1
|
Shatalov P, Falaleeva N, Bykova E, Korostin D, Belova V, Zabolotneva A, Shinkarkina A, Gorbachev AY, Potievskiy M, Surkova V, Khailova ZV, Kulemin N, Baranovskii D, Kostin A, Kaprin A, Shegai P. Genetic and therapeutic landscapes in cohort of pancreatic adenocarcinomas: next-generation sequencing and machine learning for full tumor exome analysis. Oncotarget 2024; 15:91-103. [PMID: 38329726 PMCID: PMC10852064 DOI: 10.18632/oncotarget.28512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 09/04/2023] [Indexed: 02/09/2024] Open
Abstract
About 7% of all cancer deaths are caused by pancreatic cancer (PCa). PCa is known for its lowest survival rates among all oncological diseases and heterogenic molecular profile. Enormous amount of genetic changes, including somatic mutations, exceeds the limits of routine clinical genetic laboratory tests and further stagnates the development of personalized treatments. We aimed to build a mutational landscape of PCa in the Russian population based on full exome next-generation sequencing (NGS) of the limited group of patients. Applying a machine learning model on full exome individual data we received personalized recommendations for targeted treatment options for each clinical case and summarized them in the unique therapeutic landscape.
Collapse
Affiliation(s)
- P.A. Shatalov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - N.A. Falaleeva
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - E.A. Bykova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - D.O. Korostin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - V.A. Belova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - A.A. Zabolotneva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - A.P. Shinkarkina
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - A. Yu Gorbachev
- FSBI “Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine” FMBA, Moscow 119435, Russia
| | - M.B. Potievskiy
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - V.S. Surkova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - Zh V. Khailova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - N.A. Kulemin
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - Denis Baranovskii
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
- Peoples Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - A.A. Kostin
- Peoples Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - A.D. Kaprin
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
- Peoples Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - P.V. Shegai
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| |
Collapse
|
2
|
DeCoste R, Amemiya Y, Nersesian S, Westhaver L, Lee SN, Carter MD, Sapp HL, Stueck AE, Arnason T, Boudreau J, Seth A, Huang WY. PAXgene Fixation for Pancreatic Cancer: Implications for Molecular and Surgical Pathology. J Clin Med 2022; 11:jcm11144241. [PMID: 35888003 PMCID: PMC9319620 DOI: 10.3390/jcm11144241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 12/04/2022] Open
Abstract
Genomic profiling of pancreatic cancer using small core biopsies has taken an increasingly prominent role in precision medicine. However, if not appropriately preserved, nucleic acids (NA) from pancreatic tissues are known to be susceptible to degradation due to high intrinsic levels of nucleases. PAXgene fixation (PreAnalytix, Switzerland) represents a novel formalin-free tissue preservation method. We sought to compare the NA and histomorphological preservation of pancreatic cancer tissues preserved with PAXgene-fixed paraffin-embedding (PFPE) and formalin-fixed paraffin-embedding (FFPE). Tissues from 19 patients were obtained prospectively from pancreaticoduodenectomy specimens and evaluated by four gastrointestinal pathologists. The extracted NA were quantified by Nanodrop and Qubit and assessed for quality by qPCR, targeted next-generation sequencing (NGS) assay, and RNA-sequencing. Our results demonstrated that, when assessed blindly for morphological quality, the four pathologists deemed the PFPE slides adequate for diagnostic purposes. PFPE tissues enable greater yields of less fragmented and more amplifiable DNA. PFPE tissues demonstrated significantly improved quality control (QC) metrics in a targeted NGS assay including Median Absolute Pair-wise Difference (MAPD) scores. Our results support the use of PAXgene fixative for the processing of specimens from pancreatic cancers with the potential benefits of improved yields for more amplifiable DNA in low-yield biopsy specimens and its ideal use for amplicon-based NGS assays.
Collapse
Affiliation(s)
- Ryan DeCoste
- Department of Pathology & Laboratory Medicine, QEII Health Sciences Centre, Nova Scotia Health Authority (Central Zone), Halifax, NS B3H 1V8, Canada; (R.D.); (M.D.C.); (H.L.S.); (A.E.S.); (T.A.)
- Department of Pathology, Dalhousie University, Halifax, NS B3H 1V8, Canada; (L.W.); (J.B.)
| | - Yutaka Amemiya
- Sunnybrook Research Institute Genomics Core Facility, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (Y.A.); (A.S.)
| | - Sarah Nersesian
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (S.N.); (S.N.L.)
| | - Lauren Westhaver
- Department of Pathology, Dalhousie University, Halifax, NS B3H 1V8, Canada; (L.W.); (J.B.)
| | - Stacey N. Lee
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (S.N.); (S.N.L.)
| | - Michael D. Carter
- Department of Pathology & Laboratory Medicine, QEII Health Sciences Centre, Nova Scotia Health Authority (Central Zone), Halifax, NS B3H 1V8, Canada; (R.D.); (M.D.C.); (H.L.S.); (A.E.S.); (T.A.)
