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi 590010, India
| | - Swarup S Gujarathi
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi 590010, India
| | - Harsha V Hegde
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi 590010, India
| |
Collapse
|
7
|
Small Molecule Inhibitors for Hepatocellular Carcinoma: Advances and Challenges. Molecules 2022; 27:molecules27175537. [PMID: 36080304 PMCID: PMC9457820 DOI: 10.3390/molecules27175537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 12/12/2022] Open
Abstract
According to data provided by World Health Organization, hepatocellular carcinoma (HCC) is the sixth most common cause of deaths due to cancer worldwide. Tremendous progress has been achieved over the last 10 years developing novel agents for HCC treatment, including small-molecule kinase inhibitors. Several small molecule inhibitors currently form the core of HCC treatment due to their versatility since they would be more easily absorbed and have higher oral bioavailability, thus easier to formulate and administer to patients. In addition, they can be altered structurally to have greater volumes of distribution, allowing them to block extravascular molecular targets and to accumulate in a high concentration in the tumor microenvironment. Moreover, they can be designed to have shortened half-lives to control for immune-related adverse events. Most importantly, they would spare patients, healthcare institutions, and society as a whole from the burden of high drug costs. The present review provides an overview of the pharmaceutical compounds that are licensed for HCC treatment and other emerging compounds that are still investigated in preclinical and clinical trials. These molecules are targeting different molecular targets and pathways that are proven to be involved in the pathogenesis of the disease.
Collapse
|
8
|
Kelley RK, Rimassa L, Cheng AL, Kaseb A, Qin S, Zhu AX, Chan SL, Melkadze T, Sukeepaisarnjaroen W, Breder V, Verset G, Gane E, Borbath I, Rangel JDG, Ryoo BY, Makharadze T, Merle P, Benzaghou F, Banerjee K, Hazra S, Fawcett J, Yau T. Cabozantinib plus atezolizumab versus sorafenib for advanced hepatocellular carcinoma (COSMIC-312): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2022; 23:995-1008. [PMID: 35798016 DOI: 10.1016/s1470-2045(22)00326-6] [Citation(s) in RCA: 273] [Impact Index Per Article: 136.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cabozantinib has shown clinical activity in combination with checkpoint inhibitors in solid tumours. The COSMIC-312 trial assessed cabozantinib plus atezolizumab versus sorafenib as first-line systemic treatment for advanced hepatocellular carcinoma. METHODS COSMIC-312 is an open-label, randomised, phase 3 trial that enrolled patients aged 18 years or older with advanced hepatocellular carcinoma not amenable to curative or locoregional therapy and previously untreated with systemic anticancer therapy at 178 centres in 32 countries. Patients with fibrolamellar carcinoma, sarcomatoid hepatocellular carcinoma, or combined hepatocellular cholangiocarcinoma were not eligible. Tumours involving major blood vessels, including the main portal vein, were permitted. Patients were required to have measurable disease per Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1), Barcelona Clinic Liver Cancer stage B or C disease, an Eastern Cooperative Oncology Group performance status of 0 or 1, adequate organ and marrow function, and Child-Pugh class A. Previous resection, tumour ablation, radiotherapy, or arterial chemotherapy was allowed if more than 28 days before randomisation. Patients were randomly assigned (2:1:1) via a web-based interactive response system to cabozantinib 40 mg orally once daily plus atezolizumab 1200 mg intravenously every 3 weeks, sorafenib 400 mg orally twice daily, or single-agent cabozantinib 60 mg orally once daily. Randomisation was stratified by disease aetiology, geographical region, and presence of extrahepatic disease or macrovascular invasion. Dual primary endpoints were progression-free survival per RECIST 1.1 as assessed by a blinded independent radiology committee in the first 372 patients randomly assigned to the combination treatment of cabozantinib plus atezolizumab or sorafenib (progression-free survival intention-to-treat [ITT] population), and overall survival in all patients randomly assigned to cabozantinib plus atezolizumab or sorafenib (ITT population). Final progression-free survival and concurrent interim overall survival analyses are presented. This trial is registered with ClinicalTrials.gov, NCT03755791. FINDINGS Analyses at data cut-off (March 8, 2021) included the first 837 patients randomly assigned between Dec 7, 2018, and Aug 27, 2020, to combination treatment of cabozantinib plus atezolizumab (n=432), sorafenib (n=217), or single-agent cabozantinib (n=188). Median follow-up was 15·8 months (IQR 14·5-17·2) in the progression-free survival ITT population and 13·3 months (10·5-16·0) in the ITT population. Median progression-free survival was 6·8 months (99% CI 5·6-8·3) in the combination treatment group versus 4·2 months (2·8-7·0) in the sorafenib group (hazard ratio [HR] 0·63, 99% CI 0·44-0·91, p=0·0012). Median overall survival (interim analysis) was 15·4 months (96% CI 13·7-17·7) in the combination treatment group versus 15·5 months (12·1-not estimable) in the sorafenib group (HR 0·90, 96% CI 0·69-1·18; p=0·44). The most common grade 3 or 4 adverse events were alanine aminotransferase increase (38 [9%] of 429 patients in the combination treatment group vs six [3%] of 207 in the sorafenib group vs 12 [6%] of 188 in the single-agent cabozantinib group), hypertension (37 [9%] vs 17 [8%] vs 23 [12%]), aspartate aminotransferase increase (37 [9%] vs eight [4%] vs 18 [10%]), and palmar-plantar erythrodysaesthesia (35 [8%] vs 17 [8%] vs 16 [9%]); serious treatment-related adverse events occurred in 78 (18%) patients in the combination treatment group, 16 (8%) patients in the sorafenib group, and 24 (13%) in the single-agent cabozantinib group. Treatment-related grade 5 events occurred in six (1%) patients in the combination treatment group (encephalopathy, hepatic failure, drug-induced liver injury, oesophageal varices haemorrhage, multiple organ dysfunction syndrome, and tumour lysis syndrome), one (<1%) patient in the sorafenib group (general physical health deterioration), and one (<1%) patient in the single-agent cabozantinib group (gastrointestinal haemorrhage). INTERPRETATION Cabozantinib plus atezolizumab might be a treatment option for select patients with advanced hepatocellular carcinoma, but additional studies are needed. FUNDING Exelixis and Ipsen.
Collapse
Affiliation(s)
- Robin Kate Kelley
- Department of Medicine (Hematology/Oncology), UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA.
| | - Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Ann-Lii Cheng
- National Taiwan University Cancer Center, Taipei, Taiwan; National Taiwan University Hospital, Taipei, Taiwan
| | - Ahmed Kaseb
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shukui Qin
- Cancer Center of Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Andrew X Zhu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA; Jiahui International Cancer Center, Jiahui Health, Shanghai, China
| | - Stephen L Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir Yue-Kong Pao Center for Cancer, The Chinese University of Hong Kong, Hong Kong
| | - Tamar Melkadze
- Academician Fridon Todua Medical Center-Research Institute of Clinical Medicine Tbilisi, Georgia
| | | | - Valery Breder
- FSBSI N N Blokhin Russian Cancer Research Center, Moscow, Russia
| | - Gontran Verset
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Edward Gane
- New Zealand Liver Transplant Unit, Auckland City Hospital, Auckland, New Zealand; Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Ivan Borbath
- Department of Hepato-Gastroenterology, Cliniques Universitaires St Luc, Brussels, Belgium
| | | | - Baek-Yeol Ryoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | | | - Philippe Merle
- Hepatology Unit, Hôpital de la Croix-Rousse, Groupement Hospitalier Lyon Nord, Lyon, France
| | | | | | | | | | - Thomas Yau
- Department of Medicine, University of Hong Kong, Hong Kong, China
| |
Collapse
|
9
|
Association between immunologic markers and cirrhosis in individuals with chronic hepatitis B. Sci Rep 2021; 11:21194. [PMID: 34782638 PMCID: PMC8593047 DOI: 10.1038/s41598-021-00455-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/23/2021] [Indexed: 12/20/2022] Open
Abstract
Host immune response and chronic inflammation associated with chronic hepatitis B virus (HBV) infection play a key role in the pathogenesis of liver diseases such as cirrhosis and hepatocellular carcinoma (HCC). We sampled 175 HCC, 117 cirrhotic and 165 non-cirrhotic controls from a prospective cohort study of chronically HBV-infected individuals. Multivariable polytomous logistic regression and canonical discriminant analysis (CDA) were used to compare baseline plasma levels for 102 markers in individuals who developed cirrhosis vs. controls and those who developed HCC vs. cirrhosis. Leave-one-out cross validation was used to generate receiver operating characteristic curves to compare the predictive ability of marker groups. After multivariable adjustment, HGF (Q4v1OR: 3.74; p-trend = 0.0001), SLAMF1 (Q4v1OR: 4.07; p-trend = 0.0001), CSF1 (Q4v1OR: 3.00; p-trend = 0.002), uPA (Q4v1OR: 3.36; p-trend = 0.002), IL-8 (Q4v1OR: 2.83; p-trend = 0.004), and OPG (Q4v1OR: 2.44; p-trend = 0.005) were all found to be associated with cirrhosis development compared to controls; these markers predicted cirrhosis with 69% accuracy. CDA analysis identified a nine marker model capable of predicting cirrhosis development with 79% accuracy. No markers were significantly different between HCC and cirrhotic participants. In this study, we assessed immunologic markers in relation to liver disease in chronically-HBV infected individuals. While validation in required, these findings highlight the importance of immunologic processes in HBV-related cirrhosis.
Collapse
|
10
|
Yu J, Chen GG, Lai PBS. Targeting hepatocyte growth factor/c-mesenchymal-epithelial transition factor axis in hepatocellular carcinoma: Rationale and therapeutic strategies. Med Res Rev 2020; 41:507-524. [PMID: 33026703 DOI: 10.1002/med.21738] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/27/2020] [Accepted: 09/27/2020] [Indexed: 12/27/2022]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality worldwide. The outcome of current standard treatments, as well as targeted therapies in advanced stages, are still unsatisfactory. Attention has been drawn to novel strategies for better treatment efficacy. Hepatocyte growth factor/c-mesenchymal-epithelial transition factor (HGF/c-Met) axis has been known as an essential element in the regulation of liver diseases and as an oncogenic factor in HCC. In this review, we collected the evidence of HGF/c-Met as a tumor progression and prognostic marker, discussed the anti-c-Met therapy in vitro, summarized the outcome of c-Met inhibitors in clinical trials, and identified potential impetus for future anti-c-Met treatments. We also analyzed the inconsistency of HGF/c-Met from various publications and offered reasonable explanations based on the current understanding in this area. In conclusion, HGF/c-Met plays a crucial role in the progression and growth of HCC, and the strategies to inhibit this pathway may facilitate the development of new and effective treatments for HCC patients.
Collapse
Affiliation(s)
- Jianqing Yu
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - George G Chen
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Department of Otorhinolaryngology, Head and Neck Surgery, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Paul B S Lai
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
11
|
Qin Y, Musket A, Kou J, Preiszner J, Tschida BR, Qin A, Land CA, Staal B, Kang L, Tanner K, Jiang Y, Schweitzer JB, Largaespada DA, Xie Q. Overexpression of HGF/MET axis along with p53 inhibition induces de novo glioma formation in mice. Neurooncol Adv 2020; 2:vdaa067. [PMID: 32642717 PMCID: PMC7332240 DOI: 10.1093/noajnl/vdaa067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Aberrant MET receptor tyrosine kinase (RTK) activation leads to invasive tumor growth in different types of cancer. Overexpression of MET and its ligand hepatocyte growth factor (HGF) occurs more frequently in glioblastoma (GBM) than in low-grade gliomas. Although we have shown previously that HGF-autocrine activation predicts sensitivity to MET tyrosine kinase inhibitors (TKIs) in GBM, whether it initiates tumorigenesis remains elusive. Methods Using a well-established Sleeping Beauty (SB) transposon strategy, we injected human HGF and MET cDNA together with a short hairpin siRNA against Trp53 (SB-hHgf.Met.ShP53) into the lateral ventricle of neonatal mice to induce spontaneous glioma initiation and characterized the tumors with H&E and immunohistochemistry analysis. Glioma sphere cells also were isolated for measuring the sensitivity to specific MET TKIs. Results Mixed injection of SB-hHgf.Met.ShP53 plasmids induced de novo glioma formation with invasive tumor growth accompanied by HGF and MET overexpression. While glioma stem cells (GSCs) are considered as the tumor-initiating cells in GBM, both SB-hHgf.Met.ShP53 tumor sections and glioma spheres harvested from these tumors expressed GSC markers nestin, GFAP, and Sox 2. Moreover, specific MET TKIs significantly inhibited tumor spheres' proliferation and MET/MAPK/AKT signaling. Conclusions Overexpression of the HGF/MET axis along with p53 attenuation may transform neural stem cells into GSCs, resulting in GBM formation in mice. These tumors are primarily driven by the MET RTK pathway activation and are sensitive to MET TKIs. The SB-hHgf.Met.ShP53 spontaneous mouse glioma model provides a useful tool for studying GBM tumor biology and MET-targeting therapeutics.
Collapse
Affiliation(s)
- Yuan Qin
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Anna Musket
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Jianqun Kou
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Johanna Preiszner
- Department of Pathology, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Barbara R Tschida
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anna Qin
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Craig A Land
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Ben Staal
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Liang Kang
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Kirk Tanner
- National Brain Tumor Society, Newton, Massachusetts, USA
| | - Yong Jiang
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - John B Schweitzer
- Department of Pathology, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - David A Largaespada
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Qian Xie
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| |
Collapse
|
12
|
Qin S, Chan SL, Sukeepaisarnjaroen W, Han G, Choo SP, Sriuranpong V, Pan H, Yau T, Guo Y, Chen M, Ren Z, Xu J, Yen CJ, Lin ZZ, Manenti L, Gu Y, Sun Y, Tiedt R, Hao L, Song W, Tanwandee T. A phase II study of the efficacy and safety of the MET inhibitor capmatinib (INC280) in patients with advanced hepatocellular carcinoma. Ther Adv Med Oncol 2019; 11:1758835919889001. [PMID: 31853265 PMCID: PMC6906348 DOI: 10.1177/1758835919889001] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 10/17/2019] [Indexed: 12/30/2022] Open
Abstract
Background The objectives of this phase II study were to determine the clinical activity of the MET tyrosine kinase inhibitor capmatinib (INC280) in patients with MET-dysregulated advanced hepatocellular carcinoma (HCC) and to assess the safety, pharmacokinetics, and correlation of biomarkers with the response. Methods This phase II, open-label, single-arm study evaluated twice daily (BID) oral capmatinib in a dose-determining stage, utilizing a Bayesian Logistic Regression Model (BLRM) subject to Escalation with Overdose Control criteria, safety, pharmacokinetics, and pharmacodynamic information to determine a recommended dose for expansion (RDE) evaluating efficacy in patients with MET-dysregulated HCC. Results A total of 38 patients received treatment. In the dose-determining stage, patients received capmatinib 300 mg BID capsules (n = 8), and in the expansion, patients received 600 mg BID capsules (n = 28) or 400 mg BID tablets (n = 2) based on the BLRM and other relevant clinical data. No predefined qualifying adverse events (AEs) were observed during the first 28 days of treatment, and the RDE was 600 mg BID capsules (equivalent pharmacokinetics to 400 mg BID tablets). The most common any causality AEs were nausea (42%), vomiting (37%), and diarrhea (34%). In the expansion stage, in a subgroup of 10 patients with MET-high HCC, the overall response rate was 30%, including 1 durable complete response (>600 days) and 2 partial responses [1 durable (>600 days)]. Conclusions Single agent capmatinib at the RDE is tolerable with a manageable safety profile. Antitumor activity was seen in a subset of patients with MET-dysregulated (MET-high) HCC. Trial registration ClinicalTrials.gov: NCT01737827. https://clinicaltrials.gov/ct2/show/NCT01737827.
