301
|
Bouattour M, Payancé A, Wassermann J. Evaluation of antiangiogenic efficacy in advanced hepatocellular carcinoma: Biomarkers and functional imaging. World J Hepatol 2015; 7:2245-2263. [PMID: 26380650 PMCID: PMC4568486 DOI: 10.4254/wjh.v7.i20.2245] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 05/16/2015] [Accepted: 08/30/2015] [Indexed: 02/06/2023] Open
Abstract
Many years after therapeutic wilderness, sorafenib finally showed a clinical benefit in patients with advanced hepatocellular carcinoma. After the primary general enthusiasm worldwide, some disappointments emerged particularly since no new treatment could exceed or at least match sorafenib in this setting. Without these new drugs, research focused on optimizing care of patients treated with sorafenib. One challenging research approach deals with identifying prognostic and predictive biomarkers of sorafenib in this population. The task still seems difficult; however appropriate investigations could resolve this dilemma, as observed for some malignancies where other drugs were used.
Collapse
Affiliation(s)
- Mohamed Bouattour
- Mohamed Bouattour, Audrey Payancé, Department of Hepatology, Beaujon University Hospital (AP-HP - Paris 7 Diderot), 92110 Clichy, France
| | - Audrey Payancé
- Mohamed Bouattour, Audrey Payancé, Department of Hepatology, Beaujon University Hospital (AP-HP - Paris 7 Diderot), 92110 Clichy, France
| | - Johanna Wassermann
- Mohamed Bouattour, Audrey Payancé, Department of Hepatology, Beaujon University Hospital (AP-HP - Paris 7 Diderot), 92110 Clichy, France
| |
Collapse
|
302
|
Lu S, Török HP, Gallmeier E, Kolligs FT, Rizzani A, Arena S, Göke B, Gerbes AL, De Toni EN. Tivantinib (ARQ 197) affects the apoptotic and proliferative machinery downstream of c-MET: role of Mcl-1, Bcl-xl and Cyclin B1. Oncotarget 2015; 6:22167-78. [PMID: 26259250 PMCID: PMC4673154 DOI: 10.18632/oncotarget.4240] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/28/2015] [Indexed: 01/11/2023] Open
Abstract
Tivantinib, a c-MET inhibitor, is investigated as a second-line treatment of HCC. It was shown that c-MET overexpression predicts its efficacy. Therefore, a phase-3 trial of tivantinib has been initiated to recruit "c-MET-high" patients only. However, recent evidence indicates that the anticancer activity of tivantinib is not due to c-MET inhibition, suggesting that c-MET is a predictor of response to this compound rather than its actual target. By assessing the mechanisms underlying the anticancer properties of tivantinib we showed that this agent causes apoptosis and cell cycle arrest by inhibiting the anti-apoptotic molecules Mcl-1 and Bcl-xl, and by increasing Cyclin B1 expression regardless of c-MET status. However, we found that tivantinib might antagonize the antiapoptotic effects of c-MET activation since HGF enhanced the expression of Mcl-1 and Bcl-xl. In summary, we show that the activity of tivantinib is independent of c-MET and describe Mcl-1, Bcl-xl and Cyclin B1 as effectors of its antineoplastic effects in HCC cells. We suggest that the predictive effect of c-MET expression in part reflects the c-MET-driven overexpression of Mcl-1 and Bcl-xl in c-MET-high patients and that these molecules are considered as possible response predictors.
Collapse
Affiliation(s)
- Shuai Lu
- Medizinische Klinik und Poliklinik 2, Klinikum der Universität München, Campus Grosshadern, Munich, Germany
| | - Helga-Paula Török
- Medizinische Klinik und Poliklinik 2, Klinikum der Universität München, Campus Grosshadern, Munich, Germany
| | - Eike Gallmeier
- Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University of Marburg, Marburg, Germany
| | - Frank T Kolligs
- Medizinische Klinik und Poliklinik 2, Klinikum der Universität München, Campus Grosshadern, Munich, Germany
- Department of Internal Medicine and Gastroenterology, HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | - Antonia Rizzani
- Medizinische Klinik und Poliklinik 2, Klinikum der Universität München, Campus Grosshadern, Munich, Germany
| | - Sabrina Arena
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Italy
| | - Burkhard Göke
- Medizinische Klinik und Poliklinik 2, Klinikum der Universität München, Campus Grosshadern, Munich, Germany
| | - Alexander L Gerbes
- Medizinische Klinik und Poliklinik 2, Klinikum der Universität München, Campus Grosshadern, Munich, Germany
| | - Enrico N De Toni
- Medizinische Klinik und Poliklinik 2, Klinikum der Universität München, Campus Grosshadern, Munich, Germany
| |
Collapse
|
303
|
Hepatocyte Growth Factor from a Clinical Perspective: A Pancreatic Cancer Challenge. Cancers (Basel) 2015; 7:1785-805. [PMID: 26404380 PMCID: PMC4586794 DOI: 10.3390/cancers7030861] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/07/2015] [Accepted: 08/17/2015] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer is the fourth leading cause of cancer-related deaths in the United States and incidence rates are rising. Both detection and treatment options for pancreatic cancer are limited, providing a less than 5% five-year survival advantage. The need for new biomarkers for early detection and treatment of pancreatic cancer demands the efficient translation of bench knowledge to provide clinical benefit. One source of therapeutic resistance is the pancreatic tumor microenvironment, which is characterized by desmoplasia and hypoxia making it less conducive to current therapies. A major factor regulating desmoplasia and subsequently promoting chemoresistance in pancreatic cancer is hepatocyte growth factor (HGF), the sole ligand for c-MET (mesenchymal-epithelial transition), an epithelial tyrosine kinase receptor. Binding of HGF to c-MET leads to receptor dimerization and autophosphorylation resulting in the activation of multiple cellular processes that support cancer progression. Inhibiting activation of c-MET in cancer cells, in combination with other approaches for reducing desmoplasia in the tumor microenvironment, might significantly improve the success of chemotherapy. Therefore, HGF makes a potent novel target for developing therapeutic strategies in combination with existing drugs for treating pancreatic adenocarcinoma. This review provides a comprehensive analysis of HGF and its promising potential as a chemotherapeutic target for pancreatic cancer.
Collapse
|
304
|
Abstract
Hepatocellular carcinoma (HCC) is a major health problem. Mortality owing to liver cancer has increased in the past 20 years, with recent studies reporting an incidence of 780,000 cases/year. Most patients with hepatocellular carcinoma are still diagnosed at intermediate or advanced disease stages, where curative approaches are often not feasible. Among the treatment options available, the molecular targeted agent sorafenib is able to significantly increase overall survival in these patients. Afterwards, up to 7 randomized phase III clinical trials investigating other molecular therapies in the first-line and second-line settings have failed to improve survival. Potential reasons for this include intertumor heterogeneity, issues with trial design and a lack of predictive biomarkers of response. Advance in our knowledge of the human genome has provided a comprehensive picture of commonly mutated genes in patients with HCC including mutations in the TERT promoter, CTNNB1, TP53 and ARID1A along with other amplifications (FGF19, VEGFA) or homozygous deletions (p16) as the most frequent alterations. This knowledge points toward specific drivers as candidate for druggable therapies. Thus, progressive implementation of proof-of-concept and enrichment might improve results in clinical trials testing of molecular targeted agents. Ultimately, these studies are aimed at long-term to improve current standards of care and influenced clinical decision-making and practice guidelines.
Collapse
|
305
|
|
306
|
Ettrich T, Perkhofer L, Seufferlein T. Personalisierte Tumortherapie bei gastrointestinalen Tumoren. Internist (Berl) 2015; 56:1069-78. [DOI: 10.1007/s00108-015-3752-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
307
|
Affiliation(s)
- Jin Ding
- a National Center of Liver Cancer; Eastern Hepatobiliary Surgery Hospital; Second Military Medical University ; Shanghai , China
| | | |
Collapse
|
308
|
Maida M, Iavarone M, Raineri M, Cammà C, Cabibbo G. Second line systemic therapies for hepatocellular carcinoma: Reasons for the failure. World J Hepatol 2015; 7:2053-2057. [PMID: 26301047 PMCID: PMC4539398 DOI: 10.4254/wjh.v7.i17.2053] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 06/22/2015] [Accepted: 06/30/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the main cause of death in patients with cirrhosis, with an increasing incidence worldwide. Sorafenib is the choice therapy for advanced HCC. Over time several randomized phase III trials have been performed testing sunitinib, brivanib, linifanib and other molecules in head-to-head comparison with Sorafenib as first-line treatment for advanced-stage HCC, but none of these has so far been registered in this setting. Moreover, another feared vacuum arises from the absence of molecules registered as second-line therapy for patients who have failed Sorafenib, representing an urgent unmet medical need. To date all molecules tested as second-line therapies for advanced hepatocellular carcinoma, failed to demonstrate an increased survival compared to placebo. What are the possible reasons for the failure? What we should expect in the near future?
Collapse
Affiliation(s)
- Marcello Maida
- Marcello Maida, Calogero Cammà, Giuseppe Cabibbo, Section of Gastroenterology, DIBIMIS, University of Palermo, 90127 Palermo, Italy
| | - Massimo Iavarone
- Marcello Maida, Calogero Cammà, Giuseppe Cabibbo, Section of Gastroenterology, DIBIMIS, University of Palermo, 90127 Palermo, Italy
| | - Maurizio Raineri
- Marcello Maida, Calogero Cammà, Giuseppe Cabibbo, Section of Gastroenterology, DIBIMIS, University of Palermo, 90127 Palermo, Italy
| | - Calogero Cammà
- Marcello Maida, Calogero Cammà, Giuseppe Cabibbo, Section of Gastroenterology, DIBIMIS, University of Palermo, 90127 Palermo, Italy
| | - Giuseppe Cabibbo
- Marcello Maida, Calogero Cammà, Giuseppe Cabibbo, Section of Gastroenterology, DIBIMIS, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
309
|
Ishii T, Iwasawa S, Kurimoto R, Maeda A, Takiguchi Y, Kaneda M. Crizotinib-Induced Abnormal Signal Processing in the Retina. PLoS One 2015; 10:e0135521. [PMID: 26271036 PMCID: PMC4535857 DOI: 10.1371/journal.pone.0135521] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 06/13/2015] [Indexed: 11/29/2022] Open
Abstract
Molecular target therapy for cancer is characterized by unique adverse effects that are not usually observed with cytotoxic chemotherapy. For example, the anaplastic lymphoma kinase (ALK)-tyrosine kinase inhibitor crizotinib causes characteristic visual disturbances, whereas such effects are rare when another ALK-tyrosine kinase inhibitor, alectinib, is used. To elucidate the mechanism responsible for these visual disturbances, the responses to light exhibited by retinal ganglion cells treated with these agents were evaluated using a C57BL6 mouse ex vivo model. Both crizotinib and alectinib changed the firing rate of ON and OFF type retinal ganglion cells. However, the ratio of alectinib-affected cells (15.7%) was significantly lower than that of crizotinib-affected cells (38.6%). Furthermore, these drugs changed the response properties to light stimuli of retinal ganglion cells in some of the affected cells, i.e., OFF cells responded to both ON and OFF stimuli, etc. Finally, the expressions of ALK (a target receptor of both crizotinib and alectinib) and of MET and ROS1 (additional target receptors of crizotinib) were observed at the mRNA level in the retina. Our findings suggest that these drugs might target retinal ganglion cells and that the potency of the drug actions on the light responses of retinal ganglion cells might be responsible for the difference in the frequencies of visual disturbances observed between patients treated with crizotinib and those treated with alectinib. The present experimental system might be useful for screening new molecular target agents prior to their use in clinical trials.