- Department of Pathology, Dalhousie University, Halifax, NS B3H 1V8, Canada; (L.W.); (J.B.)
| | - Heidi L. Sapp
- Department of Pathology & Laboratory Medicine, QEII Health Sciences Centre, Nova Scotia Health Authority (Central Zone), Halifax, NS B3H 1V8, Canada; (R.D.); (M.D.C.); (H.L.S.); (A.E.S.); (T.A.)
- Department of Pathology, Dalhousie University, Halifax, NS B3H 1V8, Canada; (L.W.); (J.B.)
| | - Ashley E. Stueck
- Department of Pathology & Laboratory Medicine, QEII Health Sciences Centre, Nova Scotia Health Authority (Central Zone), Halifax, NS B3H 1V8, Canada; (R.D.); (M.D.C.); (H.L.S.); (A.E.S.); (T.A.)
- Department of Pathology, Dalhousie University, Halifax, NS B3H 1V8, Canada; (L.W.); (J.B.)
| | - Thomas Arnason
- Department of Pathology & Laboratory Medicine, QEII Health Sciences Centre, Nova Scotia Health Authority (Central Zone), Halifax, NS B3H 1V8, Canada; (R.D.); (M.D.C.); (H.L.S.); (A.E.S.); (T.A.)
- Department of Pathology, Dalhousie University, Halifax, NS B3H 1V8, Canada; (L.W.); (J.B.)
| | - Jeanette Boudreau
- Department of Pathology, Dalhousie University, Halifax, NS B3H 1V8, Canada; (L.W.); (J.B.)
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (S.N.); (S.N.L.)
| | - Arun Seth
- Sunnybrook Research Institute Genomics Core Facility, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (Y.A.); (A.S.)
- Department of Laboratory Medicine & Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Weei-Yuarn Huang
- Department of Laboratory Medicine & Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence:
| |
Collapse
|
3
|
Chen Q, Su L, He X, Li J, Cao Y, Wu Q, Qin J, He Z, Huang X, Yang H, Li J. Poly(beta-amino ester)-Based Nanoparticles Enable Nonviral Delivery of Base Editors for Targeted Tumor Gene Editing. Biomacromolecules 2022; 23:2116-2125. [PMID: 35388688 DOI: 10.1021/acs.biomac.2c00137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Base editing is an emerging genome editing technology with the advantages of precise base corrections, no double-strand DNA breaks, and no need for templates, which provides an alternative treatment option for tumors with point mutations. However, effective nonviral delivery systems for base editors (BEs) are still limited. Herein, a series of poly(beta-amino esters) (PBAEs) with varying backbones, side chains, and end caps were synthesized to deliver plasmids of BEs and sgRNA. Efficient transfection and base editing were achieved in HEK-293T-sEGFP and U87-MG-sEGFP reporter cell lines by using lead PBAEs, which were superior to PEI and lipo3k. A single intratumor injection of PBAE/pDNA nanoparticles induced the robust conversion of stopped-EGFP into EGFP in mice bearing xenograft glioma tumors, indicating successful gene editing by ABEmax-NG. Overall, these results demonstrated that PBAEs can efficiently deliver BEs for tumor gene editing both in vitro and in vivo.
Collapse
Affiliation(s)
- Qimingxing Chen
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Lili Su
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaoyan He
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jinwei Li
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yan Cao
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qingxia Wu
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jianchao Qin
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zongxing He
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xingxu Huang
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Huiying Yang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jianfeng Li
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
4
|
Ikeda Y, Munekane M, Yamada Y, Kawakami M, Amano I, Sano K, Mukai T, Kambe T, Shitan N. Enhancing effect of Panax ginseng on Zip4-mediated zinc influx into the cytosol. J Ginseng Res 2021; 46:248-254. [PMID: 35509828 PMCID: PMC9058843 DOI: 10.1016/j.jgr.2021.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 06/05/2021] [Accepted: 06/09/2021] [Indexed: 11/26/2022] Open
Abstract
Background Zinc homeostasis is essential for human health and is regulated by several zinc transporters including ZIP and ZnT. ZIP4 is expressed in the small intestine and is important for zinc absorption from the diet. We investigated in the present study the effects of Panax ginseng (P. ginseng) extract on modulating Zip4 expression and cellular zinc levels in mouse Hepa cells. Methods Hepa cells were transfected with a luciferase reporter plasmid that contains metal-responsive elements, incubated with P. ginseng extract, and luciferase activity was measured. Using 65ZnCl2, zinc uptake in P. ginseng-treated cells was measured. The expression of Zip4 mRNA and protein in Hepa cells was also investigated. Finally, using a luciferase reporter assay system, the effects of several ginsenosides were monitored. Results The luciferase activity in cells incubated with P. ginseng extract was significantly higher than that of control cells cultured in normal medium. Hepa cells treated with P. ginseng extract exhibited higher zinc uptake. P. ginseng extract induced Zip4 mRNA expression, which resulted in an enhancement of Zip4 protein expression. Furthermore, some ginsenosides, such as ginsenoside Rc and Re, enhanced luciferase activity driven by intracellular zinc levels. Conclusion P. ginseng extract induced Zip4 expression at the mRNA and protein level and resulted in higher zinc uptake in Hepa cells. Some ginsenosides facilitated zinc influx. On the basis of these results, we suggest a novel effect of P. ginseng on Zip4-mediated zinc influx, which may provide a new strategy for preventing zinc deficiency.
