1
|
Zheng J, Wang S, Xia L, Sun Z, Chan KM, Bernards R, Qin W, Chen J, Xia Q, Jin H. Hepatocellular carcinoma: signaling pathways and therapeutic advances. Signal Transduct Target Ther 2025; 10:35. [PMID: 39915447 PMCID: PMC11802921 DOI: 10.1038/s41392-024-02075-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/18/2024] [Accepted: 11/14/2024] [Indexed: 02/09/2025] Open
Abstract
Liver cancer represents a major global health concern, with projections indicating that the number of new cases could surpass 1 million annually by 2025. Hepatocellular carcinoma (HCC) constitutes around 90% of liver cancer cases and is primarily linked to factors incluidng aflatoxin, hepatitis B (HBV) and C (HCV), and metabolic disorders. There are no obvious symptoms in the early stage of HCC, which often leads to delays in diagnosis. Therefore, HCC patients usually present with tumors in advanced and incurable stages. Several signaling pathways are dis-regulated in HCC and cause uncontrolled cell propagation, metastasis, and recurrence of HCC. Beyond the frequently altered and therapeutically targeted receptor tyrosine kinase (RTK) pathways in HCC, pathways involved in cell differentiation, telomere regulation, epigenetic modification and stress response also provide therapeutic potential. Investigating the key signaling pathways and their inhibitors is pivotal for achieving therapeutic advancements in the management of HCC. At present, the primary therapeutic approaches for advanced HCC are tyrosine kinase inhibitors (TKI), immune checkpoint inhibitors (ICI), and combination regimens. New trials are investigating combination therapies involving ICIs and TKIs or anti-VEGF (endothelial growth factor) therapies, as well as combinations of two immunotherapy regimens. The outcomes of these trials are expected to revolutionize HCC management across all stages. Here, we provide here a comprehensive review of cellular signaling pathways, their therapeutic potential, evidence derived from late-stage clinical trials in HCC and discuss the concepts underlying earlier clinical trials, biomarker identification, and the development of more effective therapeutics for HCC.
Collapse
Affiliation(s)
- Jiaojiao Zheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Siying Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Lei Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Zhen Sun
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Kui Ming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, PR China
| | - René Bernards
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wenxin Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jinhong Chen
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, PR China.
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Haojie Jin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
2
|
Cuesta ÁM, Palao N, Bragado P, Gutierrez-Uzquiza A, Herrera B, Sánchez A, Porras A. New and Old Key Players in Liver Cancer. Int J Mol Sci 2023; 24:17152. [PMID: 38138981 PMCID: PMC10742790 DOI: 10.3390/ijms242417152] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Liver cancer represents a major health problem worldwide with growing incidence and high mortality, hepatocellular carcinoma (HCC) being the most frequent. Hepatocytes are likely the cellular origin of most HCCs through the accumulation of genetic alterations, although hepatic progenitor cells (HPCs) might also be candidates in specific cases, as discussed here. HCC usually develops in a context of chronic inflammation, fibrosis, and cirrhosis, although the role of fibrosis is controversial. The interplay between hepatocytes, immune cells and hepatic stellate cells is a key issue. This review summarizes critical aspects of the liver tumor microenvironment paying special attention to platelets as new key players, which exert both pro- and anti-tumor effects, determined by specific contexts and a tight regulation of platelet signaling. Additionally, the relevance of specific signaling pathways, mainly HGF/MET, EGFR and TGF-β is discussed. HGF and TGF-β are produced by different liver cells and platelets and regulate not only tumor cell fate but also HPCs, inflammation and fibrosis, these being key players in these processes. The role of C3G/RAPGEF1, required for the proper function of HGF/MET signaling in HCC and HPCs, is highlighted, due to its ability to promote HCC growth and, regulate HPC fate and platelet-mediated actions on liver cancer.
Collapse
Affiliation(s)
- Ángel M. Cuesta
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (Á.M.C.); (N.P.); (P.B.); (A.G.-U.); (B.H.); (A.S.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Nerea Palao
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (Á.M.C.); (N.P.); (P.B.); (A.G.-U.); (B.H.); (A.S.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Paloma Bragado
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (Á.M.C.); (N.P.); (P.B.); (A.G.-U.); (B.H.); (A.S.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Alvaro Gutierrez-Uzquiza
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (Á.M.C.); (N.P.); (P.B.); (A.G.-U.); (B.H.); (A.S.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Blanca Herrera
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (Á.M.C.); (N.P.); (P.B.); (A.G.-U.); (B.H.); (A.S.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD-ISCIII), 28040 Madrid, Spain
| | - Aránzazu Sánchez
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (Á.M.C.); (N.P.); (P.B.); (A.G.-U.); (B.H.); (A.S.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD-ISCIII), 28040 Madrid, Spain
| | - Almudena Porras
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (Á.M.C.); (N.P.); (P.B.); (A.G.-U.); (B.H.); (A.S.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| |
Collapse
|
3
|
The Novel Action of miR-193b-3p/CDK1 Signaling in HCC Proliferation and Migration: A Study Based on Bioinformatic Analysis and Experimental Investigation. Int J Genomics 2022; 2022:8755263. [PMID: 36600989 PMCID: PMC9806689 DOI: 10.1155/2022/8755263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/30/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a common human malignancy with high mortality and dismal prognosis. A growing number of novel targets underlying HCC pathophysiology have been detected using microarray high throughput screening platforms. This study carried out bioinformatics analysis to explore underlying biomarkers in HCC and assessed the potential action of the miR-193b-3p/CDK1 signaling pathway in HCC progression. A total of 241 common differentially expressed genes (DEGs) were screened from GSE33294, GSE104310, and GSE144269. Functional analysis results implicated that DEGs are significantly associated with "cell cycle," "cell division," and "proliferation." The protein-protein interaction network analysis extracted ten hub genes from common DEGs. Ten hub genes were significantly overexpression in HCC tissues. Kaplan-Meier survival analysis revealed that 10 hub genes were linked with a poorer prognosis in HCC patients. Functional assays showed that CDK1 knockdown repressed HCC cell proliferation and migration. Luciferase reporter assay showed that miR-193b-3p could target CDK1 3' untranslated region, and miR-193b-3p negatively modulated CDK1. Enforced CDK1 expression attenuated miR-193b-3p-modulated suppressive actions on HCC cell proliferation and migration. To summarize, we performed a comprehensive bioinformatics analysis and identified 10 hub genes linked to the prognosis in HCC patients. Functional analysis revealed that CDK1, negatively regulated by miR-193b-3p, may act as an oncogene to promote HCC cell proliferation and migration and may predict poor prognosis of HCC patients. However, the role of CDK1/miR-193b-3p may still require further investigation.
Collapse
|
4
|
Liu H, Zhang L, Wang P. Complement factor H‑related 3 overexpression affects hepatocellular carcinoma proliferation and apoptosis. Mol Med Rep 2019; 20:2694-2702. [PMID: 31524260 PMCID: PMC6691229 DOI: 10.3892/mmr.2019.10514] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 12/07/2018] [Indexed: 12/14/2022] Open
Abstract
Complement factor H‑related 3 (CFHR3) belongs to the human factor H protein family and is associated with various human diseases, including nephropathy, age‑related macular degeneration and atypical hemolytic uremic syndrome. However, to the best of our knowledge, the role of CFHR3 in hepatocellular carcinoma (HCC) remains largely unknown. In the present study, reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blot analysis were performed to determine mRNA and protein expression levels of CFHR3 in HCC and normal adjacent tissue. In addition, CFHR3 was overexpressed in Huh‑7 cells and cell counting kit‑8 assay was used to determine cell viability. Cell proliferation and apoptosis were assessed using flow cytometry, RT‑qPCR and western blotting. The results demonstrated that mRNA (2‑ΔΔCq) and protein expression levels of CFHR3 were significantly lower in tumor tissue compared with in adjacent tissue. Additionally, CFHR3 overexpression decreased cell viability, inhibited cell proliferation and significantly increased apoptosis. It was also identified that CFHR3 could downregulate the expression of Ki67. The results suggested that CFHR3 induced apoptosis by downregulating the expression of survivin and B cell lymphoma 2, upregulating the expression of Bcl‑2‑associated X and promoting caspase‑3 activity. Western blotting revealed that CFHR3 significantly inhibited the protein expression levels of phosphorylated (p)‑phosphoinositide 3‑kinase (PI3K), p‑protein kinase B (Akt) and p‑mammalian target of rapamycin (mTOR). Overexpression of CFHR3 suppressed proliferation and promoted apoptosis of HCC cells by inhibiting the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Hong Liu
- Department of Hepatology, Yantai Infectious Disease Hospital, Yantai, Shandong 264001, P.R. China
| | - Ligang Zhang
- Department of Hepatology, Yantai Infectious Disease Hospital, Yantai, Shandong 264001, P.R. China
| | - Pengyan Wang
- Department of Research and Education, Yantai Infectious Disease Hospital, Yantai, Shandong 264001, P.R. China
| |
Collapse
|
5
|
Kim S, Choi S, Yoon JH, Kim Y, Lee S, Park T. Drug response prediction model using a hierarchical structural component modeling method. BMC Bioinformatics 2018; 19:288. [PMID: 30367591 PMCID: PMC6101092 DOI: 10.1186/s12859-018-2270-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Component-based structural equation modeling methods are now widely used in science, business, education, and other fields. This method uses unobservable variables, i.e., "latent" variables, and structural equation model relationships between observable variables. Here, we applied this structural equation modeling method to biologically structured data. To identify candidate drug-response biomarkers, we first used proteomic peptide-level data, as measured by multiple reaction monitoring mass spectrometry (MRM-MS), for liver cancer patients. MRM-MS is a highly sensitive and selective method for proteomic targeted quantitation of peptide abundances in complex biological samples. RESULTS We developed a component-based drug response prediction model, having the advantage that it first combines collapsed peptide-level data into protein-level information, facilitating subsequent biological interpretation. Our model also uses an alternating least squares algorithm, to efficiently estimate both coefficients of peptides and proteins. This approach also considers correlations between variables, without constraint, by a multiple testing problem. Using estimated peptide and protein coefficients, we selected significant protein biomarkers by permutation testing, resulting in our model for predicting liver cancer response to the tyrosine kinase inhibitor sorafenib. CONCLUSIONS Using data from a cohort of liver cancer patients, we then "fine-tuned" our model to successfully predict drug responses, as demonstrated by a high area under the curve (AUC) score. Such drug response prediction models may eventually find clinical translation in identifying individual patients likely to respond to specific therapies.
