1
|
Ko CJ, Li CJ, Wu MY, Chu PY. Overexpression of epithelial cell adhesion molecule as a predictor of poor outcome in patients with hepatocellular carcinoma. Exp Ther Med 2018; 16:4810-4816. [PMID: 30542436 DOI: 10.3892/etm.2018.6794] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/13/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer growth, metastasis and development are regulated by a number of genes, whose expression mediates important processes, including cellular plasticity, motility and internal interactions in the tumor microenvironment. The epithelial cell adhesion molecule (EpCAM) serves an important role in cell-cell migration and tumorigenicity, particularly metastasis. The aim of the present study was to measure EpCAM expression using immunohistochemistry and to investigate the association between clinicopathological features and prognosis in hepatocellular carcinoma (HCC). The results revealed that EpCAM expression may be a biomarker for poor prognosis in patients with HCC and may therefore be used to predict clinical outcome. The present study suggests that EpCAM expression in HCC can be considered as a routine biomarker for unfavorable prognosis and may provide a basis for the future development of anti-EpCAM-targeted therapy.
Collapse
Affiliation(s)
- Chih-Jan Ko
- Department of General Surgery, Changhua Christian Hospital, Changhua 500, Taiwan, R.O.C.,School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Chia-Jung Li
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan, R.O.C
| | - Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan, R.O.C.,Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan, R.O.C
| | - Pei-Yi Chu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan, R.O.C.,Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan, R.O.C.,National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan, R.O.C
| |
Collapse
|
2
|
Avci ME, Keskus AG, Targen S, Isilak ME, Ozturk M, Atalay RC, Adams MM, Konu O. Development of a novel zebrafish xenograft model in ache mutants using liver cancer cell lines. Sci Rep 2018; 8:1570. [PMID: 29371671 PMCID: PMC5785479 DOI: 10.1038/s41598-018-19817-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/05/2018] [Indexed: 01/09/2023] Open
Abstract
Acetylcholinesterase (AChE), an enzyme responsible for degradation of acetylcholine, has been identified as a prognostic marker in liver cancer. Although in vivo Ache tumorigenicity assays in mouse are present, no established liver cancer xenograft model in zebrafish using an ache mutant background exists. Herein, we developed an embryonic zebrafish xenograft model using epithelial (Hep3B) and mesenchymal (SKHep1) liver cancer cell lines in wild-type and ache sb55 sibling mutant larvae after characterization of cholinesterase expression and activity in cell lines and zebrafish larvae. The comparison of fluorescent signal reflecting tumor size at 3-days post-injection (dpi) revealed an enhanced tumorigenic potential and a reduced migration capacity in cancer cells injected into homozygous ache sb55 mutants when compared with the wild-type. Increased tumor load was confirmed using an ALU based tumor DNA quantification method modified for use in genotyped xenotransplanted zebrafish embryos. Confocal microscopy using the Huh7 cells stably expressing GFP helped identify the distribution of tumor cells in larvae. Our results imply that acetylcholine accumulation in the microenvironment directly or indirectly supports tumor growth in liver cancer. Use of this model system for drug screening studies holds potential in discovering new cholinergic targets for treatment of liver cancers.
Collapse
Affiliation(s)
- M Ender Avci
- Department of Molecular Biology and Genetics, Bilkent University, 06800, Ankara, Turkey.
- Izmir International Biomedicine and Genome Institute (iBG-izmir), Dokuz Eylul University, 35340, Izmir, Turkey.
| | - Ayse Gokce Keskus
- Interdisciplinary Program in Neuroscience, Bilkent University, 06800, Ankara, Turkey
| | - Seniye Targen
- Department of Molecular Biology and Genetics, Bilkent University, 06800, Ankara, Turkey
| | - M Efe Isilak
- Department of Molecular Biology and Genetics, Bilkent University, 06800, Ankara, Turkey
- UNAM-Institute of Materials Science and Nanotechnology, Bilkent University, 06800, Ankara, Turkey
| | - Mehmet Ozturk
- Department of Molecular Biology and Genetics, Bilkent University, 06800, Ankara, Turkey
- Izmir International Biomedicine and Genome Institute (iBG-izmir), Dokuz Eylul University, 35340, Izmir, Turkey
| | - Rengul Cetin Atalay
- Medical Informatics Department, Graduate School of Informatics, Middle East Technical University, 06800, Ankara, Turkey
| | - Michelle M Adams
- Department of Psychology, Bilkent University, 06800, Ankara, Turkey
- Interdisciplinary Program in Neuroscience, Bilkent University, 06800, Ankara, Turkey
- UNAM-Institute of Materials Science and Nanotechnology, Bilkent University, 06800, Ankara, Turkey
| | - Ozlen Konu
- Department of Molecular Biology and Genetics, Bilkent University, 06800, Ankara, Turkey.
- Interdisciplinary Program in Neuroscience, Bilkent University, 06800, Ankara, Turkey.
- UNAM-Institute of Materials Science and Nanotechnology, Bilkent University, 06800, Ankara, Turkey.
| |
Collapse
|
3
|
Lin C, Hu Z, Lei B, Tang B, Yu H, Qiu X, He S. Overexpression of Yes-associated protein and its association with clinicopathological features of hepatocellular carcinoma: A meta-analysis. Liver Int 2017; 37:1675-1681. [PMID: 28345185 PMCID: PMC5697662 DOI: 10.1111/liv.13428] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/18/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Yes-associated protein (YAP) overexpression is reported to be associated with risk of hepatocellular carcinoma (HCC) but current studies have not explored the relationship between YAP expression with HCC clinicopathological features. METHODS To assess these associations, a meta-analysis was performed which included four eligible studies including 391 HCC cases and 334 controls. There were eight eligible studies to investigate the association between YAP expression in HCC and clinicopathological features of liver cancer patients. Literature was obtained from PubMed, Embase, Wangfang and China National Knowledge Infrastructure. RESULTS Analysis indicated that YAP expression in HCC was greater than in adjacent non-tumour tissue (odds ratio [OR], 15.80, 95% confidence interval [CI], 10.53-23.70, P<.00001; heterogeneity=.30). YAP overexpression in HCC was significantly associated with vascular invasion (OR, 2.21, 95% CI, 11.64-2.97, P<.00001, heterogeneity=.10), less cellular differentiation (OR, 2.38, 95% CI, 1.61-3.51, P<.00001, heterogeneity=.333), tumours larger than 5 cm (OR, 2.52, 95% CI, 1.75-3.62, P<.00001; heterogeneity=.17) and TNM tumour stage I + II (OR, 0.44, 95% CI, 0.28-0.75, P=.00003, heterogeneity=.12). CONCLUSIONS Overexpression of YAP contributes to HCC formation, and its overexpression is associated with vascular invasion, low cellular differentiation tumours larger than 5 cm and TNM tumour stage III + IV.
Collapse
Affiliation(s)
- Chengjie Lin
- Department of Hepatopancreatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina,Guangxi Key Laboratory of Molecular Medicine in Liver Injury and RepairAffiliated Guilin Medical UniversityGuilinGuangxiChina
| | - Zhigao Hu
- Department of Hepatopancreatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina,Guangxi Key Laboratory of Molecular Medicine in Liver Injury and RepairAffiliated Guilin Medical UniversityGuilinGuangxiChina
| | - Biao Lei
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and RepairAffiliated Guilin Medical UniversityGuilinGuangxiChina
| | - Bo Tang
- Department of Hepatopancreatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Hongping Yu
- Department of EpidemiologySchool of Public HealthGuangxi Medical UniversityNanningChina
| | - Xiaoqiang Qiu
- Department of EpidemiologySchool of Public HealthGuangxi Medical UniversityNanningChina
| | - Songqing He
- Department of Hepatopancreatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina,Guangxi Key Laboratory of Molecular Medicine in Liver Injury and RepairAffiliated Guilin Medical UniversityGuilinGuangxiChina
| |
Collapse
|
4
|
Jin X, Tian S, Li P. Histone Acetyltransferase 1 Promotes Cell Proliferation and Induces Cisplatin Resistance in Hepatocellular Carcinoma. Oncol Res 2016; 25:939-946. [PMID: 27938492 PMCID: PMC7840994 DOI: 10.3727/096504016x14809827856524] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant diseases in the world. Mutations, overexpression, and improper recruitment of HATs can lead to tumorigenesis. HAT1 is the first histone acetyltransferase identified and is related with developing HCC, but the mechanism is still unclear. Interestingly, we found that HAT1 was upregulated in HCC patient specimens and showed that its upregulation facilitates HCC cell growth in vitro and in vivo. Moreover, we demonstrated that HAT1 promoted glycolysis in HCC cells and knockdown of HAT1 sensitized HCC cells to apoptotic death induced by cisplatin. Our results suggest that HAT1 might act as an oncogenic protein promoting cell proliferation and inducing cisplatin resistance in HCC, and targeting HAT1 represents a viable strategy for effective treatment of advanced HCC.