Collapse
Affiliation(s)
- Shukui Qin
- PLA Cancer Center, Nanjing Bayi Hospital, Nanjing 210002, China
| | - Stephen Lam Chan
- Department of Clinical Oncology, Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | | | - Guohong Han
- Department of Liver Disease and Digestive Interventional Radiology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Su Pin Choo
- Division of Medical Oncology, National Cancer Center Singapore, Singapore
| | - Virote Sriuranpong
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Thomas Yau
- Department of Surgery, Queen Mary Hospital, University of Hong Kong, Hong Kong, China
| | - Yabing Guo
- Nanfang Hospital, Guangzhou Southern Medical University, Guangzhou, China
| | - Minshan Chen
- Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhenggang Ren
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianming Xu
- Department of Gastrointestinal Oncology, 307 Hospital of People's Liberation Army, Beijing, China
| | - Chia-Jui Yen
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan City
| | - Zhong-Zhe Lin
- Department of Oncology, National Taiwan University Hospital, Taipei City
| | - Luigi Manenti
- Translational Clinical Oncology, Novartis Institutes for BioMedical Research, East Hanover, NJ, USA
| | - Yi Gu
- PK Sciences, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Yongjian Sun
- Translational Clinical Oncology, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Ralph Tiedt
- Novartis Institutes for BioMedical Research, Basel, Basel-Stadt, Switzerland
| | - Lu Hao
- Translational Clinical Oncology, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Wenjie Song
- Translational Clinical Oncology, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Tawesak Tanwandee
- Division of Gastroenterology, Department of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
13
|
Yang S, Yang L, Li X, Li B, Li Y, Zhang X, Ma Y, Peng X, Jin H, Li H. New insights into autophagy in hepatocellular carcinoma: mechanisms and therapeutic strategies. Am J Cancer Res 2019; 9:1329-1353. [PMID: 31392073 PMCID: PMC6682711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 06/13/2019] [Indexed: 06/10/2023] Open
Abstract
Autophagy is a mechanism by which cellular substances are transported to lysosomes for degradation, allowing the basic transformation of cellular components, and providing energy and macromolecular precursors. In cancer, the contradictory role of autophagy in tumor suppression and promotion has been widely acknowledged. Activation and suppression of autophagy have been proposed as cancer therapies, resulting in targeted treatment of cancer by autophagy being considered ambiguous. The dynamic effect of autophagy can also be applied to hepatocellular carcinoma (HCC), a malignant tumor with high incidence and a low survival rate. In this review, we introduce characteristics of different types of autophagy and summarize which genes, non-coding RNAs, and related signaling pathways are involved in autophagy and the regulation of the formation and progress of HCC. More importantly, we discuss the role of autophagy in the treatment of HCC, such as in traditional chemotherapy, molecular targeted drugs, and natural products.
Collapse
Affiliation(s)
- Shuo Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Bowen Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Yan Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Xiaodong Zhang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Yingbo Ma
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Hongyuan Jin
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| |
Collapse
|
14
|
Huang X, Gan G, Wang X, Xu T, Xie W. The HGF-MET axis coordinates liver cancer metabolism and autophagy for chemotherapeutic resistance. Autophagy 2019; 15:1258-1279. [PMID: 30786811 PMCID: PMC6613896 DOI: 10.1080/15548627.2019.1580105] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Notwithstanding the numerous drugs available for liver cancer, emerging evidence suggests that chemotherapeutic resistance is a significant issue. HGF and its receptor MET play critical roles in liver carcinogenesis and metastasis, mainly dependent on the activity of receptor tyrosine kinase. However, for unknown reasons, all HGF-MET kinase activity-targeted drugs have failed or have been suspended in clinical trials thus far. Macroautophagy/autophagy is a protective ‘self-eating’ process for resisting metabolic stress by recycling obsolete components, whereas the impact of autophagy-mediated reprogrammed metabolism on therapeutic resistance is largely unclear, especially in liver cancer. In the present study, we first observed that HGF stimulus facilitated the Warburg effect and glutaminolysis to promote biogenesis in multiple liver cancer cells. We then identified the pyruvate dehydrogenase complex (PDHC) and GLS/GLS1 as crucial substrates of HGF-activated MET kinase; MET-mediated phosphorylation inhibits PDHC activity but activates GLS to promote cancer cell metabolism and biogenesis. We further found that the key residues of kinase activity in MET (Y1234/1235) also constitute a conserved LC3-interacting region motif (Y1234-Y1235-x-V1237). Therefore, on inhibiting HGF-mediated MET kinase activation, Y1234/1235-dephosphorylated MET induced autophagy to maintain biogenesis for cancer cell survival. Moreover, we verified that Y1234/1235-dephosphorylated MET correlated with autophagy in clinical liver cancer. Finally, a combination of MET inhibitor and autophagy suppressor significantly improved the therapeutic efficiency of liver cancer in vitro and in mice. Together, our findings reveal an HGF-MET axis-coordinated functional interaction between tyrosine kinase signaling and autophagy, and establish a MET-autophagy double-targeted strategy to overcome chemotherapeutic resistance in liver cancer. Abbreviations: ALDO: aldolase, fructose-bisphosphate; CQ: chloroquine; DLAT/PDCE2: dihydrolipoamide S-acetyltransferase; EMT: epithelial-mesenchymal transition; ENO: enolase; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GLS/GLS1: glutaminase; GLUL/GS: glutamine-ammonia ligase; GPI/PGI: glucose-6-phosphate isomerase; HCC: hepatocellular carcinoma; HGF: hepatocyte growth factor; HK: hexokinase; LDH: lactate dehydrogenase; LIHC: liver hepatocellular carcinoma; LIR: LC3-interacting region; PDH: pyruvate dehydrogenase; PDHA1: pyruvate dehydrogenase E1 alpha 1 subunit; PDHX: pyruvate dehydrogenase complex component X; PFK: phosphofructokinase; PK: pyruvate kinase; RTK: receptor tyrosine kinase; TCGA: The Cancer Genome Atlas
Collapse
Affiliation(s)
- Xing Huang
- a The Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province , First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou , Zhejiang , China.,b The Key Laboratory of Developmental Genes and Human Disease , Institute of Life Sciences, Southeast University , Nanjing , Jiangsu , China.,c The Therapeutic Antibody Research Center of SEU-Alphamab , Southeast University , Nanjing , China
| | - Guangming Gan
- b The Key Laboratory of Developmental Genes and Human Disease , Institute of Life Sciences, Southeast University , Nanjing , Jiangsu , China
| | - Xiaoxiao Wang
- c The Therapeutic Antibody Research Center of SEU-Alphamab , Southeast University , Nanjing , China
| | - Ting Xu
- b The Key Laboratory of Developmental Genes and Human Disease , Institute of Life Sciences, Southeast University , Nanjing , Jiangsu , China.,c The Therapeutic Antibody Research Center of SEU-Alphamab , Southeast University , Nanjing , China
| | - Wei Xie
- b The Key Laboratory of Developmental Genes and Human Disease , Institute of Life Sciences, Southeast University , Nanjing , Jiangsu , China.,c The Therapeutic Antibody Research Center of SEU-Alphamab , Southeast University , Nanjing , China
| |
Collapse
|
15
|
HGF/c-MET: A Promising Therapeutic Target in the Digestive System Cancers. Int J Mol Sci 2018; 19:ijms19113295. [PMID: 30360560 PMCID: PMC6274736 DOI: 10.3390/ijms19113295] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/10/2018] [Accepted: 10/15/2018] [Indexed: 02/07/2023] Open
Abstract
The HGF/c-MET pathway is active in the development of digestive system cancers, indicating that inhibition of HGF/c-MET signaling may have therapeutic potential. Various HGF/c-MET signaling inhibitors, mainly c-MET inhibitors, have been tested in clinical trials. The observed efficacy and adverse events of some c-MET inhibitors were not very suitable for treating digestive system cancers. The development of new HGF/c-MET inhibitors in preclinical studies may bring promising treatments and synergistic combination (traditional anticancer drugs and c-MET inhibitors) strategies provided anacceptable safety and tolerability. Insights into miRNA biology and miRNA therapeutics have made miRNAs attractive tools to inhibit HGF/c-MET signaling. Recent reports show that several microRNAs participate in inhibiting HGF/c-MET signaling networks through antagonizing c-MET or HGF in digestive system cancers, and the miRNAs-HGF/c-MET axis plays crucial and novel roles for cancer treatment. In the current review, we will discuss recent findings about inhibitors of HGF/c-MET signaling in treating digestive system cancers, and how miRNAs regulate digestive system cancers via mediating HGF/c-MET pathway.