Collapse
Affiliation(s)
- Sungtae Kim
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826 South Korea
| | - Sungkyoung Choi
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826 South Korea
| | - Jung-Hwan Yoon
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, 03080 South Korea
| | - Youngsoo Kim
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, 03080 South Korea
| | - Seungyeoun Lee
- Department of Mathematics and Statistics, Sejong University, Seoul, 05006 South Korea
| | - Taesung Park
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826 South Korea
- Department of Statistics, Seoul National University, Seoul, 08826 South Korea
| |
Collapse
|
6
|
Zhu J, Yu H, Chen S, Yang P, Dong Z, Ling Y, Tang H, Bai S, Yang W, Tang L, Shen F, Wang H, Wen W. Prognostic significance of combining high mobility group Box-1 and OV-6 expression in hepatocellular carcinoma. SCIENCE CHINA-LIFE SCIENCES 2018; 61:912-923. [PMID: 29441453 DOI: 10.1007/s11427-017-9188-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/25/2017] [Indexed: 01/02/2023]
Abstract
The inflammatory environment and existence of cancer stem cells are critical for progression and intrahepatic recurrence of hepatocellular carcinoma (HCC) after curative resections. Here, we investigated the prognostic significance of combining high mobility group box 1 (HMGB1) expression and hepatic progenitor marker OV6 in hepatocellular carcinoma. Expression of HMGB1 and OV6 was evaluated using immunohistochemistry profiling in tissue microarrays containing samples from 208 HCC patients. Invasive clinical or pathological factors were found in patients with high expression of HMGB1 or OV6. Higher HMGB1 was associated with poorer clinical outcomes, and independently related to elevated 5-year recurrence incidence (85.5% vs. 62.4%, P<0.001). We also found that more OV6 positive staining was correlated with poor prognosis of HCC patients (P<0.001). Notably, expression of HMGB1 was positively correlated with OV6 in density (R2=0.032, P<0.001) and reversely related to HCC outcomes. Abnormal expression of HMGB1 in combination with positive staining of OV6 displayed poorer prognostic performance than single biomarker alone (area under curve (AUC) survival=0.696). Therefore, HMGB1 and OV6 positive staining are promising prognostic parameters for HCC, and we propose that HMGB1 and OV6 may cooperate with each other and predict poor prognosis of HCC.
Collapse
Affiliation(s)
- Jihui Zhu
- National Center for Liver Cancer, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China.,International Cooperation Laboratory on Signal Transduction of Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Han Yu
- National Center for Liver Cancer, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China.,International Cooperation Laboratory on Signal Transduction of Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Shuzhen Chen
- National Center for Liver Cancer, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China.,International Cooperation Laboratory on Signal Transduction of Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Pinghua Yang
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Zihui Dong
- National Center for Liver Cancer, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China.,International Cooperation Laboratory on Signal Transduction of Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Yan Ling
- National Center for Liver Cancer, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China.,International Cooperation Laboratory on Signal Transduction of Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Hao Tang
- Department of Respiratory Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Shilei Bai
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Wen Yang
- National Center for Liver Cancer, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China.,International Cooperation Laboratory on Signal Transduction of Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Liang Tang
- National Center for Liver Cancer, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China.,International Cooperation Laboratory on Signal Transduction of Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Feng Shen
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Hongyang Wang
- National Center for Liver Cancer, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China. .,International Cooperation Laboratory on Signal Transduction of Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200433, China. .,Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai, 200438, China.
| | - Wen Wen
- National Center for Liver Cancer, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China. .,International Cooperation Laboratory on Signal Transduction of Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200433, China. .,Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai, 200438, China.
| |
Collapse
|
7
|
Jackson R, Psarelli EE, Berhane S, Khan H, Johnson P. Impact of Viral Status on Survival in Patients Receiving Sorafenib for Advanced Hepatocellular Cancer: A Meta-Analysis of Randomized Phase III Trials. J Clin Oncol 2017; 35:622-628. [PMID: 28045619 DOI: 10.1200/jco.2016.69.5197] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Purpose Following the Sorafenib Hepatocellular Carcinoma Assessment Randomized Protocol (SHARP) trial, sorafenib has become the standard of care for patients with advanced unresectable hepatocellular carcinoma, but the relation between survival advantage and disease etiology remains unclear. To address this, we undertook an individual patient data meta-analysis of three large prospective randomized trials in which sorafenib was the control arm. Methods Of a total of 3,256 patients, 1,643 (50%) who received sorafenib were available. The primary end point was overall survival (OS). A Bayesian hierarchical approach for individual patient data meta-analyses was applied using a piecewise exponential model. Results are presented in terms of hazard ratios comparing sorafenib with alternative therapies according to hepatitis C virus (HCV) or hepatitis B virus (HBV) status. Results Hazard ratios show improved OS for sorafenib in patients who are both HBV negative and HCV positive (log [hazard ratio], -0.27; 95% CI, -0.46 to -0.06). Median unadjusted survival is 12.6 (11.15 to 13.8) months for sorafenib and 10.2 (8.88 to 12.2) months for "other" treatments in this subgroup. There was no evidence of improvement in OS for any other patient subgroups defined by HBV and HCV. Results were consistent across all trials with heterogeneity assessed using Cochran's Q statistic. Conclusion There is consistent evidence that the effect of sorafenib on OS is dependent on patients' hepatitis status. There is an improved OS for patients negative for HBV and positive for HCV when treated with sorafenib. There was no evidence of any improvement in OS attributable to sorafenib for patients positive for HBV and negative for HCV.
Collapse
Affiliation(s)
- Richard Jackson
- Richard Jackson and Eftychia-Eirini Psarelli, Liverpool Cancer Trials Unit; Sarah Berhane and Philip Johnson, University of Liverpool, Liverpool; Harun Khan, Imperial College London, London; and Philip Johnson, The Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, United Kingdom
| | - Eftychia-Eirini Psarelli
- Richard Jackson and Eftychia-Eirini Psarelli, Liverpool Cancer Trials Unit; Sarah Berhane and Philip Johnson, University of Liverpool, Liverpool; Harun Khan, Imperial College London, London; and Philip Johnson, The Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, United Kingdom
| | - Sarah Berhane
- Richard Jackson and Eftychia-Eirini Psarelli, Liverpool Cancer Trials Unit; Sarah Berhane and Philip Johnson, University of Liverpool, Liverpool; Harun Khan, Imperial College London, London; and Philip Johnson, The Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, United Kingdom
| | - Harun Khan
- Richard Jackson and Eftychia-Eirini Psarelli, Liverpool Cancer Trials Unit; Sarah Berhane and Philip Johnson, University of Liverpool, Liverpool; Harun Khan, Imperial College London, London; and Philip Johnson, The Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, United Kingdom
| | - Philip Johnson
- Richard Jackson and Eftychia-Eirini Psarelli, Liverpool Cancer Trials Unit; Sarah Berhane and Philip Johnson, University of Liverpool, Liverpool; Harun Khan, Imperial College London, London; and Philip Johnson, The Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, United Kingdom
| |
Collapse
|
8
|
Zhang Y, Ran Y, Xiong Y, Zhong ZB, Wang ZH, Fan XL, Ye QF. Effects of TMEM9 gene on cell progression in hepatocellular carcinoma by RNA interference. Oncol Rep 2016; 36:299-305. [PMID: 27220462 DOI: 10.3892/or.2016.4821] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 07/16/2015] [Indexed: 01/13/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor that has become a global health issue. The aim of the present study was to examine the role of transmembrane protein 9 (TMEM9) in cell progression, such as cell growth, cell cycle, cell metastasis of hepatoma cells, and to discuss the TMEM9 gene‑encoding protein as a potential therapy target of hepatoma. RT-qPCR was performed to examine TMEM9 expression in tumor tissues and adjacent tissues of patients with liver cancer. siRNAs were used to interfere TMEM9 in HepG2 and 7721 cells. A CCK-8 assay was performed to evaluate cell growth at 24, 48 and 72 h. Cell cycle and apoptosis were analyzed using flow cytometry. Transwell assays were used to determine cell invasion, migration and adhesion. The results showed that TMEM9 was expressed abnormally in liver cancers. TMEM9 expression increased significantly in the 34 examined patients. TMEM9 knockdown inhibited proliferation in the HepG2 and 7721 cells. The flow cytometric analysis revealed that TMEM9 knockdown by RNA interference resulted in G1 arrest and induced apoptosis. Cell invasion, migration and adhesion ability were also decreased. Western blotting indicated that expression of the cell cycle‑related proteins CDK1, EIF3H, RPL10L, S100A10, CCNB1 and CCNB2 was significantly decreased. In conclusion, TMEM9 plays an important role in the cell growth of hepatoma cells.
Collapse
Affiliation(s)
- Yi Zhang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan University, Wuchang, Hubei 430071, P.R. China
| | - Yan Ran
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Department of Anesthesiology, Wuhan University, Wuchang, Hubei 430071, P.R. China
| | - Yan Xiong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan University, Wuchang, Hubei 430071, P.R. China
| | - Zi-Biao Zhong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan University, Wuchang, Hubei 430071, P.R. China
| | - Zheng-Hua Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan University, Wuchang, Hubei 430071, P.R. China
| | - Xiao-Li Fan
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan University, Wuchang, Hubei 430071, P.R. China
| | - Qi-Fa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan University, Wuchang, Hubei 430071, P.R. China
| |
Collapse
|
9
|
Lin Z, Xu SH, Wang HQ, Cai YJ, Ying L, Song M, Wang YQ, Du SJ, Shi KQ, Zhou MT. Prognostic value of DNA repair based stratification of hepatocellular carcinoma. Sci Rep 2016; 6:25999. [PMID: 27174663 PMCID: PMC4867671 DOI: 10.1038/srep25999] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/18/2016] [Indexed: 12/14/2022] Open
Abstract
Aberrant activation of DNA repair is frequently associated with tumor progression and response to therapy in hepatocellular carcinoma (HCC). Bioinformatics analyses of HCC data in the Cancer Genome Atlas (TCGA) were performed to define DNA repair based molecular classification that could predict the prognosis of patients with HCC. Furthermore, we tested its predictive performance in 120 independent cases. Four molecular subgroups were identified on the basis of coordinate DNA repair cluster (CDRC) comprising 15 genes in TCGA dataset. Increasing expression of CDRC genes were significantly associated with TP53 mutation. High CDRC was significantly correlated with advanced tumor grades, advanced pathological stage and increased vascular invasion rate. Multivariate Cox regression analysis indicated that the molecular subgrouping was an independent prognostic parameter for both overall survival (p = 0.004, hazard ratio (HR): 2.989) and tumor-free survival (p = 0.049, HR: 3.366) in TCGA dataset. Similar results were also obtained by analyzing the independent cohort. These data suggest that distinct dysregulation of DNA repair constituents based molecular classes in HCC would be useful for predicting prognosis and designing clinical trials for targeted therapy.