Collapse
|
5
|
Talarico C, D'Antona L, Scumaci D, Barone A, Gigliotti F, Fiumara CV, Dattilo V, Gallo E, Visca P, Ortuso F, Abbruzzese C, Botta L, Schenone S, Cuda G, Alcaro S, Bianco C, Lavia P, Paggi MG, Perrotti N, Amato R. Preclinical model in HCC: the SGK1 kinase inhibitor SI113 blocks tumor progression in vitro and in vivo and synergizes with radiotherapy. Oncotarget 2016; 6:37511-25. [PMID: 26462020 PMCID: PMC4741945 DOI: 10.18632/oncotarget.5527] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 09/28/2015] [Indexed: 12/20/2022] Open
Abstract
The SGK1 kinase is pivotal in signal transduction pathways operating in cell transformation and tumor progression. Here, we characterize in depth a novel potent and selective pyrazolo[3,4-d]pyrimidine-based SGK1 inhibitor. This compound, named SI113, active in vitro in the sub-micromolar range, inhibits SGK1-dependent signaling in cell lines in a dose- and time-dependent manner. We recently showed that SI113 slows down tumor growth and induces cell death in colon carcinoma cells, when used in monotherapy or in combination with paclitaxel. We now demonstrate for the first time that SI113 inhibits tumour growth in hepatocarcinoma models in vitro and in vivo. SI113-dependent tumor inhibition is dose- and time-dependent. In vitro and in vivo SI113-dependent SGK1 inhibition determined a dramatic increase in apoptosis/necrosis, inhibited cell proliferation and altered the cell cycle profile of treated cells. Proteome-wide biochemical studies confirmed that SI113 down-regulates the abundance of proteins downstream of SGK1 with established roles in neoplastic transformation, e.g. MDM2, NDRG1 and RAN network members. Consistent with knock-down and over-expressing cellular models for SGK1, SI113 potentiated and synergized with radiotherapy in tumor killing. No short-term toxicity was observed in treated animals during in vivo SI113 administration. These data show that direct SGK1 inhibition can be effective in hepatic cancer therapy, either alone or in combination with radiotherapy.
Collapse
Affiliation(s)
- Cristina Talarico
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Lucia D'Antona
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Domenica Scumaci
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Agnese Barone
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Francesco Gigliotti
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Claudia Vincenza Fiumara
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Vincenzo Dattilo
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Enzo Gallo
- Section of Pathology, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Paolo Visca
- Section of Pathology, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Francesco Ortuso
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Claudia Abbruzzese
- Experimental Oncology, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Lorenzo Botta
- Department of Biotecnologie, Chimica e Farmacia, University of Siena, Siena, Italy
| | | | - Giovanni Cuda
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Stefano Alcaro
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Cataldo Bianco
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Patrizia Lavia
- Institute of Molecular Biology and Pathology (IBPM), National Research Council of Italy (CNR), c/o University "La Sapienza", Rome, Italy
| | - Marco G Paggi
- Experimental Oncology, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Nicola Perrotti
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Rosario Amato
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| |
Collapse
|
6
|
Lin M, Huang J, Zhang D, Jiang X, Zhang J, Yu H, Xiao Y, Shi Y, Guo T. Hepatoma-Targeted Radionuclide Immune Albumin Nanospheres: (131)I-antiAFPMcAb-GCV-BSA-NPs. Anal Cell Pathol (Amst) 2016; 2016:9142198. [PMID: 26981334 PMCID: PMC4770115 DOI: 10.1155/2016/9142198] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 01/11/2016] [Indexed: 12/21/2022] Open
Abstract
An effective strategy has been developed for synthesis of radionuclide immune albumin nanospheres ((131)I-antiAFPMcAb-GCV-BSA-NPs). In vitro as well as in vivo targeting of (131)I-antiAFPMcAb-GCV-BSA-NPs to AFP-positive hepatoma was examined. In cultured HepG2 cells, the uptake and retention rates of (131)I-antiAFPMcAb-GCV-BSA-NPs were remarkably higher than those of (131)I alone. As well, the uptake rate and retention ratios of (131)I-antiAFPMcAb-GCV-BSA-NPs in AFP-positive HepG2 cells were also significantly higher than those in AFP-negative HEK293 cells. Compared to (131)I alone, (131)I-antiAFPMcAb-GCV-BSA-NPs were much more easily taken in and retained by hepatoma tissue, with a much higher T/NT. Due to good drug-loading, high encapsulation ratio, and highly selective affinity for AFP-positive tumors, the (131)I-antiAFPMcAb-GCV-BSA-NPs are promising for further effective radiation-gene therapy of hepatoma.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/therapy
- Chemoradiotherapy/methods
- Drug Delivery Systems/methods
- Female
- Ganciclovir/administration & dosage
- Ganciclovir/therapeutic use
- Genetic Therapy/methods
- HEK293 Cells
- Hep G2 Cells
- Humans
- Iodine Radioisotopes/administration & dosage
- Iodine Radioisotopes/pharmacokinetics
- Iodine Radioisotopes/therapeutic use
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/therapy
- Mice, Inbred BALB C
- Mice, Nude
- Microscopy, Electron, Transmission
- Nanospheres/administration & dosage
- Nanospheres/chemistry
- Nanospheres/ultrastructure
- Serum Albumin, Bovine/chemistry
- Tissue Distribution
- Xenograft Model Antitumor Assays
- alpha-Fetoproteins/genetics
- alpha-Fetoproteins/immunology
Collapse
Affiliation(s)
- Mei Lin
- Clinical Medical Institute, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu 225300, China
- Medical School, Southeast University, Nanjing, Jiangsu 210009, China
| | - Junxing Huang
- Clinical Medical Institute, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu 225300, China
| | - Dongsheng Zhang
- Medical School, Southeast University, Nanjing, Jiangsu 210009, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Nanjing, Jiangsu 210009, China
| | - Xingmao Jiang
- Key Laboratory of Advanced Catalytic Material and Technology, Changzhou University, Changzhou, Jiangsu 213000, China
| | - Jia Zhang
- Medical School, Southeast University, Nanjing, Jiangsu 210009, China
| | - Hong Yu
- Clinical Medical Institute, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu 225300, China
| | - Yanhong Xiao
- Clinical Medical Institute, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu 225300, China
| | - Yujuan Shi
- Clinical Medical Institute, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu 225300, China
| | - Ting Guo
- Clinical Medical Institute, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu 225300, China
| |
Collapse
|
7
|
Rebbani K, Marchio A, Ezzikouri S, Afifi R, Kandil M, Bahri O, Triki H, El Feydi AE, Dejean A, Benjelloun S, Pineau P. TP53 R72P polymorphism modulates DNA methylation in hepatocellular carcinoma. Mol Cancer 2015; 14:74. [PMID: 25889455 PMCID: PMC4393630 DOI: 10.1186/s12943-015-0340-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 03/11/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is characterized by widespread epidemiological and molecular heterogeneity. Previous work showed that in the western part of North Africa, a region of low incidence of HCC, mutations are scarce for this tumor type. As epigenetic changes are considered possible surrogates to mutations in human cancers, we decided, thus, to characterize DNA methylation in HCC from North-African patients. METHODS A set of 11 loci was investigated in a series of 45 tumor specimens using methylation-specific and combined-bisulfite restriction assay PCR. Results obtained on clinical samples were subsequently validated in liver cancer cell lines. RESULTS DNA methylation at tumor suppressor loci is significantly higher in samples displaying chromosome instability. More importantly, DNA methylation was significantly higher in Arg/Arg when compared to Pro/Pro genotype carriers at codon 72 rs1042522 of TP53 (65% vs 20% methylated loci, p = 0.0006), a polymorphism already known to affect somatic mutation rate in human carcinomas. In vitro experiments in cell lines indicated that enzymes controlling DNA methylation were differentially regulated by codon 72 Arg or Pro isoforms of p53. Furthermore, the Arg72-carrying version of p53 was shown to re-methylate DNA more rapidly than the pro-harboring isoform. Finally, Pro-carrying cell lines were shown to be significantly more resistant to decitabine treatment (two-fold, p = 0.005). CONCLUSIONS Our data suggest that Arg72Pro polymorphism in a WT p53 context may act as a primary driver of epigenetic changes in HCC. It suggests, in addition, that rs1042522 genotype may predict sensitivity to epigenetic-targeted therapy. This model of liver tumorigenesis that associates low penetrance genetic predisposition to epigenetic changes emerges from a region of low HCC incidence and it may, therefore, apply essentially to population living in similar areas. Surveys on populations submitted to highly mutagenic conditions as perinatally-acquired chronic hepatitis B or aflatoxin B1 exposure remained to be conducted to validate our observations as a general model.
