51
|
Takiguchi S, Inoue K, Matsusue K, Furukawa M, Teramoto N, Iguchi H. Crizotinib, a MET inhibitor, prevents peritoneal dissemination in pancreatic cancer. Int J Oncol 2017; 51:184-192. [PMID: 28498406 DOI: 10.3892/ijo.2017.3992] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/18/2017] [Indexed: 11/06/2022] Open
Abstract
Peritoneal dissemination is a frequent occurrence in pancreatic cancer, which is associated with a poor prognosis. MET is associated with the progression of pancreatic cancer; therefore, we evaluated the effect of a MET inhibitor, crizotinib, on peritoneal dissemination of pancreatic cancer. Crizotinib inhibited the growth of 8 pancreatic cancer cell lines with the IC50 ranging from 1.4 to 4.3 µM. Invasion of the pancreatic cancer cell line Suit-2, was suppressed in vitro at a concentration of 1.0 µM, which is sufficient for the inhibition of MET phosphorylation. This effect on cell invasion was also recapitulated by the reduction of MET expression in Suit-2 with siRNA. Crizotinib also inhibited RhoA activation in addition to MET phosphorylation. We further evaluated the effect of crizotinib on peritoneal dissemination of pancreatic cancer in vivo. Crizotinib reduced tumor burden and ascites accumulation due to development of peritoneal dissemination after inoculation of Suit-2. Taken together, crizotinib may be a potent drug for treating peritoneal dissemination of pancreatic cancer by inhibiting cancer cell proliferation and invasion, at least in part through the suppression of HGF/MET signaling and RhoA activation.
Collapse
Affiliation(s)
- Soichi Takiguchi
- Clinical Research Institute, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Kazuko Inoue
- Clinical Research Institute, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Kimihiko Matsusue
- Department of Hygiene Chemistry, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan
| | - Masayuki Furukawa
- Department of Hepato-Biliary-Pancreatology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Norihiro Teramoto
- Division of Pathology, National Hospital Organization Shikoku Cancer Center, Matsuyama 791-0280, Japan
| | - Haruo Iguchi
- Clinical Research Institute, National Hospital Organization Shikoku Cancer Center, Matsuyama 791-0280, Japan
| |
Collapse
|
52
|
Rovithi M, Avan A, Funel N, Leon LG, Gomez VE, Wurdinger T, Griffioen AW, Verheul HMW, Giovannetti E. Development of bioluminescent chick chorioallantoic membrane (CAM) models for primary pancreatic cancer cells: a platform for drug testing. Sci Rep 2017; 7:44686. [PMID: 28304379 PMCID: PMC5356332 DOI: 10.1038/srep44686] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 02/13/2017] [Indexed: 01/17/2023] Open
Abstract
The aim of the present study was to develop chick-embryo chorioallantoic membrane (CAM) bioluminescent tumor models employing low passage cell cultures obtained from primary pancreatic ductal adenocarcinoma (PDAC) cells. Primary PDAC cells transduced with lentivirus expressing Firefly-luciferase (Fluc) were established and inoculated onto the CAM membrane, with >80% engraftment. Fluc signal reliably correlated with tumor growth. Tumor features were evaluated by immunohistochemistry and genetic analyses, including analysis of mutations and mRNA expression of PDAC pivotal genes, as well as microRNA (miRNA) profiling. These studies showed that CAM tumors had histopathological and genetic characteristic comparable to the original tumors. We subsequently tested the modulation of key miRNAs and the activity of gemcitabine and crizotinib on CAM tumors, showing that combination treatment resulted in 63% inhibition of tumor growth as compared to control (p < 0.01). These results were associated with reduced expression of miR-21 and increased expression of miR-155. Our study provides the first evidence that transduced primary PDAC cells can form tumors on the CAM, retaining several histopathological and (epi)genetic characteristics of original tumors. Moreover, our results support the use of these models for drug testing, providing insights on molecular mechanisms underlying antitumor activity of new drugs/combinations.
Collapse
Affiliation(s)
- Maria Rovithi
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Department of Internal Medicine, Agios Nikolaos General Hospital, Agios Nikolaos, Crete, Greece
| | - Amir Avan
- Molecular Medicine Group, Department of Modern Sciences and Technologies; School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niccola Funel
- Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Leticia G. Leon
- Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Valentina E. Gomez
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Thomas Wurdinger
- Department of Neurosurgery, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, Boston, Massachusetts, US
| | - Arjan W. Griffioen
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Henk M. W. Verheul
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| |
Collapse
|
53
|
Giovannetti E, Leon LG, Gómez VE, Zucali PA, Minutolo F, Peters GJ. A specific inhibitor of lactate dehydrogenase overcame the resistance toward gemcitabine in hypoxic mesothelioma cells, and modulated the expression of the human equilibrative transporter-1. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2016; 35:643-651. [PMID: 27906635 DOI: 10.1080/15257770.2016.1149193] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/26/2016] [Indexed: 02/07/2023]
Abstract
Malignant pleural mesothelioma (MPM) is a very hypoxic malignancy, and hypoxia has been associated with resistance towards gemcitabine. The muscle-isoform of lactate dehydrogenase (LDH-A) constitutes a major checkpoint for the switch to anaerobic glycolysis. Therefore we investigated the combination of a new LDH-A inhibitor (NHI-1) with gemcitabine in MPM cell lines. Under hypoxia (O2 tension of 1%) the cell growth inhibitory effects of gemcitabine, were reduced, as demonstrated by a 5- to 10-fold increase in IC50s. However, the simultaneous addition of NHI-1 was synergistic (combination index < 1). Flow cytometry demonstrated that hypoxia caused a G1 arrest, whereas the combination of NHI-1 significantly increased gemcitabine-induced cell death. Finally, the mRNA expression levels of the human equilibrative transporter-1 (hENT1) were significantly down-regulated under hypoxia, but treatment with NHI-1 was associated with a recovery of hENT1 expression. In conclusion, our data show that hypoxia increased MPM resistance to gemcitabine. However, cell death induction and modulation of the key transporter in gemcitabine uptake may contribute to the synergistic interaction of gemcitabine with the LDH-A inhibitor NHI-1 and support further studies for the rational development of this combination.