Collapse
Affiliation(s)
- Zhuo Lin
- Department of Infectious and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.,Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
| | - Shi-Hao Xu
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Hai-Qing Wang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yi-Jing Cai
- Department of Infectious and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.,Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
| | - Li Ying
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Mei Song
- Department of Infectious and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.,Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
| | - Yu-Qun Wang
- Department of Infectious and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.,Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
| | - Shan-Jie Du
- Department of Infectious and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.,Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
| | - Ke-Qing Shi
- Department of Infectious and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.,Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
| | - Meng-Tao Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Lin LL, Wang W, Hu Z, Wang LW, Chang J, Qian H. Erratum to: Negative feedback of miR-29 family TET1 involves in hepatocellular cancer. Med Oncol 2016; 32:39. [PMID: 25616722 DOI: 10.1007/s12032-014-0437-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Primary hepatocellular carcinoma (HCC) is the most common form of liver cancer and is one of the most common malignancies worldwide. Tumor suppressor gene silencing through DNA methylation contributes to cancer formation. The ten-eleven translocations (TET) family of α-ketogluta-rate-dependent dioxygenases catalyzes the sequential oxidation of 5-methylcytosine to 5-hydroxymethyl-cytosine, 5-formylcytosine and 5-carboxylcytosine, leading to eventual DNA demethylation. MicroRNAs are an abundant class of 17-25 nucleotides small noncoding RNAs, identified as important regulators of many diverse biological processes. In this study, we showed that TET1 expression was obviously reduced in the majority of examined HCC tissues. And we further investigated the expression and functional involvement of TET1 in proliferation, migration and invasion and determined that TET1 may function as a tumor suppressor. miR-29b was proved to inhibit metastasis through the targeting of TET1, indicating that downregulation of miR-29 may involve in HCC carcinogenesis and progression through potentiation of TET1 expression. Thus, we elucidated the roles of feedback of miR-29-TET1 downregulation in HCC development and suggested a potential target in identification of the prognosis and application of cancer therapy for HCC patients.
Collapse
Affiliation(s)
- Li Li Lin
- Department of Pharmacology, Wuxi Higher Health Vocational Technology School, No. 305, Xinguang Road, Wuxi, 214028, China
| | | | | | | | | | | |
Collapse
|
11
|
Li J, Dai W, Xia Y, Chen K, Li S, Liu T, Zhang R, Wang J, Lu W, Zhou Y, Yin Q, Abudumijiti H, Chen R, Zheng Y, Wang F, Lu J, Zhou Y, Guo C. Astaxanthin Inhibits Proliferation and Induces Apoptosis of Human Hepatocellular Carcinoma Cells via Inhibition of Nf-Κb P65 and Wnt/Β-Catenin in Vitro. Mar Drugs 2015; 13:6064-6081. [PMID: 26404320 PMCID: PMC4626679 DOI: 10.3390/md13106064] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/16/2015] [Accepted: 09/16/2015] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor that can cause systemic invasion; however, the exact etiology and molecular mechanism are unknown. Astaxanthin (ASX), a powerful antioxidant, has efficient anti-oxidant, anti-inflammatory, and other activities, and has great research prospects in cancer therapy. We selected the human hepatoma cell lines, LM3 and SMMC-7721, to study the anti-tumor effect and related mechanisms of ASX. The cell lines were treated with different concentrations of ASX, and its solvent DMSO as a control, for different time periods and the results were determined using CCK8, qRT-PCR, WB, apoptotic staining, and flow cytometry. ASX induced significant apoptosis of HCC cells, and its effect may have been caused by NF-κB p65 and Wnt/β-catenin down-regulation via negative activation of PI3K/Akt and ERK. Antitumor research on ASX has provided us with a potential therapy for patients with hepatomas.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Weiqi Dai
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Yujing Xia
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Kan Chen
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Sainan Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Tong Liu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Rong Zhang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
- The First Clinical Medical College of Nanjing Medical University, Nanjing 210029, China.
| | - Jianrong Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
- The First Clinical Medical College of Nanjing Medical University, Nanjing 210029, China.
| | - Wenxia Lu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
- The First Clinical Medical College of Nanjing Medical University, Nanjing 210029, China.
| | - Yuqing Zhou
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
- The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Qin Yin
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
- The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Huerxidan Abudumijiti
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Rongxia Chen
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Yuanyuan Zheng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Fan Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Jie Lu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Yingqun Zhou
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| |
Collapse
|
12
|
Govaere O, Roskams T. Pathogenesis and prognosis of hepatocellular carcinoma at the cellular and molecular levels. Clin Liver Dis 2015; 19:261-76. [PMID: 25921662 DOI: 10.1016/j.cld.2015.01.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Different approaches predict the outcome for patients with hepatocellular carcinoma (HCC). The expression of biliary-hepatic progenitor cell markers generally correlates with poor prognosis. This article focuses on the pathogenesis of HCC, how differentiation or dedifferentiation leads to a phenotype switch, and heterogeneity in the same tumor. A tumor cell decides its fate based on a complex interplay of signaling pathways. Interaction with the microenvironment decides whether it will invade, proliferate, or enter survival mode. Several signaling pathways contribute to stemness features, reflecting a small chemoresistant subpopulation of the tumor that expresses biliary-hepatic progenitor cell markers.
Collapse
Affiliation(s)
- Olivier Govaere
- Translational Cell and Tissue Research, Department of Imaging and Pathology, KULeuven and University Hospitals Leuven, Minderbroedersstraat 12, Leuven B3000, Belgium.
| | - Tania Roskams
- Translational Cell and Tissue Research, Department of Imaging and Pathology, KULeuven and University Hospitals Leuven, Minderbroedersstraat 12, Leuven B3000, Belgium.
| |
Collapse
|
13
|
Negative feedback of miR-29 family TET1 involves in hepatocellular cancer. Med Oncol 2014; 31:291. [PMID: 25367851 DOI: 10.1007/s12032-014-0291-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 10/13/2014] [Indexed: 01/04/2023]
Abstract
Primary hepatocellular carcinoma (HCC) is the most common form of liver cancer and is one of the most common malignancies worldwide. Tumor suppressor gene silencing through DNA methylation contributes to cancer formation. The ten-eleven translocations (TET) family of α-ketogluta-rate-dependent dioxygenases catalyzes the sequential oxidation of 5-methylcytosine to 5-hydroxymethyl-cytosine, 5-formylcytosine and 5-carboxylcytosine, leading to eventual DNA demethylation. MicroRNAs are an abundant class of 17-25 nucleotides small noncoding RNAs, identified as important regulators of many diverse biological processes. In this study, we showed that TET1 expression was obviously reduced in the majority of examined HCC tissues. And we further investigated the expression and functional involvement of TET1 in proliferation, migration and invasion, and determined that TET1 may function as a tumor suppressor. MiR-29b was proved to inhibit metastasis through the targeting of TET1, indicating that downregulation of miR-29 may involve in HCC carcinogenesis and progression through potentiation of TET1 expression. Thus, we elucidated the roles of feedback of miR-29-TET1 downregulation in HCC development and suggested a potential target in identification of the prognosis and application of cancer therapy for HCC patients.
Collapse
|
14
|
Negative feedback of miR-29 family TET1 involves in hepatocellular cancer. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2014. [PMID: 25367851 DOI: 10.1007/s12032- 014-0291-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Primary hepatocellular carcinoma (HCC) is the most common form of liver cancer and is one of the most common malignancies worldwide. Tumor suppressor gene silencing through DNA methylation contributes to cancer formation. The ten-eleven translocations (TET) family of α-ketogluta-rate-dependent dioxygenases catalyzes the sequential oxidation of 5-methylcytosine to 5-hydroxymethyl-cytosine, 5-formylcytosine and 5-carboxylcytosine, leading to eventual DNA demethylation. MicroRNAs are an abundant class of 17-25 nucleotides small noncoding RNAs, identified as important regulators of many diverse biological processes. In this study, we showed that TET1 expression was obviously reduced in the majority of examined HCC tissues. And we further investigated the expression and functional involvement of TET1 in proliferation, migration and invasion, and determined that TET1 may function as a tumor suppressor. MiR-29b was proved to inhibit metastasis through the targeting of TET1, indicating that downregulation of miR-29 may involve in HCC carcinogenesis and progression through potentiation of TET1 expression. Thus, we elucidated the roles of feedback of miR-29-TET1 downregulation in HCC development and suggested a potential target in identification of the prognosis and application of cancer therapy for HCC patients.
Collapse
|
15
|
Bertino G, Demma S, Ardiri A, Proiti M, Malaguarnera G, Bertino N, Malaguarnera M, Malaguarnera M. Hepatocellular carcinoma: novel molecular targets in carcinogenesis for future therapies. BIOMED RESEARCH INTERNATIONAL 2014; 2014:203693. [PMID: 25089265 PMCID: PMC4096380 DOI: 10.1155/2014/203693] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/29/2014] [Accepted: 05/30/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hepatocellular carcinoma is one of the most common and lethal malignant tumors worldwide. Over the past 15 years, the incidence of HCC has more than doubled. Due to late diagnosis and/or advanced underlying liver cirrhosis, only limited treatment options with marginal clinical benefit are available in up to 70% of patients. During the last decades, no effective conventional cytotoxic systemic therapy was available contributing to the dismal prognosis in patients with HCC. A better knowledge of molecular hepatocarcinogenesis provides today the opportunity for targeted therapy. MATERIALS AND METHODS A search of the literature was made using cancer literature, the PubMed, Scopus, and Web of Science (WOS) database for the following keywords: "hepatocellular carcinoma," "molecular hepatocarcinogenesis," "targeted therapy," and "immunotherapy." DISCUSSION AND CONCLUSION Treatment decisions are complex and dependent upon tumor staging, presence of portal hypertension, and the underlying degree of liver dysfunction. The knowledge of molecular hepatocarcinogenesis broadened the horizon for patients with advanced HCC. During the last years, several molecular targeted agents have been evaluated in clinical trials in advanced HCC. In the future, new therapeutic options will be represented by a blend of immunotherapy-like vaccines and T-cell modulators, supplemented by molecularly targeted inhibitors of tumor signaling pathways.
Collapse
Affiliation(s)
- Gaetano Bertino
- Hepatology Unit, Department of Medical and Pediatric Sciences, University of Catania, Policlinic, Via Santa Sofia No. 78, 95123 Catania, Italy
| | - Shirin Demma
- Hepatology Unit, Department of Medical and Pediatric Sciences, University of Catania, Policlinic, Via Santa Sofia No. 78, 95123 Catania, Italy
| | - Annalisa Ardiri
- Hepatology Unit, Department of Medical and Pediatric Sciences, University of Catania, Policlinic, Via Santa Sofia No. 78, 95123 Catania, Italy
| | - Maria Proiti
- Hepatology Unit, Department of Medical and Pediatric Sciences, University of Catania, Policlinic, Via Santa Sofia No. 78, 95123 Catania, Italy
| | - Giulia Malaguarnera
- Research Centre “The Great Senescence”, University of Catania, Via Messina No. 829, 95126 Catania, Italy
| | - Nicoletta Bertino
- Faculty of Pharmacy, University of Catania, Viale Andrea Doria No. 6, 95123 Catania, Italy
| | - Michele Malaguarnera
- Research Centre “The Great Senescence”, University of Catania, Via Messina No. 829, 95126 Catania, Italy
| | - Mariano Malaguarnera
- Research Centre “The Great Senescence”, University of Catania, Via Messina No. 829, 95126 Catania, Italy
| |
Collapse
|
16
|
Bertino G, Di Carlo I, Ardiri A, Calvagno GS, Demma S, Malaguarnera G, Bertino N, Malaguarnera M, Toro A, Malaguarnera M. Systemic therapies in hepatocellular carcinoma: present and future. Future Oncol 2014; 9:1533-48. [PMID: 24106903 DOI: 10.2217/fon.13.171] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is now the third leading cause of cancer deathsworldwide and is generally presented at an advanced stage, limiting patients' quality of life. The conventional cytotoxic systemic therapy has proved to be ineffective in HCC, since its induction several decades ago. Today it is possible to use our knowledge of molecular hepatocarcinogenesis to provide a targeted therapy. Sorafenib has demonstrated large improvements in overall survival in HCC. This review describes the molecular mechanisms and potential therapeutic targets, focusing on sorafenib, sunitinib, tivantinib, antiangiogenic agents, and current and future immunotherapies. Thus, it will be necessary in the future to classify HCCs into subgroups according to their genomic and proteomic profiling. The identification of key molecules/receptors/signaling pathways and the assessment of their relevance as potential targets will be the main future challenge potentially influencing response to therapy. Defining molecular targeted agents that are effective for a specific HCC subgroup will hopefully lead to personalized therapy.