Collapse
Affiliation(s)
- Toshiyuki Ishii
- Department of Physiology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113–8602, Japan
| | - Shunichiro Iwasawa
- Department of Medical Oncology, Graduate School of Medicine, Chiba University1-8-1, Inohana Chuo-ku, Chiba 260–8670, Japan
| | - Ryota Kurimoto
- Department of Medical Oncology, Graduate School of Medicine, Chiba University1-8-1, Inohana Chuo-ku, Chiba 260–8670, Japan
| | - Akemi Maeda
- Department of Physiology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113–8602, Japan
- Tokyo Medical Care and Welfare Vocational School, 1-11-11 Hatchobori, Chuo-ku, Tokyo, 104–0032, Japan
| | - Yuichi Takiguchi
- Department of Medical Oncology, Graduate School of Medicine, Chiba University1-8-1, Inohana Chuo-ku, Chiba 260–8670, Japan
- * E-mail:
| | - Makoto Kaneda
- Department of Physiology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113–8602, Japan
| |
Collapse
|
310
|
Abstract
There is a pressing need for effective therapies to treat uveal melanoma. Agents that inhibit the c-MET pathway have shown promise in multiple malignancies that overexpress c-MET. Herein, we assess c-MET expression in both primary uveal melanoma and liver metastases of uveal melanoma and evaluate the association of c-MET expression with clinical and pathologic variables. We have retrospectively identified tumor samples from primary and liver metastases of uveal melanoma from 1 January 1990 to 1 January 2012. We utilized immunohistochemistry to assess c-MET expression, and two pathologists quantified c-MET expression using an H-score (product of the intensity of staining and percentage of positive cells). The Mann-Whitney U-test, Pearson's correlation, and Cox model were used as appropriate. Thirty-nine of 40 (98%) primary tumors and nine of 10 (90%) metastatic liver lesions expressed c-MET (H-score range 0-300). There was a strong association between the percentage of positive cells and the intensity of c-MET expression (P=0.007). We found no association between c-MET H-score and clinicopathologic variables such as age, sex, or stage. c-MET expression was significantly higher in metastatic compared with primary tumors (median H-score 190 vs. 30, P=0.022). c-MET is expressed in the vast majority of primary and liver metastases of uveal melanomas; however, c-MET expression did not associate with pathologic features in our cohort. Metastatic lesions have higher expression of c-MET expression than primary tumors. Clinical trials involving c-MET inhibitors deserve further study in patients with uveal melanoma in both the adjuvant and metastatic setting.
Collapse
|
311
|
Scagliotti GV, Di Maio M. Tivantinib added to erlotinib in nonsmall-cell lung cancer: the primary end point was not MET.... Ann Oncol 2015; 26:2007-9. [PMID: 26265168 DOI: 10.1093/annonc/mdv334] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- G V Scagliotti
- Department of Oncology, University of Torino at San Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - M Di Maio
- Department of Oncology, University of Torino at San Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| |
Collapse
|
312
|
Ch'ang HJ. Optimal combination of antiangiogenic therapy for hepatocellular carcinoma. World J Hepatol 2015; 7:2029-40. [PMID: 26261692 PMCID: PMC4528276 DOI: 10.4254/wjh.v7.i16.2029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 07/21/2015] [Accepted: 07/24/2015] [Indexed: 02/06/2023] Open
Abstract
The success of sorafenib in prolonging survival of patients with hepatocellular carcinoma (HCC) makes therapeutic inhibition of angiogenesis a component of treatment for HCC. To enhance therapeutic efficacy, overcome drug resistance and reduce toxicity, combination of antiangiogenic agents with chemotherapy, radiotherapy or other targeted agents were evaluated. Nevertheless, the use of antiangiogenic therapy remains suboptimal regarding dosage, schedule and duration of therapy. The issue is further complicated by combination antiangiogenesis to other cytotoxic or biologic agents. There is no way to determine which patients are most likely respond to a given form of antiangiogenic therapy. Activation of alternative pathways associated with disease progression in patients undergoing antiangiogenic therapy has also been recognized. There is increasing importance in identifying, validating and standardizing potential response biomarkers for antiangiogenesis therapy for HCC patients. In this review, biomarkers for antiangiogenesis therapy including systemic, circulating, tissue and imaging ones are summarized. The strength and deficit of circulating and imaging biomarkers were further demonstrated by a series of studies in HCC patients receiving radiotherapy with or without thalidomide.
Collapse
Affiliation(s)
- Hui-Ju Ch'ang
- Hui-Ju Ch'ang, National Institute of Cancer Research, National Health Research Institutes, Miaoli 35053, Taiwan
| |
Collapse
|
313
|
Palmer DH, Johnson PJ. Evaluating the role of treatment-related toxicities in the challenges facing targeted therapies for advanced hepatocellular carcinoma. Cancer Metastasis Rev 2015; 34:497-509. [DOI: 10.1007/s10555-015-9580-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
314
|
Gnoni A, Santini D, Scartozzi M, Russo A, Licchetta A, Palmieri V, Lupo L, Faloppi L, Palasciano G, Memeo V, Angarano G, Brunetti O, Guarini A, Pisconti S, Lorusso V, Silvestris N. Hepatocellular carcinoma treatment over sorafenib: epigenetics, microRNAs and microenvironment. Is there a light at the end of the tunnel? Expert Opin Ther Targets 2015. [PMID: 26212068 DOI: 10.1517/14728222.2015.1071354] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Sorafenib is currently the only approved therapy in hepatocellular carcinoma (HCC). Alternative first- and second-line treatments are a significant unmet medical need, and several biologic agents have been tested in recent years, with poor results. Therefore, angiogenic pathways and the cytokine cascade remain possible targets in HCC. Recent studies suggest a role of epigenetic processes, associated with the initiation and development of HCC. In this field, DNA methylation, micro-RNAs (miRNAs) and tumor microenvironment cells became a possible new target for HCC treatment. AREAS COVERED This review explains the possible role of DNA methylation and histone deacetylase inhibitors as predictive biomarkers and target therapy, the extensive world of the promising miRNA blockade strategy, and the recent strong evidence of correlation between HCC tumors and peritumoral stroma cells. The literature and preclinic/clinic data were obtained through an electronic search. EXPERT OPINION Future research should aim to understand how best to identify patient groups that would benefit most from the prescribed therapy. To overcome the 'therapeutic stranding' of HCC, a possible way out from the current therapeutic tunnel might be to evaluate the major epigenetic and genetic processes involved in HCC carcinogenesis, not underestimating the tumor microenvironment and its 'actors' (angiogenesis, immune system, platelets). We are only at the start of a long journey towards the elucidation of HCC molecular pathways as therapeutic targets. Yet, currently this path appears to be the only one to cast some light at the end of the tunnel.
Collapse
Affiliation(s)
- Antonio Gnoni
- a 1 Hospital of Taranto, Medical Oncology Unit , Taranto, Italy
| | - Daniele Santini
- b 2 University Campus Biomedico, Medical Oncology Unit , Rome, Italy
| | - Mario Scartozzi
- c 3 University of Cagliari, Medical Oncology Unit , Cagliari, Italy
| | - Antonio Russo
- d 4 University of Palermo, Medical Oncology Unit , Palermo, Italy
| | | | - Vincenzo Palmieri
- e 5 University of Bari, Department of Biomedical Sciences and Human Oncology, Clinica Medica "A. Murri" , Bari, Italy
| | - Luigi Lupo
- f 6 University of Bari, Institute of General Surgery and Liver Transplantation, Department of Emergency and Organ Transplantation , Bari, Italy
| | - Luca Faloppi
- g 7 Polytechnic University of the Marche, Medical Oncology Unit , Ancona, Italy
| | - Giuseppe Palasciano
- e 5 University of Bari, Department of Biomedical Sciences and Human Oncology, Clinica Medica "A. Murri" , Bari, Italy
| | - Vincenzo Memeo
- f 6 University of Bari, Institute of General Surgery and Liver Transplantation, Department of Emergency and Organ Transplantation , Bari, Italy
| | | | - Oronzo Brunetti
- i 9 National Cancer Research Centre "Giovanni Paolo II", Medical Oncology Unit , Viale Orazio Flacco, 65, 70124 Bari, Italy +39 080 555 5419 ; +39 080 555 5419 ;
| | - Attilio Guarini
- j 10 National Cancer Research Centre "Giovanni Paolo II", Medical Ematology Unit , Bari, Italy
| | | | - Vito Lorusso
- i 9 National Cancer Research Centre "Giovanni Paolo II", Medical Oncology Unit , Viale Orazio Flacco, 65, 70124 Bari, Italy +39 080 555 5419 ; +39 080 555 5419 ;
| | - Nicola Silvestris
- i 9 National Cancer Research Centre "Giovanni Paolo II", Medical Oncology Unit , Viale Orazio Flacco, 65, 70124 Bari, Italy +39 080 555 5419 ; +39 080 555 5419 ;
| |
Collapse
|
315
|
Scagliotti G, von Pawel J, Novello S, Ramlau R, Favaretto A, Barlesi F, Akerley W, Orlov S, Santoro A, Spigel D, Hirsh V, Shepherd FA, Sequist LV, Sandler A, Ross JS, Wang Q, von Roemeling R, Shuster D, Schwartz B. Phase III Multinational, Randomized, Double-Blind, Placebo-Controlled Study of Tivantinib (ARQ 197) Plus Erlotinib Versus Erlotinib Alone in Previously Treated Patients With Locally Advanced or Metastatic Nonsquamous Non-Small-Cell Lung Cancer. J Clin Oncol 2015; 33:2667-74. [PMID: 26169611 DOI: 10.1200/jco.2014.60.7317] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Tivantinib, a MET receptor tyrosine kinase inhibitor, demonstrated increased anticancer activity in preclinical and early clinical studies when combined with erlotinib. Our study aimed to confirm efficacy and safety of the combination in previously treated patients with non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS Patients with advanced nonsquamous NSCLC previously treated with one to two systemic regimens, including a platinum doublet, were randomly assigned at a 1:1 ratio to receive erlotinib 150 mg daily plus oral tivantinib 360 mg twice daily (E + T) or erlotinib plus placebo (E + P) until disease progression. Tumor specimens were evaluated for EGFR and KRAS mutations, MET expression, and MET gene amplification. The primary end point was overall survival (OS). Secondary and exploratory objectives included progression-free survival (PFS), OS in molecular subgroups, and safety. RESULTS The study enrolled 1,048 patients and was discontinued for futility at the interim analysis. OS did not improve with E + T versus E + P (median OS, 8.5 v 7.8 months, respectively; hazard ratio [HR], 0.98; 95% CI, 0.84 to 1.15; P = .81), even though PFS increased (median PFS, 3.6 v 1.9 months; HR, 0.74; 95% CI, 0.62 to 0.89; P < .001). Exploratory subgroup analyses suggested OS improvement in patients with high MET expression (HR, 0.70; 95% CI, 0.49 to 1.01). Most common adverse events occurring with E + T versus E + P were rash (33.1% v 37.3%, respectively), diarrhea (34.6% v 41.0%), asthenia or fatigue (43.5% v 38.1%), and neutropenia (grade 3 to 4; 8.5% v 0.8%). CONCLUSION E + T was well tolerated and increased PFS but did not improve OS in the overall nonsquamous NSCLC population.