Collapse
Affiliation(s)
- Khadija Rebbani
- Unité d'Organisation Nucléaire et Oncogenèse, INSERM U993, Institut Pasteur, 28 rue du Docteur Roux, F-75724, Paris, Cedex 15, France. .,Laboratoire des Hépatites Virales, Institut Pasteur du Maroc, 1 Place Louis Pasteur, 20360, Casablanca, Morocco.
| | - Agnès Marchio
- Unité d'Organisation Nucléaire et Oncogenèse, INSERM U993, Institut Pasteur, 28 rue du Docteur Roux, F-75724, Paris, Cedex 15, France.
| | - Sayeh Ezzikouri
- Laboratoire des Hépatites Virales, Institut Pasteur du Maroc, 1 Place Louis Pasteur, 20360, Casablanca, Morocco.
| | - Rajaa Afifi
- Service de Médecine C-Gastroentérologie, CHU Ibn-Sina, Rabat, Morocco.
| | - Mostafa Kandil
- Equipe d'Anthropogénétique et de Biotechnologies, Faculté des Sciences Chouaib Doukkali, El Jadida, Morocco.
| | - Olfa Bahri
- Laboratoire de Virologie Clinique, Institut Pasteur de Tunis, Tunis, Tunisie.
| | - Henda Triki
- Laboratoire de Virologie Clinique, Institut Pasteur de Tunis, Tunis, Tunisie.
| | | | - Anne Dejean
- Unité d'Organisation Nucléaire et Oncogenèse, INSERM U993, Institut Pasteur, 28 rue du Docteur Roux, F-75724, Paris, Cedex 15, France.
| | - Soumaya Benjelloun
- Laboratoire des Hépatites Virales, Institut Pasteur du Maroc, 1 Place Louis Pasteur, 20360, Casablanca, Morocco.
| | - Pascal Pineau
- Unité d'Organisation Nucléaire et Oncogenèse, INSERM U993, Institut Pasteur, 28 rue du Docteur Roux, F-75724, Paris, Cedex 15, France.
| |
Collapse
|
8
|
Zhu AX, Rosmorduc O, Evans TRJ, Ross PJ, Santoro A, Carrilho FJ, Bruix J, Qin S, Thuluvath PJ, Llovet JM, Leberre MA, Jensen M, Meinhardt G, Kang YK. SEARCH: a phase III, randomized, double-blind, placebo-controlled trial of sorafenib plus erlotinib in patients with advanced hepatocellular carcinoma. J Clin Oncol 2015; 33:559-66. [PMID: 25547503 DOI: 10.1200/jco.2013.53.7746] [Citation(s) in RCA: 409] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To compare the clinical outcomes of sorafenib plus either erlotinib or placebo in patients with advanced hepatocellular carcinoma (HCC) in a multicenter, multinational, randomized, phase III trial. PATIENTS AND METHODS Patients with advanced HCC and underlying Child-Pugh class A cirrhosis, who were naive to systemic treatment (N = 720), were randomly assigned to sorafenib plus either erlotinib (n = 362) or placebo (n = 358). The primary end point was overall survival (OS). RESULTS Median OS was similar in the sorafenib plus erlotinib and sorafenib plus placebo groups (9.5 v 8.5 months, respectively; hazard ratio [HR], 0.929; P = .408), as was median time to progression (3.2 v 4.0 months, respectively; HR, 1.135; P = .18). In the sorafenib/erlotinib arm versus the sorafenib/placebo arm, the overall response rate trended higher (6.6% v 3.9%, respectively; P = .102), whereas the disease control rate was significantly lower (43.9% v 52.5%, respectively; P = .021). The median durations of treatment with sorafenib were 86 days in the sorafenib/erlotinib arm and 123 days in the sorafenib/placebo arm. In the sorafenib/erlotinib and sorafenib/placebo arms, the rates of treatment-emergent serious AEs (58.0% v 54.6%, respectively) and drug-related serious AEs (21.0% v 22.8%, respectively) were similar. AEs matched the known safety profiles of both agents, but rates of rash/desquamation, anorexia, and diarrhea were higher in the sorafenib/erlotinib arm, whereas rates of alopecia and hand-foot skin reaction were higher in the sorafenib/placebo arm. Withdrawal rates for AEs during cycles 1 to 3 were higher in the sorafenib/erlotinib arm. CONCLUSION Adding erlotinib to sorafenib did not improve survival in patients with advanced HCC.
Collapse
Affiliation(s)
- Andrew X Zhu
- Andrew X. Zhu, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Olivier Rosmorduc, Service d'Hépatologie, Hôpital Saint-Antoine, Paris; Marie-Aude Leberre, Bayer HealthCare Pharmaceuticals, Loos, France; T.R. Jeffry Evans, Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow; Paul J. Ross, King's College Hospital, London, United Kingdom; Armando Santoro, Humanitas Cancer Center, Milan, Italy; Flair Jose Carrilho, University of São Paulo School of Medicine, São Paulo, Brazil; Jordi Bruix and Josep M. Llovet, Barcelona Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Hospital Clínic Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona; Josep M. Llovet, Institució Catalana de Recerca I Estudis Avançats, Catalonia, Spain; Shukui Qin, People's Liberation Army Cancer Center of Nanjing Bayi Hospital, Jiangsu, China; Paul J. Thuluvath, Institute for Digestive Health and Liver Diseases, Mercy Medical Center, Baltimore, MD; Josep M. Llovet, Mount Sinai Liver Cancer Program, Mount Sinai School of Medicine, New York, NY; Markus Jensen, Bayer Vital GmbH, Leverkusen, Germany; Gerold Meinhardt, Bayer HealthCare Pharmaceuticals, Montville, NJ; and Yoon-Koo Kang, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea.
| | - Olivier Rosmorduc
- Andrew X. Zhu, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Olivier Rosmorduc, Service d'Hépatologie, Hôpital Saint-Antoine, Paris; Marie-Aude Leberre, Bayer HealthCare Pharmaceuticals, Loos, France; T.R. Jeffry Evans, Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow; Paul J. Ross, King's College Hospital, London, United Kingdom; Armando Santoro, Humanitas Cancer Center, Milan, Italy; Flair Jose Carrilho, University of São Paulo School of Medicine, São Paulo, Brazil; Jordi Bruix and Josep M. Llovet, Barcelona Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Hospital Clínic Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona; Josep M. Llovet, Institució Catalana de Recerca I Estudis Avançats, Catalonia, Spain; Shukui Qin, People's Liberation Army Cancer Center of Nanjing Bayi Hospital, Jiangsu, China; Paul J. Thuluvath, Institute for Digestive Health and Liver Diseases, Mercy Medical Center, Baltimore, MD; Josep M. Llovet, Mount Sinai Liver Cancer Program, Mount Sinai School of Medicine, New York, NY; Markus Jensen, Bayer Vital GmbH, Leverkusen, Germany; Gerold Meinhardt, Bayer HealthCare Pharmaceuticals, Montville, NJ; and Yoon-Koo Kang, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - T R Jeffry Evans
- Andrew X. Zhu, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Olivier Rosmorduc, Service d'Hépatologie, Hôpital Saint-Antoine, Paris; Marie-Aude Leberre, Bayer HealthCare Pharmaceuticals, Loos, France; T.R. Jeffry Evans, Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow; Paul J. Ross, King's College Hospital, London, United Kingdom; Armando Santoro, Humanitas Cancer Center, Milan, Italy; Flair Jose Carrilho, University of São Paulo School of Medicine, São Paulo, Brazil; Jordi Bruix and Josep M. Llovet, Barcelona Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Hospital Clínic Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona; Josep M. Llovet, Institució Catalana de Recerca I Estudis Avançats, Catalonia, Spain; Shukui Qin, People's Liberation Army Cancer Center of Nanjing Bayi Hospital, Jiangsu, China; Paul J. Thuluvath, Institute for Digestive Health and Liver Diseases, Mercy Medical Center, Baltimore, MD; Josep M. Llovet, Mount Sinai Liver Cancer Program, Mount Sinai School of Medicine, New York, NY; Markus Jensen, Bayer Vital GmbH, Leverkusen, Germany; Gerold Meinhardt, Bayer HealthCare Pharmaceuticals, Montville, NJ; and Yoon-Koo Kang, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Paul J Ross
- Andrew X. Zhu, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Olivier Rosmorduc, Service d'Hépatologie, Hôpital Saint-Antoine, Paris; Marie-Aude Leberre, Bayer HealthCare Pharmaceuticals, Loos, France; T.R. Jeffry Evans, Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow; Paul J. Ross, King's College Hospital, London, United Kingdom; Armando Santoro, Humanitas Cancer Center, Milan, Italy; Flair Jose Carrilho, University of São Paulo School of Medicine, São Paulo, Brazil; Jordi Bruix and Josep M. Llovet, Barcelona Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Hospital Clínic Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona; Josep M. Llovet, Institució Catalana de Recerca I Estudis Avançats, Catalonia, Spain; Shukui Qin, People's Liberation Army Cancer Center of Nanjing Bayi Hospital, Jiangsu, China; Paul J. Thuluvath, Institute for Digestive Health and Liver Diseases, Mercy Medical Center, Baltimore, MD; Josep M. Llovet, Mount Sinai Liver Cancer Program, Mount Sinai School of Medicine, New York, NY; Markus Jensen, Bayer Vital GmbH, Leverkusen, Germany; Gerold Meinhardt, Bayer HealthCare Pharmaceuticals, Montville, NJ; and Yoon-Koo Kang, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Armando Santoro
