201
|
Wu JX, Hong YH, Yang XG. Bis(acetylacetonato)-oxidovanadium(IV) and sodium metavanadate inhibit cell proliferation via ROS-induced sustained MAPK/ERK activation but with elevated AKT activity in human pancreatic cancer AsPC-1 cells. J Biol Inorg Chem 2016; 21:919-929. [PMID: 27614430 DOI: 10.1007/s00775-016-1389-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/29/2016] [Indexed: 12/12/2022]
Abstract
In this study, the antiproliferative effect of bis(acetylacetonato)-oxidovanadium(IV) and sodium metavanadate and the underlying mechanisms were investigated in human pancreatic cancer cell line AsPC-1. The results showed that both exhibited an antiproliferative effect through inducing G2/M cell cycle arrest and can also cause elevation of reactive oxygen species (ROS) levels in cells. Moreover, the two vanadium compounds induced the activation of both PI3K/AKT and MAPK/ERK signaling pathways dose- and time-dependently, which could be counteracted with the antioxidant N-acetylcysteine. In the presence of MEK-1 inhibitor, the degradation of Cdc25C, inactivation of Cdc2 and accumulation of p21 were relieved. However, the treatment of AKT inhibitor did not cause any significant effect. Therefore, it demonstrated that the ROS-induced sustained MAPK/ERK activation rather than AKT contributed to vanadium compounds-induced G2/M cell cycle arrest. The current results also exhibited that the two vanadium compounds did not induce a sustained increase of ROS generation, but the level of ROS reached a plateau instead. The results revealed that an intracellular feedback loop may be against the elevated ROS level induced by vanadate or VO(acac)2, evidenced by the increased GSH content, the unchanged level at the expression of antioxidant enzymes. Therefore, vanadium compounds can be regarded as a novel type of anticancer drugs through the prolonged activation of MAPK/ERK pathway but retained AKT activity. The present results provided a proof-of-concept evidence that vanadium-based compounds may have the potential as both antidiabetic and antipancreatic cancer agents to prevent or treat patients suffering from both diseases.
Collapse
Affiliation(s)
- Jing-Xuan Wu
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, People's Republic of China
| | - Yi-Hua Hong
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, People's Republic of China
| | - Xiao-Gai Yang
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, People's Republic of China.
| |
Collapse
|
202
|
Abstract
Cancer is an evolutionary disease, containing the hallmarks of an asexually reproducing unicellular organism subject to evolutionary paradigms. Pancreatic ductal adenocarcinoma (hereafter referred to as pancreatic cancer) is a particularly robust example of this phenomenon. Genomic features indicate that pancreatic cancer cells are selected for fitness advantages when encountering the geographic and resource-depleted constraints of the microenvironment. Phenotypic adaptations to these pressures help disseminated cells to survive in secondary sites, a major clinical problem for patients with this disease. In this Review we gather the wide-ranging aspects of pancreatic cancer research into a single concept rooted in Darwinian evolution, with the goal of identifying novel insights and opportunities for study.