Collapse
Affiliation(s)
- Elisa Giovannetti
- a Department Medical Oncology , VU University Medical Center , Amsterdam , The Netherlands
- b Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa , Pisa , Italy
| | - Leticia G Leon
- a Department Medical Oncology , VU University Medical Center , Amsterdam , The Netherlands
- b Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa , Pisa , Italy
- c University of La Laguna , La Laguna , Spain
| | - Valentina E Gómez
- a Department Medical Oncology , VU University Medical Center , Amsterdam , The Netherlands
| | - Paolo A Zucali
- d Department Oncology , Humanitas Cancer Center , Milano , Italy
| | | | - Godefridus J Peters
- a Department Medical Oncology , VU University Medical Center , Amsterdam , The Netherlands
| |
Collapse
|
54
|
Caparello C, Meijer LL, Garajova I, Falcone A, Le Large TY, Funel N, Kazemier G, Peters GJ, Vasile E, Giovannetti E. FOLFIRINOX and translational studies: Towards personalized therapy in pancreatic cancer. World J Gastroenterol 2016; 22:6987-7005. [PMID: 27610011 PMCID: PMC4988311 DOI: 10.3748/wjg.v22.i31.6987] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/09/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is an extremely aggressive disease; although progress has been made in the last few years, the prognosis of these patients remains dismal. FOLFIRINOX is now considered a standard treatment in first-line setting, since it demonstrated an improved overall and progression-free survival vs gemcitabine alone. However, the enthusiasm over the benefit of this three-drug regimen is tempered by the associated increased toxicity profile, and many efforts have been made to improve the feasibility of this schedule. After a more recent phase III trial showing an improved outcome over gemcitabine, the combination of gemcitabine/nab-paclitaxel emerged as another standard first-line treatment. However, this treatment is also associated with more side effects. In addition, despite initial promising data on the predictive role of SPARC levels, recent studies showed that these levels are not associated with nab-paclitaxel efficacy. The choice to use this treatment over FOLFIRINOX is therefore a topic of debate, also because no validated biomarkers to guide FOLFIRINOX treatment are available. In the era of actionable mutations and target agents it would be desirable to identify molecular factors or biomarkers to predict response to therapy in order to maximize the efficacy of treatment and avoid useless toxic effects for non-responding patients. However, until today the milestone of treatment for pancreatic cancer remains chemotherapy combinations, without predictive or monitoring tools existing to optimize therapy. This review analyzes the state-of-the-art treatments, promises and limitations of targeted therapies, ongoing trials and future perspectives, including potential role of microRNAs as predictive biomarkers.
Collapse
|
55
|
Delitto D, Zhang D, Han S, Black BS, Knowlton AE, Vlada AC, Sarosi GA, Behrns KE, Thomas RM, Lu X, Liu C, George TJ, Hughes SJ, Wallet SM, Trevino JG. Nicotine Reduces Survival via Augmentation of Paracrine HGF-MET Signaling in the Pancreatic Cancer Microenvironment. Clin Cancer Res 2016; 22:1787-1799. [PMID: 26667487 PMCID: PMC4818664 DOI: 10.1158/1078-0432.ccr-15-1256] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 11/26/2015] [Indexed: 01/18/2023]
Abstract
PURPOSE The relationship between smoking and pancreatic cancer biology, particularly in the context of the heterogeneous microenvironment, remains incompletely defined. We hypothesized that nicotine exposure would lead to the augmentation of paracrine growth factor signaling between tumor-associated stroma (TAS) and pancreatic cancer cells, ultimately resulting in accelerated tumor growth and metastasis. EXPERIMENTAL DESIGN The effect of tobacco use on overall survival was analyzed using a prospectively maintained database of surgically resected patients with pancreatic cancer. Nicotine exposure was evaluated in vitro using primary patient-derived TAS and pancreatic cancer cells independently and in coculture. Nicotine administration was then assessed in vivo using a patient-derived pancreatic cancer xenograft model. RESULTS Continued smoking was associated with reduced overall survival after surgical resection. In culture, nicotine-stimulated hepatocyte growth factor (HGF) secretion in primary patient-derived TAS and nicotine stimulation was required for persistent pancreatic cancer cell c-Met activation in a coculture model. c-Met activation in this manner led to the induction of inhibitor of differentiation-1 (Id1) in pancreatic cancer cells, previously established as a mediator of growth, invasion and chemoresistance. HGF-induced Id1 expression was abrogated by both epigenetic and pharmacologic c-Met inhibition. In patient-derived pancreatic cancer xenografts, nicotine treatment augmented tumor growth and metastasis; tumor lysates from nicotine-treated mice demonstrated elevated HGF expression by qRT-PCR and phospho-Met levels by ELISA. Similarly, elevated levels of phospho-Met in surgically resected pancreatic cancer specimens correlated with reduced overall survival. CONCLUSIONS Taken together, these data demonstrate a novel, microenvironment-dependent paracrine signaling mechanism by which nicotine exposure promotes the growth and metastasis of pancreatic cancer.
