51
|
Thomas RM, Gharaibeh RZ, Gauthier J, Beveridge M, Pope JL, Guijarro MV, Yu Q, He Z, Ohland C, Newsome R, Trevino J, Hughes SJ, Reinhard M, Winglee K, Fodor AA, Zajac-Kaye M, Jobin C. Intestinal microbiota enhances pancreatic carcinogenesis in preclinical models. Carcinogenesis 2018; 39:1068-1078. [PMID: 29846515 PMCID: PMC6067127 DOI: 10.1093/carcin/bgy073] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 04/12/2018] [Accepted: 05/25/2018] [Indexed: 01/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer death in the United States yet data are scant regarding host factors influencing pancreatic carcinogenesis. Increasing evidence support the role of the host microbiota in carcinogenesis but its role in PDAC is not well established. Herein, we report that antibiotic-mediated microbial depletion of KrasG12D/PTENlox/+ mice showed a decreased proportion of poorly differentiated tumors compared to microbiota-intact KrasG12D/PTENlox/+ mice. Subsequent 16S rRNA PCR showed that ~50% of KrasG12D/PTENlox/+ mice with PDAC harbored intrapancreatic bacteria. To determine if a similar observation in humans correlates with presence of PDAC, benign and malignant human pancreatic surgical specimens demonstrated a microbiota by 16S bacterial sequencing and culture confirmation. However, the microbial composition did not differentiate PDAC from non-PDAC tissue. Furthermore, murine pancreas did not naturally acquire a pancreatic microbiota, as germ-free mice transferred to specific pathogen-free housing failed to acquire intrapancreatic bacteria over time, which was not augmented by a murine model of colitis. Finally, antibiotic-mediated microbial depletion of Nod-SCID mice, compared to microbiota-intact, showed increased time to PDAC xenograft formation, smaller tumors, and attenuated growth. Interestingly, both xenograft cohorts were devoid of intratumoral bacteria by 16S rRNA PCR, suggesting that intrapancreatic/intratumoral microbiota is not the sole driver of PDAC acceleration. Xenografts from microbiota-intact mice demonstrated innate immune suppression by immunohistochemistry and differential regulation of oncogenic pathways as determined by RNA sequencing. Our work supports a long-distance role of the intestinal microbiota on PDAC progression and opens new research avenues regarding pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Ryan M Thomas
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
- Department of Surgery, North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
| | - Raad Z Gharaibeh
- Department of Medicine, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Josee Gauthier
- Department of Medicine, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Mark Beveridge
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Jillian L Pope
- Department of Medicine, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Maria V Guijarro
- Department of Anatomy and Cell Biology, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Qin Yu
- Department of Medicine, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Zhen He
- Department of Medicine, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Christina Ohland
- Department of Medicine, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Rachel Newsome
- Department of Medicine, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Jose Trevino
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Steven J Hughes
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Mary Reinhard
- Laboratory of Comparative Pathology, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Kathryn Winglee
- Department of Bioinformatics and Genomics, The University of North Carolina at Charlotte College of Computing and Informatics, Charlotte, NC, USA
| | - Anthony A Fodor
- Department of Bioinformatics and Genomics, The University of North Carolina at Charlotte College of Computing and Informatics, Charlotte, NC, USA
| | - Maria Zajac-Kaye
- Department of Anatomy and Cell Biology, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Christian Jobin
- Department of Medicine, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
- Department of Anatomy and Cell Biology, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
- Department of Infectious Disease and Immunology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| |
Collapse
|
52
|
Dou C, Liu Z, Tu K, Zhang H, Chen C, Yaqoob U, Wang Y, Wen J, van Deursen J, Sicard D, Tschumperlin D, Zou H, Huang WC, Urrutia R, Shah VH, Kang N. P300 Acetyltransferase Mediates Stiffness-Induced Activation of Hepatic Stellate Cells Into Tumor-Promoting Myofibroblasts. Gastroenterology 2018; 154:2209-2221.e14. [PMID: 29454793 PMCID: PMC6039101 DOI: 10.1053/j.gastro.2018.02.015] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 02/07/2018] [Accepted: 02/08/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Hepatic stellate cells (HSCs) contribute to desmoplasia and stiffness of liver metastases by differentiating into matrix-producing myofibroblasts. We investigated whether stiffness due to the presence of tumors increases activation of HSCs into myofibroblasts and their tumor-promoting effects, as well as the role of E1A binding protein p300, a histone acetyltransferase that regulates transcription, in these processes. METHODS HSCs were isolated from liver tissues of patients, mice in which the p300 gene was flanked by 2 loxP sites (p300F/F mice), and p300+/+ mice (controls). The HSCs were placed on polyacrylamide gels with precisely defined stiffness, and their activation (differentiation into myofibroblasts) was assessed by immunofluorescence and immunoblot analyses for alpha-smooth muscle actin. In HSCs from mice, the p300 gene was disrupted by cre recombinase. In human HSCs, levels of p300 were knocked down with small hairpin RNAs or a mutant form of p300 that is not phosphorylated by AKT (p300S1834A) was overexpressed. Human HSCs were also cultured with inhibitors of p300 (C646), PI3K signaling to AKT (LY294002), or RHOA (C3 transferase) and effects on stiffness-induced activation were measured. RNA sequencing and chromatin immunoprecipitation-quantitative polymerase chain reaction were used to identify HSC genes that changed expression levels in response to stiffness. We measured effects of HSC-conditioned media on proliferation of HT29 colon cancer cells and growth of tumors following subcutaneous injection of these cells into mice. MC38 colon cancer cells were injected into portal veins of p300F/Fcre and control mice, and liver metastases were measured. p300F/Fcre and control mice were given intraperitoneal injections of CCl4 to induce liver fibrosis. Liver tissues were collected and analyzed by immunofluorescence, immunoblot, and histology. RESULTS Substrate stiffness was sufficient to activate HSCs, leading to nuclear accumulation of p300. Disrupting p300 level or activity blocked stiffness-induced activation of HSCs. In HSCs, substrate stiffness activated AKT signaling via RHOA to induce phosphorylation of p300 at serine 1834; this caused p300 to translocate to the nucleus, where it up-regulated transcription of genes that increase activation of HSCs and metastasis, including CXCL12. MC38 cells, injected into portal veins, formed fewer metastases in livers of p300F/Fcre mice than control mice. Expression of p300 was increased in livers of mice following injection of CCl4; HSC activation and collagen deposition were reduced in livers of p300F/Fcre mice compared with control mice. CONCLUSIONS In studies of mice, we found liver stiffness to activate HSC differentiation into myofibroblasts, which required nuclear accumulation of p300. p300 increases HSC expression of genes that promote metastasis.
Collapse
Affiliation(s)
- Changwei Dou
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN,Department of Hepatobiliary Surgery, 1st Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China,Department of Hepatobiliary Surgery, Zhejiang provincial People's Hospital, Hangzhou, China
| | - Zhikui Liu
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN,Department of Hepatobiliary Surgery, 1st Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, 1st Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China,Tumor Microenvironment and Metastasis, Hormel Institute, University of Minnesota
| | - Hongbin Zhang
- Tumor Microenvironment and Metastasis, Hormel Institute, University of Minnesota
| | - Chen Chen
- Tumor Microenvironment and Metastasis, Hormel Institute, University of Minnesota
| | - Usman Yaqoob
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN
| | - Yuanguo Wang
- Tumor Microenvironment and Metastasis, Hormel Institute, University of Minnesota
| | - Jialing Wen
- Tumor Microenvironment and Metastasis, Hormel Institute, University of Minnesota
| | - Jan van Deursen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Delphine Sicard
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Daniel Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Hongzhi Zou
- Guangdong Institute of Gastroenterology, 6th Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei-Chien Huang
- Graduate Institute of Biomedical Science, China Medical University, Taiwan, R.O.C
| | - Raul Urrutia
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN
| | - Vijay H. Shah
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN,To whom correspondence should be addressed: Ningling Kang, Ph.D., Hormel Institute, 801 16th Ave NE Austin MN 55912. Fax: (507) 437-9606. Phone: (507) 437-9680. . Vijay Shah, M.D., Mayo Clinic, 200 1st ST SW Rochester MN 55915. Fax: (507) 255-6318. Phone: (507) 255-6028.
| | - Ningling Kang
- Tumor Microenvironment and Metastasis, Hormel Institute, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
53
|
Manegold P, Lai KKY, Wu Y, Teo JL, Lenz HJ, Genyk YS, Pandol SJ, Wu K, Lin DP, Chen Y, Nguyen C, Zhao Y, Kahn M. Differentiation Therapy Targeting the β-Catenin/CBP Interaction in Pancreatic Cancer. Cancers (Basel) 2018; 10:cancers10040095. [PMID: 29596326 PMCID: PMC5923350 DOI: 10.3390/cancers10040095] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/26/2018] [Accepted: 03/27/2018] [Indexed: 01/05/2023] Open
Abstract
Background: Although canonical Wnt signaling is known to promote tumorigenesis in pancreatic ductal adenocarcinoma (PDAC), a cancer driven principally by mutant K-Ras, the detailed molecular mechanisms by which the Wnt effector β-catenin regulates such tumorigenesis are largely unknown. We have previously demonstrated that β-catenin’s differential usage of the Kat3 transcriptional coactivator cyclic AMP-response element binding protein-binding protein (CBP) over its highly homologous coactivator p300 increases self-renewal and suppresses differentiation in other types of cancer. Aim/methods: To investigate Wnt-mediated carcinogenesis in PDAC, we have used the specific small molecule CBP/β-catenin antagonist, ICG-001, which our lab identified and has extensively characterized, to examine its effects in human pancreatic cancer cells and in both an orthotopic mouse model and a human patient-derived xenograft (PDX) model of PDAC. Results/conclusion: We report for the first time that K-Ras activation increases the CBP/β-catenin interaction in pancreatic cancer; and that ICG-001 specific antagonism of the CBP/β-catenin interaction sensitizes pancreatic cancer cells and tumors to gemcitabine treatment. These effects were associated with increases in the expression of let-7a microRNA; suppression of K-Ras and survivin; and the elimination of drug-resistant cancer stem/tumor-initiating cells.
Collapse
Affiliation(s)
- Philipp Manegold
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA.
| | - Keane K Y Lai
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA.
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA.
- Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Department of Pathology, University of Southern California, Los Angeles, CA 90033, USA.
| | - Yongfeng Wu
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA.
| | - Jia-Ling Teo
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA.
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.
| | - Heinz-Josef Lenz
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA.
- Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Yuri S Genyk
- Department of Surgery, University of Southern California, Los Angeles, CA 90033, USA.
| | - Stephen J Pandol
- Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Department of Pathology, University of Southern California, Los Angeles, CA 90033, USA.
- Pancreatic Research Program, Cedars-Sinai Medical Center, Veterans Affairs Greater Los Angeles Healthcare System, and Department of Medicine, University of California, Los Angeles, CA 90048, USA.
| | - Kaijin Wu
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA.
| | - David P Lin
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.
| | - Yibu Chen
- Health Sciences Libraries, University of Southern California, Los Angeles, CA 90033, USA.
| | - Cu Nguyen
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA.
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.
| | - Yi Zhao
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA.
- Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Michael Kahn
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA.
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA.
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA 90033, USA.
- Department of Molecular Pharmacology and Toxicology, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
54
|
Andricovich J, Perkail S, Kai Y, Casasanta N, Peng W, Tzatsos A. Loss of KDM6A Activates Super-Enhancers to Induce Gender-Specific Squamous-like Pancreatic Cancer and Confers Sensitivity to BET Inhibitors. Cancer Cell 2018; 33. [PMID: 29533787 PMCID: PMC5854186 DOI: 10.1016/j.ccell.2018.02.003] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
KDM6A, an X chromosome-encoded histone demethylase and member of the COMPASS-like complex, is frequently mutated in a broad spectrum of malignancies and contributes to oncogenesis with poorly characterized mechanisms. We found that KDM6A loss induced squamous-like, metastatic pancreatic cancer selectively in females through deregulation of the COMPASS-like complex and aberrant activation of super-enhancers regulating ΔNp63, MYC, and RUNX3 oncogenes. This subtype of tumor developed in males had concomitant loss of UTY and KDM6A, suggesting overlapping roles, and points to largely demethylase independent tumor suppressor functions. We also demonstrate that KDM6A-deficient pancreatic cancer is selectively sensitive to BET inhibitors, which reversed squamous differentiation and restrained tumor growth in vivo, highlighting a therapeutic niche for patient tailored therapies.
Collapse
Affiliation(s)
- Jaclyn Andricovich
- Cancer Epigenetics Laboratory, Department of Anatomy and Regenerative Biology, George Washington University (GWU) School of Medicine and Health Sciences, Washington, DC 20052, USA; GWU Cancer Center, GWU School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Stephanie Perkail
- Cancer Epigenetics Laboratory, Department of Anatomy and Regenerative Biology, George Washington University (GWU) School of Medicine and Health Sciences, Washington, DC 20052, USA; GWU Cancer Center, GWU School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Yan Kai
- Cancer Epigenetics Laboratory, Department of Anatomy and Regenerative Biology, George Washington University (GWU) School of Medicine and Health Sciences, Washington, DC 20052, USA; GWU Cancer Center, GWU School of Medicine and Health Sciences, Washington, DC 20052, USA; Department of Physics, GWU, Washington, DC 20052, USA
| | - Nicole Casasanta
- Cancer Epigenetics Laboratory, Department of Anatomy and Regenerative Biology, George Washington University (GWU) School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Weiqun Peng
- GWU Cancer Center, GWU School of Medicine and Health Sciences, Washington, DC 20052, USA; Department of Physics, GWU, Washington, DC 20052, USA
| | - Alexandros Tzatsos
- Cancer Epigenetics Laboratory, Department of Anatomy and Regenerative Biology, George Washington University (GWU) School of Medicine and Health Sciences, Washington, DC 20052, USA; GWU Cancer Center, GWU School of Medicine and Health Sciences, Washington, DC 20052, USA.
| |
Collapse
|
55
|
Okuda M, Inoue J, Fujiwara N, Kawano T, Inazawa J. Subcloning and characterization of highly metastatic cells derived from human esophageal squamous cell carcinoma KYSE150 cells by in vivo selection. Oncotarget 2018; 8:34670-34677. [PMID: 28410227 PMCID: PMC5471001 DOI: 10.18632/oncotarget.16668] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/19/2017] [Indexed: 11/25/2022] Open
Abstract
Esophageal cancer is the eighth most common cancer and the sixth most common cause of cancer-related deaths worldwide. Despite the research progress in understanding the disease, the mechanism underlying the metastasis is still unclear. Here, we successfully generated a highly metastatic cell subline, designated as KYSE150-LuM, derived from an esophageal squamous cell carcinoma cell line (KYSE150) by in vivo selection. To elucidate the mechanisms driving metastasis, we characterized the gene expression differences between LuM cells and parent KYSE150 cells. IL-6, IL-1β, and LCN2, previously associated with tumor growth and metastasis, were up-regulated in LuM cells. Recent studies on cancer have increasingly focused on the tumor microenvironment, from which these cytokines are released. The fact that these three cytokines (IL-6, IL-1β, LCN2) were up-regulated in LuM cells indicates that these highly metastatic cells obtained through in vivo selection will be a useful resource for further studies on elucidating the mechanisms underlying the tumor microenvironment which is associated with cytokine-related tumor growth and metastasis. Moreover, LuM cells could disseminate to the lung in shorter period of time in vivo, indicating their utility for in vivo experiments of metastasis and new therapeutic targets in a shorter period of time than currently possible.