Collapse
Affiliation(s)
- Gaetano Bertino
- Hepatology Unit - Department of Medical & Pediatric Science, University of Catania Policlinic, Via S Sofia 78, 95123, Catania, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
|
18
|
Rimassa L, Personeni N, Simonelli M, Santoro A. Tivantinib: a new promising mesenchymal-epithelial transition factor inhibitor in the treatment of hepatocellular carcinoma. Future Oncol 2013; 9:153-65. [PMID: 23414466 DOI: 10.2217/fon.12.188] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Tivantinib (ARQ 197) is an orally administered, selective small molecule that inhibits mesenchymal-epithelial transition factor (MET) via a novel, ATP-independent binding mechanism. Preclinical studies demonstrated that tivantinib has a broad-spectrum anti-tumor activity, especially in cells expressing high levels of MET. A randomized Phase II study in second-line hepatocellular carcinoma showed statistically significant improvement in time to progression with tivantinib compared to a placebo. Noteworthy, a significant pronounced benefit in time to progression and overall survival was observed in MET-high patients. In addition, MET expression was defined as a negative prognostic factor. The most frequent adverse events were hematologic events. A Phase III study in the MET-high hepatocellular carcinoma is actively recruiting patients. Phase II and III studies in non-small-cell lung cancer and colorectal cancer are ongoing.
Collapse
Affiliation(s)
- Lorenza Rimassa
- Medical Oncology & Hematology Unit, Humanitas Cancer Center, Istituto Clinico Humanitas, IRCCS. Via Manzoni 56, 20089 Rozzano, Milano, Italy.
| | | | | | | |
Collapse
|
19
|
Genco C, Cabibbo G, Maida M, Brancatelli G, Galia M, Alessi N, Butera G, Genova C, Romano P, Raineri M, Giarratano A, Midiri M, Cammà C. Treatment of hepatocellular carcinoma: present and future. Expert Rev Anticancer Ther 2013; 13:469-479. [PMID: 23560841 DOI: 10.1586/era.13.21] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma is a major health problem. It is the sixth most common cancer worldwide and the third most common cause of cancer-related death. Despite the availability of several treatment opportunities, diagnosis is still made in an advanced phase, limiting application of most therapeutic choices that currently are based on the Barcelona Clinic Cancer Liver Classification and include surgical resection, orthotopic liver transplantation and ablative methods for very early and early disease, arterial chemoembolization for intermediate stages and systemic therapy with sorafenib for advanced hepatocellular carcinoma. Thanks to novel advancements in knowledge of molecular pathogenesis of this tumor, many new systemic agents and locoregional treatments are in different stages of clinical development and they represent an important promise of further improvements in patients' survival.
Collapse
Affiliation(s)
- Chiara Genco
- Section of Gastroenterology, DIBIMIS, University of Palermo, Palermo, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Xu C, Liu C, Huang W, Tu S, Wan F. Effect of Mst1 overexpression on the growth of human hepatocellular carcinoma HepG2 cells and the sensitivity to cisplatin in vitro. Acta Biochim Biophys Sin (Shanghai) 2013; 45:268-79. [PMID: 23419720 DOI: 10.1093/abbs/gmt006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mammalian STE20-like kinase 1 (Mst1) is the mammalian homologue of Drosophila Hippo, a major inhibitor of cell proliferation in Drosophila. It ubiquitously encodes serine threonine kinase that belongs to the family of protein kinases related to yeast STE20, and is involved in cell proliferation, apoptosis, oncogenesis, and organ growth. Recent studies have shown that Mst1 has tumor-suppressor function, and the deletion or mutation of Mst1 is reported to be associated with tumorigenesis. To investigate the effect of overexpression of Mst1 on the growth of human liver cancer cell line HepG2 cells and the sensitivity to cisplatin in vitro, here we constructed recombinant eukaryotic expression vector pEGFP-N1-Mst1 containing Mst1 gene, and transiently transfected into HepG2 cells. The effects of Mst1 overexpression on the cell proliferation and apoptosis, the phosphorylation status of Yes-associated protein, and the mRNA transcript levels of connective tissue growth factor (CTGF), amphiregulin (AREG), and birc5 (Survivin) were determined. Results showed that overexpression of Mst1 inhibited cell proliferation, induced apoptosis of HepG2 cells, promoted YAP (Ser127) phosphorylation, and downregulated the mRNA expression of CTGF, AREG, and Survivin. We also investigated the relationship between the expression and cleavage of Mst1 and cisplatin-induced cell death. We found that Mst1 overexpression could induce cisplatin chemosensitivity, and cisplatin could promote the cleavage of Mst1 without affecting the expression of Mst1. Overall, our results indicated that Mst1 might be a promising anticancer target.
Collapse
Affiliation(s)
- Chuanming Xu
- Department of Biochemistry and Molecular Biology, Basic Medical College of Nanchang University, Nanchang 330006, China
| | | | | | | | | |
Collapse
|
21
|
Kudo M. Signaling pathway/molecular targets and new targeted agents under development in hepatocellular carcinoma. World J Gastroenterol 2012; 18:6005-17. [PMID: 23155330 PMCID: PMC3496878 DOI: 10.3748/wjg.v18.i42.6005] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 06/15/2011] [Accepted: 06/21/2011] [Indexed: 02/06/2023] Open
Abstract
Advances in molecular cell biology over the last decade have clarified the mechanisms involved in cancer growth, invasion, and metastasis, and enabled the development of molecular-targeted agents. To date, sorafenib is the only molecular-targeted agent whose survival benefit has been demonstrated in two global phase III randomized controlled trials, and has been approved worldwide. Phase III clinical trials of other molecular targeted agents comparing them with sorafenib as first-line treatment agents are ongoing. Those agents target the vascular endothelial growth factor, platelet-derived growth factor receptors, as well as target the epidermal growth factor receptor, insulin-like growth factor receptor and mammalian target of rapamycin, in addition to other molecules targeting other components of the signal transduction pathways. In addition, the combination of sorafenib with standard treatment, such as resection, ablation, transarterial embolization, and hepatic arterial infusion chemotherapy are ongoing. This review outlines the main pathways involved in the development and progression of hepatocellular carcinoma and the new agents that target these pathways. Finally, the current statuses of clinical trials of new agents or combination therapy with sorafenib and standard treatment will also be discussed.
Collapse
|
22
|
Jhaveri KS, Cleary SP, Fischer S, Haider MA, Pargoankar V, Khalidi K, Moshonov H, Gallinger S. Blood oxygen level-dependent liver MRI: can it predict microvascular invasion in HCC? J Magn Reson Imaging 2012; 37:692-9. [PMID: 23125092 DOI: 10.1002/jmri.23858] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 09/04/2012] [Indexed: 12/19/2022] Open
Abstract
PURPOSE To assess Blood Oxygen Level-Dependent (BOLD) Magnetic Resonance Imaging (MRI) for noninvasive preoperative prediction of Microvascular Invasion (MVI) in Hepatocellular Carcinoma (HCC). MATERIALS AND METHODS In this prospective, institutional review board approved study, 26 patients (21 men and 5 women age range, 34-77 years with mean age of 61 years) with HCC were evaluated preoperatively with liver MRI including baseline and post oxygen (O2) breathing BOLD MRI. Post processing of MRI data was performed to obtain R2* values (1/s) and correlated with histopathological assessment of MVI. Statistical analysis was performed to assess correlation of baseline R2*, post O2 R2* and R2* ratios to presence of MVI in HCC by binary logistic regression analysis. RESULTS MVI was present in 15/26 (58%) of HCC on histopathology. The mean R2* values ± SD at baseline and post O2 with and without MVI were 35 ± 12, 36 ± 12, 38 ± 10, 42 ± 17. The R2* values between the groups with and without MVI were not significantly different statistically. CONCLUSION BOLD MRI is unable to accurately predict MVI in HCC. The noninvasive preoperative MRI detection of MVI in HCC remains elusive.
Collapse
Affiliation(s)
- Kartik S Jhaveri
- Joint Department of Medical Imaging, University Health Network and Mount Sinai Hospital, University of Toronto, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Background Co-expression based Cancer Modules (CMs) are sets of genes that act in concert to carry out specific functions in different cancer types, and are constructed by exploiting gene expression profiles related to specific clinical conditions or expression signatures associated to specific processes altered in cancer. Unfortunately, genes involved in cancer are not always detectable using only expression signatures or co-expressed sets of genes, and in principle other types of functional interactions should be exploited to obtain a comprehensive picture of the molecular mechanisms underlying the onset and progression of cancer. Results We propose a novel semi-supervised method to rank genes with respect to CMs using networks constructed from different sources of functional information, not limited to gene expression data. It exploits on the one hand local learning strategies through score functions that extend the guilt-by-association approach, and on the other hand global learning strategies through graph kernels embedded in the score functions, able to take into account the overall topology of the network. The proposed kernelized score functions compare favorably with other state-of-the-art semi-supervised machine learning methods for gene ranking in biological networks and scales well with the number of genes, thus allowing fast processing of very large gene networks. Conclusions The modular nature of kernelized score functions provides an algorithmic scheme from which different gene ranking algorithms can be derived, and the results show that using integrated functional networks we can successfully predict CMs defined mainly through expression signatures obtained from gene expression data profiling. A preliminary analysis of top ranked "false positive" genes shows that our approach could be in perspective applied to discover novel genes involved in the onset and progression of tumors related to specific CMs.