- Andrew X. Zhu, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Olivier Rosmorduc, Service d'Hépatologie, Hôpital Saint-Antoine, Paris; Marie-Aude Leberre, Bayer HealthCare Pharmaceuticals, Loos, France; T.R. Jeffry Evans, Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow; Paul J. Ross, King's College Hospital, London, United Kingdom; Armando Santoro, Humanitas Cancer Center, Milan, Italy; Flair Jose Carrilho, University of São Paulo School of Medicine, São Paulo, Brazil; Jordi Bruix and Josep M. Llovet, Barcelona Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Hospital Clínic Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona; Josep M. Llovet, Institució Catalana de Recerca I Estudis Avançats, Catalonia, Spain; Shukui Qin, People's Liberation Army Cancer Center of Nanjing Bayi Hospital, Jiangsu, China; Paul J. Thuluvath, Institute for Digestive Health and Liver Diseases, Mercy Medical Center, Baltimore, MD; Josep M. Llovet, Mount Sinai Liver Cancer Program, Mount Sinai School of Medicine, New York, NY; Markus Jensen, Bayer Vital GmbH, Leverkusen, Germany; Gerold Meinhardt, Bayer HealthCare Pharmaceuticals, Montville, NJ; and Yoon-Koo Kang, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Flair Jose Carrilho
- Andrew X. Zhu, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Olivier Rosmorduc, Service d'Hépatologie, Hôpital Saint-Antoine, Paris; Marie-Aude Leberre, Bayer HealthCare Pharmaceuticals, Loos, France; T.R. Jeffry Evans, Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow; Paul J. Ross, King's College Hospital, London, United Kingdom; Armando Santoro, Humanitas Cancer Center, Milan, Italy; Flair Jose Carrilho, University of São Paulo School of Medicine, São Paulo, Brazil; Jordi Bruix and Josep M. Llovet, Barcelona Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Hospital Clínic Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona; Josep M. Llovet, Institució Catalana de Recerca I Estudis Avançats, Catalonia, Spain; Shukui Qin, People's Liberation Army Cancer Center of Nanjing Bayi Hospital, Jiangsu, China; Paul J. Thuluvath, Institute for Digestive Health and Liver Diseases, Mercy Medical Center, Baltimore, MD; Josep M. Llovet, Mount Sinai Liver Cancer Program, Mount Sinai School of Medicine, New York, NY; Markus Jensen, Bayer Vital GmbH, Leverkusen, Germany; Gerold Meinhardt, Bayer HealthCare Pharmaceuticals, Montville, NJ; and Yoon-Koo Kang, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jordi Bruix
- Andrew X. Zhu, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Olivier Rosmorduc, Service d'Hépatologie, Hôpital Saint-Antoine, Paris; Marie-Aude Leberre, Bayer HealthCare Pharmaceuticals, Loos, France; T.R. Jeffry Evans, Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow; Paul J. Ross, King's College Hospital, London, United Kingdom; Armando Santoro, Humanitas Cancer Center, Milan, Italy; Flair Jose Carrilho, University of São Paulo School of Medicine, São Paulo, Brazil; Jordi Bruix and Josep M. Llovet, Barcelona Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Hospital Clínic Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona; Josep M. Llovet, Institució Catalana de Recerca I Estudis Avançats, Catalonia, Spain; Shukui Qin, People's Liberation Army Cancer Center of Nanjing Bayi Hospital, Jiangsu, China; Paul J. Thuluvath, Institute for Digestive Health and Liver Diseases, Mercy Medical Center, Baltimore, MD; Josep M. Llovet, Mount Sinai Liver Cancer Program, Mount Sinai School of Medicine, New York, NY; Markus Jensen, Bayer Vital GmbH, Leverkusen, Germany; Gerold Meinhardt, Bayer HealthCare Pharmaceuticals, Montville, NJ; and Yoon-Koo Kang, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Shukui Qin
- Andrew X. Zhu, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Olivier Rosmorduc, Service d'Hépatologie, Hôpital Saint-Antoine, Paris; Marie-Aude Leberre, Bayer HealthCare Pharmaceuticals, Loos, France; T.R. Jeffry Evans, Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow; Paul J. Ross, King's College Hospital, London, United Kingdom; Armando Santoro, Humanitas Cancer Center, Milan, Italy; Flair Jose Carrilho, University of São Paulo School of Medicine, São Paulo, Brazil; Jordi Bruix and Josep M. Llovet, Barcelona Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Hospital Clínic Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona; Josep M. Llovet, Institució Catalana de Recerca I Estudis Avançats, Catalonia, Spain; Shukui Qin, People's Liberation Army Cancer Center of Nanjing Bayi Hospital, Jiangsu, China; Paul J. Thuluvath, Institute for Digestive Health and Liver Diseases, Mercy Medical Center, Baltimore, MD; Josep M. Llovet, Mount Sinai Liver Cancer Program, Mount Sinai School of Medicine, New York, NY; Markus Jensen, Bayer Vital GmbH, Leverkusen, Germany; Gerold Meinhardt, Bayer HealthCare Pharmaceuticals, Montville, NJ; and Yoon-Koo Kang, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Paul J Thuluvath
- Andrew X. Zhu, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Olivier Rosmorduc, Service d'Hépatologie, Hôpital Saint-Antoine, Paris; Marie-Aude Leberre, Bayer HealthCare Pharmaceuticals, Loos, France; T.R. Jeffry Evans, Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow; Paul J. Ross, King's College Hospital, London, United Kingdom; Armando Santoro, Humanitas Cancer Center, Milan, Italy; Flair Jose Carrilho, University of São Paulo School of Medicine, São Paulo, Brazil; Jordi Bruix and Josep M. Llovet, Barcelona Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Hospital Clínic Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona; Josep M. Llovet, Institució Catalana de Recerca I Estudis Avançats, Catalonia, Spain; Shukui Qin, People's Liberation Army Cancer Center of Nanjing Bayi Hospital, Jiangsu, China; Paul J. Thuluvath, Institute for Digestive Health and Liver Diseases, Mercy Medical Center, Baltimore, MD; Josep M. Llovet, Mount Sinai Liver Cancer Program, Mount Sinai School of Medicine, New York, NY; Markus Jensen, Bayer Vital GmbH, Leverkusen, Germany; Gerold Meinhardt, Bayer HealthCare Pharmaceuticals, Montville, NJ; and Yoon-Koo Kang, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Josep M Llovet
- Andrew X. Zhu, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Olivier Rosmorduc, Service d'Hépatologie, Hôpital Saint-Antoine, Paris; Marie-Aude Leberre, Bayer HealthCare Pharmaceuticals, Loos, France; T.R. Jeffry Evans, Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow; Paul J. Ross, King's College Hospital, London, United Kingdom; Armando Santoro, Humanitas Cancer Center, Milan, Italy; Flair Jose Carrilho, University of São Paulo School of Medicine, São Paulo, Brazil; Jordi Bruix and Josep M. Llovet, Barcelona Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Hospital Clínic Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona; Josep M. Llovet, Institució Catalana de Recerca I Estudis Avançats, Catalonia, Spain; Shukui Qin, People's Liberation Army Cancer Center of Nanjing Bayi Hospital, Jiangsu, China; Paul J. Thuluvath, Institute for Digestive Health and Liver Diseases, Mercy Medical Center, Baltimore, MD; Josep M. Llovet, Mount Sinai Liver Cancer Program, Mount Sinai School of Medicine, New York, NY; Markus Jensen, Bayer Vital GmbH, Leverkusen, Germany; Gerold Meinhardt, Bayer HealthCare Pharmaceuticals, Montville, NJ; and Yoon-Koo Kang, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Marie-Aude Leberre
- Andrew X. Zhu, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Olivier Rosmorduc, Service d'Hépatologie, Hôpital Saint-Antoine, Paris; Marie-Aude Leberre, Bayer HealthCare Pharmaceuticals, Loos, France; T.R. Jeffry Evans, Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow; Paul J. Ross, King's College Hospital, London, United Kingdom; Armando Santoro, Humanitas Cancer Center, Milan, Italy; Flair Jose Carrilho, University of São Paulo School of Medicine, São Paulo, Brazil; Jordi Bruix and Josep M. Llovet, Barcelona Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Hospital Clínic Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona; Josep M. Llovet, Institució Catalana de Recerca I Estudis Avançats, Catalonia, Spain; Shukui Qin, People's Liberation Army Cancer Center of Nanjing Bayi Hospital, Jiangsu, China; Paul J. Thuluvath, Institute for Digestive Health and Liver Diseases, Mercy Medical Center, Baltimore, MD; Josep M. Llovet, Mount Sinai Liver Cancer Program, Mount Sinai School of Medicine, New York, NY; Markus Jensen, Bayer Vital GmbH, Leverkusen, Germany; Gerold Meinhardt, Bayer HealthCare Pharmaceuticals, Montville, NJ; and Yoon-Koo Kang, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Markus Jensen