Collapse
Affiliation(s)
- Masafumi Okuda
- Department of Molecular Cytogenetics, Medical Research Institute and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Gastrointestinal Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Jun Inoue
- Department of Molecular Cytogenetics, Medical Research Institute and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Bioresource Research Center, Research and Industry-University Alliance Organization, Tokyo Medical and Dental University, Tokyo, Japan
| | - Naoto Fujiwara
- Department of Molecular Cytogenetics, Medical Research Institute and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Gastrointestinal Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tatsuyuki Kawano
- Department of Gastrointestinal Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Johji Inazawa
- Department of Molecular Cytogenetics, Medical Research Institute and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Bioresource Research Center, Research and Industry-University Alliance Organization, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
56
|
Ghadban T, Dibbern JL, Reeh M, Miro JT, Tsui TY, Wellner U, Izbicki JR, Güngör C, Vashist YK. HSP90 is a promising target in gemcitabine and 5-fluorouracil resistant pancreatic cancer. Apoptosis 2018; 22:369-380. [PMID: 27878398 DOI: 10.1007/s10495-016-1332-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Chemotherapy (CT) options in pancreatic cancer (PC) are limited to gemcitabine and 5-fluorouracil (5-FU). Several identified molecular targets in PC represent client proteins of HSP90. HSP90 is a promising target since it interferes with many oncogenic signaling pathways simultaneously. The aim of this study was to evaluate the efficacy of different HSP90 inhibitors in gemcitabine and 5-FU resistant PC. PC cell lines 5061, 5072 and 5156 were isolated and brought in to culture from patients being operated at our institution. L3.6pl cell line served as a control. Anti-proliferative efficacy of three different HSP90 inhibitors (17-AAG, 17-DMAG and 17-AEPGA) was evaluated by the MTT assay. Alterations in signaling pathway effectors and apoptosis upon HSP90 inhibition were determined by western blot analysis and annexin V/PI staining. The cell lines 5061, 5072 and 5156 were resistant to gemcitabine and 5-FU. In contrast 17-AAG and the water-soluble derivates 17-DMAG and 17-AEPGA displayed high anti-proliferative activity in all tested cell lines. The calculated IC50 was below 1 µM. Highly significant down regulation of epidermal-growth-factor-receptor, insulin-like-growth-factor-receptor-1, AKT and MAPK reflected the intracellular molecular signaling-network disruption. Furthermore, besides HSP70 also HSP27 was upregulated in all cell lines. Apoptosis occurred early under HSP90 inhibition and was determined by annexin V/PI staining and CASPASE-3 and PARP assay. In contrast, gemcitabine treated cells did not show any apoptosis. HSP90 inhibition disrupts multiple signaling cascades in gemcitabine and 5-FU resistant PC simultaneously and promotes cancer cell apoptosis. Watersoluble 17-DMAG is equally effective as 17-AAG. HSP27, besides HSP70, may represent an effective response marker of successful HSP90 inhibition.
Collapse
Affiliation(s)
- Tarik Ghadban
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Judith L Dibbern
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Matthias Reeh
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Jameel T Miro
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Tung Y Tsui
- Department of Surgery, University Medical College Rostock, Schillingallee 35, 18057, Rostock, Germany
| | - Ulrich Wellner
- Clinic for Surgery, University Clinic of Schleswig-Holstein, Campus Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany
| | - Jakob R Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Cenap Güngör
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Yogesh K Vashist
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany. .,Department of Visceral Surgery, Kantonsspital Aarau AG, Tellstrasse 25, 5001, Aarau, Switzerland.
| |
Collapse
|
57
|
Delitto D, Judge SM, Delitto AE, Nosacka RL, Rocha FG, DiVita BB, Gerber MH, George TJ, Behrns KE, Hughes SJ, Wallet SM, Judge AR, Trevino JG. Human pancreatic cancer xenografts recapitulate key aspects of cancer cachexia. Oncotarget 2018; 8:1177-1189. [PMID: 27901481 PMCID: PMC5352045 DOI: 10.18632/oncotarget.13593] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/08/2016] [Indexed: 12/27/2022] Open
Abstract
Cancer cachexia represents a debilitating syndrome that diminishes quality of life and augments the toxicities of conventional treatments. Cancer cachexia is particularly debilitating in patients with pancreatic cancer (PC). Mechanisms responsible for cancer cachexia are under investigation and are largely derived from observations in syngeneic murine models of cancer which are limited in PC. We evaluate the effect of human PC cells on both muscle wasting and the systemic inflammatory milieu potentially contributing to PC-associated cachexia. Specifically, human PC xenografts were generated by implantation of pancreatic cancer cells, L3.6pl and PANC-1, either in the flank or orthotopically within the pancreas. Mice bearing orthotopic xenografts demonstrated significant muscle wasting and atrophy-associated gene expression changes compared to controls. Further, despite the absence of adaptive immunity, splenic tissue from orthotopically engrafted mice demonstrated elevations in several pro-inflammatory cytokines associated with cancer cachexia, including TNFα, IL1β, IL6 and KC (murine IL8 homologue), when compared to controls. Therefore, data presented here support further investigation into the complexity of cancer cachexia in PC to identify potential targets for this debilitating syndrome.
Collapse
Affiliation(s)
- Daniel Delitto
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, FL 32610, USA
| | - Sarah M Judge
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, FL 32610, USA
| | - Andrea E Delitto
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL 32610, USA
| | - Rachel L Nosacka
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, FL 32610, USA
| | - Fernanda G Rocha
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL 32610, USA
| | - Bayli B DiVita
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL 32610, USA
| | - Michael H Gerber
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, FL 32610, USA
| | - Thomas J George
- Department of Medicine, College of Medicine, University of Florida Health Science Center, Gainesville, FL 32610, USA
| | - Kevin E Behrns
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, FL 32610, USA
| | - Steven J Hughes
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, FL 32610, USA
| | - Shannon M Wallet
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL 32610, USA
| | - Andrew R Judge
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, FL 32610, USA
| | - Jose G Trevino
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, FL 32610, USA
| |
Collapse
|
58
|
Kendrick AA, Schafer J, Dzieciatkowska M, Nemkov T, D'Alessandro A, Neelakantan D, Ford HL, Pearson CG, Weekes CD, Hansen KC, Eisenmesser EZ. CD147: a small molecule transporter ancillary protein at the crossroad of multiple hallmarks of cancer and metabolic reprogramming. Oncotarget 2018; 8:6742-6762. [PMID: 28039486 PMCID: PMC5341751 DOI: 10.18632/oncotarget.14272] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/30/2016] [Indexed: 02/07/2023] Open
Abstract
Increased expression of CD147 in pancreatic cancer has been proposed to play a critical role in cancer progression via CD147 chaperone function for lactate monocarboxylate transporters (MCTs). Here, we show for the first time that CD147 interacts with membrane transporters beyond MCTs and exhibits a protective role for several of its interacting partners. CD147 prevents its interacting partner's proteasome-dependent degradation and incorrect plasma membrane localization through the CD147 transmembrane (TM) region. The interactions with transmembrane small molecule and ion transporters identified here indicate a central role of CD147 in pancreatic cancer metabolic reprogramming, particularly with respect to amino acid anabolism and calcium signaling. Importantly, CD147 genetic ablation prevents pancreatic cancer cell proliferation and tumor growth in vitro and in vivo in conjunction with metabolic rewiring towards amino acid anabolism, thus paving the way for future combined pharmacological treatments.
Collapse
Affiliation(s)
- Agnieszka A Kendrick
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, CO, USA
| | - Johnathon Schafer
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, CO, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, CO, USA
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, CO, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, CO, USA
| | - Deepika Neelakantan
- Department of Pharmacology, School of Medicine, University of Colorado Denver, CO, USA
| | - Heide L Ford
- Department of Pharmacology, School of Medicine, University of Colorado Denver, CO, USA
| | - Chad G Pearson
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado Denver, CO, USA
| | - Colin D Weekes
- Division of Oncology, Department of Medicine, University of Colorado Denver, CO, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, CO, USA
| | - Elan Z Eisenmesser
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, CO, USA
| |
Collapse
|
59
|
Stark K, Schubert I, Joshi U, Kilani B, Hoseinpour P, Thakur M, Grünauer P, Pfeiler S, Schmidergall T, Stockhausen S, Bäumer M, Chandraratne S, von Brühl ML, Lorenz M, Coletti R, Reese S, Laitinen I, Wörmann SM, Algül H, Bruns CJ, Ware J, Mackman N, Engelmann B, Massberg S. Distinct Pathogenesis of Pancreatic Cancer Microvesicle-Associated Venous Thrombosis Identifies New Antithrombotic Targets In Vivo. Arterioscler Thromb Vasc Biol 2018; 38:772-786. [PMID: 29419408 DOI: 10.1161/atvbaha.117.310262] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 01/17/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Cancer patients are at high risk of developing deep venous thrombosis (DVT) and venous thromboembolism, a leading cause of mortality in this population. However, it is largely unclear how malignant tumors drive the prothrombotic cascade culminating in DVT. APPROACH AND RESULTS Here, we addressed the pathophysiology of malignant DVT compared with nonmalignant DVT and focused on the role of tumor microvesicles as potential targets to prevent cancer-associated DVT. We show that microvesicles released by pancreatic adenocarcinoma cells (pancreatic tumor-derived microvesicles [pcMV]) boost thrombus formation in a model of flow restriction of the mouse vena cava. This depends on the synergistic activation of coagulation by pcMV and host tissue factor. Unlike nonmalignant DVT, which is initiated and propagated by innate immune cells, thrombosis triggered by pcMV was largely independent of myeloid leukocytes or platelets. Instead, we identified externalization of the phospholipid phosphatidylethanolamine as a major mechanism controlling the prothrombotic activity of pcMV. Disrupting phosphatidylethanolamine-dependent activation of factor X suppressed pcMV-induced DVT without causing changes in hemostasis. CONCLUSIONS Together, we show here that the pathophysiology of pcMV-associated experimental DVT differs markedly from innate immune cell-promoted nonmalignant DVT and is therefore amenable to distinct antithrombotic strategies. Targeting phosphatidylethanolamine on tumor microvesicles could be a new strategy for prevention of cancer-associated DVT without causing bleeding complications.
Collapse
Affiliation(s)
- Konstantin Stark
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.).
| | - Irene Schubert
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Urjita Joshi
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Badr Kilani
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Parandis Hoseinpour
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Manovriti Thakur
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Petra Grünauer
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Susanne Pfeiler
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Tobias Schmidergall
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Sven Stockhausen
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Markus Bäumer
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Sue Chandraratne
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Marie-Luise von Brühl
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Michael Lorenz
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Raffaele Coletti
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Sven Reese
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Iina Laitinen
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Sonja Maria Wörmann
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Hana Algül
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Christiane J Bruns
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Jerry Ware
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Nigel Mackman
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Bernd Engelmann
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Steffen Massberg
- From the Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany (K.S., I.S., B.K., P.H., T.S., S.S., S.C., M.-L.v.B., M.L., R.C., S.M.); German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany (K.S., S.M.); Institut für Laboratoriumsmedizin (U.J., M.T., P.G., S.P., M.B., B.E.) and Lehrstuhl für Anatomie, Histologie und Embryologie, Department of Veterinary Medicine (S.R.), Ludwig-Maximilians-Universität, Munich, Germany; Nuklearmedizinische Klinik und Poliklinik (I.L.) and II. Medizinische Klinik und Poliklinik (S.M.W., H.A.), Klinikum rechts der Isar, Technische Universität München, Munich, Germany; Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Universitätsklinik Köln, Cologne, Germany (C.J.B.); Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock (J.W.); and Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| |
Collapse
|
60
|
Vasodilator-stimulated phosphoprotein promotes liver metastasis of gastrointestinal cancer by activating a β1-integrin-FAK-YAP1/TAZ signaling pathway. NPJ Precis Oncol 2018; 2:2. [PMID: 29872721 PMCID: PMC5871906 DOI: 10.1038/s41698-017-0045-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/06/2017] [Accepted: 12/28/2017] [Indexed: 12/26/2022] Open
Abstract
Extracellular matrix (ECM)-induced β1-integrin-FAK signaling promotes cell attachment, survival, and migration of cancer cells in a distant organ so as to enable cancer metastasis. However, mechanisms governing activation of the β1-integrin-FAK signaling remain incompletely understood. Here, we report that vasodilator-stimulated phosphoprotein (VASP), an actin binding protein, is required for ECM–mediated β1-integrin-FAK-YAP1/TAZ signaling in gastrointestinal (GI) cancer cells and their liver metastasis. In patient-derived samples, VASP is upregulated in 53 of 63 colorectal cancers and 43 of 53 pancreatic ductal adenocarcinomas and high VASP levels correlate with liver metastasis and reduced patient survival. In a Matrigel-based 3-dimensional (3D) culture model, short hairpin RNA (shRNA)–mediated VASP knockdown in colorectal cancer cells (KM12L4, HCT116, and HT29) and pancreatic cancer cells (L3.6 and MIA PaCa-1) suppresses the growth of 3D cancer spheroids. Mechanistic studies reveal that VASP knockdown suppresses FAK phosphorylation and YAP1/TAZ protein levels, but not Akt or Erk-related pathways and that YAP1/TAZ proteins are enhanced by the β1-integrin-FAK signaling. Additionally, VASP regulates the β1-integrin-FAK-YAP1/TAZ signaling by at least two mechanisms: (1) promoting ECM-mediated β1-integrin activation and (2) regulating YAP1/TAZ dephosphorylation at downstream of RhoA to enhance the stability of YAP1/TAZ proteins. In agreement with these, preclinical studies with two experimental liver metastasis mouse models demonstrate that VASP knockdown suppresses GI cancer liver metastasis, β1-integrin activation, and YAP1/TAZ levels of metastatic cancer cells. Together, our data support VASP as a treatment target for liver metastasis of colorectal and pancreatic cancers. A protein involved in cytoskeleton regulation and cell motility control offers a new drug target for cancer spreading to the liver. Ningling Kang Ph.D. from the Hormel Institute in Austin, Minnesota, USA, and colleagues showed that levels of this actin-binding protein, known as vasodilator-stimulated phosphoprotein (VASP), are elevated in most patients with advanced colon and pancreatic cancers and that higher VASP expression levels are linked to liver metastasis and poorer patients’ outcomes. To explore the reasons why, the researchers studied three-dimensional tumor spheroids and mouse metastasis models of these cancers, and identified the signaling pathway by which VASP promotes the survival of cancer cells in distant organs, such as the liver. What’s more, they showed that knocking down VASP of cancer cells in metastasis mouse models suppressed cancer metastatic growth in the liver, suggesting that the same might be true in patients as well.
Collapse
|
61
|
Higuchi T, Yokobori T, Naito T, Kakinuma C, Hagiwara S, Nishiyama M, Asao T. Investigation into metastatic processes and the therapeutic effects of gemcitabine on human pancreatic cancer using an orthotopic SUIT-2 pancreatic cancer mouse model. Oncol Lett 2017; 15:3091-3099. [PMID: 29435042 DOI: 10.3892/ol.2017.7722] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 11/20/2017] [Indexed: 12/26/2022] Open
Abstract
Prognosis of pancreatic cancer is poor, thus the development of novel therapeutic drugs is necessary. During preclinical studies, appropriate models are essential for evaluating drug efficacy. The present study sought to determine the ideal pancreatic cancer mouse model for reliable preclinical testing. Such a model could accurately reflect human pancreatic cancer phenotypes and predict future clinical trial results. Systemic pathology analysis was performed in an orthotopic transplantation model to prepare model mice for use in preclinical studies, mimicking the progress of human pancreatic cancer. The location and the timing of inoculated cancer cell metastases, pathogenesis and cause of fatality were analyzed. Furthermore, the efficacy of gemcitabine, a key pancreatic cancer drug, was evaluated in this model where liver metastasis and peritoneal dissemination occur. Results indicated that the SUIT-2 orthotopic pancreatic cancer model was similar to the phenotypic sequential progression of human pancreatic cancer, with extra-pancreatic invasion, intra-peritoneal dissemination and other hematogenous organ metastases. Notably, survival was prolonged by administering gemcitabine to mice with metastasized pancreatic cancer. Furthermore, the detailed effects of gemcitabine on the primary tumor and metastatic tumor lesions were pathologically evaluated in mice. The present study indicated the model accurately depicted pancreatic cancer development and metastasis. Furthermore, the detailed effects of pancreatic cancer drugs on the primary tumor and on metastatic tumor lesions. We present this model as a potential new standard for new drug development in pancreatic cancer.