Collapse
Affiliation(s)
- Daniel Delitto
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Dongyu Zhang
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Song Han
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Brian S Black
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Andrea E Knowlton
- Department of Periodontology and Oral Biology, University of Florida Health Science Center, Gainesville, Florida
| | - Adrian C Vlada
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - George A Sarosi
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida. North Florida/South Georgia Veterans Health System, University of Florida Health Science Center, Gainesville, Florida
| | - Kevin E Behrns
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Ryan M Thomas
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida. North Florida/South Georgia Veterans Health System, University of Florida Health Science Center, Gainesville, Florida
| | - Xiaomin Lu
- Department of Biostatistics and Children's Oncology Group, University of Florida Health Science Center, Gainesville, Florida
| | - Chen Liu
- Department of Pathology, Immunology, Laboratory Medicine, Colleges of Medicine, Dentistry and Public Health and Health Professions, University of Florida Health Science Center, Gainesville, Florida
| | - Thomas J George
- Department of Internal Medicine, University of Florida Health Science Center, Gainesville, Florida
| | - Steven J Hughes
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Shannon M Wallet
- Department of Periodontology and Oral Biology, University of Florida Health Science Center, Gainesville, Florida
| | - Jose G Trevino
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida.
| |
Collapse
|
56
|
Garajová I, Giovannetti E, Biasco G, Peters GJ. c-Met as a Target for Personalized Therapy. TRANSLATIONAL ONCOGENOMICS 2015; 7:13-31. [PMID: 26628860 DOI: 10.4137/togog.s30534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/20/2015] [Accepted: 09/23/2015] [Indexed: 12/18/2022]
Abstract
MET and its ligand HGF are involved in many biological processes, both physiological and pathological, making this signaling pathway an attractive therapeutic target in oncology. Downstream signaling effects are transmitted via mitogen-activated protein kinase (MAPK), PI3K (phosphoinositide 3-kinase protein kinase B)/AKT, signal transducer and activator of transcription proteins (STAT), and nuclear factor-κB. The final output of the terminal effector components of these pathways is activation of cytoplasmic and nuclear processes leading to increases in cell proliferation, survival, mobilization and invasive capacity. In addition to its role as an oncogenic driver, increasing evidence implicates MET as a common mechanism of resistance to targeted therapies including EGFR and VEGFR inhibitors. In the present review, we summarize the current knowledge on the role of the HGF-MET signaling pathway in cancer and its therapeutic targeting (HGF activation inhibitors, HGF inhibitors, MET antagonists and selective/nonselective MET kinase inhibitors). Recent advances in understanding the role of this pathway in the resistance to current anticancer strategies used in lung, kidney and pancreatic cancer are discussed.
Collapse
Affiliation(s)
- Ingrid Garajová
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands. ; Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands. ; Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Guido Biasco
- Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
57
|
Garajová I, Giovannetti E, Biasco G, Peters GJ. c-Met as a Target for Personalized Therapy. TRANSLATIONAL ONCOGENOMICS 2015; 7:13-31. [PMID: 26628860 PMCID: PMC4659440 DOI: 10.4137/tog.s30534] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/20/2015] [Accepted: 09/23/2015] [Indexed: 12/30/2022]
Abstract
MET and its ligand HGF are involved in many biological processes, both physiological and pathological, making this signaling pathway an attractive therapeutic target in oncology. Downstream signaling effects are transmitted via mitogen-activated protein kinase (MAPK), PI3K (phosphoinositide 3-kinase protein kinase B)/AKT, signal transducer and activator of transcription proteins (STAT), and nuclear factor-κB. The final output of the terminal effector components of these pathways is activation of cytoplasmic and nuclear processes leading to increases in cell proliferation, survival, mobilization and invasive capacity. In addition to its role as an oncogenic driver, increasing evidence implicates MET as a common mechanism of resistance to targeted therapies including EGFR and VEGFR inhibitors. In the present review, we summarize the current knowledge on the role of the HGF-MET signaling pathway in cancer and its therapeutic targeting (HGF activation inhibitors, HGF inhibitors, MET antagonists and selective/nonselective MET kinase inhibitors). Recent advances in understanding the role of this pathway in the resistance to current anticancer strategies used in lung, kidney and pancreatic cancer are discussed.