- Andrew X. Zhu, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Olivier Rosmorduc, Service d'Hépatologie, Hôpital Saint-Antoine, Paris; Marie-Aude Leberre, Bayer HealthCare Pharmaceuticals, Loos, France; T.R. Jeffry Evans, Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow; Paul J. Ross, King's College Hospital, London, United Kingdom; Armando Santoro, Humanitas Cancer Center, Milan, Italy; Flair Jose Carrilho, University of São Paulo School of Medicine, São Paulo, Brazil; Jordi Bruix and Josep M. Llovet, Barcelona Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Hospital Clínic Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona; Josep M. Llovet, Institució Catalana de Recerca I Estudis Avançats, Catalonia, Spain; Shukui Qin, People's Liberation Army Cancer Center of Nanjing Bayi Hospital, Jiangsu, China; Paul J. Thuluvath, Institute for Digestive Health and Liver Diseases, Mercy Medical Center, Baltimore, MD; Josep M. Llovet, Mount Sinai Liver Cancer Program, Mount Sinai School of Medicine, New York, NY; Markus Jensen, Bayer Vital GmbH, Leverkusen, Germany; Gerold Meinhardt, Bayer HealthCare Pharmaceuticals, Montville, NJ; and Yoon-Koo Kang, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Gerold Meinhardt
- Andrew X. Zhu, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Olivier Rosmorduc, Service d'Hépatologie, Hôpital Saint-Antoine, Paris; Marie-Aude Leberre, Bayer HealthCare Pharmaceuticals, Loos, France; T.R. Jeffry Evans, Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow; Paul J. Ross, King's College Hospital, London, United Kingdom; Armando Santoro, Humanitas Cancer Center, Milan, Italy; Flair Jose Carrilho, University of São Paulo School of Medicine, São Paulo, Brazil; Jordi Bruix and Josep M. Llovet, Barcelona Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Hospital Clínic Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona; Josep M. Llovet, Institució Catalana de Recerca I Estudis Avançats, Catalonia, Spain; Shukui Qin, People's Liberation Army Cancer Center of Nanjing Bayi Hospital, Jiangsu, China; Paul J. Thuluvath, Institute for Digestive Health and Liver Diseases, Mercy Medical Center, Baltimore, MD; Josep M. Llovet, Mount Sinai Liver Cancer Program, Mount Sinai School of Medicine, New York, NY; Markus Jensen, Bayer Vital GmbH, Leverkusen, Germany; Gerold Meinhardt, Bayer HealthCare Pharmaceuticals, Montville, NJ; and Yoon-Koo Kang, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Yoon-Koo Kang
- Andrew X. Zhu, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Olivier Rosmorduc, Service d'Hépatologie, Hôpital Saint-Antoine, Paris; Marie-Aude Leberre, Bayer HealthCare Pharmaceuticals, Loos, France; T.R. Jeffry Evans, Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow; Paul J. Ross, King's College Hospital, London, United Kingdom; Armando Santoro, Humanitas Cancer Center, Milan, Italy; Flair Jose Carrilho, University of São Paulo School of Medicine, São Paulo, Brazil; Jordi Bruix and Josep M. Llovet, Barcelona Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Hospital Clínic Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona; Josep M. Llovet, Institució Catalana de Recerca I Estudis Avançats, Catalonia, Spain; Shukui Qin, People's Liberation Army Cancer Center of Nanjing Bayi Hospital, Jiangsu, China; Paul J. Thuluvath, Institute for Digestive Health and Liver Diseases, Mercy Medical Center, Baltimore, MD; Josep M. Llovet, Mount Sinai Liver Cancer Program, Mount Sinai School of Medicine, New York, NY; Markus Jensen, Bayer Vital GmbH, Leverkusen, Germany; Gerold Meinhardt, Bayer HealthCare Pharmaceuticals, Montville, NJ; and Yoon-Koo Kang, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
9
|
Yao M, Wang L, Yao Y, Gu HB, Yao DF. Biomarker-based MicroRNA Therapeutic Strategies for Hepatocellular Carcinoma. J Clin Transl Hepatol 2014. [PMID: 26355266 DOI: 10.14218/jcth.2014.0002026355266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Recently, microRNAs (miRNAs) have emerged as key factors involved in a series of biological processes, ranging from embryogenesis to programmed cell death. Its link to aberrant expression profiles has rendered it a potentially attractive tool for the diagnosis, prognosis, or treatment of various diseases. Accumulating evidence has indicated that miRNAs act as tumor suppressors in hepatocyte malignant transformation by regulating development, differentiation, proliferation, and tumorigenesis. Here, we summarize recent progress in the development of novel biomarker-based miRNA therapeutic strategies for hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
- Min Yao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China ; Department of Immunology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Li Wang
- Department of Medical Informatics, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Yao Yao
- The Hospital of Nantong Maternal and Child Care Service, Nantong, Jiangsu, China
| | - Hong-Bing Gu
- The Hospital of Nantong Maternal and Child Care Service, Nantong, Jiangsu, China
| | - Deng-Fu Yao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
10
|
Yao M, Wang L, Yao Y, Gu HB, Yao DF. Biomarker-based MicroRNA Therapeutic Strategies for Hepatocellular Carcinoma. J Clin Transl Hepatol 2014; 2:253-258. [PMID: 26355266 PMCID: PMC4521238 DOI: 10.14218/jcth.2014.00020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 09/15/2014] [Accepted: 09/16/2014] [Indexed: 02/07/2023] Open
Abstract
Recently, microRNAs (miRNAs) have emerged as key factors involved in a series of biological processes, ranging from embryogenesis to programmed cell death. Its link to aberrant expression profiles has rendered it a potentially attractive tool for the diagnosis, prognosis, or treatment of various diseases. Accumulating evidence has indicated that miRNAs act as tumor suppressors in hepatocyte malignant transformation by regulating development, differentiation, proliferation, and tumorigenesis. Here, we summarize recent progress in the development of novel biomarker-based miRNA therapeutic strategies for hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
- Min Yao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Department of Immunology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Li Wang
- Department of Medical Informatics, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Yao Yao
- The Hospital of Nantong Maternal and Child Care Service, Nantong, Jiangsu, China
| | - Hong-Bing Gu
- The Hospital of Nantong Maternal and Child Care Service, Nantong, Jiangsu, China
| | - Deng-Fu Yao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
11
|
ZHANG ZHENHAI, WU YAGUANG, QIN CHENGKUN, RONG ZHONGHOU, SU ZHONGXUE, XIAN GUOZHE. Stanniocalcin 2 expression predicts poor prognosis of hepatocellular carcinoma. Oncol Lett 2014; 8:2160-2164. [PMID: 25289096 PMCID: PMC4186577 DOI: 10.3892/ol.2014.2520] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 08/12/2014] [Indexed: 12/14/2022] Open
Abstract
Previous studies have shown that the expression level of stanniocalcin 2 (STC2) is associated with tumor progression. However, to date, the association between STC2 and clinicopathological factors in hepatocellular carcinoma (HCC) has not been investigated. The clinical significance of STC2 was investigated in 30 fresh HCC samples using western blot analysis and in 240 HCC tissues using immunohistochemical analysis. The level of STC2 in cancerous tissue was higher than in the matched non-cancerous tissues. Using immunohistochemistry, the STC2-positive group exhibited a higher incidence of lymph node metastasis and venous invasion compared with the STC2-negative group. Kaplan-Meier survival analysis revealed that the positive expression of STC2 correlated with poor overall survival (OS) and disease-free survival of HCC patients (P<0.01). STC2 expression was observed to be an independent prognostic factor for OS in HCC patients by multivariate analysis (hazard ratio, 2.39; 95% confidence interval, 1.04-5.89; P=0.013). These data suggest that STC2 expression may be a useful indicator of poor prognosis in HCC patients.