Collapse
Affiliation(s)
- Tamami Higuchi
- Department of Molecular Pharmacology and Oncology, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan.,Pharmaceutical and Healthcare Research Laboratories, Research and Development Management Headquarters, Fujifilm Corporation, Kaisei-machi, Kanagawa 258-8577, Japan
| | - Takehiko Yokobori
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Gunma 371-8511, Japan.,Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), Maebashi, Gunma 371-8511, Japan
| | - Tomoharu Naito
- Pharmaceutical and Healthcare Research Laboratories, Research and Development Management Headquarters, Fujifilm Corporation, Kaisei-machi, Kanagawa 258-8577, Japan
| | - Chihaya Kakinuma
- Pharmaceutical and Healthcare Research Laboratories, Research and Development Management Headquarters, Fujifilm Corporation, Kaisei-machi, Kanagawa 258-8577, Japan
| | - Shinji Hagiwara
- Pharmaceutical and Healthcare Research Laboratories, Research and Development Management Headquarters, Fujifilm Corporation, Kaisei-machi, Kanagawa 258-8577, Japan
| | - Masahiko Nishiyama
- Department of Molecular Pharmacology and Oncology, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan.,Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), Maebashi, Gunma 371-8511, Japan
| | - Takayuki Asao
- Big Data Center for Integrative Analysis, Gunma University Initiative for Advanced Research (GIAR), Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
62
|
Pathological and functional significance of Semaphorin-5A in pancreatic cancer progression and metastasis. Oncotarget 2017; 9:5931-5943. [PMID: 29464045 PMCID: PMC5814185 DOI: 10.18632/oncotarget.23644] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/11/2017] [Indexed: 01/23/2023] Open
Abstract
Semaphorin-5A (SEMA5A) has differential cell surface expression between normal and cancer cells and represents an attractive target for therapeutic intervention in pancreatic cancer (PC). In this study, we delineated the pathological expression and significance of SEMA5A during PC progression and metastasis. We utilized human tissue microarrays and different PC mouse models (Pdx1-cre; LSL- Kras(G12D), Pdx1-Cre; LSL-Kras(G12D); LSL-p53(R172H) and RIP1-Tag2) to analyze SEMA5A expression during PC progression. Using human patients and different mouse models, we demonstrated that SEMA5A expression was highest in liver metastases, followed by primary pancreatic tumors, and the lowest expression was found in the normal pancreas. SEMA5A expression was localized on tumor cells with no staining in the surrounding stroma. To understand the functional significance of SEMA5A, we treated PC cell lines with recombinant SEMA5A. We observed an increase in migration, chemotaxis, and scattering of PC cells. To delineate the signaling axis of SEMA5A, we generated SEMA5A receptor-Plexin-B3 knockdown in T3M-4 and CD18/HPAF PC cell lines and observed that the effect of SEMA5A treatment was absent in the Plexin-B3 knockdown counterparts of T3M-4 and CD18/HPAF cells. SEMA5A treatment leads to phosphorylation of cMET in Plexin-B3 dependent manner. Our data demonstrate that there is an increase in SEMA5A expression during PC progression and the elevation of this expression takes place at metastatic sites especially the liver in both exocrine and endocrine tumors. SEMA5A can elicit a migratory response in cells by activating cMET through the Plexin-B3 receptor. In conclusion, SEMA5A signaling represents a potential molecule for targeting metastasis in pancreatic cancer.
Collapse
|
63
|
Sato S, Nakamura T, Katagiri T, Tsuchikawa T, Kushibiki T, Hontani K, Takahashi M, Inoko K, Takano H, Abe H, Takeuchi S, Ono M, Kuwabara S, Umemoto K, Suzuki T, Sato O, Nakamura Y, Hirano S. Molecular targeting of cell-permeable peptide inhibits pancreatic ductal adenocarcinoma cell proliferation. Oncotarget 2017; 8:113662-113672. [PMID: 29371937 PMCID: PMC5768354 DOI: 10.18632/oncotarget.21939] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/26/2017] [Indexed: 12/18/2022] Open
Abstract
Background Chromosome 16 open reading frame 74 (C16orf74) is highly expressed in pancreatic ductal adenocarcinoma (PDAC) and is involved in cancer cell proliferation and invasion through binding to calcineurin (CN). Therefore, C16orf74 is a good target for the development of a PDAC treatment. A cell-permeable dominant-negative (DN) peptide that can inhibit the C16orf74/CN interaction was designed to examine whether this peptide can inhibit PDAC cell proliferation in vitro and in vivo. Method TheDN-C16orf74 peptide, which corresponds to the portion of C16orf74 that interacts with CN, was synthesized, and we assessed its anti-tumor activity in proliferation assays with human PDAC cells and the underlying molecular signaling pathway. Using an orthotopic xenograft model of PDAC, we treated mice intraperitoneally with phosphate-buffered saline (PBS), control peptide, or DN-C16orf74 and analyzed the tumor-suppressive effects. Result DN-C16orf74 inhibited the binding of C16orf74 to CN in an immunoprecipitation assay. DN-C16orf74 suppressed PDAC cell proliferation, and the level of suppression depended on the expression levels of C16orf74 in vitro. DN-C16orf74 also exhibited anti-tumor effects in orthotopic xenograft model. Furthermore, the tumor-suppressive effect was associated with inhibition of the phosphorylation of Akt and mTOR. Conclusion The cell-permeable peptide DN-C16orf74 has a strong anti-tumor effect against PDAC in vitro and in vivo.
Collapse
Affiliation(s)
- Shoki Sato
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toru Nakamura
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toyomasa Katagiri
- Division of Genome Medicine, Institute for Genome Research, Tokushima University, Tokushima, Japan
| | - Takahiro Tsuchikawa
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toshihiro Kushibiki
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kouji Hontani
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Mizuna Takahashi
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kazuho Inoko
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hironobu Takano
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hirotake Abe
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shintaro Takeuchi
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masato Ono
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shota Kuwabara
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kazufumi Umemoto
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Tomohiro Suzuki
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Osamu Sato
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yusuke Nakamura
- Department of Medicine and Surgery, The University of Chicago, Chicago, IL, USA
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
64
|
Skruber K, Chaplin KJ, Phanstiel O. Synthesis and Bioevaluation of Macrocycle-Polyamine Conjugates as Cell Migration Inhibitors. J Med Chem 2017; 60:8606-8619. [PMID: 28976754 DOI: 10.1021/acs.jmedchem.7b01222] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The motuporamines are natural products isolated from the New Guinea sea sponge Xestospongia exigua. Dihydromotuporamine C contains a large macrocycle and an appended polyamine component and was shown to be both antimetastatic and cytotoxic to human L3.6pl pancreatic cancer cells. A series of macrocycle-polyamine conjugates were prepared, and the sequence of the polyamine component was varied to optimize the antimigration properties (as measured in L3.6pl cells) of this molecular class. A one-carbon spacer between the 15-membered carbocycle and the appended polyamine showed improved antimigration properties. A survey of different polyamine sequences containing two, three, or four carbon spacers revealed that the natural polyamine sequence (norspermidine, a 3,3-triamine) was superior in terms of inhibiting the migration of L3.6pl cells in vitro. An investigation of the respective ceramide and sphingomyelin populations in L3.6pl cells revealed that these molecules can modulate both ceramide and sphingomyelin pools in cells and inhibit cell migration.
Collapse
Affiliation(s)
- Kristen Skruber
- University of Central Florida, College of Medicine , 12722 Research Parkway, Orlando, Florida 32826, United States
| | - Kelvin J Chaplin
- University of Central Florida, College of Medicine , 12722 Research Parkway, Orlando, Florida 32826, United States
| | - Otto Phanstiel
- University of Central Florida, College of Medicine , 12722 Research Parkway, Orlando, Florida 32826, United States
| |
Collapse
|
65
|
Hesler RA, Huang JJ, Starr MD, Treboschi VM, Bernanke AG, Nixon AB, McCall SJ, White RR, Blobe GC. TGF-β-induced stromal CYR61 promotes resistance to gemcitabine in pancreatic ductal adenocarcinoma through downregulation of the nucleoside transporters hENT1 and hCNT3. Carcinogenesis 2017; 37:1041-1051. [PMID: 27604902 DOI: 10.1093/carcin/bgw093] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 08/16/2016] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer in part due to inherent resistance to chemotherapy, including the first-line drug gemcitabine. Although low expression of the nucleoside transporters hENT1 and hCNT3 that mediate cellular uptake of gemcitabine has been linked to gemcitabine resistance, the mechanisms regulating their expression in the PDAC tumor microenvironment are largely unknown. Here, we report that the matricellular protein cysteine-rich angiogenic inducer 61 (CYR61) negatively regulates the nucleoside transporters hENT1 and hCNT3. CRISPR/Cas9-mediated knockout of CYR61 increased expression of hENT1 and hCNT3, increased cellular uptake of gemcitabine and sensitized PDAC cells to gemcitabine-induced apoptosis. In PDAC patient samples, expression of hENT1 and hCNT3 negatively correlates with expression of CYR61 . We demonstrate that stromal pancreatic stellate cells (PSCs) are a source of CYR61 within the PDAC tumor microenvironment. Transforming growth factor-β (TGF-β) induces the expression of CYR61 in PSCs through canonical TGF-β-ALK5-Smad2/3 signaling. Activation of TGF-β signaling or expression of CYR61 in PSCs promotes resistance to gemcitabine in PDAC cells in an in vitro co-culture assay. Our results identify CYR61 as a TGF-β-induced stromal-derived factor that regulates gemcitabine sensitivity in PDAC and suggest that targeting CYR61 may improve chemotherapy response in PDAC patients.
Collapse
Affiliation(s)
| | | | - Mark D Starr
- Division of Medical Oncology, Department of Medicine
| | | | | | | | | | - Rebekah R White
- Department of Surgery, Duke University, B354 LSRC Research Drive , Box 91004, Durham, NC 27708 , USA
| | - Gerard C Blobe
- Department of Pharmacology and Cancer Biology.,Division of Medical Oncology, Department of Medicine
| |
Collapse
|
66
|
Zeleniak AE, Huang W, Brinkman MK, Fishel ML, Hill R. Loss of MTSS1 results in increased metastatic potential in pancreatic cancer. Oncotarget 2017; 8:16473-16487. [PMID: 28146435 PMCID: PMC5369978 DOI: 10.18632/oncotarget.14869] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 01/19/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a 5-year survival rate of 7%. This dismal prognosis is largely due to the inability to diagnose the disease before metastasis occurs. Tumor cell dissemination occurs early in PDAC. While it is known that inflammation facilitates this process, the underlying mechanisms responsible for this progression have not been fully characterized. Here, we functionally test the role of metastasis suppressor 1 (MTSS1) in PDAC. Despite evidence showing that MTSS1 could be important for regulating metastasis in many different cancers, its function in PDAC has not been studied. Here, we show that loss of MTSS1 leads to increased invasion and migration in PDAC cell lines. Moreover, PDAC cells treated with cancer-associated fibroblast-conditioned media also have increased metastatic potential, which is augmented by loss of MTSS1. Finally, overexpression of MTSS1 in PDAC cell lines leads to a loss of migratory potential in vitro and an increase in overall survival in vivo. Collectively, our data provide insight into an important role for MTSS1 in suppressing tumor cell invasion and migration driven by the tumor microenvironment and suggest that therapeutic strategies aimed at increasing MTSS1 levels may effectively slow the development of metastatic lesions, increasing survival of patients with PDAC.
Collapse
Affiliation(s)
- Ann E Zeleniak
- Integrated Biomedical Sciences Program, University of Notre Dame, South Bend, Indiana, USA.,Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA
| | - Wei Huang
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Mary K Brinkman
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Melissa L Fishel
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, Indianapolis, Indiana, USA.,Indiana University School of Medicine, Department of Pediatrics, Wells Center for Pediatric Research, Indianapolis, Indiana, USA.,Pancreatic Cancer Signature Center, Indianapolis, Indiana, USA
| | - Reginald Hill
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| |
Collapse
|
67
|
Mathison A, Salmonson A, Missfeldt M, Bintz J, Williams M, Kossak S, Nair A, de Assuncao TM, Christensen T, Buttar N, Iovanna J, Huebert R, Lomberk G. Combined AURKA and H3K9 Methyltransferase Targeting Inhibits Cell Growth By Inducing Mitotic Catastrophe. Mol Cancer Res 2017; 15:984-997. [PMID: 28442587 DOI: 10.1158/1541-7786.mcr-17-0063] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/31/2017] [Accepted: 04/20/2017] [Indexed: 12/18/2022]
Abstract
The current integrative pathobiologic hypothesis states that pancreatic cancer (PDAC) develops and progresses in response to an interaction between known oncogenes and downstream epigenomic regulators. Congruently, this study tests a new combinatorial therapy based on the inhibition of the Aurora kinase A (AURKA) oncogene and one of its targets, the H3K9 methylation-based epigenetic pathway. This therapeutic combination is effective at inhibiting the in vitro growth of PDAC cells both, in monolayer culture systems, and in three-dimensional spheroids and organoids. The combination also reduces the growth of PDAC xenografts in vivo Mechanistically, it was found that inhibiting methyltransferases of the H3K9 pathway in cells, which are arrested in G2-M after targeting AURKA, decreases H3K9 methylation at centromeres, induces mitotic aberrations, triggers an aberrant mitotic check point response, and ultimately leads to mitotic catastrophe. Combined, these data describe for the first time a hypothesis-driven design of an efficient combinatorial treatment that targets a dual oncogenic-epigenomic pathway to inhibit PDAC cell growth via a cytotoxic mechanism that involves perturbation of normal mitotic progression to end in mitotic catastrophe. Therefore, this new knowledge has significant mechanistic value as it relates to the development of new therapies as well as biomedical relevance.Implications: These results outline a model for the combined inhibition of a genetic-to-epigenetic pathway to inhibit cell growth and suggest an important and provocative consideration for harnessing the capacity of cell-cycle inhibitors to enhance the future use of epigenetic inhibitors. Mol Cancer Res; 15(8); 984-97. ©2017 AACR.