Collapse
Affiliation(s)
- Matteo Re
- Dipartimento di Informatica, Università degli Studi di Milano, via Comelico 39/41, 20135 Milano MI, Italia
| | | |
Collapse
|
24
|
Wang H, Wen W. Biomarkers of Hepatocellular Carcinoma. PRIMARY LIVER CANCER 2012:79-154. [DOI: 10.1007/978-3-642-28702-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
25
|
|
26
|
Abstract
The diagnosis of cancer has undergone major changes in the last 40 years. Once based purely on morphology, diagnosis has come to incorporate immunological, cytogenetic and molecular methods. Many cancers, especially leukaemias, are now defined by molecular markers. Gene expression profiling based on mRNA has led to further refinement of the classification and diagnosis of cancer. More recently, miRNAs (microRNAs), among other small non-coding RNA molecules, have been discovered and found to be major players in cell biology. miRNAs, having both oncogenic and tumour-suppressive functions, are dysregulated in many types of cancer. miRNAs also interfere with metastasis, apoptosis and invasiveness of cancer cells. In the present review, we discuss recent advances in miRNA profiling in human cancer. We discuss both frequent and rare tumour types and give an outlook on future developments.
Collapse
|
27
|
Krupp M, Maass T, Marquardt JU, Staib F, Bauer T, König R, Biesterfeld S, Galle PR, Tresch A, Teufel A. The functional cancer map: a systems-level synopsis of genetic deregulation in cancer. BMC Med Genomics 2011; 4:53. [PMID: 21718500 PMCID: PMC3148554 DOI: 10.1186/1755-8794-4-53] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 06/30/2011] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Cancer cells are characterized by massive dysegulation of physiological cell functions with considerable disruption of transcriptional regulation. Genome-wide transcriptome profiling can be utilized for early detection and molecular classification of cancers. Accurate discrimination of functionally different tumor types may help to guide selection of targeted therapy in translational research. Concise grouping of tumor types in cancer maps according to their molecular profile may further be helpful for the development of new therapeutic modalities or open new avenues for already established therapies. METHODS Complete available human tumor data of the Stanford Microarray Database was downloaded and filtered for relevance, adequacy and reliability. A total of 649 tumor samples from more than 1400 experiments and 58 different tissues were analyzed. Next, a method to score deregulation of KEGG pathway maps in different tumor entities was established, which was then used to convert hundreds of gene expression profiles into corresponding tumor-specific pathway activity profiles. Based on the latter, we defined a measure for functional similarity between tumor entities, which yielded to phylogeny of tumors. RESULTS We provide a comprehensive, easy-to-interpret functional cancer map that characterizes tumor types with respect to their biological and functional behavior. Consistently, multiple pathways commonly associated with tumor progression were revealed as common features in the majority of the tumors. However, several pathways previously not linked to carcinogenesis were identified in multiple cancers suggesting an essential role of these pathways in cancer biology. Among these pathways were 'ECM-receptor interaction', 'Complement and Coagulation cascades', and 'PPAR signaling pathway'. CONCLUSION The functional cancer map provides a systematic view on molecular similarities across different cancers by comparing tumors on the level of pathway activity. This work resulted in identification of novel superimposed functional pathways potentially linked to cancer biology. Therefore, our work may serve as a starting point for rationalizing combination of tumor therapeutics as well as for expanding the application of well-established targeted tumor therapies.
Collapse
Affiliation(s)
- Markus Krupp
- Department of Medicine I, Johannes Gutenberg University, Mainz, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Villanueva A, Llovet JM. Targeted therapies for hepatocellular carcinoma. Gastroenterology 2011; 140:1410-26. [PMID: 21406195 PMCID: PMC3682501 DOI: 10.1053/j.gastro.2011.03.006] [Citation(s) in RCA: 356] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 03/09/2011] [Accepted: 03/10/2011] [Indexed: 12/27/2022]
Abstract
Unlike most solid tumors, the incidence and mortality of hepatocellular carcinoma (HCC) have increased in the United States and Europe in the past decade. Most patients are diagnosed at advanced stages, so there is an urgent need for new systemic therapies. Sorafenib, a tyrosine kinase inhibitor (TKI), has shown clinical efficacy in patients with HCC. Studies in patients with lung, breast, or colorectal cancers have indicated that the genetic heterogeneity of cancer cells within a tumor affect its response to therapeutics designed to target specific molecules. When tumor progression requires alterations in specific oncogenes (oncogene addiction), drugs that selectively block their products might slow tumor growth. However, no specific oncogene addictions are yet known to be implicated in HCC progression, so it is important to improve our understanding of its molecular pathogenesis. There are currently many clinical trials evaluating TKIs for HCC, including those tested in combination with (eg, erlotinib) or compared with (eg, linifanib) sorafenib as a first-line therapy. For patients who do not respond or are intolerant to sorafenib, TKIs such as brivanib, everolimus, and monoclonal antibodies (eg, ramucirumab) are being tested as second-line therapies. There are early stage trials investigating the efficacy for up to 60 reagents for HCC. Together, these studies might change the management strategy for HCC, and combination therapies might be developed for patients with advanced HCC. Identification of oncogenes that mediate tumor progression, and trials that monitor their products as biomarkers, might lead to personalized therapy; reagents that interfere with signaling pathways required for HCC progression might be used to treat selected populations, and thereby maximize the efficacy and cost benefit.
Collapse
Affiliation(s)
- Augusto Villanueva
- HCC Translational Research Laboratory, Barcelona-Clinic Liver Cancer Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Liver Unit, Hospital Clinic, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto Carlos III, Madrid, Spain
| | - Josep M. Llovet
- HCC Translational Research Laboratory, Barcelona-Clinic Liver Cancer Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Liver Unit, Hospital Clinic, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto Carlos III, Madrid, Spain.,Liver Cancer Program, Division of Liver Diseases, Mount Sinai School of Medicine, New York, (NY), USA.,Institució Catalana de Recerca i Estudis Avançats, Barcelona, Catalonia, Spain
| |
Collapse
|
29
|
Abstract
Gene expression profiling using microarray technologies provides a powerful approach to understand complex biological systems and the pathogenesis of diseases. In the field of liver cancer research, a number of genome-wide profiling studies have been published. These studies have provided gene sets, that is, signature, which could classify tumors and predict clinical outcomes such as survival, recurrence, and metastasis. More recently, the application of genomic profiling has been extended to identify molecular targets, pathways, and the cellular origins of the tumors. Systemic and integrative analyses of multiple data sets and emerging new technologies also accelerate the progress of the cancer genomic studies. Here, we review the genomic signatures identified from the genomic profiling studies of hepatocellular carcinoma (HCC), and categorize and characterize them into prediction, phenotype, function, and molecular target signatures according to their utilities and properties. Our classification of the signatures would be helpful to understand and design studies with extended application of genomic profiles.
Collapse
Affiliation(s)
- Hyun Goo Woo
- Department of Physiology, Ajou University School of Medicine, Suwon, Korea
| | | | | | | |
Collapse
|
30
|
Huang P, Qiu J, Li B, Hong J, Lu C, Wang L, Wang J, Hu Y, Jia W, Yuan Y. Role of Sox2 and Oct4 in predicting survival of hepatocellular carcinoma patients after hepatectomy. Clin Biochem 2011; 44:582-9. [PMID: 21376027 DOI: 10.1016/j.clinbiochem.2011.02.012] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Revised: 02/01/2011] [Accepted: 02/18/2011] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The present study was aimed to explore the prognostic strength of Sox2 and Oct4A in hepatocellular carcinoma (HCC). DESIGN AND METHODS We investigated the expression of Sox2 and Oct4A in five hepatoma cell lines, one immortalized normal liver cell line, HCC tissues with matched nontumorous liver tissues and normal liver tissues by reverse transcription-polymerase chain reaction. RESULTS Sox2 and Oct4A mRNA were overexpressed in hepatoma cell lines and tumor tissues. Sox2 or Oct4A positive expression was significantly associated with an aggressive phenotype. Both univariate and multivariate analyses revealed that Sox2 or Oct4A was an independent prognostic factor for HCC. When using subgroup analysis, the patients with a co-expression of Sox2/Oct4A had the poorest prognosis. Further analysis demonstrated that Sox2 alone or Sox2/Oct4A could stratify outcome in HCC patients with early stage. CONCLUSIONS Sox2 and Oct4A can be novel predictors of poor prognosis for patients undergoing resection of HCC.
Collapse
Affiliation(s)
- Pinzhu Huang
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou, GD 510060, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Hepatocellular carcinoma (HCC) is a fatal disease that represents the fifth most common human cancer. Although remarkable progress has been achieved in HCC treatment in China, the overall incidence and mortality rates of HCC show no obvious changes. Pharmacological treatment can not improve the prognosis of patients with unresectable HCC. This emphasizes the need to identify new targets for early diagnosis, chemoprevention, and treatment of the disease. An effort to understand the molecular mechanisms responsible for tumor initiation and progression has led to the identification of several potential molecular targets for HCC. The majority of these targets are involved in receptor tyrosine kinase-activated pathways, such as the Raf/MEK/ERK, PI-3K/Akt/mTOR, and Jak/Stat pathways. Sorafenib is a multikinase inhibitor that has attracted wide attention. This review describes the potential targets for HCC and recent progress in targeted therapy of the disease.
Collapse
|
32
|
Kemmer N, Neff G. Recipient-based approach to tailoring immunosuppression in liver transplantation. Transplant Proc 2010; 42:1731-7. [PMID: 20620512 DOI: 10.1016/j.transproceed.2010.02.076] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2009] [Accepted: 02/02/2010] [Indexed: 01/05/2023]
Abstract
Improvements in the field of transplant immunosuppression (IS) have led to significant advances in long-term survival of liver transplant recipients. Despite this progress, survival rates vary depending on recipient, donor and/or perioperative factors. Tailoring IS based on recipient factors is of growing interest among health care providers involved in the care of organ transplant recipients. To date there is no consensus document addressing individualized IS therapy for liver transplant recipients. This review will discuss the information available on the effect of the various IS drugs on recipient-based factors such as age, ethnicity, and liver disease etiology.
Collapse
Affiliation(s)
- N Kemmer
- Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | | |
Collapse
|
33
|
Andersen JB, Factor VM, Marquardt JU, Raggi C, Lee YH, Seo D, Conner EA, Thorgeirsson SS. An integrated genomic and epigenomic approach predicts therapeutic response to zebularine in human liver cancer. Sci Transl Med 2010; 2:54ra77. [PMID: 20962331 PMCID: PMC3077922 DOI: 10.1126/scitranslmed.3001338] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Epigenomic changes such as aberrant hypermethylation and subsequent atypical gene silencing are characteristic features of human cancer. Here, we report a comprehensive characterization of epigenomic modulation caused by zebularine, an effective DNA methylation inhibitor, in human liver cancer. Using transcriptomic and epigenomic profiling, we identified a zebularine response signature that classified liver cancer cell lines into two major subtypes with different drug responses. In drug-sensitive cell lines, zebularine caused inhibition of proliferation coupled with increased apoptosis, whereas drug-resistant cell lines showed up-regulation of oncogenic networks (for example, E2F1, MYC, and TNF) that drive liver cancer growth in vitro and in preclinical mouse models. Assessment of zebularine-based therapy in xenograft mouse models demonstrated potent therapeutic effects against tumors established from zebularine-sensitive but not zebularine-resistant liver cancer cells, leading to increased survival and decreased pulmonary metastasis. Integration of the zebularine gene expression and demethylation response signatures allowed differentiation of patients with hepatocellular carcinoma according to their survival and disease recurrence. This integrated signature identified a subclass of patients within the poor-survivor group that is likely to benefit from therapeutic agents that target the cancer epigenome.