Collapse
Affiliation(s)
- ZHEN-HAI ZHANG
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - YA-GUANG WU
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - CHENG-KUN QIN
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - ZHONG-HOU RONG
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - ZHONG-XUE SU
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - GUO-ZHE XIAN
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
12
|
Li Z, Zeng Y, Zhang D, Wu M, Wu L, Huang A, Yang H, Liu X, Liu J. Glypican-3 antibody functionalized Prussian blue nanoparticles for targeted MR imaging and photothermal therapy of hepatocellular carcinoma. J Mater Chem B 2014; 2:3686-3696. [PMID: 32263805 DOI: 10.1039/c4tb00516c] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
MRI-guided photothermal therapy is becoming a more widely accepted minimally invasive technique. In this study, glypican-3 monoclonal antibody functionalized Prussian blue nanoparticles (antiGPC3-PBNPs) were developed as a novel theranostic agent for the targeted MR imaging and photothermal therapy of hepatocellular carcinoma. The physical properties of the antiGPC3-PBNPs were characterized by SEM, TEM and Vis-NIR absorption spectra, which showed that the developed nanoprobe formed well defined nanocubes with an average diameter of 21 nm. The significantly increased targeting cellular uptake efficiency in HepG2 cells via receptor-mediated endocytosis was confirmed by confocal fluorescence microscopy and ICP-MS. Furthermore, the high photothermal cytotoxicity, excellent MR imaging contrast enhancement ability and biocompatibility of the developed nanoprobe were also confirmed. Hence, the developed antiGPC3-PBNPs could be used as a promising nanoprobe for targeted MRI diagnosis and efficient photothermal therapy for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Zhenglin Li
- Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Glypican-3 as an emerging molecular target for hepatocellular carcinoma gene therapy. Tumour Biol 2014; 35:5857-68. [PMID: 24633918 DOI: 10.1007/s13277-014-1776-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 02/18/2014] [Indexed: 02/07/2023] Open
Abstract
Glypican-3 (GPC-3), a membrane-associated heparan sulfate proteoglycan, plays a crucial role in cell proliferation and metastasis, particularly in hepatocellular carcinoma (HCC) progression, and perhaps is a valuable target for its gene therapy. However, its mechanism remains to be explored. In the present study, the biological behaviors of HCC cells were investigated by interfering GPC-3 gene transcription. After the cells were transfected with specific GPC-3 short hairpin RNA (shRNA), the inhibition of GPC-3 expression was 75.6 % in MHCC-97H or 73.8 % in Huh7 cells at mRNA level; the rates of proliferation and apoptosis were 53.6 and 60.5 % in MHCC-97H or 54.9 and 54.4 % in Huh7 cells, with the cell cycles arrested in the G1 phase; the incidences of cell migration, metastasis, and invasion inhibition were 80.1, 56.4, and 69.1 % in MHCC-97H or 80.9, 59.6, and 58.3 % in Huh7 cells, respectively. The cell biological behaviors were altered by silencing GPC-3 with down-regulation of β-catenin, insulin-like growth factor-II and vascular endothelial growth factor, and Gli1 up-regulation. The cell proliferation was significantly inhibited (up to 95.11 %) by shRNA plus anti-cancer drugs, suggesting that GPC-3 gene should be a potential target for promoting hepatoma cell apoptosis and inhibiting metastasis through the Wnt/β-catenin and Hh singling pathways.
Collapse
|
14
|
Upregulation of microRNA-25 associates with prognosis in hepatocellular carcinoma. Diagn Pathol 2014; 9:47. [PMID: 24593846 PMCID: PMC4016611 DOI: 10.1186/1746-1596-9-47] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/10/2014] [Indexed: 12/31/2022] Open
Abstract
Background Accumulating evidence has shown that up-regulation of microRNA-25(miR-25) is associated with the prognosis of several types of human malignant solid tumors. However, whether miR-25 expression has influence on the prognosis of hepatocellular carcinoma (HCC) is still unknown. Methods The differentially expressed amount of the miR-25 was validated in triplicate by quantitative reverse-transcription polymerase chain reaction (qRT-PCR). Survival rate was analyzed by log-rank test, and survival curves were plotted according to Kaplan–Meier. Multivariate analysis of the prognostic factors was performed with Cox regression model. Results The expression of miR-25 was significantly upregulated in HCC tissues when compared with adjacent normal tissues (p<0.0001). Patients who had high miR-25 expression had a shorter overall survival than patients who had low miR-25 expression (median overall survival, 31.0 months versus 42.9 months, p=0.0192). The multivariate Cox regression analysis indicated that miR-25 expression (HR=2.179; p=0.001), TNM stage (HR=1.782; p=0.014), and vein invasion (HR=1.624; p=0.020) were independent prognostic factors for overall survival. Conclusion Our data suggests that the overexpression of miR-25 in HCC tissues is of predictive value on poor prognosis. Virtual slide The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/1989618421114309
Collapse
|
15
|
DONG ZHIZHEN, YAO MIN, ZHANG HAIJIAN, WANG LI, HUANG HUA, YAN MEIJUAN, WU WEI, YAO DENGFU. Inhibition of Annexin A2 gene transcription is a promising molecular target for hepatoma cell proliferation and metastasis. Oncol Lett 2014; 7:28-34. [PMID: 24348815 PMCID: PMC3861549 DOI: 10.3892/ol.2013.1663] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 10/29/2013] [Indexed: 01/07/2023] Open
Abstract
Hepatocyte Annexin A2 (ANXA2) expression is associated with the progression and metastasis of hepatocellular carcinoma (HCC). Circulating ANXA2 levels in HCC patients are significantly higher compared with that of patients with benign liver disease. ANXA2 levels have been found to correlate with hepatitis B virus infection, extrahepatic metastasis and portal vein thrombus. By contrast, ANXA2 levels do not correlate with tumour size and AFP levels. However, the underlying mechanisms of ANXA2 remain obscure. The results of the current study identified that abnormalities in hepatic ANXA2 expression were localised to the cell membrane and cytoplasm of HCC tissues and mainly in the cytoplasm of para-cancerous tissues. ANXA2 was overexpressed in MHCC97-H cells which have high metastatic potential. Following specific ANXA2-small hairpin RNA (shRNA) transfection in vitro, ANXA-2 was effectively inhibited and the S phase ratio of cells was 27.76%, compared with 36.14% in mock-treated cells. In addition, the invading cell ratio was reduced in the shRNA-treated group (52.16%) compared with the mock-treated group (86.14%). The growth and volume of xenograft tumours in vivo was significantly suppressed (P<0.05) in the shRNA group compared with that of the mock group, indicating that ANXA2 may be a novel and useful target for elucidating molecular mechanisms involving the proliferation and metastasis of HCC.
Collapse
Affiliation(s)
- ZHIZHEN DONG
- Research Centre of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - MIN YAO
- Research Centre of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - HAIJIAN ZHANG
- Research Centre of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - LI WANG
- Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - HUA HUANG
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - MEIJUAN YAN
- Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - WEI WU
- Research Centre of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - DENGFU YAO
- Research Centre of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
16
|
Fu M, Gu X, Ni H, Zhang W, Chang F, Gong L, Chen X, Li J, Qiu L, Shi C, Bao J. High expression of inositol polyphosphate phosphatase-like 1 associates with unfavorable survival in hepatocellular carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2013; 6:2515-2522. [PMID: 24228114 PMCID: PMC3816821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 09/29/2013] [Indexed: 06/02/2023]
Abstract
Inositol polyphosphate phosphatase-like 1 (INPPL1), also known as SH2-containing inositol 5'-phosphatase 2 (SHIP2), has been suggested to act downstream of the PI3K/AKT pathway and play an important function in tumor development and progression. However, the associations between SHIP2 expression and the clinical features to determine its clinicopathologic significance in hepatocellular carcinoma (HCC) have not been investigated. In the present study, one-step quantitative PCR reverse transcription-polymerase chain reaction (qPCR) and immunohistochemistry (IHC) analysis with HCC tissue microarrays (TMA) were employed to evaluate the expression of SHIP2 in HCC. The results showed that SHIP2 expression in the mRNA and protein levels was significantly higher in HCC tissue than in corresponding non-cancerous tissue (p = 0.0014 and p < 0.001, respectively). The expression of SHIP2 protein in HCC was related to tumor differentiation, α-fetoprotein level, liver cirrhosis, and five-year survival rate (all p < 0.05). Kaplan-Meier method and log-rank test indicated that high expression of SHIP2 (p = 0.017) and tumor differentiation (p = 0.036) showed significant correlations with poor prognosis of HCC patients. The data indicate that SHIP2 expression is correlated with significant characteristics of HCC, and it may be useful as an unfavorable prognostic factor in HCC.