Collapse
Affiliation(s)
- Angela Mathison
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota.,Laboratory of Epigenetics and Chromatin Dynamics, Gastroenterology Research Unit, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Ann Salmonson
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota.,Laboratory of Epigenetics and Chromatin Dynamics, Gastroenterology Research Unit, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Mckenna Missfeldt
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota.,Laboratory of Epigenetics and Chromatin Dynamics, Gastroenterology Research Unit, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Jennifer Bintz
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Monique Williams
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Sarah Kossak
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota.,Laboratory of Epigenetics and Chromatin Dynamics, Gastroenterology Research Unit, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Asha Nair
- Department of Health Science Research, Mayo Clinic, Rochester, Minnesota
| | - Thiago M de Assuncao
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Trace Christensen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Navtej Buttar
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Robert Huebert
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota.,Center for Cell Signaling in Gastroenterology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Gwen Lomberk
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota. .,Laboratory of Epigenetics and Chromatin Dynamics, Gastroenterology Research Unit, Department of Medicine, Mayo Clinic, Rochester, Minnesota.,Center for Cell Signaling in Gastroenterology, Mayo Clinic and Foundation, Rochester, Minnesota.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
68
|
Luheshi NM, Coates-Ulrichsen J, Harper J, Mullins S, Sulikowski MG, Martin P, Brown L, Lewis A, Davies G, Morrow M, Wilkinson RW. Transformation of the tumour microenvironment by a CD40 agonist antibody correlates with improved responses to PD-L1 blockade in a mouse orthotopic pancreatic tumour model. Oncotarget 2017; 7:18508-20. [PMID: 26918344 PMCID: PMC4951305 DOI: 10.18632/oncotarget.7610] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/14/2016] [Indexed: 01/05/2023] Open
Abstract
Despite the availability of recently developed chemotherapy regimens, survival times for pancreatic cancer patients remain poor. These patients also respond poorly to immune checkpoint blockade therapies (anti-CTLA-4, anti-PD-L1, anti-PD-1), which suggests the presence of additional immunosuppressive mechanisms in the pancreatic tumour microenvironment (TME). CD40 agonist antibodies (αCD40) promote antigen presenting cell (APC) maturation and enhance macrophage tumouricidal activity, and may therefore alter the pancreatic TME to increase sensitivity to immune checkpoint blockade. Here, we test whether αCD40 transforms the TME in a mouse syngeneic orthotopic model of pancreatic cancer, to increase sensitivity to PD-L1 blockade. We found that whilst mice bearing orthotopic Pan02 tumours responded poorly to PD-L1 blockade, αCD40 improved overall survival. αCD40 transformed the TME, upregulating Th1 chemokines, increasing cytotoxic T cell infiltration and promoting formation of an immune cell-rich capsule separating the tumour from the normal pancreas. Furthermore, αCD40 drove systemic APC maturation, memory T cell expansion, and upregulated tumour and systemic PD-L1 expression. Combining αCD40 with PD-L1 blockade enhanced anti-tumour immunity and improved overall survival versus either monotherapy. These data provide further support for the potential of combining αCD40 with immune checkpoint blockade to promote anti-tumour immunity in pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lee Brown
- MedImmune Ltd., Cambridge CB21 6GH, UK
| | | | | | | | | |
Collapse
|
69
|
Li L, Schmitt M, Matzke‐Ogi A, Wadhwani P, Orian‐Rousseau V, Levkin PA. CD44v6-Peptide Functionalized Nanoparticles Selectively Bind to Metastatic Cancer Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2017; 4:1600202. [PMID: 28105395 PMCID: PMC5238741 DOI: 10.1002/advs.201600202] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Indexed: 05/31/2023]
Abstract
CD44v6 peptide functionalized nanoparticles are fabricated in a facile and controllable way to selectively bind to CD44v6 positive tumor cells with highly efficient anticancer and antimetastatic properties. The reported modular synthesis and facile preparation makes this system highly potent for developing novel multifunctional nanocarriers for therapeutic and/or diagnostic anticancer applications.
Collapse
Affiliation(s)
- Linxian Li
- Institute of Toxicology and GeneticsKarlsruhe Institute of Technology76344KarlsruheGermany
- Institute of Organic ChemistryUniversity of Heidelberg69120HeidelbergGermany
| | - Mark Schmitt
- Institute of Toxicology and GeneticsKarlsruhe Institute of Technology76344KarlsruheGermany
| | - Alexandra Matzke‐Ogi
- Institute of Toxicology and GeneticsKarlsruhe Institute of Technology76344KarlsruheGermany
| | - Parvesh Wadhwani
- Institute of Biological Interfaces (IBG‐2)Karlsruhe Institute of Technology76344KarlsruheGermany
| | | | - Pavel A. Levkin
- Institute of Toxicology and GeneticsKarlsruhe Institute of Technology76344KarlsruheGermany
- Department of Applied Physical ChemistryUniversity of Heidelberg69120HeidelbergGermany
| |
Collapse
|
70
|
Calle AS, Nair N, Oo AK, Prieto-Vila M, Koga M, Khayrani AC, Hussein M, Hurley L, Vaidyanath A, Seno A, Iwasaki Y, Calle M, Kasai T, Seno M. A new PDAC mouse model originated from iPSCs-converted pancreatic cancer stem cells (CSCcm). Am J Cancer Res 2016; 6:2799-2815. [PMID: 28042501 PMCID: PMC5199755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/06/2016] [Indexed: 06/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most representative form of pancreatic cancers. PDAC solid tumours are constituted of heterogeneous populations of cells including cancer stem cells (CSCs), differentiated cancer cells, desmoplastic stroma and immune cells. The identification and consequent isolation of pancreatic CSCs facilitated the generation of genetically engineered murine models. Nonetheless, the current models may not be representative for the spontaneous tumour occurrence. In the present study, we show the generation of a novel pancreatic iPSC-converted cancer stem cell lines (CSCcm) as a cutting-edge model for the study of PDAC. The CSCcm lines were achieved only by the influence of pancreatic cancer cell lines conditioned medium and were not subjected to any genetic manipulation. The xenografts tumours from CSCcm lines displayed histopathological features of ADM, PanIN and PDAC lesions. Further molecular characterization from RNA-sequencing analysis highlighted primary culture cell lines (1st CSCcm) as potential candidates to represent the pancreatic CSCs and indicated the establishment of the pancreatic cancer molecular pattern in their subsequent progenies 2nd CSCcm and 3rd CSCcm. In addition, preliminary RNA-seq SNPs analysis showed that the distinct CSCcm lines did not harbour single point mutations for the oncogene Kras codon 12 or 13. Therefore, PDAC-CSCcm model may provide new insights about the actual occurrence of the pancreatic cancer leading to develop different approaches to target CSCs and abrogate the progression of this fatidic disease.
Collapse
Affiliation(s)
- Anna Sanchez Calle
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Neha Nair
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Aung KoKo Oo
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Marta Prieto-Vila
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Megumi Koga
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Apriliana Cahya Khayrani
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Maram Hussein
- Department of Chemistry, Faculty of Science, Menoufia UniversityShebin El-Koam 32511, Egypt
| | - Laura Hurley
- Cancer Biology Graduate Program, School of Medicine, Wayne State University10 E Warren, Avenue, Suite 2215, Detroit, Michigan 48201, USA
| | - Arun Vaidyanath
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Akimasa Seno
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Yoshiaki Iwasaki
- Department of Gastroenterology and Hepatology, Okayama University, Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayama 700-8558, Japan
| | - Malu Calle
- Department of Systems Biology, University of VicVic, Barcelona 08500, Spain
| | - Tomonari Kasai
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Masaharu Seno
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University3.1.1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| |
Collapse
|
71
|
Zubair H, Azim S, Srivastava SK, Ahmad A, Bhardwaj A, Khan MA, Patel GK, Arora S, Carter JE, Singh S, Singh AP. Glucose Metabolism Reprogrammed by Overexpression of IKKε Promotes Pancreatic Tumor Growth. Cancer Res 2016; 76:7254-7264. [PMID: 27923829 DOI: 10.1158/0008-5472.can-16-1666] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/19/2016] [Accepted: 10/04/2016] [Indexed: 02/06/2023]
Abstract
Aberrant expression of the kinase IKKε in pancreatic ductal adenocarcinoma (PDAC) has been associated with poor prognosis. In this study, we define a pathobiologic function for IKKε in reprogramming glucose metabolism and driving progression in PDAC. Silencing IKKε in PDAC cells, which overexpressed it endogenously, was sufficient to reduce malignant cell growth, clonogenic potential, glucose consumption, lactate secretion, and expression of genes involved in glucose metabolism, without impacting the basal oxygen consumption rate. IKKε silencing also attenuated c-Myc in a manner associated with diminished signaling through an AKT/GSK3β/c-MYC phosphorylation cascade that promoted MYC nuclear accumulation. In an orthotopic mouse model, IKKε-silenced PDAC exhibited a relative reduction in glucose uptake, tumorigenicity, and metastasis. Overall, our findings offer a preclinical mechanistic rationale to target IKKε to improve the therapeutic management of PDAC in patients. Cancer Res; 76(24); 7254-64. ©2016 AACR.
Collapse
Affiliation(s)
- Haseeb Zubair
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Shafquat Azim
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Sanjeev Kumar Srivastava
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Aamir Ahmad
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Arun Bhardwaj
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Mohammad Aslam Khan
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Girijesh Kumar Patel
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Sumit Arora
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - James Elliot Carter
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Seema Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama.,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Ajay Pratap Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama. .,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| |
Collapse
|
72
|
Naguib YW, Lansakara-P D, Lashinger LM, Rodriguez BL, Valdes S, Niu M, Aldayel AM, Peng L, Hursting SD, Cui Z. Synthesis, Characterization, and In Vitro and In Vivo Evaluations of 4-(N)-Docosahexaenoyl 2', 2'-Difluorodeoxycytidine with Potent and Broad-Spectrum Antitumor Activity. Neoplasia 2016; 18:33-48. [PMID: 26806350 PMCID: PMC5965255 DOI: 10.1016/j.neo.2015.11.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 11/11/2015] [Accepted: 11/11/2015] [Indexed: 12/28/2022] Open
Abstract
In this study, a new compound, 4-(N)-docosahexaenoyl 2′, 2′-difluorodeoxycytidine (DHA-dFdC), was synthesized and characterized. Its antitumor activity was evaluated in cell culture and in mouse models of pancreatic cancer. DHA-dFdC is a poorly soluble, pale yellow waxy solid, with a molecular mass of 573.3 Da and a melting point of about 96°C. The activation energy for the degradation of DHA-dFdC in an aqueous Tween 80–based solution is 12.86 kcal/mol, whereas its stability is significantly higher in the presence of vitamin E. NCI-60 DTP Human Tumor Cell Line Screening revealed that DHA-dFdC has potent and broad-spectrum antitumor activity, especially in leukemia, renal, and central nervous system cancer cell lines. In human and murine pancreatic cancer cell lines, the IC50 value of DHA-dFdC was up to 105-fold lower than that of dFdC. The elimination of DHA-dFdC in mouse plasma appeared to follow a biexponential model, with a terminal phase t1/2 of about 58 minutes. DHA-dFdC significantly extended the survival of genetically engineered mice that spontaneously develop pancreatic ductal adenocarcinoma. In nude mice with subcutaneously implanted human Panc-1 pancreatic tumors, the antitumor activity of DHA-dFdC was significantly stronger than the molar equivalent of dFdC alone, DHA alone, or the physical mixture of them (1:1, molar ratio). DHA-dFdC also significantly inhibited the growth of Panc-1 tumors orthotopically implanted in the pancreas of nude mice, whereas the molar equivalent dose of dFdC alone did not show any significant activity. DHA-dFdC is a promising compound for the potential treatment of cancers in organs such as the pancreas.
Collapse
Affiliation(s)
- Youssef W Naguib
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712
| | - Dharmika Lansakara-P
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712
| | - Laura M Lashinger
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712
| | - B Leticia Rodriguez
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712
| | - Solange Valdes
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712
| | - Mengmeng Niu
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712
| | - Abdulaziz M Aldayel
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712
| | - Lan Peng
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Stephen D Hursting
- Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599
| | - Zhengrong Cui
- Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712.
| |
Collapse
|
73
|
Kirby MK, Ramaker RC, Gertz J, Davis NS, Johnston BE, Oliver PG, Sexton KC, Greeno EW, Christein JD, Heslin MJ, Posey JA, Grizzle WE, Vickers SM, Buchsbaum DJ, Cooper SJ, Myers RM. RNA sequencing of pancreatic adenocarcinoma tumors yields novel expression patterns associated with long-term survival and reveals a role for ANGPTL4. Mol Oncol 2016; 10:1169-82. [PMID: 27282075 PMCID: PMC5423196 DOI: 10.1016/j.molonc.2016.05.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 04/25/2016] [Accepted: 05/17/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma patients have low survival rates due to late-stage diagnosis and high rates of cancer recurrence even after surgical resection. It is important to understand the molecular characteristics associated with survival differences in pancreatic adenocarcinoma tumors that may inform patient care. RESULTS RNA sequencing was performed for 51 patient tumor tissues extracted from patients undergoing surgical resection, and expression was associated with overall survival time from diagnosis. Our analysis uncovered 323 transcripts whose expression correlates with survival time in our pancreatic patient cohort. This genomic signature was validated in an independent RNA-seq dataset of 68 additional patients from the International Cancer Genome Consortium. We demonstrate that this transcriptional profile is largely independent of markers of cellular division and present a 19-transcript predictive model built from a subset of the 323 transcripts that can distinguish patients with differing survival times across both the training and validation patient cohorts. We present evidence that a subset of the survival-associated transcripts is associated with resistance to gemcitabine treatment in vitro, and reveal that reduced expression of one of the survival-associated transcripts, Angiopoietin-like 4, impairs growth of a gemcitabine-resistant pancreatic cancer cell line. CONCLUSIONS Gene expression patterns in pancreatic adenocarcinoma tumors can distinguish patients with differing survival outcomes after undergoing surgical resection, and the survival difference could be associated with the intrinsic gemcitabine sensitivity of primary patient tumors. Thus, these transcriptional differences may impact patient care by distinguishing patients who would benefit from a non-gemcitabine based therapy.
Collapse
Affiliation(s)
- Marie K Kirby
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Ryne C Ramaker
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA; University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jason Gertz
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | | | - Patsy G Oliver
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | - James A Posey
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | - Sara J Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA.
| |
Collapse
|
74
|
Huang H, Svoboda RA, Lazenby AJ, Saowapa J, Chaika N, Ding K, Wheelock MJ, Johnson KR. Up-regulation of N-cadherin by Collagen I-activated Discoidin Domain Receptor 1 in Pancreatic Cancer Requires the Adaptor Molecule Shc1. J Biol Chem 2016; 291:23208-23223. [PMID: 27605668 DOI: 10.1074/jbc.m116.740605] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinomas are highly malignant cancers characterized by extensive invasion into surrounding tissues, metastasis to distant organs, and a limited response to therapy. A main feature of pancreatic ductal adenocarcinomas is desmoplasia, which leads to extensive deposition of collagen I. We have demonstrated that collagen I can induce epithelial-mesenchymal transition (EMT) in pancreatic cancer cells. A hallmark of EMT is an increase in the expression of the mesenchymal cadherin N-cadherin. Previously we showed up-regulation of N-cadherin promotes tumor cell invasion and that collagen I-induced EMT is mediated by two collagen receptors, α2β1-integrin and discoidin domain receptor 1 (DDR1). DDR1 is a receptor-tyrosine kinase widely expressed during embryonic development and in many adult tissues and is also highly expressed in many different cancers. In the signaling pathway initiated by collagen, we have shown proline-rich tyrosine kinase 2 (Pyk2) is downstream of DDR1. In this study we found isoform b of DDR1 is responsible for collagen I-induced up-regulation of N-cadherin and tyrosine 513 of DDR1b is necessary. Knocking down Shc1, which binds to tyrosine 513 of DDR1b via its PTB (phosphotyrosine binding) domain, eliminates the up-regulation of N-cadherin. The signaling does not require a functional SH2 domain or the tyrosine residues commonly phosphorylated in Shc1 but is mediated by the interaction between a short segment of the central domain of Shc1 and the proline-rich region of Pyk2. Taken together, these data illustrate DDR1b, but not DDR1a, mediates collagen I-induced N-cadherin up-regulation, and Shc1 is involved in this process by coupling to both DDR1 and Pyk2.