Collapse
Affiliation(s)
- Jesper B. Andersen
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4262, USA
| | - Valentina M. Factor
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4262, USA
| | - Jens U. Marquardt
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4262, USA
| | - Chiara Raggi
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4262, USA
| | - Yun-Han Lee
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4262, USA
| | - Daekwan Seo
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4262, USA
| | - Elizabeth A. Conner
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4262, USA
| | - Snorri S. Thorgeirsson
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4262, USA
| |
Collapse
|
34
|
Abstract
The marked heterogeneity of hepatocellular carcinoma (HCC), particularly with regard to the etiology and severity of the underlying cirrhosis, makes clinical trial design in this disease very challenging. In addition, despite the global burden of HCC, there have been relatively few randomized studies. The major advance in medical therapy in HCC has been the benefit of sorafenib, as demonstrated in two Phase III studies. However, the benefit is small, and new therapies to augment or replace sorafenib are urgently needed. These newer therapies, as well as the progress made in two important areas – clinical trial design and molecular characterization – are the subject of this article.
Collapse
Affiliation(s)
- Austin Duffy
- Medical Oncology Branch, National Cancer Institute, 9000 Rockville Pike, 10/13N240G, Bethesda, MD 20892, USA
| | | |
Collapse
|
35
|
Villanueva A, Hoshida Y, Toffanin S, Lachenmayer A, Alsinet C, Savic R, Cornella H, Llovet JM. New strategies in hepatocellular carcinoma: genomic prognostic markers. Clin Cancer Res 2010; 16:4688-94. [PMID: 20713493 PMCID: PMC3395071 DOI: 10.1158/1078-0432.ccr-09-1811] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Accurate prognosis prediction in oncology is critical. In patients with hepatocellular carcinoma (HCC), unlike most solid tumors, the coexistence of two life-threatening conditions, cancer and cirrhosis, makes prognostic assessments difficult. Despite the usefulness of clinical staging systems for HCC in routine clinical decision making (e.g., Barcelona-Clinic Liver Cancer algorithm), there is still a need to refine and complement outcome predictions. Recent data suggest the ability of gene signatures from the tumor (e.g., EpCAM signature) and adjacent tissue (e.g., poor-survival signature) to predict outcome in HCC (either recurrence or overall survival), although independent external validation is still required. In addition, novel information is being produced by alternative genomic sources such as microRNA (miRNA; e.g., miR-26a) or epigenomics, areas in which promising preliminary data are thoroughly explored. Prognostic models need to contemplate the impact of liver dysfunction and risk of subsequent de novo tumors in a patient's life expectancy. The challenge for the future is to precisely depict genomic predictors (e.g., gene signatures, miRNA, or epigenetic biomarkers) at each stage of the disease and their specific influence to determine patient prognosis.
Collapse
Affiliation(s)
- Augusto Villanueva
- Institut d'Investigacions Biomèdiques Agusto Pi i Sunyer, Hospital Clinic, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Villanueva A, Newell P, Hoshida Y. Inherited hepatocellular carcinoma. Best Pract Res Clin Gastroenterol 2010; 24:725-34. [PMID: 20955973 DOI: 10.1016/j.bpg.2010.07.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 07/15/2010] [Accepted: 07/22/2010] [Indexed: 01/31/2023]
Abstract
Inherited liver disorders that cause chronic inflammation, fibrosis, and cirrhosis can lead to the development of liver cancer. Because of the rarity and diversity of some of these syndromes, the relative risk of developing HCC in these patients and the age at which tumours typically arise cannot be accurately estimated. Among patients with hereditary hemachromatosis (HH), the annual incidence of HCC is 4% once cirrhosis has been established. Fibrosis and portal hypertension associated with HH can be partially reversed with therapeutic phlebotomy, but it is unclear whether this treatment alters the incidence of HCC in these patients. Importantly, it seems likely that coincidence of these genetic disorders with known HCC risk factors such as alcoholism and viral hepatitis would amplify their oncogenic potential. For this reason, patients with known genetic disorders of the liver should be repeatedly counselled to avoid environmental and toxic injury to the liver. Treatment of HCC in patients with inherited liver disease mirrors that of HCC associated with other etiologies. Unfortunately, there are case series which suggest these patients with inherited liver disease and HCC tend to present at more advanced stages and are therefore not eligible for curative therapies, causing overall decreased survival relative to patients with HCC of viral or other etiologies.
Collapse
Affiliation(s)
- Augusto Villanueva
- HCC Translational Research Laboratory, Barcelona-Clinic Liver Cancer Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Liver Unit, Hospital Clinic, Barcelona, Spain.
| | | | | |
Collapse
|
37
|
Cornellà H, Villanueva A. Genomic tracing of the elusive liver cancer ancestor. J Hepatol 2010; 53:578-9. [PMID: 20561704 DOI: 10.1016/j.jhep.2010.04.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 04/21/2010] [Accepted: 04/23/2010] [Indexed: 12/11/2022]
Affiliation(s)
- Helena Cornellà
- HCC Translational Research Laboratory, BCLC Group, IDIBAPS, CIBEREHD, Liver Unit, Hospital Clinic, Barcelona, Spain
| | | |
Collapse
|
38
|
Zhang XF, Tan X, Zeng G, Misse A, Singh S, Kim Y, Klaunig JE, Monga SPS. Conditional beta-catenin loss in mice promotes chemical hepatocarcinogenesis: role of oxidative stress and platelet-derived growth factor receptor alpha/phosphoinositide 3-kinase signaling. Hepatology 2010; 52:954-65. [PMID: 20583210 PMCID: PMC3100799 DOI: 10.1002/hep.23747] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
UNLABELLED Activation of beta-catenin, the central effector of the canonical Wnt pathway and a recognized oncogene, has been implicated in hepatocellular carcinoma. We examined N-nitrosodiethylamine (DEN)-induced tumorigenesis in hepatic beta-catenin conditional knockout mice (beta-cat KO). Male beta-cat KO and age- and sex-matched littermate controls were given a single intraperitoneal DEN injection and followed for 6-12 months for hepatic tumors. Hepatic tumors were characterized for histology, proliferation, apoptosis, oxidative stress, and specific proteins by way of western blot, immunohistochemistry, and coprecipitation studies. For in vivo tumor intervention studies, specific inhibitors were administered intraperitoneally or through drinking water. Intriguingly, beta-cat KO mice showed a paradoxical increase in susceptibility to DEN-induced tumorigenesis. This accelerated tumorigenesis is due to increased injury and inflammation, unrestricted oxidative stress, fibrosis, and compensatory increase in hepatocyte proliferation secondary to platelet-derived growth factor receptor alpha (PDGFRalpha)/phosphoinositide 3-kinase (PIK3CA)/Akt activation and c-Myc overexpression. In vitro suppression of beta-catenin expression in hepatoma cells led to enhanced PDGFRalpha expression, which was abrogated in the presence of nuclear factor kappaB (NF-kappaB) inhibitor. Daily treatment of 6-month-old DEN-exposed beta-cat KO with PDGFRalpha inhibitor dramatically reduced tumor numbers and size. Inclusion of N-acetyl-L-cysteine, a known antioxidant and NF-kappaB inhibitor, in the drinking water led to complete abolition of tumorigenesis in DEN-exposed beta-cat KO. CONCLUSION Loss of beta-catenin impairs the liver's ability to counteract DEN-induced oxidative stress and enhances tumorigenesis through PDGFRalpha/PIK3CA/Akt signaling. Blockade of PDGFRalpha or oxidative stress dramatically affects beta-catenin-deficient tumorigenesis. Also, hepatoma cells use PDGFRalpha/PIK3CA signaling as an escape mechanism following beta-catenin suppression, and their sequential suppression profoundly impedes tumor proliferation.
Collapse
Affiliation(s)
- Xu-Feng Zhang
- Department of Pathology and Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Xu C, Liu S, Fu H, Li S, Tie Y, Zhu J, Xing R, Jin Y, Sun Z, Zheng X. MicroRNA-193b regulates proliferation, migration and invasion in human hepatocellular carcinoma cells. Eur J Cancer 2010; 46:2828-36. [PMID: 20655737 DOI: 10.1016/j.ejca.2010.06.127] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 06/23/2010] [Accepted: 06/23/2010] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND AIMS Recently, some miRNAs have been reported to be connected closely with the development of human hepatocellular carcinoma. However, the functions of these miRNAs in HCC remain largely undefined. METHODS The expression profiles of miR-193b were compared between HCC tissues and adjacent normal liver tissues using qRT-PCR method. This method was also be used to screen the potential target genes of miR-193b. A luciferase reporter assay was conducted to confirm target association. Finally, the functional effect of miR-193b in hepatoma cells was examined further. RESULTS miR-193b was significantly down-regulated in most of the HCC tissues compared to the matching non-tumoural liver tissues. Furthermore, ectopic expression of miR-193b dramatically suppressed the ability of hepatoma cells to form colonies in vitro and to develop tumours in nude mice. CCND1 and ETS1 were revealed to be regulated by miR-193b directly. By regulating the expressions of these oncogenes, miR-193b induced cell cycle arrest and inhibited the invasion and migration of hepatoma cells. CONCLUSIONS miR-193b may function as a tumour suppressor in the development of HCC by acting on multiple tumourigenic pathways.
Collapse
Affiliation(s)
- Chengwang Xu
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wörns MA, Galle PR. Novel inhibitors in development for hepatocellular carcinoma. Expert Opin Investig Drugs 2010; 19:615-29. [PMID: 20374038 DOI: 10.1517/13543781003767418] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE OF THE FIELD The multikinase inhibitor sorafenib was the first agent to demonstrate a survival benefit for patients with locally advanced or metastatic hepatocellular carcinoma (HCC). Although sorafenib represents a landmark in the treatment of HCC and proved molecularly targeted therapy to be effective in this disease, it represents just the first step towards an improvement in systemic therapy. Since then, novel inhibitors have been evaluated in early clinical trials, showing potential activity. AREAS COVERED IN THIS REVIEW This article aims to review novel inhibitors emerging in the field of advanced HCC. An Internet-based search was performed to identify abstracts, clinical trials ( www.clinicaltrials.gov , last accessed 30 November 2009), and original research and review articles. WHAT THE READER WILL GAIN Readers will gain a comprehensive survey of current molecularly targeted therapy approaches in advanced HCC. In addition, challenges such as the design of clinical trials, the assessment of radiological response, the role of combination therapy, and future developments in molecularly targeted therapy are discussed. TAKE HOME MESSAGE Sorafenib is the standard of care in patients with advanced HCC. However, promising novel inhibitors are under investigation. Combined molecularly targeted therapies according to an individual genomic and proteomic profiling will probably lead to more personalised medicine in advanced HCC.