Collapse
Affiliation(s)
- Maoying Fu
- Department of Infectious Diseases, The First People’s Hospital of Kunshan Affiliated with Jiangsu UniversitySuzhou 215000, China
| | - Xuefeng Gu
- Department of Infectious Diseases, The First People’s Hospital of Kunshan Affiliated with Jiangsu UniversitySuzhou 215000, China
| | - Huihui Ni
- Department of Infectious Diseases, The First People’s Hospital of Kunshan Affiliated with Jiangsu UniversitySuzhou 215000, China
| | - Wei Zhang
- Department of Infectious Diseases, The First People’s Hospital of Kunshan Affiliated with Jiangsu UniversitySuzhou 215000, China
| | - Feng Chang
- Department of Infectious Diseases, The First People’s Hospital of Kunshan Affiliated with Jiangsu UniversitySuzhou 215000, China
| | - Li Gong
- Department of Infectious Diseases, The First People’s Hospital of Kunshan Affiliated with Jiangsu UniversitySuzhou 215000, China
| | - Xiaoxiao Chen
- The Key Laboratory of Cancer Biomarkers, Prevention & Treatment Cancer Center and The Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical UniversityNanjing 210029, China
| | - Jiang Li
- Department of Pathology, Jiangsu Province Geriatric InstituteNanjing 210029, China
| | - Liang Qiu
- Department of Pathology, Jiangsu Province Geriatric InstituteNanjing 210029, China
| | - Chuanbing Shi
- Department of Pathology, The Second Affiliated Hospital of Nanjing Medical UniversityNanjing 210011, China
| | - Jun Bao
- Department of Gastroenterology, Jiangsu Province Geriatric InstituteNanjing 210029, China
| |
Collapse
|
17
|
Khan R, Zahid S, Wan YJY, Forster J, Karim ABA, Nawabi AM, Azhar A, Rahman MA, Ahmed N. Protein expression profiling of nuclear membrane protein reveals potential biomarker of human hepatocellular carcinoma. Clin Proteomics 2013; 10:6. [PMID: 23724895 PMCID: PMC3691657 DOI: 10.1186/1559-0275-10-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 05/24/2013] [Indexed: 12/14/2022] Open
Abstract
Background Complex molecular events lead to development and progression of liver cirrhosis to HCC. Differentially expressed nuclear membrane associated proteins are responsible for the functional and structural alteration during the progression from cirrhosis to carcinoma. Although alterations/ post translational modifications in protein expression have been extensively quantified, complementary analysis of nuclear membrane proteome changes have been limited. Deciphering the molecular mechanism that differentiate between normal and disease state may lead to identification of biomarkers for carcinoma. Results Many proteins displayed differential expression when nuclear membrane proteome of hepatocellular carcinoma (HCC), fibrotic liver, and HepG2 cell line were assessed using 2-DE and ESI-Q-TOF MS/MS. From the down regulated set in HCC, we have identified for the first time a 15 KDa cytochrome b5A (CYB5A), ATP synthase subunit delta (ATPD) and Hemoglobin subunit beta (HBB) with 11, 5 and 22 peptide matches respectively. Furthermore, nitrosylation studies with S-nitrosocysteine followed by immunoblotting with anti SNO-cysteine demonstrated a novel and biologically relevant post translational modification of thiols of CYB5A in HCC specimens only. Immunofluorescence images demonstrated increased protein S-nitrosylation signals in the tumor cells and fibrotic region of HCC tissues. The two other nuclear membrane proteins which were only found to be nitrosylated in case of HCC were up regulated ATP synthase subunit beta (ATPB) and down regulated HBB. The decrease in expression of CYB5A in HCC suggests their possible role in disease progression. Further insight of the functional association of the identified proteins was obtained through KEGG/ REACTOME pathway analysis databases. String 8.3 interaction network shows strong interactions with proteins at high confidence score, which is helpful in characterization of functional abnormalities that may be a causative factor of liver pathology. Conclusion These findings may have broader implications for understanding the mechanism of development of carcinoma. However, large scale studies will be required for further verification of their critical role in development and progression of HCC.
Collapse
Affiliation(s)
- Rizma Khan
- Neurochemistry Research Unit Laboratory, Department of Biochemistry, University of Karachi, Karachi, Pakistan.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Compulsory license is one of the safeguards that international IP law provides to address the undesired effects of pharmaceutical patents on access to important medicines. This article looks into three important case examples to analyze the mechanism’s effectiveness and feasibility: the first uses of the newer compulsory license regime established in 2003 under the WTO legislative framework to export medicines to third countries, which lack pharmaceutical manufacturing capacities; and further, the first compulsory license grant in India in March 2012. The case analyses are based on the historical, factual and legal background. They reveal the main challenges of the 2003 WTO regime, including the lack of economic incentives for the generic pharmaceutical companies’ participation. In the case of India’s compulsory license grant, the article takes as in depth look into possible reasons for the reluctance to use the safeguard until recently, and the important aspects and implications of the Indian authorization to manufacture and sell a generic version of a patented cancer drug.
Collapse
|
19
|
Shindoh J, Kaseb A, Vauthey JN. Surgical strategy for liver cancers in the era of effective chemotherapy. Liver Cancer 2013; 2:47-54. [PMID: 24159596 PMCID: PMC3747536 DOI: 10.1159/000346222] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Systemic chemotherapy is the only option for advanced and/or disseminated disease in patients with hepatocellular carcinoma (HCC). For decades, various systemic therapies have been explored for the treatment of advanced HCC. Nevertheless, no satisfactory results have been obtained in cytotoxic chemotherapy so far. However, with the recent introduction of effective chemotherapy agents including sorafenib, the role of systemic therapy for the treatment of HCC is changing. The goals of systemic therapy include prolongation of survival with stabilization of disease progression and, in selected patients, downsizing of primarily unresectable tumors. In the era of effective chemotherapy, patients with advanced HCC should be managed with individualized approaches to optimize outcome.
Collapse
Affiliation(s)
- Junichi Shindoh
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Tex., USA
| | - Ahmed Kaseb
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Tex., USA
| | - Jean-Nicolas Vauthey
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Tex., USA,*Jean-Nicolas Vauthey, MD, Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcomb Boulevard, Unit 1484, Houston, TX 77030 (USA), E-Mail
| |
Collapse
|
20
|
Selimovic D, El-Khattouti A, Ghozlan H, Haikel Y, Abdelkader O, Hassan M. Hepatitis C virus-related hepatocellular carcinoma: An insight into molecular mechanisms and therapeutic strategies. World J Hepatol 2012; 4:342-55. [PMID: 23355912 PMCID: PMC3554798 DOI: 10.4254/wjh.v4.i12.342] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 11/17/2012] [Accepted: 11/24/2012] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) infects more than 170 million people worldwide, and thereby becomes a series global health challenge. Chronic infection with HCV is considered one of the major causes of end-stage liver disease including cirrhosis and hepatocellular carcinoma. Although the multiple functions of the HCV proteins and their impacts on the modulation of the intracellular signaling transduction processes, the drive of carcinogenesis during the infection with HCV, is thought to result from the interactions of viral proteins with host cell proteins. Thus, the induction of mutator phenotype, in liver, by the expression of HCV proteins provides a key mechanism for the development of HCV-associated hepatocellular carcinoma (HCC). HCC is considered one of the most common malignancies worldwide with increasing incidence during the past decades. In many countries, the trend of HCC is attributed to several liver diseases including HCV infection. However, the development of HCC is very complicated and results mainly from the imbalance between tumor suppressor genes and oncogenes, as well as from the alteration of cellular factors leading to a genomic instability. Besides the poor prognosis of HCC patients, this type of tumor is quite resistance to the available therapies. Thus, understanding the molecular mechanisms, which are implicated in the development of HCC during the course of HCV infection, may help to design a general therapeutic protocol for the treatment and/or the prevention of this malignancy. This review summarizes the current knowledge of the molecular mechanisms, which are involved in the development of HCV-associated HCC and the possible therapeutic strategies.
Collapse
Affiliation(s)
- Denis Selimovic
- Denis Selimovic, Youssef Haikel, Mohamed Hassan, Institut National de la Santé et de la Recherche Médicale, U 977, 67000 Strasbourg, France
| | | | | | | | | | | |
Collapse
|
21
|
Targeted glypican-3 gene transcription inhibited the proliferation of human hepatoma cells by specific short hairpin RNA. Tumour Biol 2012. [PMID: 23192642 PMCID: PMC3597277 DOI: 10.1007/s13277-012-0593-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly chemoresistant cancer with no effective systemic therapy. Despite of surgical or locoregional therapies, prognosis remains poor because of high tumor recurrence or progression, and currently, there are no well-established effective adjuvant therapies. Glypican-3 (GPC-3) is specifically overexpressed in hepatoma and perhaps is a valuable molecular target for HCC therapy. In this present study, the effect of silencing GPC-3 gene transcription on human HepG2 cell proliferation was investigated by constructing GPC-3 short hairpin RNA (shRNA) plasmid. After HepG2 cells were transfected with the most efficient shRNA, GPC-3 mRNA expression (90.4 %) was inhibited significantly and estimated by fluorescence quantitative reverse transcriptase-polymerase chain reaction, and the result was accordance with downregulation at the protein level. The percentage of the cell proliferation was down to 28.9 % in the shRNA group and 19.9 % in the shRNA plus sorafenib group. The cell cycles were arrested in the G1 phase (65.6 %) and the apoptosis rate was increasing (66.75 %) in the shRNA1 group with significant alteration compared with that in the negative-shRNA group. Specific shRNA might intervene effectively GPC-3 activation and inhibit tumor cell proliferation, suggesting that GPC-3 gene should be a potential molecular target for HCC therapy.