Collapse
Affiliation(s)
- Huocong Huang
- From the Department of Biochemistry and Molecular Biology, College of Medicine
| | | | - Audrey J Lazenby
- Department of Pathology and Microbiology, College of Medicine, and
| | | | - Nina Chaika
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha Nebraska 68198
| | - Ke Ding
- State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Avenue, Guangzhou 510530, China, and
| | - Margaret J Wheelock
- From the Department of Biochemistry and Molecular Biology, College of Medicine.,Department of Oral Biology, College of Dentistry.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha Nebraska 68198
| | - Keith R Johnson
- From the Department of Biochemistry and Molecular Biology, College of Medicine, .,Department of Oral Biology, College of Dentistry.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha Nebraska 68198.,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
75
|
Garley M, Jabłońska E, Sawicka-Powierza J, Kłoczko J, Piszcz J, Kakareko M, Ratajczak-Wrona W, Charkiewicz A, Omeljaniuk W, Szpak A. Expression of IL-1 and IL-6 and their natural regulators in leukocytes of B-cell chronic lymphocytic leukaemia patients. Adv Med Sci 2016; 61:187-192. [PMID: 26876087 DOI: 10.1016/j.advms.2015.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/04/2015] [Accepted: 12/30/2015] [Indexed: 01/08/2023]
Abstract
PURPOSE The purpose of the study was the assessment of the expression of IL-1β and IL-6, and the proteins regulating their biological activity, namely IL-1RII, IL-1Ra, as well as sIL-6Rα, sgp-130 in leukemic lymphocytes and autologous neutrophils of B-CLL patients. MATERIAL/METHODS The study involved a group of B-cell chronic lymphocytic leukemia patients and healthy volunteer blood donors. The presence of chosen proteins and their natural regulators was confirmed by Western blot. RESULTS Western blot analysis showed a decreased expression of IL-1β and IL-6 in the leukocytes of B-CLL patients. Decreased expression of sIL-6Rα has been observed in lymphocytes, with a simultaneous increase of expression in PMNs. Lower expression of sgp-130 was found in B cells while its expression was elevated in the neutrophils of patients in early stages of the disease. CONCLUSIONS The changes observed in the expression of IL-1 and IL-6 seem to exclude their immediate involvement in the progress of B-CLL. However, the presented changes in the expression of proteins regulating IL-1β and IL-6 in PMNs indicate a potential role of early immune response cells also in advanced stages of the disease.
Collapse
|
76
|
Ilmer M, Mazurek N, Byrd JC, Ramirez K, Hafley M, Alt E, Vykoukal J, Bresalier RS. Cell surface galectin-3 defines a subset of chemoresistant gastrointestinal tumor-initiating cancer cells with heightened stem cell characteristics. Cell Death Dis 2016; 7:e2337. [PMID: 27512958 PMCID: PMC5108324 DOI: 10.1038/cddis.2016.239] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 06/13/2016] [Accepted: 07/08/2016] [Indexed: 12/12/2022]
Abstract
Recurrence of gastrointestinal adenocarcinomas after surgery and chemotherapy may be attributed, in part, to the presence of a small population of tumor-initiating cancer stem cells (CSC). The expression of galectin-3 (Gal3), a multifunctional oncolectin, has been associated with biological behaviors associated with CSC. We examined the ability of Gal3 to characterize the CSC phenotype, and to identify a clinically important gastrointestinal cancer CSC population. Human colorectal and pancreatic cancer cell lines were sorted to identify subpopulations expressing commonly used CSC markers, and Gal3-positive CSC subpopulations. The association of Gal3 with the stem cell properties and alterations of these phenotypes by manipulation of Gal3 expression was examined. Gastrointestinal cancer cell lines contain both Gal3-positive and Gal3-negative subpopulations. Gal3-positive CSCs are characterized by high ALDH activity, enhanced self-renewal ability in vitro (sphere formation) and tumor forming ability in vivo, and resistance to chemotherapeutic agents and death-receptor-mediated apoptosis compared to Gal3-negative CSCs. Silencing Gal3 modifies this behavior. Cell surface Gal3 expression identifies a subset of CSCs in gastrointestinal cancers with high levels of stem cell characteristics, including chemoresistance. This may provide a platform for developing treatment strategies that target CSC.
Collapse
Affiliation(s)
- Matthias Ilmer
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Nachman Mazurek
- Department of Gastroenterology, Hepatology and Nutrition, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - James C Byrd
- Department of Gastroenterology, Hepatology and Nutrition, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Karen Ramirez
- Department of Stem Cell Transplantation and Cellular Therapies, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Margarete Hafley
- Department of Gastroenterology, Hepatology and Nutrition, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Eckhard Alt
- Department of Medicine, Tulane University Health Science Center, New Orleans, LA USA
| | - Jody Vykoukal
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Robert S Bresalier
- Department of Gastroenterology, Hepatology and Nutrition, University of Texas MD Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
77
|
Niesen J, Sack M, Seidel M, Fendel R, Barth S, Fischer R, Stein C. SNAP-Tag Technology: A Useful Tool To Determine Affinity Constants and Other Functional Parameters of Novel Antibody Fragments. Bioconjug Chem 2016; 27:1931-41. [PMID: 27391930 DOI: 10.1021/acs.bioconjchem.6b00315] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Antibody derivatives, such as the single chain fragment variable (scFv), can be developed as diagnostic and therapeutic tools in cancer research, especially in the form of fusion proteins. Such derivatives are easier to produce and modify than monoclonal antibodies (mAbs) and achieve better tissue/tumor penetration. The genetic modification of scFvs is also much more straightforward than the challenging chemical modification of mAbs. Therefore, we constructed two scFvs derived from the approved monoclonal antibodies cetuximab (scFv2112) and panitumumab (scFv1711), both of which are specific for the epidermal growth factor receptor (EGFR), a well-characterized solid tumor antigen. Both scFvs were genetically fused to the SNAP-tag, an engineered version of the human DNA repair enzyme O(6)-alkylguanine DNA alkyltransferase that allows the covalent coupling of benzylguanine (BG)-modified substrates such as fluorescent dyes. The SNAP-tag achieves controllable and irreversible protein modification and is an important tool for experimental studies in vitro and in vivo. The affinity constant of a scFv is a key functional parameter, especially in the context of a fusion protein. Therefore, we developed a method to define the affinity constants of scFv-SNAP fusion proteins by surface plasmon resonance (SPR) spectroscopy. We could confirm that both scFvs retained their functionality after fusion to the SNAP-tag in a variety of procedures and assays, including ELISA, flow cytometry, and confocal microscopy. The experimental procedures described herein, and the new protocol for affinity determination by SPR spectroscopy, are suitable for the preclinical evaluation of diverse antibody formats and derivatives.
Collapse
Affiliation(s)
- Judith Niesen
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME , 52074 Aachen, Germany
| | - Markus Sack
- Institute of Molecular Biotechnology (Biology VII), RWTH Aachen University , 52074 Aachen, Germany
| | - Melanie Seidel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME , 52074 Aachen, Germany
| | - Rolf Fendel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME , 52074 Aachen, Germany
| | - Stefan Barth
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME , 52074 Aachen, Germany
| | - Rainer Fischer
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME , 52074 Aachen, Germany.,Institute of Molecular Biotechnology (Biology VII), RWTH Aachen University , 52074 Aachen, Germany
| | - Christoph Stein
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME , 52074 Aachen, Germany
| |
Collapse
|
78
|
Madan M, Patel A, Skruber K, Geerts D, Altomare DA, IV OP. ATP13A3 and caveolin-1 as potential biomarkers for difluoromethylornithine-based therapies in pancreatic cancers. Am J Cancer Res 2016; 6:1231-1252. [PMID: 27429841 PMCID: PMC4937730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 04/15/2016] [Indexed: 06/06/2023] Open
Abstract
The purpose of this paper was to better understand the role of polyamine transport in pancreatic cancers.This paper identifies potential biomarkers for assessing the relative tumor commitment to polyamine biosynthesis or transport. Cell lines with low polyamine import activity and low ATP13A3 protein levels appear committed to polyamine biosynthesis and required high concentrations of the polyamine biosynthesis inhibitor, difluoromethylornithine (DFMO) to inhibit their growth (e.g., AsPC-1 and Capan 1). In contrast, cell lines with high polyamine import activity and high ATP13A3 protein expression (e.g., L3.6pl) demonstrated a commitment to polyamine transport and required lower DFMO concentrations to inhibit their growth. Pancreatic cancer cell lines which were most sensitive to DFMO also gave the highest EC50 values for the polyamine transport inhibitors (PTIs) tested indicating that more PTI was needed to inhibit the active polyamine transport systems of these cell lines. Most significant is that the combination therapy of DFMO+PTI was efficacious against both cell types with the PTI showing low efficacy in cell lines with low polyamine transport activity and high efficacy in cell lines with high polyamine transport activity. High ATP13A3 protein expression and moderate to low Cav-1 protein expression was shown to be predictive of tumors which effectively escape DFMO via polyamine import. In summary, this report demonstrates for the first time the role of ATP13A3 in polyamine transport and its use as a potential biomarker along with Cav-1 to select tumors most susceptible to DFMO. These findings may help stratify patients in the ongoing clinical trials with DFMO-based therapies and help predict tumor response.
Collapse
Affiliation(s)
- Meenu Madan
- Department of Medical Education, University of Central Florida College of Medicine12722 Research Parkway, Orlando, Florida 32826, USA
| | - Arjun Patel
- Department of Medical Education, University of Central Florida College of Medicine12722 Research Parkway, Orlando, Florida 32826, USA
| | - Kristen Skruber
- Department of Medical Education, University of Central Florida College of Medicine12722 Research Parkway, Orlando, Florida 32826, USA
| | - Dirk Geerts
- Department of Pediatric Oncology, Erasmus University Medical Center, Dr. Molewaterplein 503015 GE Rotterdam, The Netherlands
| | - Deborah A Altomare
- Burnett School for Biomedical Sciences, University of Central Florida6900 Lake Nona Blvd., Orlando, FL 32827, USA
| | - Otto Phanstiel IV
- Department of Medical Education, University of Central Florida College of Medicine12722 Research Parkway, Orlando, Florida 32826, USA
| |
Collapse
|
79
|
Zhao L, Zhao Y, Schwarz B, Mysliwietz J, Hartig R, Camaj P, Bao Q, Jauch KW, Guba M, Ellwart JW, Nelson PJ, Bruns CJ. Verapamil inhibits tumor progression of chemotherapy-resistant pancreatic cancer side population cells. Int J Oncol 2016; 49:99-110. [PMID: 27177126 PMCID: PMC4902079 DOI: 10.3892/ijo.2016.3512] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/28/2016] [Indexed: 01/22/2023] Open
Abstract
Tumor side population (SP) cells display stem-like properties that can be modulated by treatment with the calcium channel blocker verapamil. Verapamil can enhance the cytotoxic effects of chemotherapeutic drugs and multi-drug resistance by targeting the transport function of the P-glycoprotein (P-gp). This study focused on the therapeutic potential of verapamil on stem-like SP tumor cells, and further investigated its chemosensitizing effects using L3.6pl and AsPC-1 pancreatic carcinoma models. As compared to parental L3.6pl cells (0.9±0.22%), L3.6pl gemcitabine-resistant cells (L3.6plGres) showed a significantly higher percentage of SP cells (5.38±0.99%) as detected by Hoechst 33342/FACS assays. The L3.6plGres SP cells showed stable gemcitabine resistance, enhanced colony formation ability and increased tumorigenicity. Verapamil effectively inhibited L3.6plGres and AsPC-1 SP cell proliferation in vitro. A pro-apoptotic effect of verapamil was observed in L3.6pl cells, but not in L3.6plGres cells, which was linked to their differential expression of P-gp and equilibrative nucleoside transporter-1 (ENT-1). In an orthotopic pancreatic cancer mouse model, both low and high dose verapamil was shown to substantially reduce L3.6plGres-SP cell tumor growth and metastasis, enhance tumor apoptosis, and reduce microvascular density.
Collapse
Affiliation(s)
- Lu Zhao
- Department of Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Yue Zhao
- Department of Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Bettina Schwarz
- Department of Surgery, Munich Medical Center, Campus Grosshadern, LMU, Munich, Germany
| | - Josef Mysliwietz
- Institute of Molecular Immunology, Helmholtz Center for Environment and Health, Munich, Germany
| | - Roland Hartig
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Camaj
- Department of Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Qi Bao
- Department of Surgery, Munich Medical Center, Campus Grosshadern, LMU, Munich, Germany
| | - Karl-Walter Jauch
- Department of Surgery, Munich Medical Center, Campus Grosshadern, LMU, Munich, Germany
| | - Makus Guba
- Department of Surgery, Munich Medical Center, Campus Grosshadern, LMU, Munich, Germany
| | - Joachim Walter Ellwart
- Institute of Molecular Immunology, Helmholtz Center for Environment and Health, Munich, Germany
| | - Peter Jon Nelson
- Clinical Biochemistry Group, Medical Clinic and Policlinic IV, Munich Medical Center, Campus Innenstadt, LMU, Munich, Germany
| | | |
Collapse
|
80
|
Yao HP, Feng L, Zhou JW, Zhang RW, Wang MH. Therapeutic evaluation of monoclonal antibody-maytansinoid conjugate as a model of RON-targeted drug delivery for pancreatic cancer treatment. Am J Cancer Res 2016; 6:937-956. [PMID: 27293990 PMCID: PMC4889711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 03/06/2016] [Indexed: 06/06/2023] Open
Abstract
Aberrant expression of the RON receptor tyrosine kinase, a member of the MET proto-oncogene family, contributes significantly to pancreatic cancer tumorigenesis and chemoresistance. Here we validate RON as a target for pancreatic cancer therapy using a novel anti-RON antibody Zt/g4-drug maytansinoid conjugates (Zt/g4-DM1) as a model for RON-targeted drug delivery to kill pancreatic cancer cells. In pancreatic cancer cell lines overexpressing RON, Zt/g4-DM1 rapidly induced receptor endocytosis, arrested cell cycle at G2/M phase, reduced cell viability, and subsequently caused massive cell death. These in vitro observations help to establish a correlation between the number of the cell surface RON receptors and the efficacy of Zt/g4-DM1 in reduction of cell viability. In mice, Zt/g4-DM1 pharmacokinetics in the linear dose range fitted into a two-compartment model with clearance in 0.21 ml/day/kg and terminal half-life at 6.05 days. These results helped to confirm a concentration-activity relationship for the BxPC-3 and other pancreatic cancer cell xenograft model with a tumoristatic dose at 3.02 mg/kg. Zt/g4-DM1 was effective in vivo against various xenograft PDAC growth but efficacy varied with individual cell lines. Combination of Zt/g4-DM1 with gemcitabine had a complete inhibition of xenograft pancreatic cancer growth. We conclude from these studies that increased RON expression in pancreatic cancer cells is a suitable targeting moiety for anti-RON ADC-directed drug delivery and anticancer therapy. Zt/g4-DM1 is highly effective alone or in combination with chemotherapeutics in inhibition of pancreatic cancer xenograft growth in preclinical models. These findings justify the use of humanized Zt/g4-DM1 for targeted pancreatic cancer therapy in the future.
Collapse
Affiliation(s)
- Hang-Ping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital of Zhejiang University School of MedicineHangzhou, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital of Zhejiang University School of MedicineHangzhou, China
| | - Liang Feng
- Cancer Biology Research Center, Texas Tech University Health Sciences Center School of PharmacyAmarillo, TX 79106, USA
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center School of PharmacyAmarillo, TX 79106, USA
| | - Jian-Wei Zhou
- Department of Molecular Cell Biology and Toxicology, Nanjing Medical University School of Public HealthNanjing, China
| | - Rui-Wen Zhang
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of PharmacyAmarillo, TX 79106, USA
| | - Ming-Hai Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital of Zhejiang University School of MedicineHangzhou, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital of Zhejiang University School of MedicineHangzhou, China
- Cancer Biology Research Center, Texas Tech University Health Sciences Center School of PharmacyAmarillo, TX 79106, USA
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center School of PharmacyAmarillo, TX 79106, USA
| |
Collapse
|
81
|
Niess H, Camaj P, Mair R, Renner A, Zhao Y, Jäckel C, Nelson PJ, Jauch KW, Bruns CJ. Overexpression of IFN-induced protein with tetratricopeptide repeats 3 (IFIT3) in pancreatic cancer: cellular "pseudoinflammation" contributing to an aggressive phenotype. Oncotarget 2016; 6:3306-18. [PMID: 25650658 PMCID: PMC4413655 DOI: 10.18632/oncotarget.2494] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 09/16/2014] [Indexed: 12/20/2022] Open
Abstract
Inflammation contributes to important traits that cancer cells acquire during malignant progression. Gene array data recently identified upregulation of interferon-induced protein with tetratricopeptide repeats 3 (IFIT3) in aggressive pancreatic cancer cells. IFIT3 belongs to the group of interferon stimulated genes (ISG), can be induced by several cellular stress stimuli and by its tetratricopeptide repeats interacts with a multitude of cellular proteins. Upregulation of IFIT3 was confirmed in the aggressive pancreatic cancer cell line L3.6pl compared with its less aggressive cell line of origin, COLO357FG. Transgenic induction of IFIT3 expression in COLO357FG resulted in greater mass of orthotopic tumors and higher prevalence of metastases. Several important traits that mediate malignancy were altered by IFIT3: increased VEGF and IL-6 secretion, chemoresistance and decreased starvation-induced apoptosis. IFIT3 showed binding to JNK and STAT1, the latter being an important inducer of IFIT3 expression. Despite still being alterable by “classical” IFN or NFκB signaling, our findings indicate constitutive - possibly auto-regulated - upregulation of IFIT3 in L3.6pl without presence of an adequate inflammatory stimulus. The transcription factor SOX9, which is linked to regulation of hypoxia-related genes, was identified as a key mediator of upregulation of the oncogene IFIT3 and thereby sustaining a “pseudoinflammatory” cellular condition.