Collapse
|
5
|
Liu SH, Yu J, Creeden JF, Sutton JM, Markowiak S, Sanchez R, Nemunaitis J, Kalinoski A, Zhang JT, Damoiseaux R, Erhardt P, Brunicardi FC. Repurposing metformin, simvastatin and digoxin as a combination for targeted therapy for pancreatic ductal adenocarcinoma. Cancer Lett 2020; 491:97-107. [PMID: 32829010 DOI: 10.1016/j.canlet.2020.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/23/2020] [Accepted: 08/01/2020] [Indexed: 12/12/2022]
Abstract
Patients with pancreatic adenocarcinoma (PDAC) have a 5-year survival rate of 8%, the lowest of any cancer in the United States. Traditional chemotherapeutic regimens, such as gemcitabine- and fluorouracil-based regimens, often only prolong survival by months. Effective precision targeted therapy is therefore urgently needed to substantially improve survival. In an effort to expedite approval and delivery of targeted therapy to patients, we utilized a platform to develop a novel combination of FDA approved drugs that would target pancreaticoduodenal homeobox1 (PDX1) and baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5) utilizing super-promoters of the target genes to interrogate an FDA approved drug library. We identified and selected metformin, simvastatin and digoxin (C3) as a novel combination of FDA approved drugs, which were shown to effectively target PDX1 and BIRC5 in human PDAC tumors in mice with no toxicity.
Collapse
Affiliation(s)
- Shi-He Liu
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA; Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA.
| | - Juehua Yu
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Justin F Creeden
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA; Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Jeffrey M Sutton
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Stephen Markowiak
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Robbi Sanchez
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - John Nemunaitis
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Andrea Kalinoski
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Jian-Ting Zhang
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Paul Erhardt
- Department of Medicinal and Biological Chemistry, University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, OH, 43614, USA
| | - F Charles Brunicardi
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA; Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| |
Collapse
|
6
|
Kaşıkcı E, Aydemir E, Bayrak ÖF, Şahin F. Inhibition of Migration, Invasion and Drug Resistance of Pancreatic Adenocarcinoma Cells - Role of Snail, Slug and Twist and Small Molecule Inhibitors. Onco Targets Ther 2020; 13:5763-5777. [PMID: 32606788 PMCID: PMC7308789 DOI: 10.2147/ott.s253418] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The main purpose of this study is to demonstrate the effects of epithelial to mesenchymal transition activating transcription factor silencing (EMT-ATF silencing) on migration, invasion, drug resistance and tumor-forming abilities of various pancreatic cancer cell lines. Additionally, the contribution of small molecule inhibitors of EMT (SD-208 and CX4945) to the effects of gene silencing was evaluated. METHODS EMT activating transcription factors "Snail, Slug and Twist" were silenced by short hairpins on Panc-1, MIA PaCa-2, BxPC-3, and AsPC-1 pancreatic cancer cell lines. The changes in migration, invasion, laminin attachment, cancer stem-like cell properties and tumor-forming abilities were investigated. Chemosensitivity assays and small molecule inhibitors of EMT were applied to the metastatic pancreatic cancer cell line AsPC-1. RESULTS EMT-ATF silencing reduced EMT and stem cell-like characteristics of pancreatic cancer cell lines. Following EMT-ATF silencing amongst the four PC cell lines, AsPC-1 showed the best response and was chosen for further chemoresistance and combinational therapy applications. EMT downregulated AsPC-1 cells showed less resistance to select chemotherapeutics compared to the control group. Both small molecule inhibitors enhanced the outcomes of EMT-ATF silencing. CONCLUSION Overall it was found that EMT-ATF silencing, either by EMT-ATF silencing or with the enhancement by small molecules, is a good candidate to treat pancreatic cancer since it simultaneously minimizes metastasis, stem cell properties, and drug resistance.