Collapse
|
16
|
Kou J, Musich PR, Staal B, Kang L, Qin Y, Yao ZQ, Zhang B, Wu W, Tam A, Huang A, Hao HX, Vande Woude GF, Xie Q. Differential responses of MET activations to MET kinase inhibitor and neutralizing antibody. J Transl Med 2018; 16:253. [PMID: 30208970 PMCID: PMC6134500 DOI: 10.1186/s12967-018-1628-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Aberrant MET tyrosine kinase signaling is known to cause cancer initiation and progression. While MET inhibitors are in clinical trials against several cancer types, the clinical efficacies are controversial and the molecular mechanisms toward sensitivity remain elusive. METHODS With the goal to investigate the molecular basis of MET amplification (METamp) and hepatocyte growth factor (HGF) autocrine-driven tumors in response to MET tyrosine kinase inhibitors (TKI) and neutralizing antibodies, we compared cancer cells harboring METamp (MKN45 and MHCCH97H) or HGF-autocrine (JHH5 and U87) for their sensitivity and downstream biological responses to a MET-TKI (INC280) and an anti-MET monoclonal antibody (MetMab) in vitro, and for tumor inhibition in vivo. RESULTS We find that cancer cells driven by METamp are more sensitive to INC280 than are those driven by HGF-autocrine activation. In METamp cells, INC280 induced a DNA damage response with activation of repair through the p53BP1/ATM signaling pathway. Although MetMab failed to inhibit METamp cell proliferation and tumor growth, both INC280 and MetMab reduced HGF-autocrine tumor growth. In addition, we also show that HGF stimulation promoted human HUVEC cell tube formation via the Src pathway, which was inhibited by either INC280 or MetMab. These observations suggest that in HGF-autocrine tumors, the endothelial cells are the secondary targets MET inhibitors. CONCLUSIONS Our results demonstrate that METamp and HGF-autocrine activation favor different molecular mechanisms. While combining MET TKIs and ATM inhibitors may enhance the efficacy for treating tumors harboring METamp, a combined inhibition of MET and angiogenesis pathways may improve the therapeutic efficacy against HGF-autocrine tumors.
Collapse
Affiliation(s)
- Jianqun Kou
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.,Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Phillip R Musich
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Ben Staal
- Center of Cell and Cancer Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Liang Kang
- Center of Cell and Cancer Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Yuan Qin
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.,Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Zhi Q Yao
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.,Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Boheng Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Weizhong Wu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Angela Tam
- Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | - Alan Huang
- Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | - Huai-Xiang Hao
- Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | - George F Vande Woude
- Center of Cell and Cancer Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Qian Xie
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA. .,Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.
| |
Collapse
|
17
|
Huang F, Cai P, Wang Y, Zhou X, Chen H, Liao W, Mao Y, Zha X, Zhang H, Hu Z. Up-regulation of brain-expressed X-linked 2 is critical for hepatitis B virus X protein-induced hepatocellular carcinoma development. Oncotarget 2017; 8:65789-65799. [PMID: 29029472 PMCID: PMC5630372 DOI: 10.18632/oncotarget.19477] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/28/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. Chronic hepatitis B virus (HBV) infection is a major cause for HCC. Hepatitis B virus X (HBx), one of four proteins encoded by HBV genome, plays a vital role in the pathogenesis of HBV-induced HCC. However, the molecular mechanisms of HBx-triggered HCC remain largely undetermined. Here we revealed that the expression of Brain-expressed X-linked 2 (BEX2) and Osteopontin (OPN) were elevated in liver tissues of HBV transgenic mice and human HCC specimens. Moreover, a positive correlation between BEX2 and OPN was exhibited in samples from HCC patients with HBV infection. The protein levels of BEX2 and OPN were both higher in HBV-positive HCC specimens compared to that of HBV-negative HCC specimens. HBx potentiated OPN expression through up-regulation of BEX2. Importantly, the depletion of BEX2 suppressed tumorigenic potential of HCC cells with highly expressed HBx. We demonstrated the important role of BEX2 in HCC pathogenesis, and BEX2 may be a novel therapeutic target for HCC patients with HBV infection. The newly identified HBx/BEX2/OPN signaling cassette is implicated in the pathogenesis of HBV-induced HCC.
Collapse
Affiliation(s)
- Fuqiang Huang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei Cai
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanan Wang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xian Zhou
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongyu Chen
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenjun Liao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Hongbing Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhongdong Hu
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.,State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Imamura R, Matsumoto K. Hepatocyte growth factor in physiology and infectious diseases. Cytokine 2017; 98:97-106. [PMID: 28094206 DOI: 10.1016/j.cyto.2016.12.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/26/2016] [Accepted: 12/26/2016] [Indexed: 01/14/2023]
Abstract
Hepatocyte growth factor (HGF) is a pleiotropic cytokine composed of an α-chain and a β-chain, and these chains contain four kringle domains and a serine protease-like structure, respectively. The receptor for HGF was identified as the c-met proto-oncogene product of transmembrane receptor tyrosine kinase. HGF-induced signaling through the receptor Met provokes dynamic biological responses that support morphogenesis, regeneration, and the survival of various cells and tissues, which includes hepatocytes, renal tubular cells, and neurons. Characterization of tissue-specific Met knockout mice has further indicated that the HGF-Met system modulates immune cell functions and also plays an inhibitory role in the progression of chronic inflammation and fibrosis. However, the biological actions that are driven by the HGF-Met pathway all play a role in the acquisition of the malignant characteristics in tumor cells, such as invasion, metastasis, and drug resistance in the tumor microenvironment. Even though oncogenic Met signaling remains the major research focus, the HGF-Met axis has also been implicated in infectious diseases. Many pathogens try to utilize host HGF-Met system to establish comfortable environment for infection. Their strategies are not only simply change the expression level of HGF or Met, but also actively hijack HGF-Met system and deregulating Met signaling using their pathogenic factors. Consequently, the monitoring of HGF and Met expression, along with real-time detection of Met activation, can be a beneficial biomarker of these infectious diseases. Preclinical studies designed to address the therapeutic significance of HGF have been performed on injury/disease models, including acute tissue injury, chronic fibrosis, and cardiovascular and neurodegenerative diseases. Likewise, manipulating the HGF-Met system with complete control will lead to a tailor made treatment for those infectious diseases.