Collapse
Affiliation(s)
- Marcus Alexander Wörns
- University Medicine of the Johannes Gutenberg-University Mainz, Department of Internal Medicine, Mainz, Germany.
| | | |
Collapse
|
41
|
Abstract
Hepatocellular carcinoma (HCC) is one of the most common and lethal malignancies worldwide. Due to late diagnosis and advanced underlying liver cirrhosis, only limited treatment options with marginal clinical benefits have been available in up to 70% of patients. However, major progress has been achieved with regard to surveillance, early diagnosis, and multimodal treatment approaches during the last years leading to an improvement in prognosis. Particularly, the increasing knowledge of molecular hepatocarcinogenesis today provides the opportunity for targeted therapy. The multikinase inhibitor sorafenib has broadened the therapeutic horizon for patients with advanced disease and is currently under investigation for patients in early and intermediate stages as adjuvant therapy after resection/local ablation or in combination with transcatheter arterial chemoembolization. Future research will continue to unravel the key signaling cascades in different subclasses of HCC patients according to their genomic and proteomic profiling. These approaches bear the potential to individualize anticancer therapy, in the end allowing treatment of those benefiting most and excluding those who do not. This article shortly reviews the current knowledge in the management of HCC and provides insights into future perspectives with a special focus on recent progress in multidisciplinary treatment modalities.
Collapse
|
42
|
Schaff Z, Kovalszky I, Lotz G, Kiss A. Hepatocellular carcinoma – from macroscopy to molecular pathology. Orv Hetil 2010; 151:982-989. [DOI: 10.1556/oh.2010.28875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
A hepatocellularis carcinoma (HCC) igen rossz prognózisú daganat, azonban diagnosztikája és kezelése területén az utóbbi években jelentős előrehaladás történt. Mindehhez hozzájárult a HCC molekuláris patogenezisének mélyebb megismerése. A cirrhosis talaján kialakult HCC praemalignus elváltozásai a nagy regeneratív nodulus, az alacsony („low”) és magas („high”) fokozatú diszplasztikus nodulus. Mikroszkóposan a WHO trabecularis (micro-, macrotrabecularis), acinaris (pseudoglandularis,), scirrhosus és szolid formát különít el, speciális altípusként a világos sejtes, fibrolamellaris és kevert cholangiohepatocellularis szöveti forma ismert. Ezen szövettani típusok prognosztikai jelentősége vitatott. A fibrolamellaris, fiatalokban előforduló, nem cirrhoticus HCC-t jobb prognózisúnak tartják, bár valószínű, hogy ez annak a következménye, hogy ezen típust cirrhosis nem kíséri. A diagnózist segíthetik egyes, a szérumban és a daganatban is kimutatható tumormarkerek, így a jól ismert alfa-fetoprotein (AFP) mellett a glipikán-3 és a survivin, az újabban leírt agrin és claudinok, valamint a májsejteredetet bizonyító hepatocytaspecifikus antigén (HSA). Újabban az úgynevezett mikro-RNS-ek diagnosztikus jelentősége, elsősorban a májsejtspecifikus mir-122-é is felmerült. A HCC molekuláris osztályozása, a kezelés irányait is megszabó barcelonai beosztás (BCLC) mellett, kulcsfontosságú molekuláris eltérések alapján csoportosítja a HCC-t. Számos olyan molekuláris alteráció észlelhető, amely minden HCC-ben megfigyelhető, míg egyes eltérések csak bizonyos tumorokban detektálhatók.
Collapse
Affiliation(s)
- Zsuzsa Schaff
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Patológiai Intézet Budapest Üllői út 93. 1091
| | - Ilona Kovalszky
- 2 Semmelweis Egyetem, Általános Orvostudományi Kar I. Patológiai és Kísérleti Rákkutató Intézet Budapest
| | - Gábor Lotz
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Patológiai Intézet Budapest Üllői út 93. 1091
| | - András Kiss
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Patológiai Intézet Budapest Üllői út 93. 1091
| |
Collapse
|
43
|
Villanueva A, Minguez B, Forner A, Reig M, Llovet JM. Hepatocellular carcinoma: novel molecular approaches for diagnosis, prognosis, and therapy. Annu Rev Med 2010; 61:317-28. [PMID: 20059340 DOI: 10.1146/annurev.med.080608.100623] [Citation(s) in RCA: 194] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The genomic era is changing the understanding of cancer, although translation of the vast amount of data available into decision-making algorithms is far from reality. Molecular profiling of hepatocellular carcinoma (HCC), the most common cause of death among cirrhotic patients and a fast-growing malignancy in Western countries, is enabling the advancement of novel approaches to disease diagnosis and management. Most HCCs arise on a cirrhotic liver, and predictably, an accurate genomic characterization will allow the identification of procarcinogenic signals amenable to selective targeting by chemopreventive strategies. Molecular diagnosis is currently feasible for small tumors, but it has not yet been formalized by scientific guidelines. Molecular treatment is a reality: Sorafenib confers unprecedented survival benefits in patients at advanced stages. Genomic information from tumor and nontumoral tissue will aid prognosis prediction and facilitate the identification of oncogene addiction loops, providing the opportunity for more personalized medicine.
Collapse
Affiliation(s)
- Augusto Villanueva
- HCC Translational Research Laboratory, Barcelona-Clínic Liver Cancer Group, Institut d'Investigacions Biomediques Agusto Pi i Sunyer (IDIBAPS), Liver Unit, Hospital Clinic, Barcelona, Spain.
| | | | | | | | | |
Collapse
|
44
|
Bhoori S, Toffanin S, Sposito C, Germini A, Pellegrinelli A, Lampis A, Mazzaferro V. Personalized molecular targeted therapy in advanced, recurrent hepatocellular carcinoma after liver transplantation: a proof of principle. J Hepatol 2010; 52:771-5. [PMID: 20347502 DOI: 10.1016/j.jhep.2010.01.025] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Revised: 01/12/2010] [Accepted: 01/18/2010] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS The advent of molecular medicine that targets specific pathways is changing the therapeutic approach to hepatocellular carcinoma. For several aberrantly activated pathways in hepatocarcinoma, surrogate markers of activation can be assessed by immunohistochemistry, although associations with in vivo response to targeted therapies are still lacking. METHODS A patient, who presented with hepatic and extra-hepatic hepatocarcinoma recurrence 11 years after liver transplantation, was assessed for beta-catenin, pERK, and pS6 in primary and secondary tumor specimens, in order to define a possible activation of the Wnt, Ras/MAPK and Akt/mTOR pathways and design a personalized targeted therapy in absence of alternative treatment options. Moreover, mutation analysis of the beta-catenin gene (CTNNB1) and DNA microsatellite analyses were performed. RESULTS The identification of the same mutation in the beta-catenin gene, as well as the same microsatellite pattern in tumor tissues taken 11 years apart, proved that the observed hepatocarcinoma was a true recurrence. Nuclear beta-catenin and pS6 in tumor cells were positive, whereas pERK was positive only in the peritumoral endothelium. This pattern of immunohistochemistry, after failure of sorafenib alone, lead to the choice to add the mTOR inhibitor, everolimus, to sorafenib. Three months later a 50% tumor reduction was observed, and after 6 months a further reduction of tumor vital components was confirmed, while a grade II gastrointestinal bleeding episode occurred. CONCLUSIONS A personalized approach aimed to treat recurrent hepatocarcinoma is possible through analysis of tumoral molecular pathways. Partial success of the selected combination of sorafenib and everolimus supports the pivotal role of mTOR signalling and highlights the importance of reliable biomarkers to route the best molecular-based therapeutic options in HCC.
Collapse
Affiliation(s)
- Sherrie Bhoori
- Liver Unit, Hepato-Oncology Group and Molecular Pathology, National Cancer Institute, Fondazione Istituto Nazionale Tumori, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
45
|
Hoshida Y, Toffanin S, Lachenmayer A, Villanueva A, Minguez B, Llovet JM. Molecular classification and novel targets in hepatocellular carcinoma: recent advancements. Semin Liver Dis 2010; 30:35-51. [PMID: 20175032 PMCID: PMC3668687 DOI: 10.1055/s-0030-1247131] [Citation(s) in RCA: 248] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of most lethal cancers worldwide. Strategic decisions for the advancement of molecular therapies in this neoplasm require a clear understanding of its molecular classification. Studies indicate aberrant activation of signaling pathways involved in cellular proliferation (e.g., epidermal growth factor and RAS/mitogen-activated protein kinase pathways), survival (e.g., Akt/mechanistic target of rapamycin pathway), differentiation (e.g., Wnt and Hedgehog pathways), and angiogenesis (e.g., vascular endothelial growth factor and platelet-derived growth factor), which is heterogeneously presented in each tumor. Integrative analysis of accumulated genomic datasets has revealed a global scheme of molecular classification of HCC tumors observed across diverse etiologic factors and geographic locations. Such a framework will allow systematic understanding of the frequently co-occurring molecular aberrations to design treatment strategy for each specific subclass of tumors. Accompanied by a growing number of clinical trials of molecular targeted drugs, diagnostic and prognostic biomarker development will be facilitated with special attention on study design and with new assay technologies specialized for archived fixed tissues. A new class of genomic information, microRNA dysregulation and epigenetic alterations, will provide insight for more precise understanding of disease mechanism and expand the opportunity of biomarker/therapeutic target discovery. These efforts will eventually enable personalized management of HCC.
Collapse
Affiliation(s)
- Yujin Hoshida
- Cancer Program, Broad Institute, Cambridge, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
46
|
Finn RS. Development of Molecularly Targeted Therapies in Hepatocellular Carcinoma: Where Do We Go Now? Clin Cancer Res 2010; 16:390-7. [DOI: 10.1158/1078-0432.ccr-09-2084] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
47
|
Zhou D, Conrad C, Xia F, Park JS, Payer B, Yin Y, Lauwers GY, Thasler W, Lee JT, Avruch J, Bardeesy N. Mst1 and Mst2 maintain hepatocyte quiescence and suppress hepatocellular carcinoma development through inactivation of the Yap1 oncogene. Cancer Cell 2009; 16:425-38. [PMID: 19878874 PMCID: PMC3023165 DOI: 10.1016/j.ccr.2009.09.026] [Citation(s) in RCA: 751] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 08/14/2009] [Accepted: 09/22/2009] [Indexed: 12/11/2022]
Abstract
Hippo-Lats-Yorkie signaling regulates tissue overgrowth and tumorigenesis in Drosophila. We show that the Mst1 and Mst2 protein kinases, the mammalian Hippo orthologs, are cleaved and constitutively activated in the mouse liver. Combined Mst1/2 deficiency in the liver results in loss of inhibitory Ser127 phosphorylation of the Yorkie ortholog, Yap1, massive overgrowth, and hepatocellular carcinoma (HCC). Reexpression of Mst1 in HCC-derived cell lines promotes Yap1 Ser127 phosphorylation and inactivation and abrogates their tumorigenicity. Notably, Mst1/2 inactivates Yap1 in liver through an intermediary kinase distinct from Lats1/2. Approximately 30% of human HCCs show low Yap1(Ser127) phosphorylation and a majority exhibit loss of cleaved, activated Mst1. Mst1/2 inhibition of Yap1 is an important pathway for tumor suppression in liver relevant to human HCC.