Collapse
Affiliation(s)
- Ezgi Kaşıkcı
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul34755, Turkey
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY10461, USA
| | - Esra Aydemir
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul34755, Turkey
| | - Ömer Faruk Bayrak
- Department of Medical Genetics, Yeditepe University Medical School and Yeditepe University Hospital, Istanbul34718, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul34755, Turkey
| |
Collapse
|
7
|
Jain A, Agostini LC, McCarthy GA, Chand SN, Ramirez A, Nevler A, Cozzitorto J, Schultz CW, Lowder CY, Smith KM, Waddell ID, Raitses-Gurevich M, Stossel C, Gorman YG, Atias D, Yeo CJ, Winter JM, Olive KP, Golan T, Pishvaian MJ, Ogilvie D, James DI, Jordan AM, Brody JR. Poly (ADP) Ribose Glycohydrolase Can Be Effectively Targeted in Pancreatic Cancer. Cancer Res 2019; 79:4491-4502. [PMID: 31273064 PMCID: PMC6816506 DOI: 10.1158/0008-5472.can-18-3645] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 05/06/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022]
Abstract
Patients with metastatic pancreatic ductal adenocarcinoma (PDAC) have an average survival of less than 1 year, underscoring the importance of evaluating novel targets with matched targeted agents. We recently identified that poly (ADP) ribose glycohydrolase (PARG) is a strong candidate target due to its dependence on the pro-oncogenic mRNA stability factor HuR (ELAVL1). Here, we evaluated PARG as a target in PDAC models using both genetic silencing of PARG and established small-molecule PARG inhibitors (PARGi), PDDX-01/04. Homologous repair-deficient cells compared with homologous repair-proficient cells were more sensitive to PARGi in vitro. In vivo, silencing of PARG significantly decreased tumor growth. PARGi synergized with DNA-damaging agents (i.e., oxaliplatin and 5-fluorouracil), but not with PARPi therapy. Mechanistically, combined PARGi and oxaliplatin treatment led to persistence of detrimental PARylation, increased expression of cleaved caspase-3, and increased γH2AX foci. In summary, these data validate PARG as a relevant target in PDAC and establish current therapies that synergize with PARGi. SIGNIFICANCE: PARG is a potential target in pancreatic cancer as a single-agent anticancer therapy or in combination with current standard of care.
Collapse
Affiliation(s)
- Aditi Jain
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Lebaron C Agostini
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Grace A McCarthy
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Saswati N Chand
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - AnnJosette Ramirez
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Avinoam Nevler
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Joseph Cozzitorto
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Christopher W Schultz
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Cinthya Yabar Lowder
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Kate M Smith
- Drug Discovery Unit, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Ian D Waddell
- Drug Discovery Unit, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | | | - Chani Stossel
- Oncology Institute, Chaim Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yulia Glick Gorman
- Oncology Institute, Chaim Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Dikla Atias
- Oncology Institute, Chaim Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Charles J Yeo
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jordan M Winter
- Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Kenneth P Olive
- Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Talia Golan
- Oncology Institute, Chaim Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michael J Pishvaian
- Department of Gastrointestinal Medical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Donald Ogilvie
- Drug Discovery Unit, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Dominic I James
- Drug Discovery Unit, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Allan M Jordan
- Drug Discovery Unit, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Jonathan R Brody
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
8
|
Zeng Q, Liu YM, Liu J, Han J, Guo JX, Lu S, Huang XM, Yi P, Lang JY, Zhang P, Wang CT. Inhibition of ZIP4 reverses epithelial-to-mesenchymal transition and enhances the radiosensitivity in human nasopharyngeal carcinoma cells. Cell Death Dis 2019; 10:588. [PMID: 31383854 PMCID: PMC6683154 DOI: 10.1038/s41419-019-1807-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/29/2019] [Accepted: 07/11/2019] [Indexed: 02/05/2023]
Abstract
ZIP4 is a zinc transporter involved in epithelial cell morphology and migration in various cancers. In the epithelial-to-mesenchymal transition (EMT), epithelial cells transition into mesenchymal cells. The EMT plays a crucial role in invasiveness and metastasis during tumorigenesis. The aim of this study was to investigate the role of ZIP4 in the invasiveness and radiosensitivity of human nasopharyngeal carcinoma (NPC). In this study, results from 99 human patients with NPC showed that ZIP4 expression levels significantly correlated with a higher TN (tumor, lymph node) classification, as well as shorter overall survival (OS), recurrence-free survival (RFS), and distant metastasis-free survival (DMFS). Forced overexpression of ZIP4 promoted the migration and invasion of C666-1 cells through regulation of the EMT process. In contrast, ZIP4 silencing by lentivirus-mediated shRNA inhibited the EMT and metastasis of C666-1 cells in vitro and in vivo. Importantly, protein microarray analyses showed that downregulation of ZIP4 in C666-1 cells resulted in the decreased abundance of phosphoinositide 3-kinase (PI3K) p85 (Tyr607), phosphorylated (p)-Akt (Ser473), phosphorylated (p)-Akt (Thr308), and phosphorylated glycogen synthase kinase 3β (pGSK3β; Ser9). These data suggest that ZIP4 induces the EMT and promotes migration and invasion via the PI3K/Akt signaling pathway in NPC. Moreover, ZIP4 silencing significantly enhanced radiation-induced apoptosis and growth inhibition of human C666-1 cells in vitro and enhanced the antitumor activity of ionizing radiation (IR), leading to tumor growth inhibition in vivo. These results demonstrate that ZIP4 is a novel prognostic factor for malignant NPC progression. More importantly, targeting ZIP4, along with radiotherapy, may be an effective new treatment for NPC.