Collapse
Affiliation(s)
- Alvin Makohon-Moore
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Christine A Iacobuzio-Donahue
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
203
|
Lin PY, Peng SJ, Shen CN, Pasricha PJ, Tang SC. PanIN-associated pericyte, glial, and islet remodeling in mice revealed by 3D pancreatic duct lesion histology. Am J Physiol Gastrointest Liver Physiol 2016; 311:G412-22. [PMID: 27340125 PMCID: PMC6347070 DOI: 10.1152/ajpgi.00071.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/20/2016] [Indexed: 01/31/2023]
Abstract
Pericytes and glial cells are accessory cells of neurovascular networks, which have been reported to participate in scar formation after tissue injury. However, it remains unclear whether similar reactive cellular responses occur in pancreatic intraepithelial neoplasia (PanIN). In this study we developed three-dimensional (3D) duct lesion histology to investigate PanIN and the associated pericyte, glial, and islet remodeling. Transparent mouse pancreata with a Kras(G12D) mutation were used to develop 3D duct lesion histology. Deep-tissue, tile-scanning microscopy was performed to generate panoramic views of the diseased pancreas for global examination of early stage and advanced duct lesion formation. Fluorescence signals of ductal and neurovascular networks were simultaneously detected to reveal associated remodeling. Significantly, in Kras(G12D)-mutant mice, when the low-grade PanINs emerge, duct lesions appear as epithelial buds with perilesional pericyte and glial activation. When PanINs occur in large scale (induced by cerulein injections to the mutant mice), the 3D image data identifies 1) aggregation of PanINs in clusters in space; 2) overexpression of the pericyte marker NG2 in the PanIN microenvironment; and 3) epithelial in-growth to islets, forming the PanIN-islet complexes. Particularly, the PanIN-islet complexes associate with proliferating epithelial and stromal cells and receive substantial neurovascular supplies, making them landmarks in the atrophic lobe. Overall, perilesional pericyte and glial activation and formation of the PanIN-islet complex underline cellular heterogeneity in the duct lesion microenvironment. The results also illustrate the advantage of using 3D histology to reveal previously unknown details of neurovascular and endocrine links to the disease.
Collapse
Affiliation(s)
- Pei-Yu Lin
- 1Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan; ,2Genomics Research Center, Academia Sinica, Taipei, Taiwan;
| | - Shih-Jung Peng
- 3Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan; ,4Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan; and
| | - Chia-Ning Shen
- 1Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan; ,2Genomics Research Center, Academia Sinica, Taipei, Taiwan;
| | - Pankaj J. Pasricha
- 5Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Shiue-Cheng Tang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan; and
| |
Collapse
|
204
|
Abstract
Pancreatic cancer is now the third leading cause of cancer related deaths in the United States, yet advances in treatment options have been minimal over the past decade. In this review, we summarize the evaluation and treatments for this disease. We highlight molecular advances that hopefully will soon translate into improved outcomes.
Collapse
Affiliation(s)
- Cinthya S Yabar
- Department of Surgery, Thomas Jefferson University Hospital, Sidney Kimmel Medical College, 1015 Walnut Street, Curtis Building, Suite 620, Philadelphia, PA 19107, USA
| | - Jordan M Winter
- Department of Surgery, Thomas Jefferson University Hospital, Sidney Kimmel Medical College, 1015 Walnut Street, Curtis Building, Suite 620, Philadelphia, PA 19107, USA.
| |
Collapse
|
205
|
Li W, Ma Z, Ma J, Li X, Xu Q, Duan W, Chen X, Lv Y, Zhou S, Wu E, Ma Q, Huo X. Hydrogen peroxide mediates hyperglycemia-induced invasive activity via ERK and p38 MAPK in human pancreatic cancer. Oncotarget 2016; 6:31119-33. [PMID: 26439801 PMCID: PMC4741592 DOI: 10.18632/oncotarget.5045] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/26/2015] [Indexed: 12/11/2022] Open
Abstract
Diabetes mellitus and pancreatic cancer are intimately related, as approximately 85% of pancreatic cancer patients suffer from glucose intolerance or even diabetes. In this study, we evaluate the underlying mechanism by which hyperglycemia modulates the invasive potential of cancer cells and contributes to their enhanced metastatic behavior. Here we show that hyperglycemia increases the hydrogen peroxide (H2O2) concentration through up-regulation of manganese superoxide dismutase (SOD2) expression, which further activates the ERK and p38 MAPK pathways, as well as the transcription factors NF-κB and AP-1, in a time-dependent manner. The invasion of pancreatic cancer cells resulting from the activation of the H2O2/MAPK axis under high glucose conditions is effectively inhibited by PD 98059 (ERK inhibitor), SB 203580 (p38 MAPK inhibitor), polyethylene glycol-conjugated catalase (PEG-CAT), or the siRNA specific to SOD2. In addition, streptozotocin-treated diabetic nude mice exhibit a stronger tumor invasive ability in renal capsule xenografts which could be suppressed by PEG-CAT treatment. Furthermore, the integrated optical density (IOD) of SOD2 and uPA stainings is higher in the tumor tissues of pancreatic cancer patients with diabetes compared with pancreatic cancer patients with euglycemia. Taken together, our results demonstrate that hyperglycemia enhances cell invasive ability through the SOD2/H2O2/MAPK axis in human pancreatic cancer. Thus, SOD2/H2O2/MAPK axis may represent a promising therapeutic target for pancreatic cancer patients combined with diabetes mellitus.