Collapse
Affiliation(s)
- Ingrid Garajová
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, Sant’Orsola-Malpighi Hospital, Bologna, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Guido Biasco
- Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, Sant’Orsola-Malpighi Hospital, Bologna, Italy
| | - Godefridus J. Peters
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
58
|
TOMIZAWA MINORU, SHINOZAKI FUMINOBU, MOTOYOSHI YASUFUMI, SUGIYAMA TAKAO, YAMAMOTO SHIGENORI, ISHIGE NAOKI. SU11274 suppresses proliferation and motility of pancreatic cancer cells. Oncol Lett 2015; 10:1468-1472. [PMID: 26622692 PMCID: PMC4533741 DOI: 10.3892/ol.2015.3452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 06/16/2015] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal-epithelial transition factor (c-Met) is associated with the proliferation and motility of cancer cells. c-Met expression has been detected in surgical pancreatic cancer specimens, and its overexpression is associated with a poor prognosis. SU11274 is a specific inhibitor of c-Met. In the present study, the cell proliferation and motility of pancreatic cancer cells treated with SU11274 was investigated. The PANC-1, MIA-Paca2, NOR-P1, PK-45H, PK-1 and PK-59 pancreatic cancer cell lines were used. The expression of c-Met and cyclin D1 was analyzed by quantitative polymerase chain reaction. In addition, a 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium inner salt assay was performed to assess cell proliferation, and a scratch assay was performed to assess cell motility. c-Met expression was higher in PANC-1, PK-45H, PK-1 and PK-59 cell lines compared with that in normal pancreatic tissue. Following treatment with 30 µM SU11274, the proliferation of MIA-Paca2 and PK-45H cells was suppressed to 19.8±10.7% (P<0.05) and 45.8±14.8% (P<0.05) of the control level, respectively. Furthermore, cyclin D1 expression was downregulated to 43.7±17.9% (P<0.05) and 53.2±18.6% (P<0.05) of the control level in the MIA-Paca2 and PK-45H cell lines, respectively, following treatment with 30 µM SU11274. In addition, cell motility was reduced to 1.0±0.3% in MIA-Paca2 (P<0.05) and 14.7±3.5% in PK-45H (P<0.05) following treatment with 30 µM SU11274, compared with the motility of untreated cells. These results indicated that SU11274 suppresses the proliferation of pancreatic cancer cells via the downregulation of cyclin D1. The present study also demonstrated that cell motility was suppressed by treatment with SU11274.
Collapse
Affiliation(s)
- MINORU TOMIZAWA
- Department of Gastroenterology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - FUMINOBU SHINOZAKI
- Department of Radiology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - YASUFUMI MOTOYOSHI
- Department of Neurology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - TAKAO SUGIYAMA
- Department of Rheumatology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - SHIGENORI YAMAMOTO
- Department of Pediatrics, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - NAOKI ISHIGE
- Department of Neurosurgery, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| |
Collapse
|
59
|
Yan HH, Jung KH, Son MK, Fang Z, Kim SJ, Ryu YL, Kim J, Kim MH, Hong SS. Crizotinib exhibits antitumor activity by targeting ALK signaling not c-MET in pancreatic cancer. Oncotarget 2015; 5:9150-68. [PMID: 25193856 PMCID: PMC4253425 DOI: 10.18632/oncotarget.2363] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Crizotinib, a c-MET/ALK inhibitor, has exhibited antitumor efficacy in different types of cancers. However, studies regarding Crizotinib in pancreatic cancer have been limited. Thus, we investigated the effect of Crizotinib on pancreatic cancer and its mechanism of action. Crizotinib strongly suppressed the growth and proliferation of pancreatic cancer cells in a dose-dependent manner. Also, it induced apoptosis by modulating its related factors. In the study, with regard to the mechanism of action, Crizotinib did not inhibit c-MET expression on pancreatic cancer cells; instead, it specifically inhibited the activity of ALK, which was identified to be highly expressed on various pancreatic cancer cells and tissues in our study. In 42 different receptor tyrosine kinase (RTKs) array, Crizotinib also strongly inhibited the expression of activated ALK in pancreatic cancer cells, modulating its downstream mediators such as STAT3, AKT, and ERK. Furthermore, Crizotinib inhibited angiogenesis in a mouse Matrigel plug assay as well as the progression of tumor growth in a mouse xenograft model. Taken together, our investigation shows that Crizotinib inhibits the ALK signaling pathway in pancreatic cancer, resulting in cell growth/angiogenesis inhibition and apoptosis induction. We suggest that Crizotinib might be used as a novel therapeutic drug for treating pancreatic cancer.
Collapse
Affiliation(s)
- Hong Hua Yan
- College of Medicine, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea; These authors equally contributed to this work
| | - Kyung Hee Jung
- College of Medicine, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea; These authors equally contributed to this work
| | - Mi Kwon Son
- College of Medicine, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Zhenghuan Fang
- College of Medicine, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Soo Jung Kim
- College of Medicine, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Ye-Lim Ryu
- College of Medicine, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Juyoung Kim
- College of Medicine, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Mi-Hyun Kim
- School of Biological & Chemical Engineering, Yanbian University of Science & Technology, Beishan St., Yanji City, Jilin Prov., 133000, China
| | - Soon-Sun Hong
- College of Medicine, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| |
Collapse
|
60
|
ShahidSales S, Mobarhan MG, Ghasemi F, Gholamin S, Avan A. Low expression of chloride channel accessory 1 predicts a poor prognosis in colorectal cancer: The question is still open. Cancer 2015. [DOI: 10.1002/cncr.29598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Soodabeh ShahidSales
- School of Medicine; Cancer Research Center, Mashhad University of Medical Sciences; Mashhad Iran
| | - Majid Ghayour Mobarhan
- Department of Modern Sciences and Technologies; Biochemistry of Nutrition Research Center, School of Medicine, Mashhad University of Medical Sciences; Mashhad Iran
| | - Faezeh Ghasemi
- Department of Modern Sciences and Technologies; Biochemistry of Nutrition Research Center, School of Medicine, Mashhad University of Medical Sciences; Mashhad Iran
| | - Sharareh Gholamin
- Institute of Stem Cell Biology, and Regenerative Medicine, Stanford University School of Medicine; Stanford California
| | - Amir Avan
- Department of Modern Sciences and Technologies; Biochemistry of Nutrition Research Center, School of Medicine, Mashhad University of Medical Sciences; Mashhad Iran
- Department of Medical Oncology; VU University Medical Center Amsterdam; Amsterdam the Netherlands
| |
Collapse
|
61
|
Seicean A, Petrusel L, Seicean R. New targeted therapies in pancreatic cancer. World J Gastroenterol 2015; 21:6127-6145. [PMID: 26034349 PMCID: PMC4445091 DOI: 10.3748/wjg.v21.i20.6127] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/26/2015] [Accepted: 04/16/2015] [Indexed: 02/06/2023] Open
Abstract
Patients with pancreatic cancer have a poor prognosis with a median survival of 4-6 mo and a 5-year survival of less than 5%. Despite therapy with gemcitabine, patient survival does not exceed 6 mo, likely due to natural resistance to gemcitabine. Therefore, it is hoped that more favorable results can be obtained by using guided immunotherapy against molecular targets. This review summarizes the new leading targeted therapies in pancreatic cancers, focusing on passive and specific immunotherapies. Passive immunotherapy may have a role for treatment in combination with radiochemotherapy, which otherwise destroys the immune system along with tumor cells. It includes mainly therapies targeting against kinases, including epidermal growth factor receptor, Ras/Raf/mitogen-activated protein kinase cascade, human epidermal growth factor receptor 2, insulin growth factor-1 receptor, phosphoinositide 3-kinase/Akt/mTOR and hepatocyte growth factor receptor. Therapies against DNA repair genes, histone deacetylases, microRNA, and pancreatic tumor tissue stromal elements (stromal extracellular matric and stromal pathways) are also discussed. Specific immunotherapies, such as vaccines (whole cell recombinant, peptide, and dendritic cell vaccines), adoptive cell therapy and immunotherapy targeting tumor stem cells, have the role of activating antitumor immune responses. In the future, treatments will likely include personalized medicine, tailored for numerous molecular therapeutic targets of multiple pathogenetic pathways.