Collapse
Affiliation(s)
- Ryu Imamura
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan.
| |
Collapse
|
19
|
Lau EYT, Lo J, Cheng BYL, Ma MKF, Lee JMF, Ng JKY, Chai S, Lin CH, Tsang SY, Ma S, Ng IOL, Lee TKW. Cancer-Associated Fibroblasts Regulate Tumor-Initiating Cell Plasticity in Hepatocellular Carcinoma through c-Met/FRA1/HEY1 Signaling. Cell Rep 2016; 15:1175-89. [PMID: 27134167 DOI: 10.1016/j.celrep.2016.04.019] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 01/06/2016] [Accepted: 03/31/2016] [Indexed: 12/12/2022] Open
Abstract
Like normal stem cells, tumor-initiating cells (T-ICs) are regulated extrinsically within the tumor microenvironment. Because HCC develops primarily in the context of cirrhosis, in which there is an enrichment of activated fibroblasts, we hypothesized that cancer-associated fibroblasts (CAFs) would regulate liver T-ICs. We found that the presence of α-SMA(+) CAFs correlates with poor clinical outcome. CAF-derived HGF regulates liver T-ICs via activation of FRA1 in an Erk1,2-dependent manner. Further functional analysis identifies HEY1 as a direct downstream effector of FRA1. Using the STAM NASH-HCC mouse model, we find that HGF-induced FRA1 activation is associated with the fibrosis-dependent development of HCC. Thus, targeting the CAF-derived, HGF-mediated c-Met/FRA1/HEY1 cascade may be a therapeutic strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Eunice Yuen Ting Lau
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, PRC; Department of Pathology, The University of Hong Kong, Hong Kong, PRC
| | - Jessica Lo
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, PRC; Department of Pathology, The University of Hong Kong, Hong Kong, PRC
| | - Bowie Yik Ling Cheng
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, PRC; Department of Pathology, The University of Hong Kong, Hong Kong, PRC
| | - Mark Kin Fai Ma
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, PRC; Department of Pathology, The University of Hong Kong, Hong Kong, PRC
| | - Joyce Man Fong Lee
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, PRC; Department of Pathology, The University of Hong Kong, Hong Kong, PRC
| | - Johnson Kai Yu Ng
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, PRC; School of Biomedical Sciences, The University of Hong Kong, Hong Kong, PRC
| | - Stella Chai
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, PRC; School of Biomedical Sciences, The University of Hong Kong, Hong Kong, PRC
| | - Chi Ho Lin
- Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, PRC
| | - Suk Ying Tsang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Stephanie Ma
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, PRC; School of Biomedical Sciences, The University of Hong Kong, Hong Kong, PRC
| | - Irene Oi Lin Ng
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, PRC; Department of Pathology, The University of Hong Kong, Hong Kong, PRC.
| | - Terence Kin Wah Lee
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, PRC; Department of Pathology, The University of Hong Kong, Hong Kong, PRC.
| |
Collapse
|
20
|
Gao R, Cai C, Gan J, Yang X, Shuang Z, Liu M, Li S, Tang H. miR-1236 down-regulates alpha-fetoprotein, thus causing PTEN accumulation, which inhibits the PI3K/Akt pathway and malignant phenotype in hepatoma cells. Oncotarget 2016; 6:6014-28. [PMID: 25714026 PMCID: PMC4467418 DOI: 10.18632/oncotarget.3338] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 01/14/2015] [Indexed: 12/21/2022] Open
Abstract
Alpha fetoprotein (AFP) is a clinical biomarker of hepatocellular carcinoma (HCC). Here, we found that miR-1236 is down-regulated, whereas AFP is highly expressed in HCC tissues and cells. We demonstrated that miR-1236 directly targets the 3′UTR of AFP and down-regulates its expression. Also, miR-1236 inhibited and AFP stimulated proliferation, migration, invasion and vasculogenic mimicry (VM) of HCC. In agreement, AFP over-expression counteracted the inhibitory effect of miR-1236. We demonstrated that AFP promoted the ubiquitination of PTEN, thus decreasing PTEN levels, while miR-1236 inhibited the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Rui Gao
- Tianjin Life Science Research Center and Basic Medical School, Tianjin Medical University, Tianjin, China
| | - Chunli Cai
- Tianjin Life Science Research Center and Basic Medical School, Tianjin Medical University, Tianjin, China
| | - Jiancheng Gan
- Department of Surgery, Secondary Hospital of Tianjin Medical University, Tianjin, China
| | - Xi Yang
- Tianjin Life Science Research Center and Basic Medical School, Tianjin Medical University, Tianjin, China
| | - Zeyu Shuang
- State Key Laboratory of Oncology in Southern China, Department of Hepatobiliary Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Min Liu
- Tianjin Life Science Research Center and Basic Medical School, Tianjin Medical University, Tianjin, China
| | - Shengping Li
- State Key Laboratory of Oncology in Southern China, Department of Hepatobiliary Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Hua Tang
- Tianjin Life Science Research Center and Basic Medical School, Tianjin Medical University, Tianjin, China
| |
Collapse
|
21
|
Targeting the microRNA-21/AP1 axis by 5-fluorouracil and pirarubicin in human hepatocellular carcinoma. Oncotarget 2016; 6:2302-14. [PMID: 25544773 PMCID: PMC4385853 DOI: 10.18632/oncotarget.2955] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/09/2014] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs function as oncomiRs and tumor suppressors in diverse cancers. However, the utility of specific microRNAs in predicting the clinical benefit of chemotherapy has not been well-established. Here, we investigated the correlation between microRNA-21 expression and hepatic arterial infusion chemotherapy with 5-fluorouracil and pirarubicin (HAIC) for hepatocellular carcinoma (HCC). We found that HCC patients with low microRNA-21 levels in tumors tended to have a longer time to recurrence and disease-free survival. We demonstrated that microRNA-21 suppression in combination with 5-fluorouracil and pirarubicin treatment inhibited tumor growth in subcutaneous xenograft mice models. Mechanistically, the AP-1 and microRNA-21-mediated axis was verified to be a therapeutic target of cytotoxic drugs and deregulation of this axis led to an enhanced cell growth in HCC. Taken together, our findings demonstrate that microRNA-21 is a chemotherapy responsive microRNA and can serve as a prognostic biomarker for HCC patients undergoing HAIC. Targeting microRNA-21 enhances the effect of chemotherapeutic drugs, thereby suggesting that microRNA-21 suppression in combination with HAIC may be a novel approach for HCC treatment.
Collapse
|
22
|
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: 5.1] [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
|
23
|
Sarin H. Conserved molecular mechanisms underlying the effects of small molecule xenobiotic chemotherapeutics on cells. Mol Clin Oncol 2015; 4:326-368. [PMID: 26998284 DOI: 10.3892/mco.2015.714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 12/08/2015] [Indexed: 12/14/2022] Open
Abstract
For proper determination of the apoptotic potential of chemoxenobiotics in synergism, it is important to understand the modes, levels and character of interactions of chemoxenobiotics with cells in the context of predicted conserved biophysical properties. Chemoxenobiotic structures are studied with respect to atom distribution over molecular space, the predicted overall octanol-to-water partition coefficient (Log OWPC; unitless) and molecular size viz a viz van der Waals diameter (vdWD). The Log OWPC-to-vdWD (nm-1 ) parameter is determined, and where applicable, hydrophilic interacting moiety/core-to-vdWD (nm-1 ) and lipophilic incorporating hydrophobic moiety/core-to-vdWD (nm-1 ) parameters of their part-structures are determined. The cellular and sub-cellular level interactions of the spectrum of xenobiotic chemotherapies have been characterized, for which a classification system has been developed based on predicted conserved biophysical properties with respect to the mode of chemotherapeutic effect. The findings of this study are applicable towards improving the effectiveness of existing combination chemotherapy regimens and the predictive accuracy of personalized cancer treatment algorithms as well as towards the selection of appropriate novel xenobiotics with the potential to be potent chemotherapeutics for dendrimer nanoparticle-based effective transvascular delivery.