Collapse
Affiliation(s)
- Hanno Niess
- Department of Surgery, Medical Center of the Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Peter Camaj
- Department of Surgery, Medical Center of the Otto-von-Guericke-University, Magdeburg, Germany
| | - Ruth Mair
- Department of Surgery, Medical Center of the Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Andrea Renner
- Department of Surgery, Medical Center of the Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Yue Zhao
- Department of Surgery, Medical Center of the Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Carsten Jäckel
- Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Universitaet Muenchen, Arbeitsgruppe Klinische Biochemie, Munich, Germany
| | - Peter J Nelson
- Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Universitaet Muenchen, Arbeitsgruppe Klinische Biochemie, Munich, Germany
| | - Karl-Walter Jauch
- Department of Surgery, Medical Center of the Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | - Christiane J Bruns
- Department of Surgery, Medical Center of the Otto-von-Guericke-University, Magdeburg, Germany
| |
Collapse
|
82
|
Principe M, Ceruti P, Shih NY, Chattaragada MS, Rolla S, Conti L, Bestagno M, Zentilin L, Yang SH, Migliorini P, Cappello P, Burrone O, Novelli F. Targeting of surface alpha-enolase inhibits the invasiveness of pancreatic cancer cells. Oncotarget 2016; 6:11098-113. [PMID: 25860938 PMCID: PMC4484442 DOI: 10.18632/oncotarget.3572] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 02/22/2015] [Indexed: 12/22/2022] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a highly aggressive malignancy characterized by rapid progression, invasiveness and resistance to treatment. We have previously demonstrated that most PDAC patients have circulating antibodies against the glycolytic enzyme alpha-enolase (ENO1), which correlates with a better response to therapy and survival. ENO1 is a metabolic enzyme, also expressed on the cell surface where it acts as a plasminogen receptor. ENO1 play a crucial role in cell invasion and metastasis by promoting plasminogen activation into plasmin, a serine-protease involved in extracellular matrix degradation. The aim of this study was to investigate the role of ENO1 in PDAC cell invasion. We observed that ENO1 was expressed on the cell surface of most PDAC cell lines. Mouse anti-human ENO1 monoclonal antibodies inhibited plasminogen-dependent invasion of human PDAC cells, and their metastatic spreading in immunosuppressed mice was inhibited. Notably, a single administration of Adeno-Associated Virus (AAV)-expressing cDNA coding for 72/1 anti-ENO1 mAb reduced the number of lung metastases in immunosuppressed mice injected with PDAC cells. Overall, these data indicate that ENO1 is involved in PDAC cell invasion, and that administration of an anti-ENO1 mAb can be exploited as a novel therapeutic option to increase the survival of metastatic PDAC patients.
Collapse
Affiliation(s)
- Moitza Principe
- Center for Experimental Research and Medical Studies (CeRMS), Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Patrizia Ceruti
- Center for Experimental Research and Medical Studies (CeRMS), Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Neng-Yao Shih
- National Institute of Cancer Research, National Health Research Institutes, Tainan City, Taiwan
| | - Michelle S Chattaragada
- Center for Experimental Research and Medical Studies (CeRMS), Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Simona Rolla
- Center for Experimental Research and Medical Studies (CeRMS), Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy.,Molecular Biotechnology Center (MBC), University of Turin, Turin, Italy
| | - Marco Bestagno
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Sheng-Hui Yang
- College of Medical Science and Technology, Taipei Medical University, Taipei City, Taiwan
| | - Paola Migliorini
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Paola Cappello
- Center for Experimental Research and Medical Studies (CeRMS), Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Oscar Burrone
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Francesco Novelli
- Center for Experimental Research and Medical Studies (CeRMS), Azienda Universitaria Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| |
Collapse
|
83
|
Matzke-Ogi A, Jannasch K, Shatirishvili M, Fuchs B, Chiblak S, Morton J, Tawk B, Lindner T, Sansom O, Alves F, Warth A, Schwager C, Mier W, Kleeff J, Ponta H, Abdollahi A, Orian-Rousseau V. Inhibition of Tumor Growth and Metastasis in Pancreatic Cancer Models by Interference With CD44v6 Signaling. Gastroenterology 2016; 150:513-25.e10. [PMID: 26597578 DOI: 10.1053/j.gastro.2015.10.020] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Cancer cells with high metastatic potential and stem cell-like characteristics express the cell surface marker CD44. CD44 isoforms that include the v6 exon are co-receptors for the receptor tyrosine kinases MET and Vascular Endothelial Growth factor Receptor-2 (VEGFR-2). We studied CD44v6 signaling in several pancreatic cancer cell lines, and its role in tumor growth and metastasis in several models of pancreatic cancer. METHODS We analyzed the effects of v6 peptides that interfere with the co-receptor functions of CD44v6 for MET and VEGFR-2 in tumors and metastases grown from cells that express different CD44 isoforms, including CD44v6. The peptides were injected into rats with syngeneic tumors and mice with orthotopic or xenograft tumors. We also tested the effects of the peptides in mice with xenograft tumors grown from patient tumor samples and mice that express an oncogenic form of RAS and develop spontaneous pancreatic cancer (KPC mice). We measured levels of CD44v6 messenger RNA (mRNA) in pancreatic cancer tissues from 136 patients. RESULTS Xenograft tumors grown from human cancer cells injected with v6 peptides were smaller and formed fewer metastases in mice. The v6 peptide was more efficient than the MET inhibitor crizotinib and/or the VEGFR-2 inhibitor pazopanib in reducing xenograft tumor growth and metastasis. Injection of KPC mice with the v6 peptide increased their survival time. Injection of mice and rats bearing metastases with the v6 peptide induced regression of metastases. Higher levels of CD44v6 mRNA in human pancreatic tumor tissues were associated with increased expression of MET, tumor metastasis, and shorter patient survival times. CONCLUSIONS Peptide inhibitors of CD44v6 isoforms block tumor growth and metastasis in several independent models of pancreatic cancer. The v6 peptides induced regression of metastases. Levels of CD44v6 mRNA are increased, along with those of MET mRNA, in patients with metastatic pancreatic tumors, compared with nonmetastatic tumors; the increased levels correlated with shorter patient survival time.
Collapse
Affiliation(s)
- Alexandra Matzke-Ogi
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Eggenstein-Leopoldshafen, Germany; Amcure GmbH, Eggenstein-Leopoldshafen, Germany
| | - Katharina Jannasch
- Department of Hematology and Oncology, University Medicine Göttingen, Göttingen, Germany
| | - Marine Shatirishvili
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Eggenstein-Leopoldshafen, Germany
| | - Beatrix Fuchs
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Eggenstein-Leopoldshafen, Germany
| | - Sara Chiblak
- Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology, University of Heidelberg Medical School and German Cancer Research Center, Heidelberg, Germany; The German Cancer Consortium, Heidelberg, Germany
| | - Jennifer Morton
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Bouchra Tawk
- Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology, University of Heidelberg Medical School and German Cancer Research Center, Heidelberg, Germany; The German Cancer Consortium, Heidelberg, Germany
| | - Thomas Lindner
- Department of Nuclear Medicine, University of Heidelberg, Heidelberg, Germany
| | - Owen Sansom
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Frauke Alves
- Department of Hematology and Oncology, University Medicine Göttingen, Göttingen, Germany
| | - Arne Warth
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Christian Schwager
- Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology, University of Heidelberg Medical School and German Cancer Research Center, Heidelberg, Germany; The German Cancer Consortium, Heidelberg, Germany
| | - Walter Mier
- Department of Nuclear Medicine, University of Heidelberg, Heidelberg, Germany
| | - Jörg Kleeff
- Department of Surgery, Technische Universität München, Munich, Germany
| | | | - Amir Abdollahi
- Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology, University of Heidelberg Medical School and German Cancer Research Center, Heidelberg, Germany; The German Cancer Consortium, Heidelberg, Germany
| | - Véronique Orian-Rousseau
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Eggenstein-Leopoldshafen, Germany.
| |
Collapse
|
84
|
Niccolai E, Taddei A, Ricci F, Rolla S, D'Elios MM, Benagiano M, Bechi P, Bencini L, Ringressi MN, Pini A, Castiglione F, Giordano D, Satolli MA, Coratti A, Cianchi F, Bani D, Prisco D, Novelli F, Amedei A. Intra-tumoral IFN-γ-producing Th22 cells correlate with TNM staging and the worst outcomes in pancreatic cancer. Clin Sci (Lond) 2016; 130:247-58. [PMID: 26590104 DOI: 10.1042/cs20150437] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 11/20/2015] [Indexed: 12/11/2022]
Abstract
PDAC (pancreatic ductal adenocarcinoma) is the fifth leading cause of cancer-related death. The causes of this cancer remain unknown, but increasing evidence indicates a key role of the host immune response and cytokines in human carcinogenesis. Intra-tumoral IL (interleukin)-22 levels have been shown to be elevated in PDAC patients. However, little is known regarding the expression and clinical relevance of Th22 cells in human PDAC and, furthermore, which TILs (tumour-infiltrating lymphocytes) are the main producers of IL-22 is unknown. In the present study, we characterized the functional proprieties of the different subsets of IL-22-producing TILs and analysed their relationship with the TNM staging system and patient survival. We have demonstrated for the first time that, in PDAC patients, the T-cells co-producing IFN-γ (interferon γ) and exerting perforin-mediated cytotoxicity are the major intra-tumoral source of IL-22. In addition, isolated Th22 cells were able to induce apoptosis, which was antagonized by IL-22. Finally, we observed that the IL-22-producing T-cells were significantly increased in tumour tissue and that this increase was positively correlated with TNM staging of PDAC and poorer patient survival. These novel findings support the dual role of the anti-tumour immune system and that IL-22-producing cells may participate in PDAC pathogenesis. Therefore monitoring Th22 levels could be a good diagnostic parameter, and blocking IL-22 signalling may represent a viable method for anti-PDAC therapies.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Apoptosis
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/pathology
- Cell Line, Tumor
- Coculture Techniques
- Cytotoxicity, Immunologic
- Female
- Granzymes/metabolism
- Humans
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Interleukins/immunology
- Interleukins/metabolism
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Male
- Middle Aged
- Neoplasm Staging
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/mortality
- Pancreatic Neoplasms/pathology
- Paracrine Communication
- Perforin/metabolism
- Phenotype
- Signal Transduction
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- T-Lymphocytes, Helper-Inducer/pathology
- Interleukin-22
Collapse
Affiliation(s)
- Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Antonio Taddei
- Immunogenetics and Transplant Biology Service, Azienda Ospedaliera Città della Salute e della Scienza di Torino, via Santena 19, 10126 Turin, Italy
| | - Federica Ricci
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Simona Rolla
- Centre for Experimental Research and Medical Studies (CERMS), Azienda Ospedaliera Città della Salute e della Scienza di Torino, via Cherasco 15, 10126 Turin, Italy Molecular Biology Center and Department of Molecular Biotechnology and Health Sciences, University of Turin, via Nizza 52, 10126 Turin, Italy
| | - Mario Milco D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy Department of Neuro-Skeletal Muscle and Sensory Organs, Interdisciplinary Internal Medicine Unit, Azienda Ospedaliero Universitaria Careggi, 50134 Florence, Italy
| | - Marisa Benagiano
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Paolo Bechi
- Department of Surgery and Translational Medicine, University of Florence, Viale Michelangiolo 41, 50125 Florence, Italy Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Largo Brambilla 3, 50134 Florence, Italy
| | - Lapo Bencini
- Department of Oncology, Division of General and Oncologic Surgery, Azienda Ospedaliera Universitaria Careggi (AOUC), Largo Brambilla 3, 50134 Florence, Italy
| | - Maria Novella Ringressi
- Department of Surgery and Translational Medicine, University of Florence, Viale Michelangiolo 41, 50125 Florence, Italy Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Largo Brambilla 3, 50134 Florence, Italy
| | - Alessandro Pini
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Francesca Castiglione
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Daniele Giordano
- Centre for Experimental Research and Medical Studies (CERMS), Azienda Ospedaliera Città della Salute e della Scienza di Torino, via Cherasco 15, 10126 Turin, Italy Molecular Biology Center and Department of Molecular Biotechnology and Health Sciences, University of Turin, via Nizza 52, 10126 Turin, Italy
| | - Maria Antonietta Satolli
- Centro Oncologico Ematologico Subalpino (COES), AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Andrea Coratti
- Department of Oncology, Division of General and Oncologic Surgery, Azienda Ospedaliera Universitaria Careggi (AOUC), Largo Brambilla 3, 50134 Florence, Italy
| | - Fabio Cianchi
- Department of Surgery and Translational Medicine, University of Florence, Viale Michelangiolo 41, 50125 Florence, Italy Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Largo Brambilla 3, 50134 Florence, Italy
| | - Daniele Bani
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Domenico Prisco
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy Department of Neuro-Skeletal Muscle and Sensory Organs, Interdisciplinary Internal Medicine Unit, Azienda Ospedaliero Universitaria Careggi, 50134 Florence, Italy
| | - Francesco Novelli
- Immunogenetics and Transplant Biology Service, Azienda Ospedaliera Città della Salute e della Scienza di Torino, via Santena 19, 10126 Turin, Italy Centre for Experimental Research and Medical Studies (CERMS), Azienda Ospedaliera Città della Salute e della Scienza di Torino, via Cherasco 15, 10126 Turin, Italy Molecular Biology Center and Department of Molecular Biotechnology and Health Sciences, University of Turin, via Nizza 52, 10126 Turin, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy Department of Neuro-Skeletal Muscle and Sensory Organs, Interdisciplinary Internal Medicine Unit, Azienda Ospedaliero Universitaria Careggi, 50134 Florence, Italy
| |
Collapse
|
85
|
Unkel S, Belka C, Lauber K. On the analysis of clonogenic survival data: Statistical alternatives to the linear-quadratic model. Radiat Oncol 2016; 11:11. [PMID: 26822015 PMCID: PMC4730743 DOI: 10.1186/s13014-016-0584-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/05/2016] [Indexed: 12/28/2022] Open
Abstract
Background The most frequently used method to quantitatively describe the response to ionizing irradiation in terms of clonogenic survival is the linear-quadratic (LQ) model. In the LQ model, the logarithm of the surviving fraction is regressed linearly on the radiation dose by means of a second-degree polynomial. The ratio of the estimated parameters for the linear and quadratic term, respectively, represents the dose at which both terms have the same weight in the abrogation of clonogenic survival. This ratio is known as the α/β ratio. However, there are plausible scenarios in which the α/β ratio fails to sufficiently reflect differences between dose-response curves, for example when curves with similar α/β ratio but different overall steepness are being compared. In such situations, the interpretation of the LQ model is severely limited. Methods Colony formation assays were performed in order to measure the clonogenic survival of nine human pancreatic cancer cell lines and immortalized human pancreatic ductal epithelial cells upon irradiation at 0-10 Gy. The resulting dataset was subjected to LQ regression and non-linear log-logistic regression. Dimensionality reduction of the data was performed by cluster analysis and principal component analysis. Results Both the LQ model and the non-linear log-logistic regression model resulted in accurate approximations of the observed dose-response relationships in the dataset of clonogenic survival. However, in contrast to the LQ model the non-linear regression model allowed the discrimination of curves with different overall steepness but similar α/β ratio and revealed an improved goodness-of-fit. Additionally, the estimated parameters in the non-linear model exhibit a more direct interpretation than the α/β ratio. Dimensionality reduction of clonogenic survival data by means of cluster analysis was shown to be a useful tool for classifying radioresistant and sensitive cell lines. More quantitatively, principal component analysis allowed the extraction of scores of radioresistance, which displayed significant correlations with the estimated parameters of the regression models. Conclusions Undoubtedly, LQ regression is a robust method for the analysis of clonogenic survival data. Nevertheless, alternative approaches including non-linear regression and multivariate techniques such as cluster analysis and principal component analysis represent versatile tools for the extraction of parameters and/or scores of the cellular response towards ionizing irradiation with a more intuitive biological interpretation. The latter are highly informative for correlation analyses with other types of data, including functional genomics data that are increasingly beinggenerated.