Collapse
|
62
|
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
|
63
|
Avan A, Avan A, Maftouh M, Mobarhan MG, Gholamin S. Pharmacogenomic modeling in pancreatic cancer—letter. Clin Cancer Res 2015; 21:1497. [PMID: 25770295 DOI: 10.1158/1078-0432.ccr-14-2951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Amir Avan
- Molecular Medicine Group, Department of Modern Sciences and Technologies and Biochemistry of Nutrition Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. Department of Medical Oncology, VU University Medical Center Amsterdam, Amsterdam, the Netherlands.
| | - Abolfazl Avan
- Department of Medical Oncology, VU University Medical Center Amsterdam, Amsterdam, the Netherlands
| | - Mina Maftouh
- Department of Medical Oncology, VU University Medical Center Amsterdam, Amsterdam, the Netherlands
| | - Majid Ghayour Mobarhan
- Molecular Medicine Group, Department of Modern Sciences and Technologies and Biochemistry of Nutrition Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. Brighton and Sussex Medical School, Division of Medical Education, Falmer, Brighton, United Kingdom
| | - Sharareh Gholamin
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
64
|
Targeting cMET with INC280 impairs tumour growth and improves efficacy of gemcitabine in a pancreatic cancer model. BMC Cancer 2015; 15:71. [PMID: 25884642 PMCID: PMC4340491 DOI: 10.1186/s12885-015-1064-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/30/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Expression and activation of the cMET receptor have been implicated in tumor progression and resistance to chemotherapy in human pancreatic cancer. In this regard we assessed the effects of targeting cMET in pancreatic cancer models in vitro and in vivo. METHODS Human (L3.6pl, BxP3, HPAF-II, MiaPaCa2) and murine (Panc02) pancreatic cancer cell lines, endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) were used for the experiments. Furthermore, the human pancreatic cancer cell line MiaPaCa2 with acquired resistance to gemcitabine was employed (MiaPaCa2(G250)). For targeting the cMET receptor, the oral available, ATP-competitive inhibitor INC280 was used. Effects of cMET inhibition on cancer and stromal cells were determined by growth assays, western blotting, motility assays and ELISA. Moreover, orthotopic xenogeneic and syngeneic mouse (BALB-C nu/nu; C57BL/6) models were used to assess in vivo efficacy of targeting cMET alone and in combination with gemcitabine. RESULTS Treatment with INC280 impairs activation of signaling intermediates in pancreatic cancer cells and ECs, particularly when cells were stimulated with hepatocyte growth factor (HGF). Moreover, motility of cancer cells and ECs in response to HGF was reduced upon treatment with INC280. Only minor effects on VSMCs were detected. Interestingly, MiaPaCa2(G250) showed an increase in cMET expression and cMET inhibition abrogated HGF-induced effects on growth, motility and signaling as well as DFX-hypoxia HIF-1alpha and MDR-1 expression in vitro. In vivo, therapy with INC280 alone led to inhibition of orthotopic tumor growth in xenogeneic and syngeneic models. Similar to in vitro results, cMET expression was increased upon treatment with gemcitabine, and combination of the cMET inhibitor with gemcitabine improved anti-neoplastic capacity in an orthotopic syngeneic model. Immunohistochemical analysis revealed a significant inhibition of tumor cell proliferation (Ki67) and tumor vascularization (CD31). Finally, combination of gemcitabine with INC280 significantly prolonged survival in the orthotopic syngeneic tumor model even when treatment with the cMET inhibitor was initiated at an advanced stage of disease. CONCLUSIONS These data provide evidence that targeting cMET in combination with gemcitabine may be effective in human pancreatic cancer and warrants further clinical evaluation.