Collapse
Affiliation(s)
- Wei Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhenhua Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jiguang Ma
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xuqi Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Qinhong Xu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xin Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yunfu Lv
- Department of General Surgery, People's Hospital of Hainan Province, Haikou, 570311, China
| | - Shuang Zhou
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, 58108, ND, USA
| | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, 58108, ND, USA
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiongwei Huo
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
206
|
Dugnani E, Gandolfi A, Balzano G, Scavini M, Pasquale V, Aleotti F, Liberati D, Di Terlizzi G, Petrella G, Reni M, Doglioni C, Bosi E, Falconi M, Piemonti L. Diabetes associated with pancreatic ductal adenocarcinoma is just diabetes: Results of a prospective observational study in surgical patients. Pancreatology 2016; 16:844-52. [PMID: 27546476 DOI: 10.1016/j.pan.2016.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/27/2016] [Accepted: 08/09/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Identification of a specific diabetes signature associated to pancreatic ductal carcinoma (PDAC) could be a key to detect asymptomatic, early stage tumors. We aim to characterize the clinical signature and the pathogenetic factors of the different types of diabetes associated with PDAC, based on the time between diabetes and cancer diagnosis. METHODS Prospective observational study on 364 PDAC patients admitted to a referral center for pancreatic disease. Hospital and/or outpatient medical records were reviewed. Blood biochemical values including fasting blood glucose, insulin and/or C-peptide, glycosylated hemoglobin and anti-islet antibodies were determined. Diabetes onset was assessed after surgery and during follow-up. RESULTS The prevalence of diabetes in patients was 67%. Considering 174 patients (47.8%) already having diabetes when diagnosed with PDAC (long duration, short duration, concomitant), the clinical and biochemical profile was similar to that of patients with type 2 diabetes (T2D). Diabetes was associated with known risk factors (i.e., age, sex, family history for diabetes and increased BMI) and both beta-cell dysfunction and insulin resistance were present. Considering 70 patients (19.2%) with onset of diabetes after PDAC diagnosis (early and late onset), the strongest predictor was the loss of beta-cell mass following pancreatectomy in patients with risk factors for T2D. CONCLUSION Different types of diabetes according to the time between diabetes and PDAC diagnosis are clinical entities widely overlapping with T2D. Therefore, the success of a strategy considering diabetes onset as a marker of asymptomatic PDAC will largely depend on our ability to identify new diabetes-unrelated biomarkers of PDAC.
Collapse
Affiliation(s)
- Erica Dugnani
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Alessandra Gandolfi
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Gianpaolo Balzano
- Pancreatic Surgery Unit, Pancreas Translational & Clinical Research Center, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Marina Scavini
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Valentina Pasquale
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Francesca Aleotti
- Pancreatic Surgery Unit, Pancreas Translational & Clinical Research Center, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Daniela Liberati
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Gaetano Di Terlizzi
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Giovanna Petrella
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Michele Reni
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Claudio Doglioni
- Department of Pathology, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy; Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Emanuele Bosi
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy; Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Massimo Falconi
- Pancreatic Surgery Unit, Pancreas Translational & Clinical Research Center, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy; Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
207
|
Gerdtsson AS, Wingren C, Persson H, Delfani P, Nordström M, Ren H, Wen X, Ringdahl U, Borrebaeck CAK, Hao J. Plasma protein profiling in a stage defined pancreatic cancer cohort - Implications for early diagnosis. Mol Oncol 2016; 10:1305-16. [PMID: 27522951 DOI: 10.1016/j.molonc.2016.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/01/2016] [Accepted: 07/04/2016] [Indexed: 12/30/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a disease where detection preceding clinical symptoms significantly increases the life expectancy of patients. In this study, a recombinant antibody microarray platform was used to analyze 213 Chinese plasma samples from PDAC patients and normal control (NC) individuals. The cohort was stratified according to disease stage, i.e. resectable disease (stage I/II), locally advanced (stage III) and metastatic disease (stage IV). Support vector machine analysis showed that all PDAC stages could be discriminated from controls and that the accuracy increased with disease progression, from stage I to IV. Patients with stage I/II PDAC could be discriminated from NC with high accuracy based on a plasma protein signature, indicating a possibility for early diagnosis and increased detection rate of surgically resectable tumors.