Collapse
|
22
|
Cabrera-Benitez NE, Pérez-Roth E, Casula M, Ramos-Nuez Á, Ríos-Luci C, Rodríguez-Gallego C, Sologuren I, Jakubkiene V, Slutsky AS, Padrón JM, Villar J. Anti-inflammatory activity of a novel family of aryl ureas compounds in an endotoxin-induced airway epithelial cell injury model. PLoS One 2012; 7:e48468. [PMID: 23144889 PMCID: PMC3493555 DOI: 10.1371/journal.pone.0048468] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 09/25/2012] [Indexed: 01/11/2023] Open
Abstract
Background Despite our increased understanding of the mechanisms involved in acute lung injury (ALI) and the acute respiratory distress syndrome (ARDS), there is no specific pharmacological treatment of proven benefit. We used a novel screening methodology to examine potential anti-inflammatory effects of a small structure-focused library of synthetic carbamate and urea derivatives in a well established cell model of lipopolysaccharide (LPS)-induced ALI/ARDS. Methodology/Principal Findings After a pilot study to develop an in vitro LPS-induced airway epithelial cell injury model, a library of synthetic carbamate and urea derivates was screened against representative panels of human solid tumor cell lines and bacterial and fungal strains. Molecules that were non-cytotoxic and were inactive in terms of antiproliferative and antimicrobial activities were selected to study the effects on LPS-induced inflammatory response in an in vitro cell culture model using A549 human alveolar and BEAS-2B human bronchial cells. These cells were exposed for 18 h to LPS obtained from Escherichia coli, either alone or in combination with the test compounds. The LPS antagonists rhein and emodin were used as reference compounds. The most active compound (CKT0103) was selected as the lead compound and the impact of CKT0103 on pro-inflammatory IL-6 and IL-8 cytokine levels, expression of toll-like receptor-4 (TLR4) and nuclear factor kappa B inhibitor alpha (IκBα) was measured. CKT0103 significantly inhibited the synthesis and release of IL-6 and IL-8 induced by LPS. This suppression was associated with inhibition of TLR4 up-regulation and IκBα down-regulation. Immunocytochemical staining for TLR4 and IκBα supported these findings. Conclusions/Significance Using a novel screening methodology, we identified a compound – CKT0103 – with potent anti-inflammatory effects. These findings suggest that CKT0103 is a potential target for the treatment of the acute phase of sepsis and sepsis-induced ALI/ARDS.
Collapse
Affiliation(s)
- Nuria E. Cabrera-Benitez
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Multidisciplinary Organ Dysfunction Evaluation Research Network, Research Unit, Hospital Universitario Dr. Negrin, Las Palmas de Gran Canaria, Spain
| | - Eduardo Pérez-Roth
- BioLab, Instituto Universitario de Bio-Orgánica “Antonio González” (IUBO-AG), Universidad de La Laguna, La Laguna, Spain
| | - Milena Casula
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Multidisciplinary Organ Dysfunction Evaluation Research Network, Research Unit, Hospital Universitario Dr. Negrin, Las Palmas de Gran Canaria, Spain
| | - Ángela Ramos-Nuez
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Multidisciplinary Organ Dysfunction Evaluation Research Network, Research Unit, Hospital Universitario Dr. Negrin, Las Palmas de Gran Canaria, Spain
| | - Carla Ríos-Luci
- BioLab, Instituto Universitario de Bio-Orgánica “Antonio González” (IUBO-AG), Universidad de La Laguna, La Laguna, Spain
| | | | - Ithaisa Sologuren
- Department of Immunology, Hospital Universitario Dr. Negrin, Las Palmas de Gran Canaria, Spain
| | - Virginija Jakubkiene
- Department of Organic Chemistry, Faculty of Chemistry, Vilnius University, Vilnius, Lithuania
| | - Arthur S. Slutsky
- Keenan Research Center at the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Canada
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada
| | - José M. Padrón
- BioLab, Instituto Universitario de Bio-Orgánica “Antonio González” (IUBO-AG), Universidad de La Laguna, La Laguna, Spain
| | - Jesús Villar
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Multidisciplinary Organ Dysfunction Evaluation Research Network, Research Unit, Hospital Universitario Dr. Negrin, Las Palmas de Gran Canaria, Spain
- Keenan Research Center at the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Canada
- * E-mail:
| |
Collapse
|
23
|
Zhou YM, Zhang XF, Cao L, Li B, Sui CJ, Li YM, Yin ZF. MCM7 expression predicts post-operative prognosis for hepatocellular carcinoma. Liver Int 2012; 32:1505-9. [PMID: 22784096 DOI: 10.1111/j.1478-3231.2012.02846.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 06/04/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND Dysregulation of minichromosome maintenance protein 7 (MCM7) was previously identified in multiple human malignancies. The clinical significance of MCM7 expression is yet to be delineated in patients with hepatocellular carcinoma (HCC). METHODS Paired cancerous and non-cancerous specimens from 87 patients with HCC who underwent resection were used for the immunohistochemical evaluation of MCM7 expression. Effect of sorafenib on the expression of MCM7 was tested in two human HCC cell lines SMMC-7721 and PLC/PRF/5. RESULTS Non-cancerous tissues were negative for immunohistochemical staining for MCM7 expression. Nuclear MCM7 was expressed in 42 of 87 HCC (48.2%) and was correlated with hepatitis B virus infection (P = 0.020), intrahepatic metastasis (P = 0.022) and vascular invasion (P = 0.013). Moreover, its expression was correlated with shorter overall survival (P = 0.033). Multivariate analysis showed that MCM7 expression was an independent prognostic factor for overall survival(P = 0.041). Sorafenib inhibited the expression of MCM7 in a concentration-dependent manner in vitro. CONCLUSIONS The current findings suggested that MCM7 expression may be a useful predictor of prognosis in patients with HCC after resection. Adjuvant therapy with sorafenib might be a valuable therapeutic strategy for MCM7-positive HCC patients.
Collapse
Affiliation(s)
- Yan-Ming Zhou
- Department of Hepato-Biliary-Pancreato-Vascular Surgery, First affiliated Hospital of Xiamen University, Xiamen, China
| | | | | | | | | | | | | |
Collapse
|
24
|
Tsai SC, Yang JS, Peng SF, Lu CC, Chiang JH, Chung JG, Lin MW, Lin JK, Amagaya S, Wai-Shan Chung C, Tung TT, Huang WW, Tseng MT. Bufalin increases sensitivity to AKT/mTOR-induced autophagic cell death in SK-HEP-1 human hepatocellular carcinoma cells. Int J Oncol 2012; 41:1431-42. [PMID: 22858649 DOI: 10.3892/ijo.2012.1579] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 07/10/2012] [Indexed: 11/06/2022] Open
Abstract
Bufalin is the major component of Chan-Su (a traditional Chinese medicine, TCM) extracts from the venom of Bufo bufo gargarizan. In the present study, we investigated the pharmacological mechanisms of cell cycle arrest and autophagic cell death induced by bufalin in SK-HEP-1 human hepatocellular carcinoma cells in vitro. Bufalin inhibited cell survival by MTT assay and increased cell death by trypan blue exclusion assay in a concentration-dependent manner. In addition, bufalin induced G2/M phase arrest by reducing CDK1 activity. Bufalin triggered DNA fragmentation and apoptotic cell death in SK-HEP-1 cells by DNA gel electrophoresis, TUNEL and caspase-3 activity assay, while bufalin induced autophagic cell death by double-membrane vacuoles (transmission electron microscopy, TEM), acidic vesicular organelles (acridine orange staining) and cleavage of microtubule-associated protein 1 light chain 3 (LC3). Protein expression levels of cyclin A and B, CDK1, phospho-CDK1 (Thr161), Cdc25c, phospho-Cdc25c (Ser198), phospho-AKT (Thr308), phospho-AKT (Ser473), phospho‑mTOR (Ser2481) were downregulated. In contrast, protein expression levels of the Chk1, Wee1, LC3-II, Beclin-1, Atg 5, Atg 7 and Atg 12 were upregulated in SK-HEP-1 cells after bufalin treatment. Inhibition of autophagy by 3-methyladenine (an inhibitor of class III phosphatidylinositol-3 kinase; 3-MA) or bafilomycin A1 (an inhibitor of the vacuolar proton pump of lysosomes and endosomes) reduced the effect of bufalin on cell viability and enhanced the effect of bufalin on apoptosis. In conclusion, bufalin triggered autophagic cell death and G2/M phase arrest through the AKT/mTOR signaling pathway in SK-HEP-1 cells. Our findings showed that bufalin may be potentially efficacious in the treatment of human hepatocellular carcinoma.
Collapse
Affiliation(s)
- Shih-Chang Tsai
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan, R.O.C
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|