Collapse
Affiliation(s)
- Giorgio Scagliotti
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ.
| | - Joachim von Pawel
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Silvia Novello
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Rodryg Ramlau
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Adolfo Favaretto
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Fabrice Barlesi
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Wallace Akerley
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Sergey Orlov
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Armando Santoro
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - David Spigel
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Vera Hirsh
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Frances A Shepherd
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Lecia V Sequist
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Alan Sandler
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Jeffrey S Ross
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Qiang Wang
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Reinhard von Roemeling
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Dale Shuster
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| | - Brian Schwartz
- Giorgio Scagliotti and Silvia Novello, University of Turin, Orbassano, Torino; Adolfo Favaretto, Istituto Oncologico Veneto, Padova; Armando Santoro, Istituto Clinico Humanitas, Milan, Italy; Joachim von Pawel, Asklepios-Fachkliniken München-Gauting, Munich, Germany; Rodryg Ramlau, Poznań University of Medical Sciences, Poznań, Poland; Fabrice Barlesi, Aix Marseille University, Assistance Publique Hôpitaux de Marseille, Marseille, France; Wallace Akerley, Huntsman Cancer Institute, Salt Lake City, UT; Sergey Orlov, St Petersburg State Medical University, St Petersburg, Russian Federation; David Spigel, Clinical Locations, Nashville, TN; Vera Hirsh, McGill University Health Centre, Montreal, Quebec; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Lecia V. Sequist, Massachusetts General Hospital, Boston; Jeffrey S. Ross, Foundation Medicine, Cambridge; Brian Schwartz, ArQule, Woburn, MA; Alan Sandler, Genentech, San Francisco, CA; and Qiang Wang, Reinhard von Roemeling, and Dale Shuster, Daiichi Sankyo, Edison, NJ
| |
Collapse
|
316
|
Abstract
The heterogeneous nature of hepatocellular carcinoma (HCC) poses a challenge to effective therapy. Significant strides have been made in molecular profiling. Telomerase promoter mutations are now known to represent early events in linear carcinogenesis. However, the translation of these advances into clinical practice has been limited. Although further work is needed to extrapolate genetic information to patient care, progress has been made in using genetics and stratification for HCC therapy. Targeted therapies for patients with c-MET overexpression, VEGF-A amplification and Wnt β-catenin-driven tumors offer promise.
Collapse
Affiliation(s)
- Sumera Rizvi
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minn., USA
| | | |
Collapse
|
317
|
Abstract
From its approval in 2008, sorafenib is the recommended treatment option for advanced-stage patients and its safety and efficacy has been confirmed by several studies. However, its mechanism of action is not completely understood and many efforts have been dedicated to investigating possible treatment response predictors. Dermatological adverse events occurring within the first 2 months of treatment are predictors of longer survival, while the same role for hypertension and diarrhea still needs a prospective confirmation. This association is opposite to the strategy of starting at a low dose as it may imply suboptimal drug exposure. In case of radiological progression, the appearance of new extrahepatic metastasis or vascular invasion significantly worsens life expectancy if compared to other patterns of progression. To date no genetic or biologic marker is available to predict response, even if some encouraging results have been reported by the study of polymorphism of VEGF and its receptor. Currently, data are conflicting about the possible predictive role of α-fetoprotein. Due to failure or the progression of therapies for earlier evolutionary stages (BCLC B) some patients in such a clinical profile may be treated with sorafenib. Indeed, almost 50% of the sorafenib-treated patients belong to this class. Patients with severely decompensated liver disease (jaundice, ascites in need of intense diuretic therapy/paracentesis) may not benefit from treatment. The use of sorafenib in the waiting list for liver transplantation is controversial, while its use at an advanced age requires careful evaluation of existing comorbidities that may increase the risk of adverse events. Many strides have been made in the field of hepatocellular carcinoma systemic therapy, and many remain to be realized. Considering the disappointing results of the trials conducted on new agents, a more dynamic interpretation of events together with the development of new strategies is key to enriching new and hopefully more successful trials.
Collapse
Affiliation(s)
- Alessia Gazzola
- Barcelona Clinic Liver Cancer (BCLC) Group, Liver Unit, Hospital Clinic, IDIBAPS, University of Barcelona, Barcelona, Spain
| | | | | | | |
Collapse
|
318
|
Tolaney SM, Tan S, Guo H, Barry W, Van Allen E, Wagle N, Brock J, Larrabee K, Paweletz C, Ivanova E, Janne P, Overmoyer B, Wright JJ, Shapiro GI, Winer EP, Krop IE. Phase II study of tivantinib (ARQ 197) in patients with metastatic triple-negative breast cancer. Invest New Drugs 2015; 33:1108-14. [PMID: 26123926 PMCID: PMC4608248 DOI: 10.1007/s10637-015-0269-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 06/19/2015] [Indexed: 12/21/2022]
Abstract
Background MET expression and activation appear to be important for initiation and progression of triple-negative breast cancer. Tivantinib (ARQ 197) is an orally administered agent that targets MET, although recent preclinical data suggests the agent may have mechanisms of action that are independent of MET signaling. We conducted a phase 2 study of tivantinib monotherapy in patients with metastatic triple-negative breast cancer. Methods Patients with metastatic triple-negative breast cancer who had received 1 to 3 prior lines of chemotherapy in the metastatic setting were enrolled into this two-stage, single arm phase 2 study. Treatment consisted of twice daily oral dosing of tivantinib (360 mg po bid) during a 21-day cycle. Patients underwent restaging scans at 6 weeks, and then every 9 weeks. Tumor biomarkers that might predict response to tivantinib were explored. Results 22 patients were enrolled. The overall response rate was 5 % (95 % CI 0–25 %) and the 6-month progression-free survival (PFS) was 5 % (95 % CI 0–25 %), with one patient achieving a partial response (PR). Toxicity was minimal with only 5 grade ≥3 adverse events (one grade 3 anemia, one grade 3 fatigue, and 3 patients with grade 3/4 neutropenia). Conclusion This study represents the first evaluation of tivantinib for the treatment of metastatic triple-negative breast cancer. These results suggest that single agent tivantinib is well tolerated, but did not meet prespecified statistical targets for efficacy.
Collapse
Affiliation(s)
- Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Yawkey 1257, Boston, MA, 02215, USA.
| | - Sally Tan
- Harvard Medical School, Boston, MA, USA
| | - Hao Guo
- Department of Biostatistics and Computation Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - William Barry
- Department of Biostatistics and Computation Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Eliezer Van Allen
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nikhil Wagle
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Yawkey 1257, Boston, MA, 02215, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jane Brock
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Katherine Larrabee
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Yawkey 1257, Boston, MA, 02215, USA
| | - Cloud Paweletz
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Yawkey 1257, Boston, MA, 02215, USA
- Belfer Institute for Applied Cancer Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Elena Ivanova
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Yawkey 1257, Boston, MA, 02215, USA
- Belfer Institute for Applied Cancer Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Pasi Janne
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Yawkey 1257, Boston, MA, 02215, USA
- Belfer Institute for Applied Cancer Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Beth Overmoyer
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Yawkey 1257, Boston, MA, 02215, USA
| | - John J Wright
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, USA
| | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Yawkey 1257, Boston, MA, 02215, USA
| | - Eric P Winer
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Yawkey 1257, Boston, MA, 02215, USA
| | - Ian E Krop
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Yawkey 1257, Boston, MA, 02215, USA
| |
Collapse
|
319
|
Facciorusso A, Licinio R, Carr BI, Di Leo A, Barone M. MEK 1/2 inhibitors in the treatment of hepatocellular carcinoma. Expert Rev Gastroenterol Hepatol 2015; 9:993-1003. [PMID: 25915713 DOI: 10.1586/17474124.2015.1040763] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Sorafenib is the only approved systemic treatment for advanced hepatocellular carcinoma patients and all the recently published randomized controlled trials on new systemic drugs have been unsuccessful. This is likely due to a lack of understanding of tumor progression, molecular drivers, and liver toxicity, as well as flaws in trial design. An important signaling pathway in hepatocarcinogenesis is the MEK cascade involved in various cellular responses, including adaptation and survival. A key role in this cascade is played by MEK, of which MEK 1/2 represent the prototypes and an interesting target for new oncological drugs. This review analyzes recent developments and future perspectives on the role of MEK inhibitors in hepatocellular carcinoma treatment.
Collapse
Affiliation(s)
- Antonio Facciorusso
- Gastroenterology Unit, Department of Medical and Surgical Sciences, University of Foggia, Ospedali Riuniti Foggia, Italy
| | | | | | | | | |
Collapse
|
320
|
Bupathi M, Kaseb A, Meric-Bernstam F, Naing A. Hepatocellular carcinoma: Where there is unmet need. Mol Oncol 2015; 9:1501-9. [PMID: 26160430 DOI: 10.1016/j.molonc.2015.06.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 05/05/2015] [Accepted: 06/17/2015] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a complex and heterogeneous tumor most commonly associated with underlying chronic liver disease, especially hepatitis. It is a growing problem in the United States and worldwide. There are two potential ways to prevent HCC. Primary prevention which is based on vaccination or secondary prevention involving agents that slow down carcinogenesis. Several pathways have been thought to play a role in the development of HCC; specifically, those involving vascular endothelial growth factor (VEGF)-mediated angiogenesis, WNT, phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR), AMP-activated protein kinase (AMPK), and c-MET. Currently, there are only a limited number of drugs which have been proven as effective treatment options for HCC and several clinical trials are testing drugs which target aberrations in the pathways mentioned above. In this review, we discuss currently approved therapies, monotherapies and combination therapy for the treatment of HCC.
Collapse
Affiliation(s)
- Manojkumar Bupathi
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ahmed Kaseb
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
321
|
Baccelli I, Stenzinger A, Vogel V, Pfitzner BM, Klein C, Wallwiener M, Scharpff M, Saini M, Holland-Letz T, Sinn HP, Schneeweiss A, Denkert C, Weichert W, Trumpp A. Co-expression of MET and CD47 is a novel prognosticator for survival of luminal breast cancer patients. Oncotarget 2015; 5:8147-60. [PMID: 25230070 PMCID: PMC4226673 DOI: 10.18632/oncotarget.2385] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Although luminal-type primary breast cancer can be efficiently treated, development of metastatic disease remains a significant clinical problem. We have previously shown that luminal-type circulating tumor cells (CTCs) co-expressing the tyrosine-kinase MET and CD47, a ligand involved in cancer cell evasion from macrophage scavenging, are able to initiate metastasis in xenografts. Here, we investigated the clinical relevance of MET-CD47 co-expression in 255 hormone receptor positive breast tumors by immunohistochemistry and found a 10.3-year mean overall-survival difference between MET-CD47 double-positive and double-negative patients (p<0.001). MET-CD47 co-expression defined a novel independent prognosticator for overall-survival by multivariate analysis (Cox proportional hazards model: HR: 4.1, p<0.002) and CD47 expression alone or in combination with MET was strongly associated with lymph node metastasis. Furthermore, flow cytometric analysis of metastatic patient blood revealed consistent presence of MET+CD47+ CTCs (range 0.8 – 33.3% of CTCs) and their frequency was associated with increased metastatic spread. Finally, primary uncultured CTCs with high MET+CD47+ content showed an enhanced capacity to initiate metastasis in mice. Detection and targeting of MET and CD47 may thus provide a rational basis for risk stratification and treatment of patients with luminal-type breast cancer.