Collapse
Affiliation(s)
- Anna Sandström Gerdtsson
- Department of Immunotechnology, CREATE Health Translational Cancer Center, Medicon Village bldg. 406, Lund University, SE 223 81 Lund, Sweden.
| | - Christer Wingren
- Department of Immunotechnology, CREATE Health Translational Cancer Center, Medicon Village bldg. 406, Lund University, SE 223 81 Lund, Sweden.
| | - Helena Persson
- Department of Immunotechnology, CREATE Health Translational Cancer Center, Medicon Village bldg. 406, Lund University, SE 223 81 Lund, Sweden.
| | - Payam Delfani
- Department of Immunotechnology, CREATE Health Translational Cancer Center, Medicon Village bldg. 406, Lund University, SE 223 81 Lund, Sweden.
| | | | - He Ren
- Tianjin Medical University Cancer Institute & Hospital, Huan-Hu-Xi Road, Ti-Huan-Bei, He Xi District, Tianjin 300060, PR China.
| | - Xin Wen
- Tianjin Medical University Cancer Institute & Hospital, Huan-Hu-Xi Road, Ti-Huan-Bei, He Xi District, Tianjin 300060, PR China.
| | - Ulrika Ringdahl
- Department of Immunotechnology, CREATE Health Translational Cancer Center, Medicon Village bldg. 406, Lund University, SE 223 81 Lund, Sweden.
| | - Carl A K Borrebaeck
- Department of Immunotechnology, CREATE Health Translational Cancer Center, Medicon Village bldg. 406, Lund University, SE 223 81 Lund, Sweden.
| | - Jihui Hao
- Tianjin Medical University Cancer Institute & Hospital, Huan-Hu-Xi Road, Ti-Huan-Bei, He Xi District, Tianjin 300060, PR China.
| |
Collapse
|
208
|
Wahutu M, Vesely SK, Campbell J, Pate A, Salvatore AL, Janitz AE. Pancreatic Cancer: A Survival Analysis Study in Oklahoma. THE JOURNAL OF THE OKLAHOMA STATE MEDICAL ASSOCIATION 2016; 109:391-398. [PMID: 27885308 PMCID: PMC5119763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
BACKGROUND Pancreatic cancer is among the most deadly cancers. Risk factors associated with the disease include age, race, sex, smoking status, and diabetes status. METHOD We conducted a prospective analysis of risk factors and length of survival among pancreatic cancer patients living in Oklahoma between 1997 and 2012 (n=6,291). Kaplan-Meier survival curves were created followed by the log-rank test to compare difference in the survival time. Cox proportional hazard regression models were used to examine the strength of association through the estimated hazard ratios. RESULTS The median survival time of the cohort was three months. Significant risk factors for reduced survival times included age, stage at diagnosis, and year of diagnosis. CONCLUSION Results are in agreement with previous research findings. There have been small but noteworthy improvements in survival times for pancreatic cancer patients in Oklahoma. Length of survival during the study period was significantly associated with known risk factors such as age and stage of diagnosis.