Collapse
Affiliation(s)
- Qi Zeng
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China.,Department of Gynaecology and Obstetrics, Institute of Surgery Research, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Yi-Min Liu
- Department of Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jun Liu
- Department of Otorhinolaryngology, Head and Neck Surgey, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Jian Han
- Department of Gynaecology and Obstetrics, Institute of Surgery Research, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Jian-Xin Guo
- Department of Gynaecology and Obstetrics, Institute of Surgery Research, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Shun Lu
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Radiation Oncology Key Laboratory of Sichuan Province, 610041, Chengdu, China
| | - Xue-Mei Huang
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Radiation Oncology Key Laboratory of Sichuan Province, 610041, Chengdu, China
| | - Ping Yi
- Department of Gynaecology and Obstetrics, Institute of Surgery Research, Daping Hospital, Army Medical University (Third Military Medical University), 400042, Chongqing, China
| | - Jin-Yi Lang
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Radiation Oncology Key Laboratory of Sichuan Province, 610041, Chengdu, China
| | - Peng Zhang
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Radiation Oncology Key Laboratory of Sichuan Province, 610041, Chengdu, China.
| | - Chun-Ting Wang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
9
|
Liu J, Hu G, Gong Y, Yu Q, He B, Li W, He Z, Hao W, He Z, Liu Y. Silencing of TRPM8 inhibits aggressive tumor phenotypes and enhances gemcitabine sensitivity in pancreatic cancer. Pancreatology 2018; 18:935-944. [PMID: 30316690 DOI: 10.1016/j.pan.2018.08.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 07/04/2018] [Accepted: 08/21/2018] [Indexed: 12/11/2022]
Abstract
The transient receptor potential TRPM8 ion channel is required for cellular proliferation in pancreatic epithelia and adenocarcinoma. To elucidate the mechanism that mediates the function of TRPM8, we examined its role in the proliferation and invasion of pancreatic cancer (PC) cells. TRPM8 expression increased in both the PC tissues and cell lines; a high TRPM8 expression was correlated with poorer prognosis in patients with PC. In PC cell lines, PACN-1 and BxPC-3, Ca2+ influxes could be evoked by TRPM8; the sensitivity of PC cells to gemcitabine was increased, while the proliferation and invasion of PC cells were suppressed after RNA interference-mediated silencing of TRPM8. The mechanism of TRPM8 in gemcitabine-based chemotherapy was then investigated. The expression and activity of multidrug resistance-associated proteins, P-gp, MRP-2, LRP, was significantly reduced in response to TRPM8 silence. Moreover, TRPM8 knockdown significantly increased hENT1 protein levels and the ratio of Bax/Bcl-2 while decreased the protein levels of RRM1. Thus, TRPM8 is required for PC cell proliferation and invasion and was closely related to the gemcitabine sensitivity of PC. The modulation of TRPM8 expression may help improve treatment response of PC by combining with traditional chemotherapy.
Collapse
Affiliation(s)
- JieFeng Liu
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - GuoHuang Hu
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - YuJing Gong
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - QianLe Yu
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - Bin He
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - WangHao Li
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - ZhiGuo He
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - WenJie Hao
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - ZiChao He
- Department of General Surgery, The Fourth Hospital of Changsha, Hunan Normal University, Changsha, 410006, People's Republic of China
| | - YiPing Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410078, People's Republic of China.
| |
Collapse
|
10
|
The Effect of GPRC5a on the Proliferation, Migration Ability, Chemotherapy Resistance, and Phosphorylation of GSK-3β in Pancreatic Cancer. Int J Mol Sci 2018; 19:ijms19071870. [PMID: 29949874 PMCID: PMC6073545 DOI: 10.3390/ijms19071870] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 06/17/2018] [Accepted: 06/25/2018] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer (PaCa) is the fourth leading cause of cancer-related death, and personalized targeted cancer therapy is becoming a promising treatment strategy for PaCa. The central approach of targeted therapy is to find a targetable key and an effective targeting method. In this study, the importance of GPRC5a (the G-protein-coupled receptor family C, member 5, group A) was identified using data mining methods based on published datasets. After analysis of the basic expression of GPRC5a in normal pancreas tissue and various PaCa cell lines, gene editing of GPRC5a in the human PaCa cell line MIA PaCa-2 and the mouse PaCa cell line TB32047 was performed using CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated proteins 9) to investigate the influence of GPRC5a on the proliferation and migration of PaCa cells as well as its effects on chemotherapy drug resistance. The results showed that GPRC5a was upregulated in PaCa tissues and various PaCa cell lines. Knockout of GPRC5a reduced the proliferation and migration ability of PaCa cell lines and suppressed the chemotherapy drug resistance of gemcitabine, oxaliplatin, and fluorouracil in PaCa cells. The phosphorylation of GSK-3β (Glycogen synthase kinase-3β) was found to be upregulated in the MIA PaCa-2 and TB32047 cells after GPRC5a knockout. In conclusion, GPRC5a was upregulated in PaCa leading to an enhanced drug resistance in PaCa cells. These results provide for the first time a theoretical basis for the development of an improved PaCa targeted therapy.