Collapse
Affiliation(s)
- Dawang Zhou
- Department of Molecular Biology, Harvard Medical School, Boston, MA USA 02114
- Diabetes unit, Harvard Medical School, Boston, MA USA 02114
| | - Claudius Conrad
- Medical services, Cancer Center, Harvard Medical School, Boston, MA USA 02114
- Surgical services, Harvard Medical School, Boston, MA USA 02114
| | - Fan Xia
- Department of Molecular Biology, Harvard Medical School, Boston, MA USA 02114
- Diabetes unit, Harvard Medical School, Boston, MA USA 02114
| | - Ji-Sun Park
- Medical services, Cancer Center, Harvard Medical School, Boston, MA USA 02114
| | - Bernhard Payer
- Department of Molecular Biology, Harvard Medical School, Boston, MA USA 02114
- Howard Hughes Medical Institute, Massachusetts General Hospital; Departments of Medicine, Pathology and Genetics, Harvard Medical School, Boston, MA USA 02114
| | - Yi Yin
- Department of Molecular Biology, Harvard Medical School, Boston, MA USA 02114
- Diabetes unit, Harvard Medical School, Boston, MA USA 02114
| | | | - Wolfgang Thasler
- Department of Surgery, LM University Munich, Hospital Grosshadern, Munich Germany
| | - Jeannie T. Lee
- Department of Molecular Biology, Harvard Medical School, Boston, MA USA 02114
- Howard Hughes Medical Institute, Massachusetts General Hospital; Departments of Medicine, Pathology and Genetics, Harvard Medical School, Boston, MA USA 02114
| | - Joseph Avruch
- Department of Molecular Biology, Harvard Medical School, Boston, MA USA 02114
- Diabetes unit, Harvard Medical School, Boston, MA USA 02114
- Address correspondence to: Nabeel Bardeesy Ph.D., Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, CPZN4216, 185 Cambridge Street, Boston, Massachusetts 02114, Tel:617-643-2579, Fax:617-643-3170, or Joseph Avruch M.D., Diabetes Research Lab, Department of Molecular Biology, Massachusetts General Hospital, Simches Research Bldg, 6408, 185 Cambridge St., Boston, MA 02114, Tel:617-726-6909, Fax:617-726-5649,
| | - Nabeel Bardeesy
- Medical services, Cancer Center, Harvard Medical School, Boston, MA USA 02114
- Address correspondence to: Nabeel Bardeesy Ph.D., Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, CPZN4216, 185 Cambridge Street, Boston, Massachusetts 02114, Tel:617-643-2579, Fax:617-643-3170, or Joseph Avruch M.D., Diabetes Research Lab, Department of Molecular Biology, Massachusetts General Hospital, Simches Research Bldg, 6408, 185 Cambridge St., Boston, MA 02114, Tel:617-726-6909, Fax:617-726-5649,
| |
Collapse
|
48
|
Newell P, Toffanin S, Villanueva A, Chiang DY, Minguez B, Cabellos L, Savic R, Hoshida Y, Lim KH, Melgar-Lesmes P, Yea S, Peix J, Deniz K, Fiel MI, Thung S, Alsinet C, Tovar V, Mazzaferro V, Bruix J, Roayaie S, Schwartz M, Friedman SL, Llovet JM. Ras pathway activation in hepatocellular carcinoma and anti-tumoral effect of combined sorafenib and rapamycin in vivo. J Hepatol 2009; 51:725-33. [PMID: 19665249 PMCID: PMC2970800 DOI: 10.1016/j.jhep.2009.03.028] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Revised: 03/04/2009] [Accepted: 03/16/2009] [Indexed: 02/08/2023]
Abstract
BACKGROUND/AIMS The success of sorafenib in the treatment of advanced hepatocellular carcinoma (HCC) has focused interest on the role of Ras signaling in this malignancy. We investigated the molecular alterations of the Ras pathway in HCC and the antineoplastic effects of sorafenib in combination with rapamycin, an inhibitor of mTOR pathway, in experimental models. METHODS Gene expression (qRT-PCR, oligonucleotide microarray), DNA copy number changes (SNP-array), methylation of tumor suppressor genes (methylation-specific PCR) and protein activation (immunohistochemistry) were analysed in 351 samples. Anti-tumoral effects of combined therapy targeting the Ras and mTOR pathways were evaluated in cell lines and HCC xenografts. RESULTS Different mechanisms accounted for Ras pathway activation in HCC. H-ras was up-regulated during different steps of hepatocarcinogenesis. B-raf was overexpressed in advanced tumors and its expression was associated with genomic amplification. Partial methylation of RASSF1A and NORE1A was detected in 89% and 44% of tumors respectively, and complete methylation was found in 11 and 4% of HCCs. Activation of the pathway (pERK immunostaining) was identified in 10.3% of HCC. Blockade of Ras and mTOR pathways with sorafenib and rapamycin reduced cell proliferation and induced apoptosis in cell lines. In vivo, the combination of both compounds enhanced tumor necrosis and ulceration when compared with sorafenib alone. CONCLUSIONS Ras activation results from several molecular alterations, such as methylation of tumor suppressors and amplification of oncogenes (B-raf). Sorafenib blocks signaling and synergizes with rapamycin in vivo, preventing tumor progression. These data provide the rationale for testing this combination in clinical studies.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Benzenesulfonates/administration & dosage
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- DNA Methylation/drug effects
- Drug Synergism
- Female
- Gene Dosage/drug effects
- Genes, ras/drug effects
- Humans
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/genetics
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/pathology
- Mice
- Mice, Nude
- Neoplasm Transplantation
- Niacinamide/analogs & derivatives
- Phenylurea Compounds
- Promoter Regions, Genetic/drug effects
- Protein Kinases/drug effects
- Pyridines/administration & dosage
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Signal Transduction/drug effects
- Sirolimus/administration & dosage
- Sorafenib
- TOR Serine-Threonine Kinases
- Transplantation, Heterologous
- ras Proteins/metabolism
Collapse
Affiliation(s)
- Pippa Newell
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | - Sara Toffanin
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
- Gastrointestinal Surgery and Liver Transplantation Unit, National Cancer Institute, IRCSS Foundation, Milan, Italy
| | - Augusto Villanueva
- BCLC Group [HCC Translational Research Laboratory, Liver Unit, and Department of Pathology], IDIBAPS, CIBERehd, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Derek Y. Chiang
- Department of Medical Oncology and Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA, USA
- Cancer Program, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Beatriz Minguez
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | - Laia Cabellos
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | - Radoslav Savic
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | - Yujin Hoshida
- Cancer Program, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kiat Hon Lim
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | - Pedro Melgar-Lesmes
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | - Steven Yea
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | - Judit Peix
- BCLC Group [HCC Translational Research Laboratory, Liver Unit, and Department of Pathology], IDIBAPS, CIBERehd, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Kemal Deniz
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | - M. Isabel Fiel
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | - Swan Thung
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | - Clara Alsinet
- BCLC Group [HCC Translational Research Laboratory, Liver Unit, and Department of Pathology], IDIBAPS, CIBERehd, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Victoria Tovar
- BCLC Group [HCC Translational Research Laboratory, Liver Unit, and Department of Pathology], IDIBAPS, CIBERehd, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Vincenzo Mazzaferro
- Gastrointestinal Surgery and Liver Transplantation Unit, National Cancer Institute, IRCSS Foundation, Milan, Italy
| | - Jordi Bruix
- BCLC Group [HCC Translational Research Laboratory, Liver Unit, and Department of Pathology], IDIBAPS, CIBERehd, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Sasan Roayaie
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | - Myron Schwartz
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | - Scott L. Friedman
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | - Josep M. Llovet
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases and Hemato/Oncology; Department of Medicine; Surgical Oncology, Department of Surgery; Department of Pathology), Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
- BCLC Group [HCC Translational Research Laboratory, Liver Unit, and Department of Pathology], IDIBAPS, CIBERehd, Hospital Clínic, Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Catalonia, Spain
| |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Over the past decades, advances in the knowledge of the molecular pathogenesis of hepatocellular carcinoma (HCC) have allowed significant improvements in the therapeutic management of this devastating disease. Several investigations have established the role of aberrant activation of major intracellular signaling pathways during human hepatocarcinogenesis. Genome-wide analysis of DNA copy number changes and gene expression led to the identification of gene signatures and novel targets for cancer treatment. Numerous attempts have tried to develop a molecular classification of HCC. This review aims to summarize the most relevant genetic alterations and pathways involved in the development and progression of HCC, providing an overview of the molecular targeted therapies tested so far in human HCC. RECENT FINDINGS The discovery of sorafenib, a multikinase inhibitor, as a treatment with survival benefits in patients with advanced HCC, has become a major breakthrough in the clinical management of HCC. For the first time, a molecular therapy was able to demonstrate significant efficacy for the treatment of HCC patients. New guidelines have established the ideal endpoints for the design of clinical trials for HCC. At last, a molecular classification of HCC based on genome-wide investigations, able to identify patient subclasses according to drug sensitivity will lead to a more personalized medicine. SUMMARY In this review, we provide a comprehensive analysis of the underlying molecular mechanisms leading to human hepatocarcinogenesis, providing the scientific rationale for the development of new therapeutic targets.
Collapse
|
50
|
Gramantieri L, Fornari F, Callegari E, Sabbioni S, Lanza G, Croce CM, Bolondi L, Negrini M. MicroRNA involvement in hepatocellular carcinoma. J Cell Mol Med 2009; 12:2189-204. [PMID: 19120703 PMCID: PMC4514099 DOI: 10.1111/j.1582-4934.2008.00533.x] [Citation(s) in RCA: 212] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the third cause of cancer-related death worldwide. Curative options for HCC are limited and exclusively available for patients carrying an early stage HCC. In advanced stages, traditional chemotherapy proved to be only marginally effective or even toxic. Thus, the identification of new treatment options is needed. New targets for non-conventional treatment will necessarily take advantage of progresses on the molecular pathogenesis of HCC. MicroRNAs (miRNAs) are a group of tiny RNAs with a fundamental role in the regulation of gene expression. Aberrant expression of several miRNAs was found to be involved in human hepatocarcinogenesis. miRNA expression signatures were correlated with bio-pathological and clinical features of HCC. In some cases, aberrantly expressed miRNAs could be linked to cancer-associated pathways, indicating a direct role in liver tumourigenesis. For example, up-regulation of mir-221 and mir-21 could promote cell cycle progression, reduce cell death and favour angiogenesis and invasion. These findings suggest that miRNAs could become novel molecular targets for HCC treatment. The demonstration of in vivo efficacy and safety of anti-miRNA compounds has opened the way to their use in clinical trials.
Collapse
Affiliation(s)
- Laura Gramantieri
- Department of Internal Medicine and Gastroenterology, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | |
Collapse
|