Collapse
Affiliation(s)
- Steffen Unkel
- Department of Medical Statistics University Medical Centre, Georg-August-University Goettingen, Goettingen, Germany.
| | - Claus Belka
- Clinic for Radiotherapy and Radiation Oncology, LMU Munich, Munich, Germany. .,Clinic Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer', Helmholtz Center Munich, Munich, Germany.
| | - Kirsten Lauber
- Clinic for Radiotherapy and Radiation Oncology, LMU Munich, Munich, Germany. .,Clinic Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer', Helmholtz Center Munich, Munich, Germany.
| |
Collapse
|
86
|
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 2015; 22:1787-99. [PMID: 26667487 DOI: 10.1158/1078-0432.ccr-15-1256] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [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
|
87
|
Fujimura K, Choi S, Wyse M, Strnadel J, Wright T, Klemke R. Eukaryotic Translation Initiation Factor 5A (EIF5A) Regulates Pancreatic Cancer Metastasis by Modulating RhoA and Rho-associated Kinase (ROCK) Protein Expression Levels. J Biol Chem 2015; 290:29907-19. [PMID: 26483550 PMCID: PMC4706006 DOI: 10.1074/jbc.m115.687418] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 10/15/2015] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers with an overall survival rate of less than 5%. The poor patient outcome in PDAC is largely due to the high prevalence of systemic metastasis at the time of diagnosis and lack of effective therapeutics that target disseminated cells. The fact that the underlying mechanisms driving PDAC cell migration and dissemination are poorly understood have hindered drug development and compounded the lack of clinical success in this disease. Recent evidence indicates that mutational activation of K-Ras up-regulates eIF5A, a component of the cellular translational machinery that is critical for PDAC progression. However, the role of eIF5A in PDAC cell migration and metastasis has not been investigated. We report here that pharmacological inhibition or genetic knockdown of eIF5A reduces PDAC cell migration, invasion, and metastasis in vitro and in vivo. Proteomic profiling and bioinformatic analyses revealed that eIF5A controls an integrated network of cytoskeleton-regulatory proteins involved in cell migration. Functional interrogation of this network uncovered a critical RhoA/ROCK signaling node that operates downstream of eIF5A in invasive PDAC cells. Importantly, eIF5A mediates PDAC cell migration and invasion by modulating RhoA/ROCK protein expression levels. Together our findings implicate eIF5A as a cytoskeletal rheostat controlling RhoA/ROCK protein expression during PDAC cell migration and metastasis. Our findings also implicate the eIF5A/RhoA/ROCK module as a potential new therapeutic target to treat metastatic PDAC cells.
Collapse
Affiliation(s)
- Ken Fujimura
- From the Department of Pathology, Moores Cancer Center, University of California, San Diego, La Jolla, California 92093
| | - Sunkyu Choi
- From the Department of Pathology, Moores Cancer Center, University of California, San Diego, La Jolla, California 92093
| | - Meghan Wyse
- From the Department of Pathology, Moores Cancer Center, University of California, San Diego, La Jolla, California 92093
| | - Jan Strnadel
- From the Department of Pathology, Moores Cancer Center, University of California, San Diego, La Jolla, California 92093
| | - Tracy Wright
- From the Department of Pathology, Moores Cancer Center, University of California, San Diego, La Jolla, California 92093
| | - Richard Klemke
- From the Department of Pathology, Moores Cancer Center, University of California, San Diego, La Jolla, California 92093
| |
Collapse
|
88
|
Niesen J, Stein C, Brehm H, Hehmann-Titt G, Fendel R, Melmer G, Fischer R, Barth S. Novel EGFR-specific immunotoxins based on panitumumab and cetuximab show in vitro and ex vivo activity against different tumor entities. J Cancer Res Clin Oncol 2015; 141:2079-95. [PMID: 25899161 DOI: 10.1007/s00432-015-1975-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 04/15/2015] [Indexed: 12/15/2022]
Abstract
PURPOSE The epidermal growth factor receptor (EGFR) is overexpressed in many solid tumors. EGFR-specific monoclonal antibodies (mAbs), such as cetuximab and panitumumab, have been approved for the treatment of colorectal and head and neck cancer. To increase tissue penetration, we constructed single-chain fragment variable (scFv) antibodies derived from these mAbs and evaluated their potential for targeted cancer therapy. The resulting scFv-based EGFR-specific immunotoxins (ITs) combine target specificity of the full-size mAb with the cell-killing activity of a toxic effector domain, a truncated version of Pseudomonas exotoxin A (ETA'). METHODS The ITs and corresponding imaging probes were tested in vitro against four solid tumor entities (rhabdomyosarcoma, breast, prostate and pancreatic cancer). Specific binding and internalization of the ITs scFv2112-ETA' (from cetuximab) and scFv1711-ETA' (from panitumumab) were demonstrated by flow cytometry and for the scFv-SNAP-tag imaging probes by live cell imaging. Cytotoxic potential of the ITs was analyzed in cell viability and apoptosis assays. Binding of the ITs was proofed ex vivo on rhabdomyosarcoma, prostate and breast cancer formalin-fixed paraffin-embedded biopsies. RESULTS Both novel ITs showed significant pro-apoptotic and anti-proliferative effects toward the target cells, achieving IC50 values of 4 pM (high EGFR expression) to 460 pM (moderate EGFR expression). Additionally, rapid internalization and specific in vitro and ex vivo binding on patient tissue were confirmed. CONCLUSIONS These data demonstrate the potent therapeutic activity of two novel EGFR-specific ETA'-based ITs. Both molecules are promising candidates for further development toward clinical use in the treatment of various solid tumors to supplement the existing therapeutic regimes.
Collapse
Affiliation(s)
- Judith Niesen
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074, Aachen, Germany.
| | - Christoph Stein
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074, Aachen, Germany
- Department of Experimental Medicine and Immunotherapy, Institute of Applied Medical Engineering, RWTH Aachen University Clinic, Aachen, Germany
| | - Hannes Brehm
- Department of Experimental Medicine and Immunotherapy, Institute of Applied Medical Engineering, RWTH Aachen University Clinic, Aachen, Germany
| | | | - Rolf Fendel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074, Aachen, Germany
- Department of Experimental Medicine and Immunotherapy, Institute of Applied Medical Engineering, RWTH Aachen University Clinic, Aachen, Germany
| | | | - Rainer Fischer
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074, Aachen, Germany
- Institute of Molecular Biotechnology (Biology VII), RWTH Aachen University, Aachen, Germany
| | - Stefan Barth
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074, Aachen, Germany
- Department of Experimental Medicine and Immunotherapy, Institute of Applied Medical Engineering, RWTH Aachen University Clinic, Aachen, Germany
| |
Collapse
|
89
|
Primary outgrowth cultures are a reliable source of human pancreatic stellate cells. J Transl Med 2015; 95:1331-40. [PMID: 26322418 DOI: 10.1038/labinvest.2015.117] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/20/2015] [Indexed: 01/18/2023] Open
Abstract
Recent advances demonstrate a critical yet poorly understood role for the pancreatic stellate cell (PSC) in the pathogenesis of chronic pancreatitis (CP) and pancreatic cancer (PC). Progress in this area has been hampered by the availability, fidelity, and/or reliability of in vitro models of PSCs. We examined whether outgrowth cultures from human surgical specimens exhibited reproducible phenotypic and functional characteristics of PSCs. PSCs were cultured from surgical specimens of healthy pancreas, CP and PC. Growth dynamics, phenotypic characteristics, soluble mediator secretion profiles and co-culture with PC cells both in vitro and in vivo were assessed. Forty-seven primary cultures were established from 52 attempts, demonstrating universal α-smooth muscle actin and glial fibrillary acidic protein but negligible epithelial surface antigen expression. Modification of culture conditions consistently led to cytoplasmic lipid accumulation, suggesting induction of a quiescent phenotype. Secretion of growth factors, chemokines and cytokines did not significantly differ between donor pathologies, but did evolve over time in culture. Co-culture of PSCs with established PC cell lines resulted in significant changes in levels of multiple secreted mediators. Primary PSCs co-inoculated with PC cells in a xenograft model led to augmented tumor growth and metastasis. Therefore, regardless of donor pathology, outgrowth cultures produce PSCs that demonstrate consistent growth and protein secretion properties. Primary cultures from pancreatic surgical specimens, including malignancies, may represent a reliable source of human PSCs.
Collapse
|
90
|
Nakano H, Hasegawa T, Saito N, Furukawa K, Mukaida N, Kojima H, Okabe T, Nagano T. Design and synthesis of an in vivo-efficacious PIM3 kinase inhibitor as a candidate anti-pancreatic cancer agent. Bioorg Med Chem Lett 2015; 25:5687-93. [PMID: 26547690 DOI: 10.1016/j.bmcl.2015.10.098] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 10/29/2015] [Accepted: 10/31/2015] [Indexed: 12/16/2022]
Abstract
Serine/threonine kinase PIM3 is a potential therapeutic target for pancreatic cancer. Here, we describe the evolution of our previous PIM1 inhibitor 1 into PIM3 inhibitor 11 guided by use of the crystal structure of PIM1 as a surrogate to provide a basis for rational modification. Compound 11 potently inhibits PIM3 kinase activity, as well as growth of several pancreatic cancer cell lines. In a mouse xenograft model, 11 inhibited growth of human pancreatic cancer cell line PCI66 with negligible body weight loss. Thus, 11 appears to be a promising lead compound for further optimization to develop new anti-pancreatic cancer agents.
Collapse
Affiliation(s)
- Hirofumi Nakano
- Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Tsukasa Hasegawa
- Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Nae Saito
- Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Kaoru Furukawa
- Division of Molecular Bioregulation, Cancer Microenvironment Research Program, Cancer Research Institute, Kanazawa University, 13-1 Kakuma-machi, Kanazawa, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Microenvironment Research Program, Cancer Research Institute, Kanazawa University, 13-1 Kakuma-machi, Kanazawa, Japan
| | - Hirotatsu Kojima
- Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Takayoshi Okabe
- Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Tetsuo Nagano
- Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
91
|
Downstream mediators of the intratumoral interferon response suppress antitumor immunity, induce gemcitabine resistance and associate with poor survival in human pancreatic cancer. Cancer Immunol Immunother 2015; 64:1553-63. [PMID: 26423423 DOI: 10.1007/s00262-015-1760-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/17/2015] [Indexed: 02/07/2023]
Abstract
The cancer microenvironment allows tumor cells to evade immune surveillance through a variety of mechanisms. While interferon-γ (IFNγ) is central to effective antitumor immunity, its effects on the microenvironment are not as clear and have in some cancers been shown to induce immune checkpoint ligands. The heterogeneity of these responses to IFNγ remains poorly characterized in desmoplastic malignancies with minimal inflammatory cell infiltration, such as pancreatic cancer (PC). Thus, the IFNγ response within and on key cells of the PC microenvironment was evaluated. IFNγ induced expression of human leukocyte antigen (HLA) class I and II on PC cell lines, primary pancreatic cancer epithelial cells (PPCE) and patient-derived tumor-associated stroma, concomitant with an upregulation of PDL1 in the absence of CD80 and CD86 expression. As expected, IFNγ also induced high levels of CXCL10 from all cell types. In addition, significantly higher levels of CXCL10 were observed in PC specimens compared to those from chronic pancreatitis, whereby intratumoral CXCL10 concentration was an independent predictor of poor survival. Immunohistochemical analysis revealed a subset of CXCR3-positive cancer cells in over 90 % of PC specimens, as well as on a subset of cultured PC cell lines and PPCE, whereby exposure to CXCL10 induced resistance to the chemotherapeutic gemcitabine. These findings suggest that IFNγ has multiple effects on many cell types within the PC microenvironment that may lead to immune evasion, chemoresistance and shortened survival.
Collapse
|
92
|
Logsdon CD, Arumugam T, Ramachandran V. Animal Models of Gastrointestinal and Liver Diseases. The difficulty of animal modeling of pancreatic cancer for preclinical evaluation of therapeutics. Am J Physiol Gastrointest Liver Physiol 2015; 309:G283-91. [PMID: 26159697 PMCID: PMC4556944 DOI: 10.1152/ajpgi.00169.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/25/2015] [Indexed: 01/31/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is relatively rare but extremely lethal. Standard cytotoxic therapeutics provide little benefit. To date, newer targeted therapeutics have also not been highly successful. Often novel therapeutics that have appeared to perform well in preclinical models have failed in the clinic. Many factors contribute to these failures, but the one most often attributed is the shortcomings of the preclinical models. A plethora of animal models now exist for PDAC, including cell line xenografts, patient-derived xenografts, a wide variety of genetic mouse models, and syngeneic xenografts. These models have generated a tremendous amount of information useful for the understanding of PDAC. Yet none seems to well predict clinical outcomes of new treatments. This review will discuss how genetic instability and cellular heterogeneity make this disease so difficult to model accurately. We will also discuss the strengths and weaknesses of many of the popular models. Ultimately we will argue that there is no perfect model and that the best approach to understanding clinical performance is the use of multiple preclinical models with an understanding of their salient features.
Collapse
Affiliation(s)
- Craig D. Logsdon
- 1Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas and ,2Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Thiruvengadam Arumugam
- 1Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas and
| | - Vijaya Ramachandran
- 1Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas and
| |
Collapse
|
93
|
Zhang J, He DH, Zajac-Kaye M, Hochwald SN. A small molecule FAK kinase inhibitor, GSK2256098, inhibits growth and survival of pancreatic ductal adenocarcinoma cells. Cell Cycle 2015; 13:3143-9. [PMID: 25486573 DOI: 10.4161/15384101.2014.949550] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Focal adhesion kinase (FAK) hyperactivation is common in pancreatic ductal adenocarcinoma (PDAC). A small molecule, GSK2256098 (GlaxoSmithKline), has been developed to inhibit FAK activity through targeting the phosphorylation site of FAK, tyrosine (Y) 397. We sought to determine whether GSK2256098 inhibition of FAK Y397 phosphorylation attenuates PDAC-associated cell proliferation, motility and survival. Cultured PDAC cells were used as cellular models of GSK2256098-impaired abnormal growth. Western blot analysis, cell viability analysis, clonogenic survival, soft-agar and wound healing assays were performed. The responses of 6 PDAC cell lines in regards to FAK Y397 phosphorylation or activity to GSK2256098 treatments (0.1-10 μM) ranged from low (less than 20% inhibition) to high (more than 90% inhibition). The least and most sensitive cell lines (PANC-1 and L3.6P1) were selected for further analysis. GSK2256098 inhibition of FAK Y397 phosphorylation correlated with decreased levels of phosphorylated Akt and ERK in L3.6P1 cells. GSK2256098 decreased cell viability, anchorage-independent growth, and motility in a dose dependent manner. Current studies demonstrate that small molecule kinase inhibitors targeting FAK Y397 phosphorylation can inhibit PDAC cell growth. Assessments of FAK Y397 phosphorylation in biopsies may be used as a biomarker to select the subgroup of responsive patients and/or monitor the effects of GSK2256098 on FAK-modulated tumor growth during treatment.