Collapse
|
65
|
Avan A, Avan A, Le Large TYS, Mambrini A, Funel N, Maftouh M, Ghayour-Mobarhan M, Cantore M, Boggi U, Peters GJ, Pacetti P, Giovannetti E. AKT1 and SELP polymorphisms predict the risk of developing cachexia in pancreatic cancer patients. PLoS One 2014; 9:e108057. [PMID: 25238546 PMCID: PMC4169595 DOI: 10.1371/journal.pone.0108057] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 08/19/2014] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) patients have the highest risk of developing cachexia, which is a direct cause of reduced quality of life and shorter survival. Novel biomarkers to identify patients at risk of cachexia are needed and might have a substantial impact on clinical management. Here we investigated the prognostic value and association of SELP-rs6136, IL6-rs1800796 and AKT1-rs1130233 polymorphisms with cachexia in PDAC. Genotyping was performed in DNA from blood samples of a test and validation cohorts of 151 and 152 chemo-naive locally-advanced/metastatic PDAC patients, respectively. The association of SELP-rs6136, IL6-rs1800796 and AKT1-rs1130233 polymorphisms with cachexia as well as the correlation between cachexia and the candidate polymorphisms and overall survival were analyzed. Akt expression and phosphorylation in muscle biopsies were evaluated by specific ELISA assays. SELP-rs6136-AA and AKT1-rs1130233-AA/GA genotypes were associated with increased risk of developing cachexia in both cohorts (SELP: p = 0.011 and p = 0.045; AKT1: p = 0.004 and p = 0.019 for the first and second cohorts, respectively), while patients carrying AKT1-rs1130233-GG survived significantly longer (p = 0.002 and p = 0.004 for the first and second cohorts, respectively). In the multivariate analysis AKT1-rs1130233-AA/GA genotypes were significant predictors for shorter survival, with an increased risk of death of 1.7 (p = 0.002) and 1.6 (p = 0.004), in the first and second cohorts, respectively. This might be explained by the reduced phosphorylation of Akt1 in muscle biopsies from patients harboring AKT1-rs1130233-AA/GA (p = 0.003), favoring apoptosis induction. In conclusion, SELP and AKT1 polymorphisms may play a role in the risk of cachexia and death in PDAC patients, and should be further evaluated in larger prospective studies.
Collapse
Affiliation(s)
- Abolfazl Avan
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Amir Avan
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
- Biochemistry of Nutrition Research Center, and Department of New Sciences and Technology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tessa Y. S. Le Large
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Andrea Mambrini
- Department of Medical Oncology, Carrara Civic Hospital, Carrara, Italy
| | | | - Mina Maftouh
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Majid Ghayour-Mobarhan
- Biochemistry of Nutrition Research Center, and Department of New Sciences and Technology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maurizio Cantore
- Department of Medical Oncology, Carrara Civic Hospital, Carrara, Italy
| | - Ugo Boggi
- Start-Up Unit, University of Pisa, Pisa, Italy
| | - Godefridus J. Peters
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Paola Pacetti
- Department of Medical Oncology, Carrara Civic Hospital, Carrara, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
- Start-Up Unit, University of Pisa, Pisa, Italy
| |
Collapse
|
66
|
Ormanns S, Assmann G, Reu S, Gallmeier E, Bader DC, Kleespies A, Haas M, Kruger S, Heinemann V, Kirchner T, Boeck S. ALK expression is absent in pancreatic ductal adenocarcinoma. J Cancer Res Clin Oncol 2014; 140:1625-8. [PMID: 25017418 DOI: 10.1007/s00432-014-1774-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 07/03/2014] [Indexed: 10/25/2022]
Abstract
PURPOSE It has not yet been clearly defined whether anaplastic lymphoma kinase (ALK) expression can be detected in pancreatic ductal adenocarcinoma (PDAC). METHODS Within a retrospective study, archival PDAC surgical specimens were screened for ALK expression in tumor and normal tissue by immunohistochemistry (IHC) with the use of a specific ALK detection kit on a tissue microarray (TMA). RESULTS PDAC tumor tissue was available from 99 resected cases: fifty-eight out of 99 patients (59 %) had nodal-positive disease, and 80 patients (81 %) had pT3 tumors. Forty-nine patients underwent R0 resection, and in 48 cases, resection status was classified R1. Regarding ALK expression, five cases showed faint immunoreactivity on TMA, which was negative on whole mount sections. All other 94 cases showed no ALK expression. CONCLUSION In 99 PDAC cases, no ALK expression was detected by IHC; ALK thus may not serve as a relevant drug target in PDAC.
Collapse
Affiliation(s)
- Steffen Ormanns
- Institute of Pathology, Ludwig-Maximilians-University of Munich, Thalkirchnerstr. 36, 80337, Munich, Germany,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Maftouh M, Belo AI, Avan A, Funel N, Peters GJ, Giovannetti E, van Die I. Galectin-4 expression is associated with reduced lymph node metastasis and modulation of Wnt/β-catenin signalling in pancreatic adenocarcinoma. Oncotarget 2014; 5:5335-5349. [PMID: 24977327 PMCID: PMC4170638 DOI: 10.18632/oncotarget.2104] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 06/12/2014] [Indexed: 12/19/2022] Open
Abstract
Galectin-4 (Gal-4) has been recently identified as a pivotal factor in the migratory capabilities of a set of defined pancreatic ductal adenocarcinoma (PDAC) cell lines using zebrafish as a model system. Here we evaluated the expression of Gal-4 in PDAC tissues selected according to their lymph node metastatic status (N0 vs. N1), and investigated the therapeutic potential of targeting the cross-link with the Wnt signaling pathway in primary PDAC cells. Analysis of Gal-4 expression in PDACs showed high expression of Gal-4 in 80% of patients without lymph node metastasis, whereas 70% of patients with lymph node metastases had low Gal-4 expression. Accordingly, in primary PDAC cells high Gal-4 expression was negatively associated with migratory and invasive ability in vitro and in vivo. Knockdown of Gal-4 in primary PDAC cells with high Gal-4 expression resulted in significant increase of invasion (40%) and migration (50%, P<0.05), whereas enforced expression of Gal-4 in primary cells with low Gal-4 expression reduced the migratory and invasive behavior compared to the control cells. Gal-4 markedly reduces β-catenin levels in the cell, counteracting the function of Wnt signaling, as was assessed by down-regulation of survivin and cyclin D1. Furthermore, Gal-4 sensitizes PDAC cells to the Wnt inhibitor ICG-001, which interferes with the interaction between CREB binding protein (CBP) and β-catenin. Collectively, our data suggest that Gal-4 lowers the levels of cytoplasmic β-catenin, which may lead to lowered availability of nuclear β-catenin, and consequently diminished levels of nuclear CBP-β-catenin complex and reduced activation of the Wnt target genes. Our findings provide novel insights into the role of Gal-4 in PDAC migration and invasion, and support the analysis of Gal-4 for rational targeting of Wnt/β-catenin signaling in the treatment of PDAC.