Collapse
Affiliation(s)
- Hemant Sarin
- Freelance Investigator in Translational Science and Medicine, Charleston, WV 25314, USA
| |
Collapse
|
24
|
Genomic profiling of a Hepatocyte growth factor-dependent signature for MET-targeted therapy in glioblastoma. J Transl Med 2015; 13:306. [PMID: 26381735 PMCID: PMC4574608 DOI: 10.1186/s12967-015-0667-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 09/09/2015] [Indexed: 11/10/2022] Open
Abstract
Background Constitutive MET signaling promotes invasiveness in most primary and recurrent GBM. However, deployment of available MET-targeting agents is confounded by lack of effective biomarkers for selecting suitable patients for treatment. Because endogenous HGF overexpression often causes autocrine MET activation, and also indicates sensitivity to MET inhibitors, we investigated whether it drives the expression of distinct genes which could serve as a signature indicating vulnerability to MET-targeted therapy in GBM. Methods Interrogation of genomic data from TCGA GBM (Student’s t test, GBM patients with high and low HGF expression, p ≤ 0.00001) referenced against patient-derived xenograft (PDX) models (Student’s t test, sensitive vs. insensitive models, p ≤ 0.005) was used to identify the HGF-dependent signature. Genomic analysis of GBM xenograft models using both human and mouse gene expression microarrays (Student’s t test, treated vs. vehicle tumors, p ≤ 0.01) were performed to elucidate the tumor and microenvironment cross talk. A PDX model with EGFRamp was tested for MET activation as a mechanism of erlotinib resistance. Results We identified a group of 20 genes highly associated with HGF overexpression in GBM and were up- or down-regulated only in tumors sensitive to MET inhibitor. The MET inhibitors regulate tumor (human) and host (mouse) cells within the tumor via distinct molecular processes, but overall impede tumor growth by inhibiting cell cycle progression. EGFRamp tumors undergo erlotinib resistance responded to a combination of MET and EGFR inhibitors. Conclusions Combining TCGA primary tumor datasets (human) and xenograft tumor model datasets (human tumor grown in mice) using therapeutic efficacy as an endpoint may serve as a useful approach to discover and develop molecular signatures as therapeutic biomarkers for targeted therapy. The HGF dependent signature may serve as a candidate predictive signature for patient enrollment in clinical trials using MET inhibitors. Human and mouse microarrays maybe used to dissect the tumor-host interactions. Targeting MET in EGFRamp GBM may delay the acquired resistance developed during treatment with erlotinib. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0667-x) contains supplementary material, which is available to authorized users.
Collapse
|
25
|
Jo JC, Choi EK, Shin JS, Moon JH, Hong SW, Lee HR, Kim SM, Jung SA, Lee DH, Jung SH, Lee SH, Kim JE, Kim KP, Hong YS, Suh YA, Jang SJ, Choi EK, Lee JS, Jin DH, Kim TW. Targeting FGFR Pathway in Human Hepatocellular Carcinoma: Expressing pFGFR and pMET for Antitumor Activity. Mol Cancer Ther 2015; 14:2613-22. [PMID: 26351320 DOI: 10.1158/1535-7163.mct-14-0780] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 08/24/2015] [Indexed: 12/18/2022]
Abstract
The MET receptor tyrosine kinase, the receptor for hepatocyte growth factor (HGF), has been implicated in cancer growth, invasion, migration, angiogenesis, and metastasis in a broad variety of human cancers, including human hepatocellular carcinoma (HCC). Recently, MET was suggested to be a potential target for the personalized treatment of HCC with an active HGF-MET signaling pathway. However, the mechanisms of resistance to MET inhibitors need to be elucidated to provide effective treatment. Here, we show that HCC cells exhibit different sensitivities to the MET inhibitor PHA665752, depending on the phosphorylation status of FGFR. Treatment of cells expressing both phospho-FGFR and phospho-MET with the inhibitor PHA665752 did not cause growth inhibition and cell death, whereas treatment with AZD4547, a pan-FGFR inhibitor, resulted in decreased colony formation and cleavage of caspase-3. Moreover, silencing of endogenous FGFR1 and FGFR2 by RNAi of HCC cells expressing phospho-FGFR, phospho-FGFR2, and phospho-MET overcame the resistance to PHA665752 treatment. Treatment of primary cancer cells from patients with HCC expressing both phospho-FGFR and phospho-MET with PHA665752 did not induce cell death, whereas AZD4547 treatment induced cell death through the cleavage of caspase-3. In addition, treatment of cells resistant to PHA665752 with AZD4547 abrogated the activation of downstream effectors of cell growth, proliferation, and survival. On the basis of these results, we conclude that the FGFR pathway is critical for HCC survival, and that targeting this pathway with AZD4547 may be beneficial for the treatment of patients with HCC-expressing phospho-FGFR and phospho-MET.
Collapse
Affiliation(s)
- Jae-Cheol Jo
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea. Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Eun Kyoung Choi
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jae-Sik Shin
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jai-Hee Moon
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Seung-Woo Hong
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Ha-Reum Lee
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea. Department of Life Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Seung-Mi Kim
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Soo-A Jung
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Dae-Hee Lee
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Seang Hwan Jung
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Sun-Hye Lee
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jeong Eun Kim
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Kyu-pyo Kim
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Yong Sang Hong
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Young-Ah Suh
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Se Jin Jang
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Eun Kyung Choi
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Radiation Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jung Shin Lee
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Dong-Hoon Jin
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea. Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea.
| | - Tae Won Kim
- Innovative Cancer Research, ASAN Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea. Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
26
|
Wei T, Zhang LN, Lv Y, Ma XY, Zhi L, Liu C, Ma F, Zhang XF. Overexpression of platelet-derived growth factor receptor alpha promotes tumor progression and indicates poor prognosis in hepatocellular carcinoma. Oncotarget 2015; 5:10307-17. [PMID: 25333264 PMCID: PMC4279374 DOI: 10.18632/oncotarget.2537] [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: 05/09/2014] [Accepted: 09/29/2014] [Indexed: 12/23/2022] Open
Abstract
Dysregulation of platelet-derived growth factor receptor alpha (PDGFRα) has been documented in various cancers. However, its role in hepatocellular carcinoma (HCC) remains unknown. We and others have examined that upregulation of PDGFRα might be involved in hepatocarcinogenesis. Here, we report that PDGFRα plays a critical role in HCC progression and prognosis. The expression of PDGFRα was markedly higher in human HCC compared to adjacent liver tissues. Although PDGFRA mRNA was decreased in HCC, PDGF-A mRNA was dramatically increased in HCC. Overexpression of PDGFRα was strongly correlated with microvessel density (MVD) of HCC (p<0.05), as well as macroscopic vascular invasion of the tumors (p<0.05). HCC patients with high PDGFRα expression displayed a shorter overall survival and a higher recurrence rate than those with low PDGFRα expression (p<0.05, respectively). Additionally, stable overexpression of PDGFRα in hepatoma cells promoted cell proliferation, migration, invasion and epithelial-mesenchymal transition in vitro. Similarly, an in vivo assay showed that PDGFRα overexpression in hepatoma cells exhibited remarkably tumorigenic potential in tumor size and weight in vivo, which displayed markedly elevated MVD than controls. Thus, our study provided the evidence that PDGFRα may serve as a candidate prognostic marker and a novel therapeutic target for HCC.