Collapse
Affiliation(s)
- Irène Baccelli
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany. Divison of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Vanessa Vogel
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany. Divison of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany. Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Berit Maria Pfitzner
- Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Corinna Klein
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany. Divison of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Markus Wallwiener
- National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Martina Scharpff
- National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Massimo Saini
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany. Divison of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Tim Holland-Letz
- Department of Biostatistics, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld TP4, 69120 Heidelberg, Germany
| | - Hans-Peter Sinn
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Carsten Denkert
- Institute of Pathology, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany. German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany. National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany. German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany. Divison of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany. German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| |
Collapse
|
322
|
Waidmann O, Trojan J. Novel drugs in clinical development for hepatocellular carcinoma. Expert Opin Investig Drugs 2015; 24:1075-82. [PMID: 26108356 DOI: 10.1517/13543784.2015.1058776] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Sorafenib is the only systemic drug approved for the treatment of advanced hepatocellular carcinoma (HCC). Within recent years, several investigational agents mainly targeting angiogenesis failed in late-phase clinical development either due to toxicity or lack of benefit. AREAS COVERED This review covers recent clinical data on systemic agents and ongoing trials in patients with advanced HCC. EXPERT OPINION In unselected patients with advanced HCC, disappointing results have been reported from several large trials. However, in two subgroups encouraging results have been achieved. Treatment with the MET inhibitor tivantinib resulted in a substantial survival benefit in the subgroup of MET overexpressing tumors in a randomized Phase II trial. Furthermore, the vascular endothelial growth factor receptor 2 antibody ramucirumab resulted in improved overall survival in patients with baseline α-fetoprotein (AFP) ≥ 400 ng/ml in a Phase III trial. These two agents, and several others, will be further developed in HCC. Moreover, immunotherapeutics such as checkpoint inhibitors, programmed death receptor-1 blocking antibodies and oncolytic viruses are under investigation in advanced HCC.
Collapse
Affiliation(s)
- Oliver Waidmann
- Universitätsklinikum Frankfurt, Medizinische Klinik 1 , Theodor-Stern-Kai 7, Frankfurt am Main , Germany +49 0 69 6301 7860 ; +49 0 69 6301 6448 ;
| | | |
Collapse
|
323
|
Forner A, Reig M. Does ramucirumab deserve a second chance for liver cancer? Lancet Oncol 2015; 16:751-2. [PMID: 26095780 DOI: 10.1016/s1470-2045(15)00053-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 05/18/2015] [Indexed: 02/07/2023]
Affiliation(s)
- Alejandro Forner
- Barcelona Clinic Liver Cancer (BCLC) group, Liver Unit, Hospital Clinic Barcelona, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain.
| | - María Reig
- Barcelona Clinic Liver Cancer (BCLC) group, Liver Unit, Hospital Clinic Barcelona, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
| |
Collapse
|
324
|
Yeh MM, Yeung RS, Apisarnthanarax S, Bhattacharya R, Cuevas C, Harris WP, Hon TLK, Padia SA, Park JO, Riggle KM, Daoud SS. Multidisciplinary perspective of hepatocellular carcinoma: A Pacific Northwest experience. World J Hepatol 2015; 7:1460-83. [PMID: 26085907 PMCID: PMC4462686 DOI: 10.4254/wjh.v7.i11.1460] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 04/03/2015] [Accepted: 04/27/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most rapidly increasing type of cancer in the United States. HCC is a highly malignant cancer, accounting for at least 14000 deaths in the United States annually, and it ranks third as a cause of cancer mortality in men. One major difficulty is that most patients with HCC are diagnosed when the disease is already at an advanced stage, and the cancer cannot be surgically removed. Furthermore, because almost all patients have cirrhosis, neither chemotherapy nor major resections are well tolerated. Clearly there is need of a multidisciplinary approach for the management of HCC. For example, there is a need for better understanding of the fundamental etiologic mechanisms that are involved in hepatocarcinogenesis, which could lead to the development of successful preventive and therapeutic modalities. It is also essential to define the cellular and molecular bases for malignant transformation of hepatocytes. Such knowledge would: (1) greatly facilitate the identification of patients at risk; (2) prompt efforts to decrease risk factors; and (3) improve surveillance and early diagnosis through diagnostic imaging modalities. Possible benefits extend also to the clinical management of this disease. Because there are many factors involved in pathogenesis of HCC, this paper reviews a multidisciplinary perspective of recent advances in basic and clinical understanding of HCC that include: molecular hepatocarcinogenesis, non-invasive diagnostics modalities, diagnostic pathology, surgical modality, transplantation, local therapy and oncological/target therapeutics.
Collapse
Affiliation(s)
- Matthew M Yeh
- Matthew M Yeh, Raymond S Yeung, Department of Pathology, University of Washington School of Medicine, Seattle, WA 99210, United States
| | - Raymond S Yeung
- Matthew M Yeh, Raymond S Yeung, Department of Pathology, University of Washington School of Medicine, Seattle, WA 99210, United States
| | - Smith Apisarnthanarax
- Matthew M Yeh, Raymond S Yeung, Department of Pathology, University of Washington School of Medicine, Seattle, WA 99210, United States
| | - Renuka Bhattacharya
- Matthew M Yeh, Raymond S Yeung, Department of Pathology, University of Washington School of Medicine, Seattle, WA 99210, United States
| | - Carlos Cuevas
- Matthew M Yeh, Raymond S Yeung, Department of Pathology, University of Washington School of Medicine, Seattle, WA 99210, United States
| | - William P Harris
- Matthew M Yeh, Raymond S Yeung, Department of Pathology, University of Washington School of Medicine, Seattle, WA 99210, United States
| | - Tony Lim Kiat Hon
- Matthew M Yeh, Raymond S Yeung, Department of Pathology, University of Washington School of Medicine, Seattle, WA 99210, United States
| | - Siddharth A Padia
- Matthew M Yeh, Raymond S Yeung, Department of Pathology, University of Washington School of Medicine, Seattle, WA 99210, United States
| | - James O Park
- Matthew M Yeh, Raymond S Yeung, Department of Pathology, University of Washington School of Medicine, Seattle, WA 99210, United States
| | - Kevin M Riggle
- Matthew M Yeh, Raymond S Yeung, Department of Pathology, University of Washington School of Medicine, Seattle, WA 99210, United States
| | - Sayed S Daoud
- Matthew M Yeh, Raymond S Yeung, Department of Pathology, University of Washington School of Medicine, Seattle, WA 99210, United States
| |
Collapse
|
325
|
Ono A, Fujimoto A, Yamamoto Y, Akamatsu S, Hiraga N, Imamura M, Kawaoka T, Tsuge M, Abe H, Hayes CN, Miki D, Furuta M, Tsunoda T, Miyano S, Kubo M, Aikata H, Ochi H, Kawakami YI, Arihiro K, Ohdan H, Nakagawa H, Chayama K. Circulating Tumor DNA Analysis for Liver Cancers and Its Usefulness as a Liquid Biopsy. Cell Mol Gastroenterol Hepatol 2015; 1:516-534. [PMID: 28210698 PMCID: PMC5301414 DOI: 10.1016/j.jcmgh.2015.06.009] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 06/03/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Circulating tumor DNA (ctDNA) carrying tumor-specific sequence alterations has been found in the cell-free fraction of blood. Liver cancer tumor specimens are difficult to obtain, and noninvasive methods are required to assess cancer progression and characterize underlying genomic features. METHODS We analyzed 46 patients with hepatocellular carcinoma who underwent hepatectomy or liver transplantation and for whom whole-genome sequencing data was available. We designed personalized assays targeting somatic rearrangements of each tumor to quantify serum ctDNA. Exome sequencing was performed using cell-free DNA paired primary tumor tissue DNA from a patient with recurrent liver cancer after transcatheter arterial chemoembolization (TACE). RESULTS We successfully detected ctDNA from 100 μL of serum samples in 7 of the 46 patients before surgery, increasing with disease progression. The cumulative incidence of recurrence and extrahepatic metastasis in the ctDNA-positive group were statistically significantly worse than in the ctDNA-negative group (P = .0102 and .0386, respectively). Multivariate analysis identified ctDNA (OR 6.10; 95% CI, 1.11-33.33, P = .038) as an independent predictor of microscopic vascular invasion of the portal vein (VP). We identified 45 nonsynonymous somatic mutations in cell-free DNA after TACE and 71 nonsynonymous somatic mutations in primary tumor tissue by exome sequencing. We identified 25 common mutations in both samples, and 83% of mutations identified in the primary tumor could be detected in the cell-free DNA. CONCLUSIONS The presence of ctDNA reflects tumor progression, and detection of ctDNA can predict VP and recurrence, especially extrahepatic metastasis within 2 years. Our study demonstrated the usefulness of ctDNA detection and sequencing analysis of cell-free DNA for personalized treatment of liver cancer.
Collapse
Key Words
- AFP, α-fetoprotein
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- Circulating Tumor DNA
- DCP, des-γ-carboxy prothrombin
- Exome Sequencing
- HAIC, hepatic arterial infusion chemotherapy
- HBV, hepatitis B virus
- HCC, hepatocellular carcinoma
- HCV, hepatitis C virus
- Hepatocellular Carcinoma
- PCR, polymerase-chain-reaction
- TACE, transcatheter arterial chemoembolization
- VP, microscopic vascular invasion to portal vein
- Whole-Genome Sequencing
- cHCC/CC, combined hepatocellular and cholangiocarcinoma
- ctDNA, circulating tumor DNA
Collapse
Affiliation(s)
- Atsushi Ono
- Department of Gastroenterology and Metabolism, Applied Life Science, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan,Liver Research Project Center, Hiroshima University, Hiroshima, Japan,Laboratory for Digestive Diseases, RIKEN Center for Integrative Medical Sciences, Hiroshima, Japan
| | - Akihiro Fujimoto
- Laboratory for Genome Sequencing Analysis, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan,Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yujiro Yamamoto
- Laboratory for Genome Sequencing Analysis, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Sakura Akamatsu
- Department of Gastroenterology and Metabolism, Applied Life Science, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan,Liver Research Project Center, Hiroshima University, Hiroshima, Japan,Laboratory for Digestive Diseases, RIKEN Center for Integrative Medical Sciences, Hiroshima, Japan
| | - Nobuhiko Hiraga
- Department of Gastroenterology and Metabolism, Applied Life Science, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Michio Imamura
- Department of Gastroenterology and Metabolism, Applied Life Science, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Tomokazu Kawaoka
- Department of Gastroenterology and Metabolism, Applied Life Science, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Masataka Tsuge
- Department of Gastroenterology and Metabolism, Applied Life Science, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Hiromi Abe
- Department of Gastroenterology and Metabolism, Applied Life Science, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan,Liver Research Project Center, Hiroshima University, Hiroshima, Japan,Laboratory for Digestive Diseases, RIKEN Center for Integrative Medical Sciences, Hiroshima, Japan
| | - C. Nelson Hayes
- Department of Gastroenterology and Metabolism, Applied Life Science, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan,Liver Research Project Center, Hiroshima University, Hiroshima, Japan,Laboratory for Digestive Diseases, RIKEN Center for Integrative Medical Sciences, Hiroshima, Japan
| | - Daiki Miki
- Liver Research Project Center, Hiroshima University, Hiroshima, Japan,Laboratory for Digestive Diseases, RIKEN Center for Integrative Medical Sciences, Hiroshima, Japan
| | - Mayuko Furuta
- Laboratory for Genome Sequencing Analysis, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Tatsuhiko Tsunoda
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Satoru Miyano
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Michiaki Kubo
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hiroshi Aikata
- Department of Gastroenterology and Metabolism, Applied Life Science, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Hidenori Ochi
- Department of Gastroenterology and Metabolism, Applied Life Science, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan,Liver Research Project Center, Hiroshima University, Hiroshima, Japan,Laboratory for Digestive Diseases, RIKEN Center for Integrative Medical Sciences, Hiroshima, Japan
| | - Yoshi-iku Kawakami
- Department of Gastroenterology and Metabolism, Applied Life Science, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Koji Arihiro
- Department of Anatomical Pathology, Hiroshima University School of Medicine, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastroenterological Surgery, Hiroshima University School of Medicine, Hiroshima, Japan
| | - Hidewaki Nakagawa
- Laboratory for Genome Sequencing Analysis, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan,Hidewaki Nakagawa, MD, PhD, Laboratory for Genome Sequencing Analysis, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan. fax: +81-3-5449-5785.