Collapse
Affiliation(s)
| | - Sara K Vesely
- College of Public Health, University of Oklahoma Health Sciences Center
| | - Janis Campbell
- College of Public Health, University of Oklahoma Health Sciences Center
| | - Anne Pate
- School of Nursing and Allied Health Sciences, Southwestern Oklahoma State University
| | | | - Amanda E Janitz
- College of Public Health, University of Oklahoma Health Sciences Center
| |
Collapse
|
209
|
Inaishi J, Saisho Y, Sato S, Kou K, Murakami R, Watanabe Y, Kitago M, Kitagawa Y, Yamada T, Itoh H. Effects of Obesity and Diabetes on α- and β-Cell Mass in Surgically Resected Human Pancreas. J Clin Endocrinol Metab 2016; 101:2874-82. [PMID: 27070277 PMCID: PMC4929842 DOI: 10.1210/jc.2016-1374] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 04/07/2016] [Indexed: 12/16/2022]
Abstract
CONTEXT The ethnic difference in β-cell regenerative capacity in response to obesity may be attributable to different phenotypes of type 2 diabetes among ethnicities. OBJECTIVE This study aimed to clarify the effects of diabetes and obesity on β- (BCM) and α-cell mass (ACM) in the Japanese population. DESIGN, SETTING, AND PARTICIPANTS We obtained the pancreases of 99 individuals who underwent pancreatic surgery and whose resected pancreas sample contained adequate normal pancreas for histological analysis. Questionnaires on a family history of diabetes and history of obesity were conducted in 59 patients. Pancreatic sections were stained for insulin or glucagon, and fractional β- and α-cell area were measured. Islet size and density as well as β-cell turnover were also quantified. RESULTS In patients with diabetes, BCM was decreased by 46% compared with age- and body mass index-matched nondiabetic patients (1.48% ± 1.08% vs 0.80% ± 0.54%, P < .001), whereas there was no difference in ACM between the groups. There was no effect of obesity or history of obesity on BCM and ACM irrespective of the presence or absence of diabetes. There was a negative correlation between BCM, but not ACM, and glycated hemoglobin before and after pancreatic surgery. In addition, reduced BCM was observed in patients with pancreatic cancer compared with those with other pancreatic tumors. CONCLUSIONS These findings suggest that the increase in BCM in the face of insulin resistance is extremely limited in the Japanese, and BCM rather than ACM has a major role in regulating blood glucose level in humans.
Collapse
Affiliation(s)
- Jun Inaishi
- Departments of Internal Medicine (J.I., Y.S., S.S., K.K., R.M., Y.W., H.I.), Surgery (M.K., Y.K.), and Pathology (T.Y.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Pathology (T.Y.), Saitama Medical University, Saitama 350-0495, Japan
| | - Yoshifumi Saisho
- Departments of Internal Medicine (J.I., Y.S., S.S., K.K., R.M., Y.W., H.I.), Surgery (M.K., Y.K.), and Pathology (T.Y.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Pathology (T.Y.), Saitama Medical University, Saitama 350-0495, Japan
| | - Seiji Sato
- Departments of Internal Medicine (J.I., Y.S., S.S., K.K., R.M., Y.W., H.I.), Surgery (M.K., Y.K.), and Pathology (T.Y.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Pathology (T.Y.), Saitama Medical University, Saitama 350-0495, Japan
| | - Kinsei Kou
- Departments of Internal Medicine (J.I., Y.S., S.S., K.K., R.M., Y.W., H.I.), Surgery (M.K., Y.K.), and Pathology (T.Y.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Pathology (T.Y.), Saitama Medical University, Saitama 350-0495, Japan
| | - Rie Murakami
- Departments of Internal Medicine (J.I., Y.S., S.S., K.K., R.M., Y.W., H.I.), Surgery (M.K., Y.K.), and Pathology (T.Y.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Pathology (T.Y.), Saitama Medical University, Saitama 350-0495, Japan
| | - Yuusuke Watanabe