Collapse
Affiliation(s)
- Tao Wei
- Department of Hepatobiliary Surgery and Institute of Advanced Surgical Technology and Engineering, the 1st Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China. Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Li-Na Zhang
- Department of Pharmacy, the 2nd Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710004, China
| | - Yi Lv
- Department of Hepatobiliary Surgery and Institute of Advanced Surgical Technology and Engineering, the 1st Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Xiao-Ya Ma
- Department of Pharmacy, the 2nd Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710004, China
| | - Lei Zhi
- Department of General Surgery, General Hospital of Ningxia Medical College, Yinchuan, Ningxia Hui Autonomous Region, 750004, China
| | - Chang Liu
- Department of Hepatobiliary Surgery and Institute of Advanced Surgical Technology and Engineering, the 1st Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Feng Ma
- Department of Hepatobiliary Surgery and Institute of Advanced Surgical Technology and Engineering, the 1st Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Xu-Feng Zhang
- Department of Hepatobiliary Surgery and Institute of Advanced Surgical Technology and Engineering, the 1st Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| |
Collapse
|
27
|
Zhao B, Zhao W, Wang Y, Xu Y, Xu J, Tang K, Zhang S, Yin Z, Wu Q, Wang X. Connexin32 regulates hepatoma cell metastasis and proliferation via the p53 and Akt pathways. Oncotarget 2015; 6:10116-33. [PMID: 25426556 PMCID: PMC4496344 DOI: 10.18632/oncotarget.2687] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 11/02/2014] [Indexed: 12/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC) progresses rapidly and is frequently associated with vascular invasion, metastasis, recurrence, and poor prognosis. The expression of connexin32 (Cx32) is frequently downregulated in HCC tissues. In this study, the role of Cx32 in HCC metastasis and proliferation was investigated. The reduction of Cx32 in HCC tissues was significantly associated with increased vascular invasion, increased tumor size, and poor survival. In vitro assays revealed that Cx32 not only suppressed the invasion and migration of HCC cells, but also repressed HCC cell proliferation. Subsequent investigations revealed that Cx32 directly enhanced the acetylation and transcriptional activity of p53, thus upregulating the expression of the tumor metastasis suppressor protein KAI1/CD82, which is a p53 target gene. Additionally, Cx32 negatively regulated the phosphorylation of Akt and the expression of the cell cycle regulation protein cyclin D1, thereby inhibiting the proliferation of HCC cells. Our in vivo nude mice model further confirmed that Cx32 is able to suppress HCC tumor growth and metastasis in nude mice. Our results imply that Cx32 downregulation contributes to the proliferation and metastasis of HCC, and the restoration of Cx32 expression may be a promising strategy for HCC therapy.
Collapse
Affiliation(s)
- Bixing Zhao
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University. Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen University Affiliated Zhongshan Hospital
- Research Institute of Digestive Disease, Xiamen University, Xiamen, Fujian, China
| | - Wenxiu Zhao
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University. Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen University Affiliated Zhongshan Hospital
- Research Institute of Digestive Disease, Xiamen University, Xiamen, Fujian, China
| | - Yu Wang
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University. Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen University Affiliated Zhongshan Hospital
- Research Institute of Digestive Disease, Xiamen University, Xiamen, Fujian, China
| | - Yaping Xu
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University. Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen University Affiliated Zhongshan Hospital
- Research Institute of Digestive Disease, Xiamen University, Xiamen, Fujian, China
| | - Jianfeng Xu
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University. Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen University Affiliated Zhongshan Hospital
- Research Institute of Digestive Disease, Xiamen University, Xiamen, Fujian, China
| | - Kai Tang
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University. Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen University Affiliated Zhongshan Hospital
- Research Institute of Digestive Disease, Xiamen University, Xiamen, Fujian, China
| | - Sheng Zhang
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University. Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen University Affiliated Zhongshan Hospital
- Research Institute of Digestive Disease, Xiamen University, Xiamen, Fujian, China
| | - Zhenyu Yin
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University. Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen University Affiliated Zhongshan Hospital
- Research Institute of Digestive Disease, Xiamen University, Xiamen, Fujian, China
| | - Qiao Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian Province, China
| | - Xiaomin Wang
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University. Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen University Affiliated Zhongshan Hospital
- Research Institute of Digestive Disease, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
28
|
Huang MJ, Hu RH, Chou CH, Hsu CL, Liu YW, Huang J, Hung JS, Lai IR, Juan HF, Yu SL, Wu YM, Huang MC. Knockdown of GALNT1 suppresses malignant phenotype of hepatocellular carcinoma by suppressing EGFR signaling. Oncotarget 2015; 6:5650-65. [PMID: 25730904 PMCID: PMC4467392 DOI: 10.18632/oncotarget.3117] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 01/08/2015] [Indexed: 12/21/2022] Open
Abstract
O-glycosylation is a common protein modification. Aberrant O-glycosylation is associated with many cancers. GALNT1 is a GalNAc-transferase that initiates protein O-glycosylation. We found that GALNT1 is frequently up-regulated in hepatocellular carcinoma (HCC) and is associated with poor patient survival. Overexpression of GALNT1 increased and knockdown decreased HCC cell migration and invasion. Knockdown of GALNT1 inhibited EGF-induced migration and invasion. Knockdown of GALNT1 decreased EGFR activation and increased EGFR degradation, by decreasing EGFR O-glycosylation. This study demonstrates that down-regulation of GALNT1 is sufficient to suppress malignant phenotype of HCC cells by decreasing EGFR signaling. Thus, GALNT1 is a potential target in HCC.
Collapse
Affiliation(s)
- Miao-Juei Huang
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Rey-Heng Hu
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Hsing Chou
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Ya-Wen Liu
- Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - John Huang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Ji-Shiang Hung
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - I-Rue Lai
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsueh-Fen Juan
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Sung-Liang Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yao-Ming Wu
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Min-Chuan Huang
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
29
|
Zhang Y, Jain RK, Zhu M. Recent Progress and Advances in HGF/MET-Targeted Therapeutic Agents for Cancer Treatment. Biomedicines 2015; 3:149-181. [PMID: 28536405 PMCID: PMC5344234 DOI: 10.3390/biomedicines3010149] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 02/25/2015] [Accepted: 03/03/2015] [Indexed: 12/31/2022] Open
Abstract
The hepatocyte growth factor (HGF): MET axis is a ligand-mediated receptor tyrosine kinase pathway that is involved in multiple cellular functions, including proliferation, survival, motility, and morphogenesis. Aberrancy in the HGF/MET pathway has been reported in multiple tumor types and is associated with tumor stage and prognosis. Thus, targeting the HGF/MET pathway has become a potential therapeutic strategy in oncology development in the last two decades. A number of novel therapeutic agents-either as therapeutic proteins or small molecules that target the HGF/MET pathway-have been tested in patients with different tumor types in clinical studies. In this review, recent progress in HGF/MET pathway-targeted therapy for cancer treatment, the therapeutic potential of HGF/MET-targeted agents, and challenges in the development of such agents will be discussed.