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Applied Life Science, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan,Liver Research Project Center, Hiroshima University, Hiroshima, Japan,Laboratory for Digestive Diseases, RIKEN Center for Integrative Medical Sciences, Hiroshima, Japan,Correspondence Address correspondence to: Kazuaki Chayama, MD, PhD, Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734–8551, Japan. fax: +81-82-255-6220.
| |
Collapse
|
326
|
Rota Caremoli E, Labianca R. Tivantinib: critical review with a focus on hepatocellular carcinoma. Expert Opin Investig Drugs 2015; 23:1563-74. [PMID: 25307444 DOI: 10.1517/13543784.2014.949339] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Sorafenib is the only approved agent for the treatment of advanced hepatocellular carcinoma (HCC). Sorafenib is an oral multikinase inhibitor that blocks several receptors involved in tumor cell proliferation and angiogenesis. The hepatocyte-growth factor/mesenchymal-epithelial transition (MET) factor pathway represents another emerging target in HCC. Tivantinib (ARQ 197) is an oral, selective small MET tyrosine kinase inhibitor with antitumor activity, especially in MET-high patients. Recent clinical data exhibit promising activity in HCC. AREAS COVERED This article reviews the preclinical and clinical data of tivantinib (published and ongoing trials), focusing on development in advanced HCC. Comments regarding the failure of trials with nonspecific drugs reported in the past 2 years are also included. EXPERT OPINION A randomized Phase II trial in second-line HCC showed a significant improvement in time to progression with tivantinib treatment in MET-high patients. Tivantinib remains in clinical development and has not yet been approved for any indication. A Phase III study in MET-high HCC is ongoing in a second-line setting, after sorafenib failure. In case of a survival benefit, tivantinib might become the first treatment for selected patients, based on MET status as a predictor. Therefore, there is a need for identifying HCC molecular subclasses and for developing a trial design based on molecular biomarkers.
Collapse
Affiliation(s)
- Elena Rota Caremoli
- Cancer Center Azienda Ospedaliera Papa Giovanni XXIII - Unit of Medical Oncology , Piazza OMS 1, 24127 Bergamo , Italy +39 035 267 3694, +39 035 267 3691 ; +39 035 267 4985 ;
| | | |
Collapse
|
327
|
Abstract
Mortality owing to liver cancer has increased in the past 20 years, and the latest estimates indicate that the global health burden of this disease will continue to grow. Most patients with hepatocellular carcinoma (HCC) are still diagnosed at intermediate or advanced disease stages, where curative approaches are often not feasible. Among the treatment options available, the molecular targeted agent sorafenib is able to significantly increase overall survival in these patients. Thereafter, up to seven large, randomized phase III clinical trials investigating other molecular therapies in the first-line and second-line settings have failed to improve on the results observed with this agent. Potential reasons for this include intertumour heterogeneity, issues with trial design and a lack of predictive biomarkers of response. During the past 5 years, substantial advances in our knowledge of the human genome have provided a comprehensive picture of commonly mutated genes in patients with HCC. This knowledge has not yet influenced clinical decision-making or current clinical practice guidelines. In this Review the authors summarize the molecular concepts of progression, discuss the potential reasons for clinical trial failure and propose new concepts of drug development, which might lead to clinical implementation of emerging targeted agents.
Collapse
|
328
|
Chen KW, Ou TM, Hsu CW, Horng CT, Lee CC, Tsai YY, Tsai CC, Liou YS, Yang CC, Hsueh CW, Kuo WH. Current systemic treatment of hepatocellular carcinoma: A review of the literature. World J Hepatol 2015; 7:1412-20. [PMID: 26052386 PMCID: PMC4450204 DOI: 10.4254/wjh.v7.i10.1412] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/29/2014] [Accepted: 03/30/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common form of human cancer worldwide and the third most common cause of cancer-related deaths. The strategies of various treatments for HCC depend on the stage of tumor, the status of patient's performance and the reserved hepatic function. The Barcelona Clinic Liver Cancer (BCLC) staging system is currently used most for patients with HCC. For example, for patients with BCLC stage 0 (very early stage) and stage A (early stage) HCC, the curable treatment modalities, including resection, transplantation and radiofrequency ablation, are taken into consideration. If the patients are in BCLC stage B (intermediate stage) and stage C (advanced stage) HCC, they may need the palliative transarterial chemoembolization and even the target medication of sorafenib. In addition, symptomatic treatment is always recommended for patients with BCLC stage D (end stage) HCC. In this review, we will attempt to summarize the historical perspective and the current developments of systemic therapies in BCLC stage B and C in HCC.
Collapse
Affiliation(s)
- Kai-Wen Chen
- Kai-Wen Chen, Department of Internal Medicine, Hualien Armed Forces General Hospital, Hualien 97144, Taiwan
| | - Tzu-Ming Ou
- Kai-Wen Chen, Department of Internal Medicine, Hualien Armed Forces General Hospital, Hualien 97144, Taiwan
| | - Chin-Wen Hsu
- Kai-Wen Chen, Department of Internal Medicine, Hualien Armed Forces General Hospital, Hualien 97144, Taiwan
| | - Chi-Ting Horng
- Kai-Wen Chen, Department of Internal Medicine, Hualien Armed Forces General Hospital, Hualien 97144, Taiwan
| | - Ching-Chang Lee
- Kai-Wen Chen, Department of Internal Medicine, Hualien Armed Forces General Hospital, Hualien 97144, Taiwan
| | - Yuh-Yuan Tsai
- Kai-Wen Chen, Department of Internal Medicine, Hualien Armed Forces General Hospital, Hualien 97144, Taiwan
| | - Chi-Chang Tsai
- Kai-Wen Chen, Department of Internal Medicine, Hualien Armed Forces General Hospital, Hualien 97144, Taiwan
| | - Yi-Sheng Liou
- Kai-Wen Chen, Department of Internal Medicine, Hualien Armed Forces General Hospital, Hualien 97144, Taiwan
| | - Chen-Chieh Yang
- Kai-Wen Chen, Department of Internal Medicine, Hualien Armed Forces General Hospital, Hualien 97144, Taiwan
| | - Chao-Wen Hsueh
- Kai-Wen Chen, Department of Internal Medicine, Hualien Armed Forces General Hospital, Hualien 97144, Taiwan
| | - Wu-Hsien Kuo
- Kai-Wen Chen, Department of Internal Medicine, Hualien Armed Forces General Hospital, Hualien 97144, Taiwan
| |
Collapse
|
329
|
Porta C, Giglione P, Ferrari A, Reversi F, Liguigli W, Imarisio I, Ganini C. Tivantinib (ARQ197) in hepatocellular carcinoma. Expert Rev Anticancer Ther 2015; 15:615-622. [DOI: 10.1586/14737140.2015.1050383] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
330
|
Granito A, Marinelli S, Terzi E, Piscaglia F, Renzulli M, Venerandi L, Benevento F, Bolondi L. Metronomic capecitabine as second-line treatment in hepatocellular carcinoma after sorafenib failure. Dig Liver Dis 2015; 47:518-522. [PMID: 25861840 DOI: 10.1016/j.dld.2015.03.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/27/2015] [Accepted: 03/10/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND No standard second-line treatments are available for hepatocellular carcinoma patients who fail sorafenib therapy. We assessed the safety and efficacy of metronomic capecitabine after first-line sorafenib failure. METHODS Retrospective analysis of consecutive hepatocellular carcinoma patients receiving metronomic capecitabine between January 2012 and November 2014. The primary end-point was safety, secondary end-point was efficacy, including time-to-progression and overall survival. RESULTS Twenty-six patients (80% Child-Pugh A, 80% Barcelona Clinic Liver Cancer stage C) received metronomic capecitabine (500 mg/bid). Median treatment duration was 3.2 months (range 0.6-31). Fourteen (53%) patients experienced at least one adverse event. The most frequent drug-related adverse events were bilirubin elevation (23%), fatigue (15%), anaemia (11%), lymphoedema (11%), and hand-foot syndrome (7.6%). Treatment was interrupted in 19 (73%) for disease progression, in 4 (15%) for liver deterioration, and in 1 (3.8%) for adverse event. Disease control was achieved in 6 (23%) patients. Median time-to-progression was 4 months (95% confidence interval 3.2-4.7). Median overall survival was 8 months (95% confidence interval 3.7-12.3). CONCLUSIONS Metronomic capecitabine was well tolerated in hepatocellular carcinoma patients who had been treated with sorafenib. Preliminary data show potential anti-tumour activity with long-lasting disease control in a subgroup of patients that warrants further evaluation in a phase III study.
Collapse
Affiliation(s)
- Alessandro Granito
- Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy.
| | - Sara Marinelli
- Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Eleonora Terzi
- Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Fabio Piscaglia
- Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Matteo Renzulli
- Radiology Unit, Department of Digestive Diseases and Internal Medicine, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Laura Venerandi
- Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Francesca Benevento
- Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Luigi Bolondi
- Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| |
Collapse
|
331
|
Slotta JE, Kollmar O, Ellenrieder V, Ghadimi BM, Homayounfar K. Hepatocellular carcinoma: Surgeon's view on latest findings and future perspectives. World J Hepatol 2015; 7:1168-1183. [PMID: 26019733 PMCID: PMC4438492 DOI: 10.4254/wjh.v7.i9.1168] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 11/14/2014] [Accepted: 03/20/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common liver-derived malignancy with a high fatality rate. Risk factors for the development of HCC have been identified and are clearly described. However, due to the lack of tumor-specific symptoms, HCC are diagnosed at progressed tumor stages in most patients, and thus curative therapeutic options are limited. The focus of this review is on surgical therapeutic options which can be offered to patients with HCC with special regard to recent findings, not exclusively focused on surgical therapy, but also to other treatment modalities. Further, potential promising future perspectives for the treatment of HCC are discussed.
Collapse
|
332
|
Patel A, Sun W. Molecular targeted therapy in hepatocellular carcinoma: from biology to clinical practice and future. Curr Treat Options Oncol 2015; 15:380-94. [PMID: 24838298 DOI: 10.1007/s11864-014-0291-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OPINION STATEMENT Hepatocellular carcinoma (HCC) is one of the most lethal cancers globally, particularly in certain regions of the world. Although the major risk factors for HCC have been identified, the specific mechanisms driving hepatocarcinogenesis remain unclear. Sorafenib is the only systemic therapy that has demonstrated an overall survival benefit in patients with advanced HCC and does so primarily through antiangiogenic activity. However, that actual benefit is still relatively small. Extensive research has focused on targeting dysfunctional molecular pathways in HCC. Despite promising preclinical and early-phase studies, other agents have failed to expand upon the efficacy of sorafenib in large-scale randomized trials. As the development of treatment options in the post-sorafenib setting is ongoing, more efforts are being focused on (1) evaluation of molecular agents targeting pathogenic, HCC-specific pathways; (2) the combination of targeted and cytotoxic therapies in selected subgroups; and (3) the combination of systemic and locoregional therapies in various settings. This article provides a review of recently completed and ongoing studies of molecular targeted agents in HCC, including a brief description of the biologic rationale behind these agents.