Collapse
Affiliation(s)
- Mina Maftouh
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Ana I. Belo
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Amir Avan
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
- Biochemistry of Nutrition Research Center, and Department of New Sciences and Technology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Godefridus J. Peters
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
- Start-Up Unit, University of Pisa, Pisa, Italy
| | - Irma van Die
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
68
|
Schneider M, Schüler J, Höfflin R, Korzeniewski N, Grüllich C, Roth W, Teber D, Hadaschik B, Pahernik S, Hohenfellner M, Duensing S. Phenotypic drug screening and target validation for improved personalized therapy reveal the complexity of phenotype-genotype correlations in clear cell renal cell carcinoma. Urol Oncol 2014; 32:877-84. [PMID: 24929890 DOI: 10.1016/j.urolonc.2014.03.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 03/11/2014] [Accepted: 03/11/2014] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Novel personalized therapeutic approaches are urgently needed for patients with metastatic clear cell renal cell carcinoma (ccRCC). METHODS AND MATERIALS We combined the development of a primary patient-derived ccRCC cell line with a phenotypic drug screen consisting of 101 approved anticancer compounds. RESULTS We identified the MNNG HOS transforming gene (MET)-anaplastic lymphoma receptor tyrosine kinase (ALK) inhibitor crizotinib as the top hit of our drug screen, whereas compounds targeting the vascular endothelial growth factor (VEGF) or mammalian target of rapamycin (mTOR) pathway showed no or only minor in vitro activity. Among the known major crizotinib targets MET, ALK, and ROS-1, only MET was expressed in our ccRCC cell line. Subsequent sequence analysis revealed a heterozygous R988C mutation of the MET gene and a VHL deletion in both the primary tumor and the tumor-derived ccRCC cell line. However, we were unable to show an activation of MET and, further, MET knockdown did not result in increased apoptosis or cytotoxicity. Therefore, our results suggest that MET R988C does not function as a major oncogenic driver mutation but rather represents a sequence variant. However, we provide evidence that the cytotoxic effect of crizotinib in our cell line model correlates with its ability to inhibit P-glycoprotein (ABCB1)-associated transport functions. CONCLUSIONS Our study shows that a phenotypic screen of a patient-derived tumor cell line can identify compounds with antitumor activity but with an unexpected mode of action. Our results underscore that target validation and phenotype-genotype correlations remain a major experimental challenge. The implications of our findings for a personalized management of patients with cancer are discussed.
Collapse
Affiliation(s)
- Meike Schneider
- Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany; Molecular Urooncology, Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany
| | | | - Rouven Höfflin
- Molecular Urooncology, Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany
| | - Nina Korzeniewski
- Molecular Urooncology, Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany
| | - Carsten Grüllich
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg, Germany; Center for Kidney Tumors, National Center for Tumor Disease and University of Heidelberg School of Medicine, Heidelberg, Germany
| | - Wilfried Roth
- Center for Kidney Tumors, National Center for Tumor Disease and University of Heidelberg School of Medicine, Heidelberg, Germany; Department of Pathology, University of Heidelberg School of Medicine, Heidelberg, Germany
| | - Dogu Teber
- Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany; Center for Kidney Tumors, National Center for Tumor Disease and University of Heidelberg School of Medicine, Heidelberg, Germany
| | - Boris Hadaschik
- Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany; Center for Kidney Tumors, National Center for Tumor Disease and University of Heidelberg School of Medicine, Heidelberg, Germany
| | - Sascha Pahernik
- Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany; Center for Kidney Tumors, National Center for Tumor Disease and University of Heidelberg School of Medicine, Heidelberg, Germany
| | - Markus Hohenfellner
- Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany; Center for Kidney Tumors, National Center for Tumor Disease and University of Heidelberg School of Medicine, Heidelberg, Germany
| | - Stefan Duensing
- Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany; Molecular Urooncology, Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany; Center for Kidney Tumors, National Center for Tumor Disease and University of Heidelberg School of Medicine, Heidelberg, Germany.