- Departments of Internal Medicine (J.I., Y.S., S.S., K.K., R.M., Y.W., H.I.), Surgery (M.K., Y.K.), and Pathology (T.Y.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Pathology (T.Y.), Saitama Medical University, Saitama 350-0495, Japan
| | - Minoru Kitago
- Departments of Internal Medicine (J.I., Y.S., S.S., K.K., R.M., Y.W., H.I.), Surgery (M.K., Y.K.), and Pathology (T.Y.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Pathology (T.Y.), Saitama Medical University, Saitama 350-0495, Japan
| | - Yuko Kitagawa
- Departments of Internal Medicine (J.I., Y.S., S.S., K.K., R.M., Y.W., H.I.), Surgery (M.K., Y.K.), and Pathology (T.Y.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Pathology (T.Y.), Saitama Medical University, Saitama 350-0495, Japan
| | - Taketo Yamada
- Departments of Internal Medicine (J.I., Y.S., S.S., K.K., R.M., Y.W., H.I.), Surgery (M.K., Y.K.), and Pathology (T.Y.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Pathology (T.Y.), Saitama Medical University, Saitama 350-0495, Japan
| | - Hiroshi Itoh
- Departments of Internal Medicine (J.I., Y.S., S.S., K.K., R.M., Y.W., H.I.), Surgery (M.K., Y.K.), and Pathology (T.Y.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Pathology (T.Y.), Saitama Medical University, Saitama 350-0495, Japan
| |
Collapse
|
210
|
Cao L, Chen X, Xiao X, Ma Q, Li W. Resveratrol inhibits hyperglycemia-driven ROS-induced invasion and migration of pancreatic cancer cells via suppression of the ERK and p38 MAPK signaling pathways. Int J Oncol 2016; 49:735-43. [PMID: 27278736 DOI: 10.3892/ijo.2016.3559] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 05/26/2016] [Indexed: 11/06/2022] Open
Abstract
Increasing evidence suggests that there is a strong relationship between diabetes mellitus (DM) and pancreatic cancer. Our previous study revealed that hyperglycemia could enhance the invasive and migratory activities of pancreatic cancer cells. Resveratrol, a natural polyphenolic phytoalexin, has many biological and pharmaceutical properties, including antioxidant and anti-tumorigenic capabilities. The aim of the present study was to evaluate whether resveratrol affects hyperglycemia-induced reactive oxygen species (ROS) production as well as the invasion and migration of pancreatic cancer and its underlying mechanisms. Human pancreatic cancer Panc-1 cells were exposed to high glucose condition with or without resveratrol, N-acetylcysteine (NAC, a scavenger of free radicals), PD 98059 (an ERK inhibitor) or SB 203580 (a p38 MAPK inhibitor). The intracellular ROS and hydrogen peroxide (H2O2) were determined using 2,7-dichlorodihydrofluorecein diacetate and H2O2 assay. MTT, wound healing assay and transwell matrigel invasion assay were used to detect the proliferation, migration and invasion potential of cancer cells. The expressions of uPA, E-cadherin and Glut-1 were examined using QT-PCR and western blot analysis at mRNA and protein levels. The activation of p-ERK, p-p38 and p-NF-κB were measured by western blot analysis. The results of the present study showed that resveratrol could significantly decrease high glucose-induced production of ROS and H2O2 in Panc-1 cells. Resveratrol was also able to inhibit high glucose-induced proliferation, migration and invasion of pancreatic cancer cells. High glucose-modulated expression of uPA, E-cadherin and Glut-1 were inhibited by resveratrol. In addition, high glucose-induced activation of ERK and p38 MAPK signaling pathways as well as the transcription factor NF-κB could also be suppressed by resveratrol. Furthermore, resveratrol was able to suppress H2O2-induced migration and invasion abilities of pancreatic cancer cells. Taken together, these data indicate that resveratrol plays an important role in suppressing hyperglycemia-driven ROS-induced pancreatic cancer progression by inhibiting the ERK and p38 MAPK signaling pathways, providing evidence that resveratrol might be a potential candidate for chemoprevention of pancreatic cancer.