Collapse
Affiliation(s)
- Anuj Patel
- University of Pittsburgh Cancer Institute, 5150 Centre Ave, 5th Floor, Pittsburgh, PA, 19232, USA
| | | |
Collapse
|
333
|
Steinway SN, Dang H, You H, Rountree CB, Ding W. The EGFR/ErbB3 Pathway Acts as a Compensatory Survival Mechanism upon c-Met Inhibition in Human c-Met+ Hepatocellular Carcinoma. PLoS One 2015; 10:e0128159. [PMID: 26000702 PMCID: PMC4441360 DOI: 10.1371/journal.pone.0128159] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 04/22/2015] [Indexed: 12/22/2022] Open
Abstract
Background c-Met, a high-affinity receptor for Hepatocyte Growth Factor (HGF), plays a critical role in tumor growth, invasion, and metastasis. Hepatocellular carcinoma (HCC) patients with activated HGF/c-Met signaling have a significantly worse prognosis. Targeted therapies using c-Met tyrosine kinase inhibitors are currently in clinical trials for HCC, although receptor tyrosine kinase inhibition in other cancers has demonstrated early success. Unfortunately, therapeutic effect is frequently not durable due to acquired resistance. Methods We utilized the human MHCC97-H c-Met positive (c-Met+) HCC cell line to explore the compensatory survival mechanisms that are acquired after c-Met inhibition. MHCC97-H cells with stable c-Met knockdown (MHCC97-H c-Met KD cells) were generated using a c-Met shRNA vector with puromycin selection and stably transfected scrambled shRNA as a control. Gene expression profiling was conducted, and protein expression was analyzed to characterize MHCC97-H cells after blockade of the c-Met oncogene. A high-throughput siRNA screen was performed to find putative compensatory survival proteins, which could drive HCC growth in the absence of c-Met. Findings from this screen were validated through subsequent analyses. Results We have previously demonstrated that treatment of MHCC97-H cells with a c-Met inhibitor, PHA665752, results in stasis of tumor growth in vivo. MHCC97-H c-Met KD cells demonstrate slower growth kinetics, similar to c-Met inhibitor treated tumors. Using gene expression profiling and siRNA screening against 873 kinases and phosphatases, we identified ErbB3 and TGF-α as compensatory survival factors that are upregulated after c-Met inhibition. Suppressing these factors in c-Met KD MHCC97-H cells suppresses tumor growth in vitro. In addition, we found that the PI3K/Akt signaling pathway serves as a negative feedback signal responsible for the ErbB3 upregulation after c-Met inhibition. Furthermore, in vitro studies demonstrate that combination therapy with PHA665752 and Gefitinib (an EGFR inhibitor) significantly reduced cell viability and increased apoptosis compared with either PHA665752 or Gefitinib treatment alone. Conclusion c-Met inhibition monotherapy is not sufficient to eliminate c-Met+ HCC tumor growth. Inhibition of both c-Met and EGFR oncogenic pathways provides superior suppression of HCC tumor growth. Thus, combination of c-Met and EGFR inhibition may represent a superior therapeutic regimen for c-Met+ HCC.
Collapse
Affiliation(s)
- Steven N. Steinway
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Hien Dang
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Hanning You
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - C. Bart Rountree
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Wei Ding
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
334
|
Colagrande S, Regini F, Taliani GG, Nardi C, Inghilesi AL. Advanced hepatocellular carcinoma and sorafenib: Diagnosis, indications, clinical and radiological follow-up. World J Hepatol 2015; 7:1041-1053. [PMID: 26052393 PMCID: PMC4450181 DOI: 10.4254/wjh.v7.i8.1041] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 12/27/2014] [Accepted: 01/30/2015] [Indexed: 02/06/2023] Open
Abstract
Advanced stage hepatocellular carcinoma (HCC) is a category of disease defined by radiological, clinical and hepatic function parameters, comprehending a wide range of patients with different general conditions. The main therapeutic option is represented by sorafenib treatment, a multi-kinase inhibitor with anti-proliferative and anti-angiogenic effect. Trans-arterial Radio Embolization also represents a promising new approach to intermediate/advanced HCC. Post-marketing clinical studies showed that only a portion of patients actually benefits from sorafenib treatment, and an even smaller percentage of patients treated shows partial/complete response on follow-up examinations, up against relevant costs and an incidence of drug related adverse effects. Although the treatment with sorafenib has shown a significant increase in mean overall survival in different studies, only a part of patients actually shows real benefits, while the incidence of drug related significant adverse effects and the economic costs are relatively high. Moreover, only a small percentage of patients also shows a response in terms of lesion dimensions reduction. Being able to properly differentiate patients who are responding to the therapy from non-responders as early as possible is then still difficult and could be a pivotal challenge for the future; in fact it could spare several patients a therapy often difficult to bear, directing them to other second line treatments (many of which are at the moment still under investigation). For this reason, some supplemental criteria to be added to the standard modified Response Evaluation Criteria in Solid Tumors evaluation are being searched for. In particular, finding some parameters (cellular density, perfusion grade and enhancement rate) able to predict the sensitivity of the lesions to anti-angiogenic agents could help in stratifying patients in terms of treatment responsiveness before the beginning of the therapy itself, or in the first weeks of sorafenib treatment. This would bring a strongly desirable help in clinical managements of these patients.
Collapse
Affiliation(s)
- Stefano Colagrande
- Stefano Colagrande, Francesco Regini, Gian Giacomo Taliani, Cosimo Nardi, Dipartimento di Scienze Biomediche, Sperimentali e Cliniche, Università degli Studi di Firenze-Azienda Ospedaliero-Universitaria Careggi, 50134 Florence, Italy
| | - Francesco Regini
- Stefano Colagrande, Francesco Regini, Gian Giacomo Taliani, Cosimo Nardi, Dipartimento di Scienze Biomediche, Sperimentali e Cliniche, Università degli Studi di Firenze-Azienda Ospedaliero-Universitaria Careggi, 50134 Florence, Italy
| | - Gian Giacomo Taliani
- Stefano Colagrande, Francesco Regini, Gian Giacomo Taliani, Cosimo Nardi, Dipartimento di Scienze Biomediche, Sperimentali e Cliniche, Università degli Studi di Firenze-Azienda Ospedaliero-Universitaria Careggi, 50134 Florence, Italy
| | - Cosimo Nardi
- Stefano Colagrande, Francesco Regini, Gian Giacomo Taliani, Cosimo Nardi, Dipartimento di Scienze Biomediche, Sperimentali e Cliniche, Università degli Studi di Firenze-Azienda Ospedaliero-Universitaria Careggi, 50134 Florence, Italy
| | - Andrea Lorenzo Inghilesi
- Stefano Colagrande, Francesco Regini, Gian Giacomo Taliani, Cosimo Nardi, Dipartimento di Scienze Biomediche, Sperimentali e Cliniche, Università degli Studi di Firenze-Azienda Ospedaliero-Universitaria Careggi, 50134 Florence, Italy
| |
Collapse
|
335
|
Factors Associated with Post-Progression Survival in Patients with Advanced Hepatocellular Carcinoma Treated with Sorafenib. Diseases 2015; 3:68-77. [PMID: 28943609 PMCID: PMC5548234 DOI: 10.3390/diseases3020068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 04/19/2015] [Accepted: 05/06/2015] [Indexed: 12/16/2022] Open
Abstract
Sorafenib exerts modest antitumor activity in patients with advanced hepatocellular carcinoma (HCC), and radiological progressive disease (rPD) does not always correspond to so-called clinical progressive disease (cPD). We evaluated 101 patients who initiated sorafenib treatment for HCC and assessed post-progression survival (PPS) using the Cox proportional hazards model. PPS was calculated from the date of the first rPD until the date of death or the last follow-up. Using Cox model analysis of the 76 patients who experienced first rPD, we identified the Child-Pugh class, Eastern Cooperative Oncology Group performance status, the best antitumor response during treatment (using Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1) and α-fetoprotein levels as independent factors affecting PPS. When these factors were used to define scores ranging from zero to five with a cutoff value of two, PPS of patients who received best supportive care (BSC) after rPD was not statistically significantly different from that of patients who received post-rPD therapy with scores ≥2 (p = 0.220). In contrast, the PPS for the post-rPD therapy group was significantly longer compared with the BSC patients with scores <2 (p < 0.001). Patients who scored ≥2 at their first rPD were judged cPD and as candidates for BSC.
Collapse
|
336
|
Qi XS, Guo XZ, Han GH, Li HY, Chen J. MET inhibitors for treatment of advanced hepatocellular carcinoma: A review. World J Gastroenterol 2015; 21:5445-5453. [PMID: 25987766 PMCID: PMC4427665 DOI: 10.3748/wjg.v21.i18.5445] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/09/2015] [Accepted: 02/11/2015] [Indexed: 02/06/2023] Open
Abstract
The current standard treatment option for advanced hepatocellular carcinoma (HCC) is sorafenib, but its clinical benefit is modest. In spite of many attempts, few drugs can provide any significant improvement of survival as the first- or second-line therapy of choice in phase III randomized controlled trials. Recently, the subgroup analysis of a phase II randomized controlled trial has shown that tivantinib, a selective MET inhibitor, can significantly improve the overall survival in patients with MET-positive advanced HCC after the failure or intolerance of a prior systemic therapy. These findings enlighten the role of MET inhibitors in the treatment of advanced HCC. In this paper, we review all ongoing and completed clinical trials regarding this topic. As for the first-line therapy of advanced HCC, INC280 and foretinib are being evaluated in 2 phase II single-arm trials; and MSC2156119J and golvatinib plus sorafenib are being compared with sorafenib alone in 2 phase II randomized controlled trials. As for the second-line therapy of advanced HCC, tivantinib and cabozantinib are being compared with placebo in 2 phase III randomized controlled trials.
Collapse
|
337
|
Neuzillet C, Couvelard A, Tijeras-Raballand A, de Mestier L, de Gramont A, Bédossa P, Paradis V, Sauvanet A, Bachet JB, Ruszniewski P, Raymond E, Hammel P, Cros J. High c-Met expression in stage I-II pancreatic adenocarcinoma: proposal for an immunostaining scoring method and correlation with poor prognosis. Histopathology 2015; 67:664-76. [PMID: 25809563 DOI: 10.1111/his.12691] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/14/2015] [Indexed: 12/11/2022]
Abstract
AIMS c-Met is an emerging biomarker in pancreatic ductal adenocarcinoma (PDAC); there is no consensus regarding the immunostaining scoring method for this marker. We aimed to assess the prognostic value of c-Met overexpression in resected PDAC, and to elaborate a robust and reproducible scoring method for c-Met immunostaining in this setting. METHODS AND RESULTS c-Met immunostaining was graded according to the validated MetMab score, a classic visual scale combining surface and intensity (SI score), or a simplified score (high c-Met: ≥ 20% of tumour cells with strong membranous staining), in stage I-II PDAC. A computer-assisted classification method (Aperio software) was developed. Clinicopathological parameters were correlated with disease-free survival (DFS) and overall survival(OS). One hundred and forty-nine patients were analysed retrospectively in a two-step process. Thirty-seven samples (whole slides) were analysed as a pre-run test. Reproducibility values were optimal with the simplified score (kappa = 0.773); high c-Met expression (7/37) was associated with shorter DFS [hazard ratio (HR) 3.456, P = 0.0036] and OS (HR 4.257, P = 0.0004). c-Met expression was concordant on whole slides and tissue microarrays in 87.9% of samples, and quantifiable with a specific computer-assisted algorithm. In the whole cohort (n = 131), patients with c-Met(high) tumours (36/131) had significantly shorter DFS (9.3 versus 20.0 months, HR 2.165, P = 0.0005) and OS (18.2 versus 35.0 months, HR 1.832, P = 0.0098) in univariate and multivariate analysis. CONCLUSIONS Simplified c-Met expression is an independent prognostic marker in stage I-II PDAC that may help to identify patients with a high risk of tumour relapse and poor survival.