Collapse
|
11
|
Rosemurgy A. Special issue editorial-Cancer Genetics. Cancer Genet 2017; 209:535-536. [PMID: 27931696 DOI: 10.1016/j.cancergen.2016.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
12
|
Zhang X, Yu J, Li M. Precision regimen for personalized pancreatic cancer therapy. PRECISION RADIATION ONCOLOGY 2017. [DOI: 10.1002/pro6.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Xiaoli Zhang
- Department of Medicine; The University of Oklahoma Health Sciences Center; Oklahoma City Oklahoma USA
- Department of Surgery; The University of Oklahoma Health Sciences Center; Oklahoma City Oklahoma USA
- Department of Radiation Oncology; Shandong Cancer Hospital Affiliated to Shandong University; Jinan Shandong China
| | - Jinming Yu
- Department of Radiation Oncology; Shandong Cancer Hospital Affiliated to Shandong University; Jinan Shandong China
| | - Min Li
- Department of Medicine; The University of Oklahoma Health Sciences Center; Oklahoma City Oklahoma USA
- Department of Surgery; The University of Oklahoma Health Sciences Center; Oklahoma City Oklahoma USA
| |
Collapse
|
13
|
Prognostic Implications of Expression Profiling for Gemcitabine-Related Genes (hENT1, dCK, RRM1, RRM2) in Patients With Resectable Pancreatic Adenocarcinoma Receiving Adjuvant Chemotherapy. Pancreas 2017; 46:684-689. [PMID: 28196013 DOI: 10.1097/mpa.0000000000000807] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The aim of this study was to examine the relevance of expression profiling of 4 genes involved in the action of gemcitabine among patients with pancreatic ductal-cell adenocarcinoma (PDAC). METHODS A group of 100 patients who underwent pancreatic resections for PDAC and received adjuvant chemotherapy with gemcitabine between 2007 and 2010 was identified. Expression of mRNAs for human equilibrative nucleoside transporter 1 (hENT1), ribonucleotide reductase subunits (RRM1, RRM2), and deoxycytidine kinase (dCK) was examined by quantitative real-time polymerase chain reaction, normalized to glyceraldehyde-3-phosphate dehydrogenase (GAPDH), and dichotomized into groups of low and moderate/high expression levels grouped by tertiles. RESULTS Significantly better median survival times were found for high/moderate expression levels of hENT1 (27.9 vs 12.4 months, P = 0.001) and dCK (19.7 vs 10.5 months, P = 0.003), as well as low expression of RRM1 (23.4 vs 11.4 months, P = 0.027). A Cox proportional hazards model identified low expression of hENT1 (hazard ratio [HR], 3.38; 95% confidence intervals [CI], 2.28-10.50) and dCK (HR, 2.24; 95% CI, 1.63-3.39), and high/moderate levels of RRM1 (HR, 1.65; 95% CI, 1.23-2.45) as negative prognostic factors. CONCLUSIONS Expression of hENT, RRM1, and dCK genes provides important prognostic information for PDAC patients treated with adjuvant gemcitabine.
Collapse
|
14
|
Johnson MD, Stone B, Thibodeau BJ, Baschnagel AM, Galoforo S, Fortier LE, Ketelsen B, Ahmed S, Kelley Z, Hana A, Wilson TG, Robertson JM, Jury RP, Wilson GD. The significance of Trk receptors in pancreatic cancer. Tumour Biol 2017; 39:1010428317692256. [PMID: 28218045 DOI: 10.1177/1010428317692256] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
This study investigated the Trk receptor family as a therapeutic target in pancreatic ductal adenocarcinoma and assessed their prognostic significance. Global gene expression analysis was investigated in prospectively collected pancreatic ductal adenocarcinomas that had either undergone neoadjuvant chemoradiation or were treated by surgery. PANC-1 and MIA-PaCa-2 cell lines were investigated to establish whether fractionated radiation altered expression of four neuroendocrine genes and whether this resulted in subsequent changes in radiosensitivity. A specific inhibitor of TrkA, B, and C, AstraZeneca 1332, was investigated in vitro and in vivo in combination with radiation. A tissue microarray was constructed from 77 pancreatic ductal adenocarcinoma patients who had undergone neoadjuvant chemoradiation and the Trk receptor, and neurogenic differentiation 1 expression was assessed and correlated with overall survival. A total of 99 genes were identified that were differentially expressed in the chemoradiation patients with neuroendocrine genes and pathways, in particular the neurogenic differentiation 1 and Trk receptor family, being prominent. Fractionated radiation upregulated the expression of neuroendocrine genes, and AstraZeneca 1332 treatment in vitro enhanced radiosensitivity. No added effect of AstraZeneca 1332 was observed in vivo. Trk receptor expression varied between isoforms but did not correlate significantly with clinical outcome. Radiation treatment upregulated neuroendocrine gene expression but the Trk receptor family does not appear to be a promising treatment target.