Collapse
Affiliation(s)
- Lei Cao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Xin Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xue Xiao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wei Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
211
|
Sakai A, Suzuki M, Kobayashi T, Nishiumi S, Yamanaka K, Hirata Y, Nakagawa T, Azuma T, Yoshida M. Pancreatic cancer screening using a multiplatform human serum metabolomics system. Biomark Med 2016; 10:577-86. [DOI: 10.2217/bmm-2016-0020] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: To examine a novel screening method for pancreatic cancer involving gas chromatography/mass spectrometry and liquid chromatography/mass spectrometry-based metabolomics analysis. Materials & methods: Sera from pancreatic cancer patients (n = 59) and healthy volunteers (n = 59) were allocated to the training set or validation set. Serum metabolome analysis was carried out using our multiplatform metabolomics system. A diagnostic model was constructed using a two-phase screening method that was newly advocated. Results: When the training set was used, the constructed diagnostic model exhibited high sensitivity (100%) and specificity (80%) for pancreatic cancer. When the validation set was used, the model displayed high sensitivity (84.1%) and specificity (84.1%). Conclusion: We successfully developed a diagnostic model for pancreatic cancer using a multiplatform serum metabolomics system.
Collapse
Affiliation(s)
- Arata Sakai
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Makoto Suzuki
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Takashi Kobayashi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Shin Nishiumi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Kodai Yamanaka
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Yuichi Hirata
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Takashi Nakagawa
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Takeshi Azuma
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Masaru Yoshida
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
- Division of Metabolomics Research, Department of Internal Related, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
- AMED-CREST, AMED, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| |
Collapse
|
212
|
Lu Y, Rodríguez LAG, Malgerud L, González-Pérez A, Martín-Pérez M, Lagergren J, Bexelius TS. Reply to 'Comment on 'New-onset type 2 diabetes, elevated HbA1c, anti-diabetic medications, and risk of pancreatic cancer''. Br J Cancer 2016; 114:e12. [PMID: 27219289 PMCID: PMC4891496 DOI: 10.1038/bjc.2016.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Yunxia Lu
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 76, Sweden.,Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
| | | | - Linnéa Malgerud
- Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm 171 77, Sweden
| | | | - Mar Martín-Pérez
- Centro Español de Investigación Farmacoepidemiológica, Madrid 28004, Spain
| | - Jesper Lagergren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 76, Sweden.,King's College London, Division of Cancer Studies, London WC2R 2LS, UK
| | - Tomas S Bexelius
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 76, Sweden.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm 171 77, Sweden
| |
Collapse
|
213
|
Comment on 'New-onset type 2 diabetes, elevated HbA1c, anti-diabetic medications, and risk of pancreatic cancer'. Br J Cancer 2016; 114:e11. [PMID: 27219288 PMCID: PMC4891518 DOI: 10.1038/bjc.2016.99] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
214
|
Dai X, Pang W, Zhou Y, Yao W, Xia L, Wang C, Chen X, Zen K, Zhang CY, Yuan Y. Altered profile of serum microRNAs in pancreatic cancer-associated new-onset diabetes mellitus. J Diabetes 2016; 8:422-33. [PMID: 25991015 DOI: 10.1111/1753-0407.12313] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 05/06/2015] [Accepted: 05/08/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND New-onset diabetes mellitus in pancreatic cancer has been recognized as a paraneoplastic phenomenon caused by the existence of the tumor. Circulating microRNAs (miRNAs) are emerging as non-invasive biomarkers for the detection of various cancers. In the present study, we hypothesized that a specific serum miRNA profile exists in pancreatic cancer-associated new-onset diabetes mellitus (PaC-DM). METHODS Initial screening of differentially expressed miRNAs in pooled serum samples from 25 PaC-DM patients, 25 non-cancer new-onset type 2 diabetes mellitus (T2DM) patients, and 25 healthy controls was performed by TaqMan low-density arrays (TLDA). A stem-loop quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was conducted to confirm the relative concentrations of candidate miRNAs in 80 PaC-DM, 85 non-cancer new-onset T2DM patients, and 80 healthy controls. RESULTS The TLDA identified 16 serum miRNAs that were significantly increased in PaC-DM samples. A combination of six serum miRNAs (miR-483-5p, miR-19a, miR-29a, miR-20a, miR-24, miR-25) was selected by qRT-PCR as a biomarker for PaC-DM. The area under the receiver operating characteristic curve (AUC) for the six-miRNA panel training and validation sets was 0.959 (95% confidence interval [CI] 0.890-1.028) and 0.902 (95% CI 0.844-0.955), respectively. The combination of these six miRNAs enabled the discrimination of PaC-DM from non-cancer new-onset T2DM with an AUC of 0.885 (95% CI 0.784-0.986) and 0.887 (95% CI 0.823-0.952) for the training and validation sets, respectively. CONCLUSION The six-serum miRNA panel may have potential as a biomarker for the accurate diagnosis and discrimination of PaC-DM from healthy controls and non-cancer new-onset T2DM.