Collapse
Affiliation(s)
- Cindy Neuzillet
- INSERM UMR1149.,Department of Digestive Oncology, Beaujon University Hospital, Clichy, France.,Paris 7 Denis Diderot University, Paris, France
| | - Anne Couvelard
- INSERM UMR1149.,Paris 7 Denis Diderot University, Paris, France.,Department of Pathology, Bichat-Beaujon University Hospital, Paris-Clichy, France
| | | | | | - Armand de Gramont
- Department of Medical Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Pierre Bédossa
- INSERM UMR1149.,Paris 7 Denis Diderot University, Paris, France.,Department of Pathology, Bichat-Beaujon University Hospital, Paris-Clichy, France
| | - Valérie Paradis
- INSERM UMR1149.,Paris 7 Denis Diderot University, Paris, France.,Department of Pathology, Bichat-Beaujon University Hospital, Paris-Clichy, France
| | - Alain Sauvanet
- Paris 7 Denis Diderot University, Paris, France.,Department of Biliary and Pancreatic Surgery, Beaujon University Hospital, Clichy, France
| | - Jean-Baptiste Bachet
- Department of Gastroenterology, Pitié-Salpêtrière University Hospital, Paris, France
| | - Philippe Ruszniewski
- INSERM UMR1149.,Paris 7 Denis Diderot University, Paris, France.,Department of Gastroenterology and Pancreatology, Beaujon University Hospital, Clichy, France
| | - Eric Raymond
- Department of Medical Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Pascal Hammel
- INSERM UMR1149.,Department of Digestive Oncology, Beaujon University Hospital, Clichy, France.,Paris 7 Denis Diderot University, Paris, France
| | - Jérôme Cros
- INSERM UMR1149.,Paris 7 Denis Diderot University, Paris, France.,Department of Pathology, Bichat-Beaujon University Hospital, Paris-Clichy, France
| |
Collapse
|
338
|
Valcz G, Sipos F, Tulassay Z, Molnar B, Yagi Y. Republished: Importance of carcinoma-associated fibroblast-derived proteins in clinical oncology. Postgrad Med J 2015; 91:1026-1031. [PMID: 25976496 DOI: 10.1136/postgradmedj-2014-202561rep] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Carcinoma-associated fibroblast (CAF) as prominent cell type of the tumour microenvironment has complex interaction with both the cancer cells and other non-neoplastic surrounding cells. The CAF-derived regulators and extracellular matrix proteins can support cancer progression by providing a protective microenvironment for the cancer cells via reduction of chemotherapy sensitivity. On the other hand, these proteins may act as powerful prognostic markers as well as potential targets of anticancer therapy. In this review, we summarise the clinical importance of the major CAF-derived signals influencing tumour behaviour and determining the outcome of chemotherapy.
Collapse
Affiliation(s)
- Gabor Valcz
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Ferenc Sipos
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Zsolt Tulassay
- Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary
| | - Bela Molnar
- Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary
| | - Yukako Yagi
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
339
|
Granito A, Guidetti E, Gramantieri L. c-MET receptor tyrosine kinase as a molecular target in advanced hepatocellular carcinoma. J Hepatocell Carcinoma 2015; 2:29-38. [PMID: 27508192 PMCID: PMC4918282 DOI: 10.2147/jhc.s77038] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
c-MET is the membrane receptor for hepatocyte growth factor (HGF), also known as scatter factor or tumor cytotoxic factor, a mitogenic growth factor for hepatocytes. HGF is mainly produced by cells of mesenchymal origin and it mainly acts on neighboring epidermal and endothelial cells, regulating epithelial growth and morphogenesis. HGF/MET signaling has been identified among the drivers of tumorigenesis in human cancers. As such, c-MET is a recognized druggable target, and against it, targeted agents are currently under clinical investigation. c-MET overexpression is a common event in a wide range of human malignancies, including gastric, lung, breast, ovary, colon, kidney, thyroid, and liver carcinomas. Despite c-MET overexpression being reported by a large majority of studies, no evidence for a c-MET oncogenic addiction exists in hepatocellular carcinoma (HCC). In particular, c-MET amplification is a rare event, accounting for 4%–5% of cases while no mutation has been identified in c-MET oncogene in HCC. Thus, the selection of patient subgroups more likely to benefit from c-MET inhibition is challenging. Notwithstanding, c-MET overexpression was reported to be associated with increased metastatic potential and poor prognosis in patients with HCC, providing a rationale for its therapeutic inhibition. Here we summarize the role of activated HGF/MET signaling in HCC, its prognostic relevance, and the implications for therapeutic approaches in HCC.
Collapse
Affiliation(s)
- Alessandro Granito
- Dipartimento di Scienze Mediche e Chirurgiche Università di Bologna, Bologna, Italy
| | - Elena Guidetti
- Dipartimento di Scienze Mediche e Chirurgiche Università di Bologna, Bologna, Italy
| | - Laura Gramantieri
- Dipartimento dell'Apparato Digerente, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Centro di Ricerca Biomedica Applicata (CRBA), Azienda Ospedaliero-Universitaria Policlinico S Orsola-Malpighi e Università di Bologna, Bologna, Italy
| |
Collapse
|
340
|
Deng GL, Zeng S, Shen H. Chemotherapy and target therapy for hepatocellular carcinoma: New advances and challenges. World J Hepatol 2015; 7:787-798. [PMID: 25914779 PMCID: PMC4404384 DOI: 10.4254/wjh.v7.i5.787] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/26/2014] [Accepted: 01/20/2015] [Indexed: 02/06/2023] Open
Abstract
Primary liver cancer is one of the commonest causes of death. Hepatocellular carcinoma (HCC) accounts for 90% of primary liver cancers. For patients with unresectable or metastatic HCC, conventional chemotherapy is of limited or no benefit. Sorafenib is the only systemic treatment to demonstrate a statistically significant but modest overall survival benefit, leading to an era of targeted agents. Many clinical trials of targeted drugs have been carried out with many more in progress. Some drugs like PTK787 showed potential benefits in the treatment of HCC. Despite these promising breakthroughs, patients with HCC still have a dismal prognosis. Recently, both a phase III trial of everolimus and a phase II clinical trial of trebananib failed to demonstrate effective antitumor activity in advanced HCC. Sorafenib still plays a pivotal role in advanced HCC, leading to further explorations to exert its maximum efficacy. Combinations targeted with chemotherapy or transarterial chemoembolization is now being tested and might bring about advances. New targeted agents such as mammalian target of rapamycin inhibitors are under investigation, as well as further exploration of the mechanism of hepatocarcinogenesis.
Collapse
|
341
|
Okusaka T, Aramaki T, Inaba Y, Nakamura S, Morimoto M, Moriguchi M, Sato T, Ikawa Y, Ikeda M, Furuse J. Phase I study of tivantinib in Japanese patients with advanced hepatocellular carcinoma: Distinctive pharmacokinetic profiles from other solid tumors. Cancer Sci 2015; 106:611-7. [PMID: 25711511 PMCID: PMC4452163 DOI: 10.1111/cas.12644] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/17/2015] [Accepted: 02/20/2015] [Indexed: 12/12/2022] Open
Abstract
A c-Met inhibitor tivantinib is a candidate anticancer agent for patients with hepatocellular carcinoma (HCC), and CYP2C19 is the key metabolic enzyme for tivantinib. Previous Japanese phase I studies in patients with solid tumors (except HCC) recommend 360 mg twice daily (BID) and 240 mg BID for CYP2C19 extensive metabolizers (EM) and poor metabolizers (PM), respectively. In this study, Japanese patients with HCC in whom sorafenib treatment has failed were enrolled to evaluate the safety, tolerability and pharmacokinetics of oral tivantinib as a single agent. The dose was escalated separately in EM and PM, from 120 mg BID to 240 mg BID, in both capsule and tablet formulations. A total of 28 patients (EM: 21, PM: 7) received tivantinib. At a dose of 120 mg BID, dose-limiting toxicities (DLT) did not develop in 12 EM (capsule: 6, tablet: 6) and 7 PM (capsule: 4, tablet: 3) during the DLT-observation period (for 29 days after first dosing). At this dose, the pharmacokinetic profiles of tivantinib (AUC0–12 and Cmax) did not remarkably differ between EM and PM. When treated with 240 mg BID, 5 of 9 EM (capsule: 4 of 6, tablet: 1 of 3) developed neutropenia-related DLT accompanying plasma tivantinib concentration higher than expected from the previous studies. Consequently, PM did not receive 240 mg BID. In conclusion, 120 mg BID of tivantinib is recommended among Japanese patients with HCC regardless of CYP2C19 phenotype. This would be one of the first manuscripts demonstrating that the recommended dose of an anti-cancer agent for patients with hepatocellular carcinoma (HCC) could be distinct from its recommended dose for patients with other solid tumors.
Collapse
Affiliation(s)
- Takuji Okusaka
- Division of Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Chuo-ku, Japan
| | - Takeshi Aramaki
- Division of Interventional Radiology, Shizuoka Cancer Center, Shunto-gun, Japan
| | - Yoshitaka Inaba
- Department of Diagnostic and Interventional Radiology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Shinichiro Nakamura
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Manabu Morimoto
- Division of Hepatobiliary and Pancreatic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Michihisa Moriguchi
- Division of Interventional Radiology, Shizuoka Cancer Center, Shunto-gun, Japan
| | - Takashi Sato
- R&D Division, Kyowa Hakko Kirin Co., Ltd., Chiyoda-ku, Japan
| | - Yuta Ikawa
- R&D Division, Kyowa Hakko Kirin Co., Ltd., Chiyoda-ku, Japan
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Junji Furuse
- Department of Medical Oncology, Kyorin University School of Medicine, Mitaka, Japan
| |
Collapse
|
342
|
Zhai B, Jiang X, He C, Zhao D, Ma L, Xu L, Jiang H, Sun X. Arsenic trioxide potentiates the anti-cancer activities of sorafenib against hepatocellular carcinoma by inhibiting Akt activation. Tumour Biol 2015; 36:2323-2334. [PMID: 25416439 DOI: 10.1007/s13277-014-2839-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 11/11/2014] [Indexed: 01/05/2023] Open
Abstract
Sorafenib is the standard first-line systemic drug for advanced hepatocellular carcinoma (HCC), but it also induces the activation of Akt, which contributes to the mechanisms for the resistance to sorafenib. Arsenic trioxide (ATO) is a currently clinically used anticancer drug and displays its anticancer activities by inhibiting Akt activation. Therefore, we hypothesized that ATO may potentiate the anti-cancer activities of sorafenib against HCC. The results have demonstrated that ATO synergized with sorafenib to inhibit the proliferation and promote the apoptosis of HCC cells by diminishing the increased activation of Akt by sorafenib. ATO was shown to inhibit the expression or activation of Akt downstream factors, including glycogen synthase kinase (GSK)-3β, mammalian target of rapamycin (mTOR), ribosomal protein S6 kinase (S6K), and eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1), which regulate cell apoptosis and were upregulated or activated by sorafenib. Both sorafenib and ATO downregulated the expression of cyclin D1, resulting in HCC cells arrested at G0/G1 phase. ATO downregulated the expression of Bcl-2 and Bcl-xL and upregulated the expression of Bax, indicating that ATO could induce the apoptosis of HCC cells through the intrinsic pathways; but sorafenib showed little effects on these proteins of Bcl-2 family. ATO synergized with sorafenib to suppress the growth of HCC tumors established in mice by inhibiting the proliferation and inducing the apoptosis of HCC cells in situ. These results indicate that ATO may be a potential agent that given in combination with sorafenib acts synergistically for treating HCC.