Collapse
Affiliation(s)
- Matthew D Johnson
- 1 Department of Radiation Oncology, William Beaumont Hospital, Beaumont Health System, Royal Oak, MI, USA
| | - Brandon Stone
- 1 Department of Radiation Oncology, William Beaumont Hospital, Beaumont Health System, Royal Oak, MI, USA
| | - Bryan J Thibodeau
- 2 Beaumont BioBank, William Beaumont Hospital, Beaumont Health System, Royal Oak, MI, USA
| | - Andrew M Baschnagel
- 3 Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Sandra Galoforo
- 1 Department of Radiation Oncology, William Beaumont Hospital, Beaumont Health System, Royal Oak, MI, USA
| | - Laura E Fortier
- 2 Beaumont BioBank, William Beaumont Hospital, Beaumont Health System, Royal Oak, MI, USA
| | - Billie Ketelsen
- 2 Beaumont BioBank, William Beaumont Hospital, Beaumont Health System, Royal Oak, MI, USA
| | - Samreen Ahmed
- 2 Beaumont BioBank, William Beaumont Hospital, Beaumont Health System, Royal Oak, MI, USA
| | - Zakiya Kelley
- 2 Beaumont BioBank, William Beaumont Hospital, Beaumont Health System, Royal Oak, MI, USA
| | - Alaa Hana
- 1 Department of Radiation Oncology, William Beaumont Hospital, Beaumont Health System, Royal Oak, MI, USA
| | - Thomas G Wilson
- 1 Department of Radiation Oncology, William Beaumont Hospital, Beaumont Health System, Royal Oak, MI, USA
| | - John M Robertson
- 1 Department of Radiation Oncology, William Beaumont Hospital, Beaumont Health System, Royal Oak, MI, USA
| | - Robert P Jury
- 4 Department of General Surgery, William Beaumont Hospital, Beaumont Health System, Royal Oak, MI, USA
| | - George D Wilson
- 1 Department of Radiation Oncology, William Beaumont Hospital, Beaumont Health System, Royal Oak, MI, USA
| |
Collapse
|
15
|
Li Y, Wu C, Chen T, Zhang J, Liu G, Pu Y, Zhu J, Shen C, Zhang Y, Zeng N, Zhang X. Effects of RNAi-mediated MUC4 gene silencing on the proliferation and migration of human pancreatic carcinoma BxPC-3 cells. Oncol Rep 2016; 36:3449-3455. [PMID: 27748843 DOI: 10.3892/or.2016.5152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 05/27/2016] [Indexed: 11/05/2022] Open
Abstract
It was previously demonstrated that mucin 4 (MUC4) is not expressed in normal pancreatic tissues or in chronic pancreatitis tissue but is highly expressed in pancreatic cancer (PC) tissue. Effective MUC4 gene knockdown in PC may contribute to the elucidation of pancreatic tumor development and metastasis, and may be valuable in new therapeutic approaches. Thus to confirm this, in the present study, the BxPC-3 cell line was transfected with eight pairs of shRNA lentiviral vectors for MUC4. The qPCR results showed that expression of MUC4 mRNA in the BxPC-3 cells was significantly decreased at 96 h after transfection. One of these shRNA lentiviral vectors (shRNA‑A141) had showed the strongest suppressive effect on MUC4 mRNA expression and was used for MUC4 knockdown in BxPC-3 cells. After stable transfection, BxPC-3 cells showed a significantly lower expression of MUC4 mRNA and MUC4 protein, and were suppressed on cell growth and migration. In vivo, lower tumor growth rates and tumor volume were observed in the tumors derived from the MUC4-knockdown cells, whereas the transplanted tumors derived from the control group cells, grew rapidly. Thus, inhibition of MUC4 expression may be an effective means for mitigating metastasis and invasion of PC.