Collapse
Affiliation(s)
- Yilong Zhang
- Department of Clinical Pharmacology, Modeling and Simulation, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | - Rajul K Jain
- Kite Pharma, Inc., 2225 Colorado Avenue, Santa Monica, CA 90404, USA.
| | - Min Zhu
- Department of Clinical Pharmacology, Modeling and Simulation, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| |
Collapse
|
30
|
Hu CT, Cheng CC, Wu JR, Pan SM, Wu WS. PKCε-mediated c-Met endosomal processing directs fluctuant c-Met-JNK-paxillin signaling for tumor progression of HepG2. Cell Signal 2015; 27:1544-55. [PMID: 25778903 DOI: 10.1016/j.cellsig.2015.02.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 02/28/2015] [Indexed: 12/16/2022]
Abstract
Hepatocyte growth factor (HGF) induced c-Met signaling play critical roles in the progression of hepatocellular carcinoma (HCC). However, c-Met targeting approaches suffered resistance and side effect, thus identification of more suitable downstream targets is needed. Recently, we demonstrated HGF-induced fluctuant ERK/paxillin signaling within 24h. We further examined the underlying mechanisms for fluctuant c-Met/JNK/paxillin signal cascade within 12h. HGF-induced phosphorylation of c-Met, JNK, and paxillin (Ser178) shared a common fluctuation pattern characterized by an initial peak at 0.5h, a middle drop at 4h, and a later peak at 10h. Dynasore, the inhibitor of dynamin, suppressed HGF-induced c-Met internalization and phosphorylation of JNK and paxillin (Ser178) at 0.5h, indicating that endosome formation is required for initial signal enhancement. Further, depletion of PKCε not only enhanced HGF-induced phosphorylation of JNK and paxillin (Ser178) but also prevented c-Met degradation at 0.5h, suggesting that PKCε mediated c-Met degradation for signal declination. On the other hand, HGF induced colocalizations of both phosphorylated JNK and paxillin with the endosomal recycling protein GGA3 at 10h and depletion of GGA3 abolished membrane recycling of c-Met and phosphorylation of JNK/paxillin at the same time point. Interestingly, HGF induced GGA3 phosphorylation in a PKCε-dependent manner during 0.5-4h, which is associated with c-Met degradation in the same period. Finally, HGF-induced cell migration, invasion and intrahepatic metastasis of HepG2 were prevented by the inhibitors of endocytosis. Our results suggest that critical endosomal components are promising therapeutic targets for preventing HGF-induced progression of HCC.
Collapse
Affiliation(s)
- Chi-Tan Hu
- Research Centre for Hepatology, Department of Internal Medicine, Buddhist Tzu Chi General Hospital and School of Medicine Tzu Chi University, Taiwan
| | - Chuan-Chu Cheng
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Jia-Ru Wu
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Siou-Mei Pan
- Research Centre for Hepatology, Department of Internal Medicine, Buddhist Tzu Chi General Hospital and School of Medicine Tzu Chi University, Taiwan
| | - Wen-Sheng Wu
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
31
|
Blum D, LaBarge S. Registered report: Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. eLife 2014; 3. [PMID: 25490933 PMCID: PMC4270138 DOI: 10.7554/elife.04034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 11/17/2014] [Indexed: 01/16/2023] Open
Abstract
The Reproducibility Project: Cancer
Biology seeks to address growing concerns about reproducibility in
scientific research by conducting replications of 50 papers in the field of cancer
biology published between 2010 and 2012. This Registered Report describes the
proposed replication plan of key experiments from ‘Tumour micro-environment
elicits innate resistance to RAF inhibitors through HGF secretion’ by
Straussman and colleagues, published in Nature in 2012 (Straussman et al., 2012). The key experiments being replicated
in this study are from Figure 2A, C, and D (and Supplemental Figure 11) and Figure 4C
(and Supplemental Figure 19) (Straussman et al.,
2012). Figure 2 demonstrates resistance to drug sensitivity conferred by
co-culture with some stromal cell lines and identifies the secreted factor
responsible as HGF. In Figure 4, Straussman and colleagues show that blocking the HGF
receptor MET abrogates HGF’s rescue of drug sensitivity. The Reproducibility
Project: Cancer Biology is a collaboration between the Center for Open Science
and Science Exchange, and the
results of the replications will be published by eLife. DOI:http://dx.doi.org/10.7554/eLife.04034.001
Collapse
Affiliation(s)
- David Blum
- Bioexpression and Fermentation Facility, University of Georgia, Athens, United States
| | | | | |
Collapse
|
32
|
CSBF/C10orf99, a novel potential cytokine, inhibits colon cancer cell growth through inducing G1 arrest. Sci Rep 2014; 4:6812. [PMID: 25351403 PMCID: PMC4212244 DOI: 10.1038/srep06812] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 10/09/2014] [Indexed: 12/16/2022] Open
Abstract
Cytokines are soluble proteins that exert their functions by binding specific receptors. Many cytokines play essential roles in carcinogenesis and have been developed for the treatment of cancer. In this study, we identified a novel potential cytokine using immunogenomics designated colon-derived SUSD2 binding factor (CSBF), also known as chromosome 10 open reading frame 99 (C10orf99). CSBF/C10orf99 is a classical secreted protein with predicted molecular mass of 6.5 kDa, and a functional ligand of Sushi Domain Containing 2 (SUSD2). CSBF/C10orf99 has the highest expression level in colon tissue. Both CSBF/C10orf99 and SUSD2 are down-regulated in colon cancer tissues and cell lines with different regulation mechanisms. CSBF/C10orf99 interacts with SUSD2 to inhibit colon cancer cell growth and induce G1 cell cycle arrest by down-regulating cyclin D and cyclin-dependent kinase 6 (CDK6). CSBF/C10orf99 displays a bell-shaped activity curve with the optimal effect at ~10 ng/ml. Its growth inhibitory effects can be blocked by sSUSD2-Fc soluble protein. Our results suggest that CSBF/C10orf99 is a novel potential cytokine with tumor suppressor functions.
Collapse
|
33
|
Bladt F, Friese-Hamim M, Ihling C, Wilm C, Blaukat A. The c-Met Inhibitor MSC2156119J Effectively Inhibits Tumor Growth in Liver Cancer Models. Cancers (Basel) 2014; 6:1736-52. [PMID: 25256830 PMCID: PMC4190565 DOI: 10.3390/cancers6031736] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/22/2014] [Accepted: 07/31/2014] [Indexed: 12/16/2022] Open
Abstract
The mesenchymal-epithelial transition factor (c-Met) is a receptor tyrosine kinase with hepatocyte growth factor (HGF) as its only high-affinity ligand. Aberrant activation of c-Met is associated with many human malignancies, including hepatocellular carcinoma (HCC). We investigated the in vivo antitumor and antimetastatic efficacy of the c-Met inhibitor MSC2156119J (EMD 1214063) in patient-derived tumor explants. BALB/c nude mice were inoculated with MHCC97H cells or with tumor fragments of 10 patient-derived primary liver cancer explants selected according to c-Met/HGF expression levels. MSC2156119J (10, 30, and 100 mg/kg) and sorafenib (50 mg/kg) were administered orally as single-agent treatment or in combination, with vehicle as control. Tumor response, metastases formation, and alpha fetoprotein (AFP) levels were measured. MSC2156119J inhibited tumor growth and induced complete regression in mice bearing subcutaneous and orthotopic MHCC97H tumors. AFP levels were undetectable after 5 weeks of MSC2156119J treatment, and the number of metastatic lung foci was reduced. Primary liver explant models with strong c-Met/HGF activation showed increased responsiveness to MSC2156119J, with MSC2156119J showing similar or superior activity to sorafenib. Tumors characterized by low c-Met expression were less sensitive to MSC2156119J. MSC2156119J was better tolerated than sorafenib, and combination therapy did not improve efficacy. These findings indicate that selective c-Met/HGF inhibition with MSC2156119J is associated with marked regression of c-Met high-expressing tumors, supporting its clinical development as an antitumor treatment for HCC patients with active c-Met signaling.
Collapse
Affiliation(s)
- Friedhelm Bladt
- EMD Serono, and Merck Serono Research and Development, Merck KGaA, Darmstadt 64293, Germany.
| | - Manja Friese-Hamim
- EMD Serono, and Merck Serono Research and Development, Merck KGaA, Darmstadt 64293, Germany.
| | - Christian Ihling
- EMD Serono, and Merck Serono Research and Development, Merck KGaA, Darmstadt 64293, Germany.
| | - Claudia Wilm
- EMD Serono, and Merck Serono Research and Development, Merck KGaA, Darmstadt 64293, Germany.
| | - Andree Blaukat
- EMD Serono, and Merck Serono Research and Development, Merck KGaA, Darmstadt 64293, Germany.
| |
Collapse
|