Collapse
Affiliation(s)
- Bo Zhai
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | | | | | | | | | | | | | | |
Collapse
|
343
|
Bruix J, Han KH, Gores G, Llovet JM, Mazzaferro V. Liver cancer: Approaching a personalized care. J Hepatol 2015; 62:S144-56. [PMID: 25920083 PMCID: PMC4520430 DOI: 10.1016/j.jhep.2015.02.007] [Citation(s) in RCA: 228] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/03/2015] [Accepted: 02/04/2015] [Indexed: 12/04/2022]
Abstract
The knowledge and understanding of all aspects of liver cancer [this including hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA)] have experienced a major improvement in the last decades. New laboratory technologies have identified several molecular abnormalities that, at the very end, should provide an accurate stratification and optimal treatment of patients diagnosed with liver cancer. The seminal discovery of the TP53 hotspot mutation [1 ,2 ] was an initial landmark step for the future classification and treatment decision using conventional clinical criteria blended with molecular data. At the same time, the development of ultrasound, computed tomography (CT) and magnetic resonance (MR) has been instrumental for earlier diagnosis, accurate staging and treatment advances. Several treatment options with proven survival benefit if properly applied are now available. Major highlights include: i) acceptance of liver transplantation for HCC if within the Milan criteria [3 ], ii) recognition of ablation as a potentially curative option [4 ,5 ], iii) proof of benefit of chemoembolization (TACE), [6 ] and iv) incorporation of sorafenib as an effective systemic therapy [7 ]. These options are part of the widely endorsed BCLC staging and treatment model (Fig. 1 ) [8 ,9 ]. This is clinically useful and it will certainly keep evolving to accommodate new scientific evidence. This review summarises the data which are the basis for the current recommendations for clinical practice, while simultaneously exposes the areas where more research is needed to fulfil the still unmet needs (Table 1 ).
Collapse
Affiliation(s)
- Jordi Bruix
- Barcelona Clinic Liver Cancer Group (BCLC), Liver Unit, IDIBAPS, CIBERehd, Hospital Clínic, Universitat de Barcelona, Catalonia, Spain.
| | - Kwang-Hyub Han
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gregory Gores
- Mayo Clinic, Mayo College of Medicine, Rochester, MN, USA
| | - Josep Maria Llovet
- Barcelona Clinic Liver Cancer Group (BCLC), Liver Unit, IDIBAPS, CIBERehd, Hospital Clínic, Universitat de Barcelona, Catalonia, Spain; Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
| | - Vincenzo Mazzaferro
- Gastrointestinal Surgery and Liver Transplantation, Istituto Nazionale Tumori IRCCS (National Cancer Institute), Milan 20133, Italy
| |
Collapse
|
344
|
Tivantinib, a new option for second-line treatment of advanced hepatocellular carcinoma? The experience of Italian centers. TUMORI JOURNAL 2015; 101:139-43. [PMID: 25838254 DOI: 10.5301/tj.5000217] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2014] [Indexed: 12/22/2022]
Abstract
In the last decades the management of hepatocellular carcinoma (HCC) has undergone significant changes following the introduction of novel therapies such as sorafenib, which have improved patient survival. Nevertheless, HCC is still the third most common cause of cancer-related death worldwide. The evidence-based therapy for advanced HCC that is unsuitable for locoregional treatment is limited to sorafenib, with no second-line option available. This article focuses on the development of the MET inhibitor tivantinib in HCC as a promising treatment option for patients who failed sorafenib. A randomized, placebo-controlled phase II study showed activity of tivantinib in patients with high MET expression. Based on these results, the METIV-HCC phase III study in second-line treatment for MET-high patients was initiated to demonstrate the survival advantage of tivantinib compared to placebo.
Collapse
|
345
|
Dang H, Steinway SN, Ding W, Rountree CB. Induction of tumor initiation is dependent on CD44s in c-Met⁺ hepatocellular carcinoma. BMC Cancer 2015; 15:161. [PMID: 25886575 PMCID: PMC4380258 DOI: 10.1186/s12885-015-1166-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 03/05/2015] [Indexed: 12/16/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) patients with active hepatocyte growth factor (HGF)/c-Met signaling have a significantly worse prognosis. c-Met, a high affinity receptor for HGF, plays a critical role in cancer growth, invasion and metastasis. c-Met and CD44 have been utilized as cell surface markers to identify mesenchymal tumor-initiating stem-like cells (TISC) in several cancers including HCC. In this work, we examine the complex relationship between c-Met and CD44s (standard form), and investigate the specific role of CD44s as a tumor initiator and stemness marker in HCC. Methods Gene and protein expression assays were utilized to investigate the relationship between CD44s and c-Met in HCC cell lines. Tumor-sphere assays and in vivo tumor assays were performed to investigate the role of CD44+ cells as TISCs. Student’s t-test or one-way ANOVA with Tukeys post-hoc test was performed to test for differences amongst groups with a p < .05 as significant. Results In an immunohistochemical and immunoblot analysis of human HCC samples, we observed that more than 39% of human HCC samples express c-Met and CD44. To study the relationship between c-Met and CD44, we used MHCC97-H cells, which are CD44+/c-Met+. The knockdown of c-Met in MHCC97-H cells decreased CD44s, reduced TISC characteristics and decreased tumorsphere formation. Furthermore, we demonstrate that the inhibition of PI3K/AKT signaling decreased CD44s expression and subsequently decreased tumorsphere formation. The down-regulation of CD44s leads to a significant loss of a TISC and mesenchymal phenotype. Finally, the down-regulation of CD44s in MHCC97-H cells decreased tumor initiation in vivo compared with the scrambled control. Conclusions In summary, our data suggest that CD44s is modulated by the c-Met-PI3K-AKT signaling cascade to support a mesenchymal and TISC phenotype in HCC cells. Moreover, c-Met could be a potential therapeutic drug for targeting HCC cells with TISC and mesenchymal phenotypes. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1166-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hien Dang
- Department of Pediatrics and Pharmacology, The Pennsylvania State University, College of Medicine, Penn State Children's Hospital, Hershey, PA, USA. .,Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, 37 Convent Drive, Bethesda, MD, 20892, USA.
| | - Steven N Steinway
- Department of Pediatrics and Pharmacology, The Pennsylvania State University, College of Medicine, Penn State Children's Hospital, Hershey, PA, USA.
| | - Wei Ding
- Department of Pediatrics and Pharmacology, The Pennsylvania State University, College of Medicine, Penn State Children's Hospital, Hershey, PA, USA.
| | - Carl B Rountree
- Department of Pediatrics and Pharmacology, The Pennsylvania State University, College of Medicine, Penn State Children's Hospital, Hershey, PA, USA. .,Bon Secours St. Mary's Hospital, 5875 Bremo Road, MOB South Suite 303, Richmond, VA, 23226, USA.
| |
Collapse
|
346
|
Genome-Wide Expression of MicroRNAs Is Regulated by DNA Methylation in Hepatocarcinogenesis. Gastroenterol Res Pract 2015; 2015:230642. [PMID: 25861255 PMCID: PMC4377534 DOI: 10.1155/2015/230642] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 02/17/2015] [Accepted: 02/20/2015] [Indexed: 12/12/2022] Open
Abstract
Background. Previous studies, including ours, have examined the regulation of microRNAs (miRNAs) by DNA methylation, but whether this regulation occurs at a genome-wide level in hepatocellular carcinoma (HCC) is unclear. Subjects/Methods. Using a two-phase study design, we conducted genome-wide screening for DNA methylation and miRNA expression to explore the potential role of methylation alterations in miRNAs regulation. Results. We found that expressions of 25 miRNAs were statistically significantly different between tumor and nontumor tissues and perfectly differentiated HCC tumor from nontumor. Six miRNAs were overexpressed, and 19 were repressed in tumors. Among 133 miRNAs with inverse correlations between methylation and expression, 8 miRNAs (6%) showed statistically significant differences in expression between tumor and nontumor tissues. Six miRNAs were validated in 56 additional paired HCC tissues, and significant inverse correlations were observed for miR-125b and miR-199a, which is consistent with the inactive chromatin pattern found in HepG2 cells. Conclusion. These data suggest that the expressions of miR-125b and miR-199a are dramatically regulated by DNA hypermethylation that plays a key role in hepatocarcinogenesis.
Collapse
|
347
|
Cuestas ML, Oubiña JR, Mathet VL. Hepatocellular carcinoma and multidrug resistance: Past, present and new challenges for therapy improvement. World J Pharmacol 2015; 4:96-116. [DOI: 10.5497/wjp.v4.i1.96] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/02/2014] [Accepted: 12/01/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most frequent form of liver cancer and the third most common cause of cancer-related death in the world. The main risk factor worldwide for this type of malignancy is chronic hepatitis caused by hepatitis B virus and hepatitis C virus infections. Advances in early detection and treatment have improved life expectancy of patients with HCC. However, this disorder remains as a disease with poor prognosis. In fact, epidemiological studies have revealed that there is an 8-mo median survival rate in patients, approximately 20% of whom survive one year while only 5% remain alive after three years. Additionally, HCC is particularly difficult to treat because of its high recurrence rate, and its resistance to conventional chemotherapy is due, among other mechanisms, to several members of the ATP-Binding Cassette protein family involved in drug transport being overexpressed. Fortunately, there is evidence that these patients may benefit from alternative molecular-targeted therapies. This manuscript intends to provide further insight into the etiology and molecular mechanisms related to HCC development and the latest therapeutic approaches to treat this malignancy. The development of effective delivery systems of antitumor drugs able to target the liver parenchyma is also assessed. Finally, the prospects in the development of more efficient drug therapies to overcome multidrug resistance are also examined.
Collapse
|
348
|
FOLFIRI and Cetuximab Every Second Week for First-Line Treatment of KRAS Wild-Type Metastatic Colorectal Cancer According to Phosphatase and Tensin Homolog Expression: A Phase II Study. Clin Colorectal Cancer 2015; 14:162-9. [PMID: 25861836 DOI: 10.1016/j.clcc.2015.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 02/26/2015] [Accepted: 02/26/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Retrospective studies have suggested that phosphatase and tensin homolog (PTEN) expression might predict the efficacy of cetuximab in patients with KRAS wild-type metastatic colorectal cancer (mCRC). The present study was designed to prospectively evaluate the efficacy of first-line irinotecan, fluorouracil, and folinate (FOLFIRI) plus cetuximab every second week according to PTEN expression. PATIENTS AND METHODS Originally, patients with KRAS wild-type mCRC were randomly assigned to receive either FOLFIRI or cetuximab plus FOLFIRI (FOLFIRI-C). After a protocol amendment, the FOLFIRI arm was discontinued, and additional patients received FOLFIRI-C. Cox proportional hazard models were used to investigate the effect of PTEN and MET expression and BRAF and phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit α mutations on progression-free survival (PFS) and overall survival (OS). RESULTS A total of 35 and 54 patients received FOLFIRI and FOLFIRI-C, respectively. For the patients assigned to FOLFIRI and FOLFIRI-C, the median OS was 17.7 and 23.3 months and the median PFS was 8.2 and 6.6 months, respectively. For patients receiving FOLFIRI-C, the loss of PTEN expression did not affect PFS or OS. Significant interactions for PFS were detected between the MET expression levels (P = .047) and BRAF mutation (P = .018) and treatment. On univariate analysis, BRAF mutation was significantly associated with shorter OS for patients receiving either FOLFIRI-C (P = .016) or FOLFIRI (P = .035). Multivariate analysis confirmed the independent prognostic value of BRAF mutation on OS and that of MET expression levels on PFS (P = .025) and OS (P = .028) but only in the patients receiving FOLFIRI alone. Adverse events with FOLFIRI-C were consistent with those expected from FOLFIRI plus weekly cetuximab. CONCLUSION Although prospective analysis of PTEN did not allow a validation of the prognostic value of this biomarker, an every second week cetuximab schedule, in addition to first-line FOLFIRI, was effective and well tolerated. The possible predictive value of MET expression levels warrants additional investigation.
Collapse
|
349
|
Rimassa L, Personeni N, Santoro A. Tivantinib for hepatocellular carcinoma. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1009038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
350
|
Larson BK, Dhall D, Guindi M. Immunohistochemistry as a surrogate for molecular diagnosis in hepatic tumours. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.mpdhp.2015.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|