Collapse
Affiliation(s)
- Yong Li
- Sichuan Key Laboratory of Medical Imaging and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Changqiang Wu
- Sichuan Key Laboratory of Medical Imaging and School of Medical Imaging, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Tianwu Chen
- Sichuan Key Laboratory of Medical Imaging and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Juanjuan Zhang
- Sichuan Key Laboratory of Medical Imaging, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Yu Pu
- Sichuan Key Laboratory of Medical Imaging and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Jiang Zhu
- Sichuan Key Laboratory of Medical Imaging and Department of Chemistry, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Chengyi Shen
- Sichuan Key Laboratory of Medical Imaging and Department of Pathophysiology, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Yang Zhang
- Sichuan Key Laboratory of Medical Imaging and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Nanlin Zeng
- Sichuan Key Laboratory of Medical Imaging and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Xiaoming Zhang
- Sichuan Key Laboratory of Medical Imaging and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
16
|
Hu B, Zhang K, Li S, Li H, Yan Z, Huang L, Wu J, Han X, Jiang W, Mulatibieke T, Zheng L, Wan R, Wang X, Hu G. HIC1 attenuates invasion and metastasis by inhibiting the IL-6/STAT3 signalling pathway in human pancreatic cancer. Cancer Lett 2016; 376:387-98. [DOI: 10.1016/j.canlet.2016.04.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/07/2016] [Accepted: 04/11/2016] [Indexed: 12/18/2022]
|
17
|
Fotopoulos G, Syrigos K, Saif MW. Genetic factors affecting patient responses to pancreatic cancer treatment. Ann Gastroenterol 2016; 29:466-476. [PMID: 27708512 PMCID: PMC5049553 DOI: 10.20524/aog.2016.0056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/05/2016] [Indexed: 12/15/2022] Open
Abstract
Cancer of the exocrine pancreas is a malignancy with a high lethal rate. Surgical resection is the only possible curative mode of treatment. Metastatic pancreatic cancer is incurable with modest results from the current treatment options. New genomic information could prove treatment efficacy. An independent review of PubMed and ScienceDirect databases was performed up to March 2016, using combinations of terms such pancreatic exocrine cancer, chemotherapy, genomic profile, pancreatic cancer pharmacogenomics, genomics, molecular pancreatic pathogenesis, and targeted therapy. Recent genetic studies have identified new markers and therapeutic targets. Our current knowledge of pancreatic cancer genetics must be further advanced to elucidate the molecular basis and pathogenesis of the disease, improve the accuracy of diagnosis, and guide tailor-made therapies.
Collapse
Affiliation(s)
- George Fotopoulos
- Oncology Unit, Third Department of Medicine, University of Athens, Athens, Greece (George Fotopoulos, Konstantinos Syrigos)
| | - Konstantinos Syrigos
- Oncology Unit, Third Department of Medicine, University of Athens, Athens, Greece (George Fotopoulos, Konstantinos Syrigos); Yale School of Medicine, New Haven, CT, USA (Konstantinos Syrigos)
| | - Muhammad Wasif Saif
- Tufts University School of Medicine, Boston, Massachusetts, USA (Muhammad Wasif Saif)
| |
Collapse
|
18
|
Erb U, Zöller M. Progress and potential of exosome analysis for early pancreatic cancer detection. Expert Rev Mol Diagn 2016; 16:757-67. [PMID: 27206554 DOI: 10.1080/14737159.2016.1187563] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Pancreatic cancer (PaCa) is the most deadly malignancy, due to late diagnosis prohibiting surgery. Thus, strong efforts are taken improving early diagnosis via biomarkers recovered in the serum of PaCa patients. AREAS COVERED One promising option are PaCa-derived exosomes in patients' sera. Exosomes, small vesicles delivered by live cells and recovered in all body fluids, are a powerful diagnostic tool due to relative stability and composition covering the whole range of cancer-related biomarkers including proteins, metabolites, DNA, DNA modifications, coding and noncoding RNA. We discuss the mechanisms accounting for the condensed packaging of biomarkers, refer to studies using PaCa serum-exosomes for diagnosis. Based on an extensive literature search, we outline questions that answers may help establishing a serum-exosome-based screening for early PaCa detection. Expert commentary: Improved proteomic and genomic characterization and progress in the biogenesis of exosomes will allow for optimized and unified screening panels for PaCa diagnosis via TEX in body fluids.
Collapse
Affiliation(s)
- Ulrike Erb
- a Department of Tumor Cell Biology , University Hospital of Surgery , Heidelberg , Germany
| | - Margot Zöller
- a Department of Tumor Cell Biology , University Hospital of Surgery , Heidelberg , Germany
| |
Collapse
|
19
|
Mantripragada KC, Safran H. Optimizing initial chemotherapy for metastatic pancreatic cancer. Future Oncol 2016; 12:1125-33. [PMID: 26939741 DOI: 10.2217/fon-2015-0006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The two combination chemotherapy regimens FOLFIRINOX and gemcitabine plus nab-paclitaxel represent major breakthroughs in the management of metastatic pancreatic cancer. Both regimens showed unprecedented survival advantage in the setting of front-line therapy. However, their application for treatment of patients in the community is challenging because of significant toxicities, thus limiting potential benefits to a narrow population of patients. Modifications to the dose intensity or schedule of those regimens improve their tolerability, while likely retaining survival advantage over single-agent chemotherapy. Newer strategies to optimize these two active regimens in advanced pancreatic cancer are being explored that can help personalize treatment to individual patients.
Collapse
Affiliation(s)
- Kalyan C Mantripragada
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Howard Safran
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| |
Collapse
|