| |
Collapse
|
69
|
Samore WR, Gondi CS. Brief overview of selected approaches in targeting pancreatic adenocarcinoma. Expert Opin Investig Drugs 2014; 23:793-807. [PMID: 24673265 DOI: 10.1517/13543784.2014.902933] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Pancreatic adenocarcinoma (PDAC) has the worst prognosis of any major malignancy, with 5-year survival painfully inadequate at under 5%. Investigators have struggled to target and exploit PDAC unique biology, failing to bring meaningful results from bench to bedside. Nonetheless, in recent years, several promising targets have emerged. AREAS COVERED This review will discuss novel drug approaches in development for use in PDAC. The authors examine the continued efforts to target Kirsten rat sarcoma viral oncogene homolog (KRas), which have recently been successfully abated using novel small interfering RNA (siRNA) eluting devices. The authors also discuss other targets relevant to PDAC including those downstream of mutated KRas, such as MAPK kinase and phosphatidylinositol 3-kinase. EXPERT OPINION Although studies into novel biomarkers and advanced imaging have highlighted the potential new avenues toward discovering localized tumors earlier, the current therapeutic options highlight the fact that PDAC is a highly metastatic and chemoresistant cancer that often must be fought with virulent, systemic therapies. Several newer approaches, including siRNA targeting of mutated KRas and enzymatic depletion of hyaluronan with PEGylated hyaluronidase are particularly exciting given their early stage results. Further research should help in elucidating their potential impact as therapeutic options.
Collapse
Affiliation(s)
- Wesley R Samore
- M3 student, University of Illinois College of Medicine , One Illini Drive Peoria, IL 61605 , USA
| | | |
Collapse
|
70
|
Colvin EK, Scarlett CJ. A historical perspective of pancreatic cancer mouse models. Semin Cell Dev Biol 2014; 27:96-105. [PMID: 24685616 DOI: 10.1016/j.semcdb.2014.03.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 03/20/2014] [Accepted: 03/21/2014] [Indexed: 12/22/2022]
Abstract
Pancreatic cancer is an inherently aggressive disease with an extremely poor prognosis and lack of effective treatments. Over the past few decades, much has been uncovered regarding the pathogenesis of pancreatic cancer and the underlying genetic alterations necessary for tumour initiation and progression. Much of what we know about pancreatic cancer has come from mouse models of this disease. This review focusses on the development of genetically engineered mouse models that phenotypically and genetically recapitulate human pancreatic cancer, as well as the increasing use of patient-derived xenografts for preclinical studies and the development of personalised medicine strategies.
Collapse
Affiliation(s)
- Emily K Colvin
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St Leonards, NSW, Australia.
| | - Christopher J Scarlett
- Pancreatic Cancer Research, Nutrition, Food and Health Research Group, School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW, Australia.
| |
Collapse
|
71
|
Giovannetti E, Wang Q, Avan A, Funel N, Lagerweij T, Lee JH, Caretti V, van der Velde A, Boggi U, Wang Y, Vasile E, Peters GJ, Wurdinger T, Giaccone G. Role of CYB5A in pancreatic cancer prognosis and autophagy modulation. J Natl Cancer Inst 2014; 106:djt346. [PMID: 24301457 PMCID: PMC3906994 DOI: 10.1093/jnci/djt346] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 10/28/2013] [Accepted: 10/29/2013] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Loss of 18q22.3 is a prognostic marker in pancreatic ductal adenocarcinoma (PDAC). This study investigated genes encoded by this cytoband. METHODS We studied mRNA/protein expression in radically resected (n = 130) and metastatic patients (n = 50). The role of CYB5A was tested in 11 PDAC cell lines and five primary cultures through retrovirus-mediated upregulation and small interfering RNA using wound-healing, invasion, annexin-V, electron microscopy, and autophagic assays, as well as autophagy genes and kinases arrays. CYB5A+ orthotopic models (n = 6 mice/group) were monitored by Firefly and Gaussia-luciferase bioluminescence, magnetic resonance imaging, and high-frequency ultrasound. Data were analyzed by t test, Fisher exact-test, log-rank test and Cox proportional hazards models. All statistical tests were two-sided. RESULTS Both resected and metastatic patients with low mRNA or protein expression of CYB5A had statistically significantly shorter survival (eg, median = 16.7 months, 95% confidence interval [CI] = 13.5 to 19.9; vs median = 24.8 months, 95% CI = 12.8 to 36.9; P = .02, two-sided log-rank test; n = 82 radically resected PDACs), and multivariable analyses confirmed prognostic relevance. Moreover, we characterized a novel function to CYB5A, autophagy induction, concomitant with reduced proliferation and migration/invasion of PDAC cells. Network analysis of proautophagic pathways suggested CYB5A interaction with TRAF6, which was confirmed by TRAF6 downregulation after CYB5A reconstitution (-69% in SU.86.86-CYB5A+; P = .005, two-sided t test). CYB5A silencing had opposite effects, restoring TRAF6 expression and wound healing. In vivo studies showed that CYB5A induced autophagy while inhibiting tumor growth/metastasis and increasing survival (median = 57 days, 95% CI = 52 to 61; vs median = 44 days, 95% CI = 21 to 57; P = .03, two-sided log-rank test). CONCLUSIONS These results define CYB5A as a novel prognostic factor for PDAC that exerts its tumor-suppressor function through autophagy induction and TRAF6 modulation.
Collapse
Affiliation(s)
- Elisa Giovannetti
- Affiliations of authors: Department of Medical Oncology (EG, AA, GJP) and Department of Neurosurgery (TL, VC, TW), VU University Medical Center, and Centre for Integrative Bioinformatics (AvdV), VU University, Amsterdam, the Netherlands; Department of Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, University of Pisa, Pisa, Italy (NF, UB, EV); Department of Neurology, Stanford University, Stanford, CA (VC); Department of Neurology, Massachusetts General Hospital and Neuroscience Program, Harvard Medical School, Boston, MA (TW); Medical Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD (QW, J-HL, YW, GG)
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|