Collapse
Affiliation(s)
- Jianliang Zhang
- a Department of Surgical Oncology ; Roswell Park Cancer Institute ; Elm and Carlton Streets ; Buffalo , NY USA
| | | | | | | |
Collapse
|
94
|
Neural Regulation of Pancreatic Cancer: A Novel Target for Intervention. Cancers (Basel) 2015; 7:1292-312. [PMID: 26193320 PMCID: PMC4586771 DOI: 10.3390/cancers7030838] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 07/07/2015] [Accepted: 07/13/2015] [Indexed: 12/16/2022] Open
Abstract
The tumor microenvironment is known to play a pivotal role in driving cancer progression and governing response to therapy. This is of significance in pancreatic cancer where the unique pancreatic tumor microenvironment, characterized by its pronounced desmoplasia and fibrosis, drives early stages of tumor progression and dissemination, and contributes to its associated low survival rates. Several molecular factors that regulate interactions between pancreatic tumors and their surrounding stroma are beginning to be identified. Yet broader physiological factors that influence these interactions remain unclear. Here, we discuss a series of preclinical and mechanistic studies that highlight the important role chronic stress plays as a physiological regulator of neural-tumor interactions in driving the progression of pancreatic cancer. These studies propose several approaches to target stress signaling via the β-adrenergic signaling pathway in order to slow pancreatic tumor growth and metastasis. They also provide evidence to support the use of β-blockers as a novel therapeutic intervention to complement current clinical strategies to improve cancer outcome in patients with pancreatic cancer.
Collapse
|
95
|
Abstract
OBJECTIVES Survivin, an antiapoptotic gene inhibited by p53, is overexpressed in human cancers and correlates with chemotherapy resistance. Here, we investigated the mutual regulatory mechanism between MGMT (O-methylguanine DNA methyltransferase) and survivin. METHODS This study used standard techniques for protein and messenger RNA levels, promoter activity, protein-DNA interaction, cell viability, and correlative animal model. RESULTS O-benzylguanine (BG), a potent inhibitor of MGMT (a DNA repair protein), curtails the expression of survivin in pancreatic cancer. Silencing MGMT by small interfering RNA down-regulates survivin transcription. p53 inhibition enhances MGMT and survivin expressions. When p53 was silenced, BG-induced MGMT inhibition was not associated with the down-regulation of survivin, underscoring the regulatory role of p53 in the MGMT-survivin axis. O-benzylguanine inhibits survivin and PCNA (proliferating cell nuclear antigen) at messenger RNA and protein levels in PANC-1 and L3.6pl cells and decreases survivin promoter activity via increased p53 recruitment to the survivin promoter. In orthotopic pancreatic xenografts established in nude mice, BG ± gemcitabine (GEM) decrease survivin expression in tumor tissue; protein levels and immunohistochemistry show significant decrease in survivin and PCNA levels, which correlate with increased sensitivity to GEM. CONCLUSIONS MGMT inhibition is associated with decrease in survivin expression and increase in sensitivity to GEM in pancreatic cancer.
Collapse
|
96
|
Kashatus JA, Nascimento A, Myers LJ, Sher A, Byrne FL, Hoehn KL, Counter CM, Kashatus DF. Erk2 phosphorylation of Drp1 promotes mitochondrial fission and MAPK-driven tumor growth. Mol Cell 2015; 57:537-51. [PMID: 25658205 DOI: 10.1016/j.molcel.2015.01.002] [Citation(s) in RCA: 509] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 11/20/2014] [Accepted: 12/29/2014] [Indexed: 02/08/2023]
Abstract
Ras is mutated in up to 30% of cancers, including 90% of pancreatic ductal adenocarcinomas, causing it to be constitutively GTP-bound, and leading to activation of downstream effectors that promote a tumorigenic phenotype. As targeting Ras directly is difficult, there is a significant effort to understand the downstream biological processes that underlie its protumorigenic activity. Here, we show that expression of oncogenic Ras or direct activation of the MAPK pathway leads to increased mitochondrial fragmentation and that blocking this phenotype, through knockdown of the mitochondrial fission-mediating GTPase Drp1, inhibits tumor growth. This fission is driven by Erk2-mediated phosphorylation of Drp1 on Serine 616, and both this phosphorylation and mitochondrial fragmentation are increased in human pancreatic cancer. Finally, this phosphorylation is required for Ras-associated mitochondrial fission, and its inhibition is sufficient to block xenograft growth. Collectively, these data suggest mitochondrial fission may be a target for treating MAPK-driven malignancies.
Collapse
Affiliation(s)
- Jennifer A Kashatus
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Aldo Nascimento
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Lindsey J Myers
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Annie Sher
- Department of Pharmacology and Cancer Biology, Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Frances L Byrne
- Department of Pharmacology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Kyle L Hoehn
- Department of Pharmacology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Christopher M Counter
- Department of Pharmacology and Cancer Biology, Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - David F Kashatus
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA.
| |
Collapse
|
97
|
Rychahou P, Haque F, Shu Y, Zaytseva Y, Weiss HL, Lee EY, Mustain W, Valentino J, Guo P, Evers BM. Delivery of RNA nanoparticles into colorectal cancer metastases following systemic administration. ACS NANO 2015; 9:1108-16. [PMID: 25652125 PMCID: PMC4613746 DOI: 10.1021/acsnano.5b00067] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The majority of deaths from all cancers, including colorectal cancer (CRC), is a result of tumor metastasis to distant organs. To date, an effective and safe system capable of exclusively targeting metastatic cancers that have spread to distant organs or lymph nodes does not exist. Here, we constructed multifunctional RNA nanoparticles, derived from the three-way junction (3WJ) of bacteriophage phi29 motor pRNA, to target metastatic cancer cells in a clinically relevant mouse model of CRC metastasis. The RNA nanoparticles demonstrated metastatic tumor homing without accumulation in normal organ tissues surrounding metastatic tumors. The RNA nanoparticles simultaneously targeted CRC cancer cells in major sites of metastasis, such as liver, lymph nodes, and lung. Our results demonstrate the therapeutic potential of these RNA nanoparticles as a delivery system for the treatment of CRC metastasis.
Collapse
Affiliation(s)
- Piotr Rychahou
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, United States
- Department of Surgery, University of Kentucky, Lexington, KY 40536, United States
| | - Farzin Haque
- Nanobiotechnology Center, University of Kentucky, Lexington, KY 40536, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, United States
| | - Yi Shu
- Nanobiotechnology Center, University of Kentucky, Lexington, KY 40536, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, United States
| | - Yekaterina Zaytseva
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, United States
| | - Heidi L. Weiss
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, United States
| | - Eun Y. Lee
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, United States
- Department of Surgery, University of Kentucky, Lexington, KY 40536, United States
- Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40536, United States
| | - William Mustain
- Department of Surgery, University of Kentucky, Lexington, KY 40536, United States
| | - Joseph Valentino
- Department of Surgery, University of Kentucky, Lexington, KY 40536, United States
| | - Peixuan Guo
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, United States
- Nanobiotechnology Center, University of Kentucky, Lexington, KY 40536, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, United States
| | - B. Mark Evers
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, United States
- Department of Surgery, University of Kentucky, Lexington, KY 40536, United States
- Corresponding Author: B. Mark Evers, M.D., Markey Cancer Center, University of Kentucky, 800 Rose Street, CC140, Lexington, KY 40536, Phone: 859-323-6556, Fax: 859-323-2074,
| |
Collapse
|
98
|
Zhao Y, Zhao L, Ischenko I, Bao Q, Schwarz B, Nieß H, Wang Y, Renner A, Mysliwietz J, Jauch KW, Nelson PJ, Ellwart JW, Bruns CJ, Camaj P. Antisense inhibition of microRNA-21 and microRNA-221 in tumor-initiating stem-like cells modulates tumorigenesis, metastasis, and chemotherapy resistance in pancreatic cancer. Target Oncol 2015; 10:535-48. [PMID: 25639539 DOI: 10.1007/s11523-015-0360-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/15/2015] [Indexed: 02/07/2023]
Abstract
Our preliminary studies identified a small population side population (SP) cells in pancreatic cancer cells with stem cell-like properties, which were able to induce fast and aggressive tumor formation in nude mice. Gene expression analysis showed a significant difference in the expression of more than 1,300 genes in SP cells, among which a highly significant difference in microRNA expression of miR-21 and miR-221 between SP and NSP cells was identified. SP cells were identified and characterized by flow cytometry using Hoechst 33342 dye staining from a highly metastatic human pancreatic cancer cell line (L3.6pl). Antagomir transfection was performed using miRNA-21 and miRNA-221 antisense oligonucleotides (ASOs) and followed by detection of cell apoptosis, cell cycle progression, chemosensitivity, and invasion. Sorted SP cells from gemcitabine-resistant L3.6pl cells (L3.6pl(Gres)-SP) cells were orthotopically implanted in nude mice with or without miRNA-21 and miRNA-221 ASOs mono- and combination therapy. The administration of antagomir-21 and antagomir-221 significantly reduced the SP cell fraction, decreased SP cell differentiation, and downstream gene regulation, and thereby induced reduction of L3.6pl cell proliferation, invasion, and chemoresistance against gemcitabine and 5-Fluorouracil. Combination of ASOs therapy against miRNA-21 and miRNA-221 significantly inhibited primary tumor growth and metastasis compared to single antagomir treatment, especially, in L3.6plGres-SP-induced pancreatic tumor growth in vivo. These findings further indicate that the inhibition of miR-21 and miR-221 appear particularly suitable to target stem-like subpopulations and address their specific biological function to promote tumor progression in pancreatic cancer.
Collapse
Affiliation(s)
- Yue Zhao
- Department of General, Visceral und Vascular Surgery, Otto-von-Guericke University, Leipziger Strasse 44, 39120, Magdeburg, Germany.
| | - Lu Zhao
- Department of General, Visceral und Vascular Surgery, University of Munich, Campus Grosshadern, Munich, Germany
| | - Ivan Ischenko
- Department of General, Visceral und Vascular Surgery, University of Munich, Campus Grosshadern, Munich, Germany
| | - Qi Bao
- Department of General, Visceral und Vascular Surgery, University of Munich, Campus Grosshadern, Munich, Germany
| | - Bettina Schwarz
- Department of General, Visceral und Vascular Surgery, University of Munich, Campus Grosshadern, Munich, Germany
| | - Hanno Nieß
- Department of General, Visceral und Vascular Surgery, University of Munich, Campus Grosshadern, Munich, Germany
| | - Yan Wang
- Department of General, Visceral und Vascular Surgery, Otto-von-Guericke University, Leipziger Strasse 44, 39120, Magdeburg, Germany
| | - Andrea Renner
- Department of General, Visceral und Vascular Surgery, University of Munich, Campus Grosshadern, Munich, Germany
| | - Josef Mysliwietz
- Institute of Molecular Immunology, Helmholtz Center Munich, Munich, Germany
| | - Karl-Walter Jauch
- Department of General, Visceral und Vascular Surgery, University of Munich, Campus Grosshadern, Munich, Germany
| | - Peter J Nelson
- Clinical Biochemistry Group, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Joachim W Ellwart
- Institute of Molecular Immunology, Helmholtz Center Munich, Munich, Germany
| | - Christiane J Bruns
- Department of General, Visceral und Vascular Surgery, Otto-von-Guericke University, Leipziger Strasse 44, 39120, Magdeburg, Germany
| | - Peter Camaj
- Department of General, Visceral und Vascular Surgery, Otto-von-Guericke University, Leipziger Strasse 44, 39120, Magdeburg, Germany.
| |
Collapse
|
99
|
Loo JM, Scherl A, Nguyen A, Man FY, Weinberg E, Zeng Z, Saltz L, Paty PB, Tavazoie SF. Extracellular metabolic energetics can promote cancer progression. Cell 2015; 160:393-406. [PMID: 25601461 DOI: 10.1016/j.cell.2014.12.018] [Citation(s) in RCA: 262] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 09/25/2014] [Accepted: 11/19/2014] [Indexed: 12/12/2022]
Abstract
Colorectal cancer primarily metastasizes to the liver and globally kills over 600,000 people annually. By functionally screening 661 microRNAs (miRNAs) in parallel during liver colonization, we have identified miR-551a and miR-483 as robust endogenous suppressors of liver colonization and metastasis. These miRNAs convergently target creatine kinase, brain-type (CKB), which phosphorylates the metabolite creatine, to generate phosphocreatine. CKB is released into the extracellular space by metastatic cells encountering hepatic hypoxia and catalyzes production of phosphocreatine, which is imported through the SLC6A8 transporter and used to generate ATP—fueling metastatic survival. Combinatorial therapeutic viral delivery of miR-551a and miR-483-5p through single-dose adeno-associated viral (AAV) delivery significantly suppressed colon cancer metastasis, as did CKB inhibition with a small-molecule inhibitor. Importantly, human liver metastases express higher CKB and SLC6A8 levels and reduced miR-551a/miR-483 levels relative to primary tumors. We identify the extracellular space as an important compartment for malignant energetic catalysis and therapeutic targeting.
Collapse
Affiliation(s)
- Jia Min Loo
- Laboratory of Systems Cancer Biology, Rockefeller University, New York, NY 10065, USA
| | - Alexis Scherl
- Laboratory of Systems Cancer Biology, Rockefeller University, New York, NY 10065, USA
| | - Alexander Nguyen
- Laboratory of Systems Cancer Biology, Rockefeller University, New York, NY 10065, USA
| | - Fung Ying Man
- Laboratory of Systems Cancer Biology, Rockefeller University, New York, NY 10065, USA
| | - Ethan Weinberg
- Laboratory of Systems Cancer Biology, Rockefeller University, New York, NY 10065, USA
| | - Zhaoshi Zeng
- Department of Surgery, Memorial-Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Leonard Saltz
- Department of Medicine, Memorial-Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Philip B Paty
- Department of Surgery, Memorial-Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sohail F Tavazoie
- Laboratory of Systems Cancer Biology, Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
100
|
Longitudinal Bioluminescence Imaging of Primary Versus Abdominal Metastatic Tumor Growth in Orthotopic Pancreatic Tumor Models in NSG Mice. Pancreas 2015; 44:64-75. [PMID: 25406955 PMCID: PMC4262664 DOI: 10.1097/mpa.0000000000000238] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES The purpose of the present study was to develop and validate noninvasive bioluminescence imaging methods for differentially monitoring primary and abdominal metastatic tumor growth in mouse orthotopic models of pancreatic cancer. METHODS A semiautomated maximum entropy segmentation method was implemented for the primary tumor region of interest, and a rule-based method for manually drawing a region of interest for the abdominal metastatic region was developed for monitoring tumor growth in orthotopic models of pancreatic cancer. The 2 region-of-interest methods were validated by having 2 observers independently segment Panc-1 tumors, and the results were compared with the number of mesenteric lymph node nodules and histopathologic assessment of liver metastases. The findings were extended to orthotopic tumors of the more metastatic MIA PaCa-2 and AsPC-1 cells where separate groups of animals were implanted with different numbers of cells. RESULTS The results demonstrated that the segmentation methods were highly reliable, reproducible, and robust and allowed statistically significant discrimination in the growth rates of primary and abdominal metastatic tumors of different cell lines implanted with different numbers of cells. CONCLUSIONS The present results demonstrate that primary tumors and abdominal metastatic foci in orthotopic pancreatic cancer models can be reliably quantified separately and noninvasively over time with bioluminescence imaging.
Collapse
|