Collapse
Affiliation(s)
- Xin Dai
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenjing Pang
- Department of Gastroenterology, Tianyou Hospital, Shanghai Tongji University School of Medicine, Shanghai, China
| | - Yufeng Zhou
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weiyan Yao
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lu Xia
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Cheng Wang
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xi Chen
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ke Zen
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Chen-Yu Zhang
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yaozong Yuan
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
215
|
Antwi SO, Oberg AL, Shivappa N, Bamlet WR, Chaffee KG, Steck SE, Hébert JR, Petersen GM. Pancreatic cancer: associations of inflammatory potential of diet, cigarette smoking and long-standing diabetes. Carcinogenesis 2016; 37:481-90. [PMID: 26905587 PMCID: PMC4843052 DOI: 10.1093/carcin/bgw022] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 02/04/2016] [Accepted: 02/11/2016] [Indexed: 01/02/2023] Open
Abstract
Epidemiologic studies show strong associations between pancreatic cancer (PC) and inflammatory stimuli or conditions such as cigarette smoking and diabetes, suggesting that inflammation may play a key role in PC. Studies of dietary patterns and cancer outcomes also suggest that diet might influence an individual's risk of PC by modulating inflammation. We therefore examined independent and joint associations between inflammatory potential of diet, cigarette smoking and long-standing (≥5 years) type II diabetes in relation to risk of PC. Analyses included data from 817 cases and 1756 controls. Inflammatory potential of diet was measured using the dietary inflammatory index (DII), calculated from dietary intake assessed via a 144-item food frequency questionnaire, and adjusted for energy intake. Information on smoking and diabetes were obtained via risk factor questionnaires. Associations were examined using multivariable-adjusted logistic regression. Higher DII scores, reflecting a more proinflammatory diet, were associated with increased risk of PC [odds ratio (OR)Quintile 5 versus 1 = 2.54, 95% confidence interval (CI) = 1.87-3.46, P trend < 0.0001]. Excess risk of PC also was observed among former (OR = 1.29, 95% CI = 1.07-1.54) and current (OR = 3.40, 95% CI = 2.28-5.07) smokers compared with never smokers, and among participants with long-standing diabetes (OR = 3.09, 95% CI = 2.02-4.72) compared with nondiabetics. Joint associations were observed for the combined effects of having greater than median DII score, and being a current smoker (OR = 4.79, 95% CI = 3.00-7.65) or having long-standing diabetes (OR = 6.03, 95% CI = 3.41-10.85). These findings suggest that a proinflammatory diet may act as cofactor with cigarette smoking and diabetes to increase risk of PC beyond the risk of any of these factors alone.
Collapse
Affiliation(s)
- Samuel O. Antwi
- Division of Epidemiology and
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Charlton 6-243, Rochester, MN 55905, USA and
- Cancer Prevention and Control Program and
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Ann L. Oberg
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Charlton 6-243, Rochester, MN 55905, USA and
| | - Nitin Shivappa
- Cancer Prevention and Control Program and
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - William R. Bamlet
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Charlton 6-243, Rochester, MN 55905, USA and
| | - Kari G. Chaffee
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Charlton 6-243, Rochester, MN 55905, USA and
| | - Susan E. Steck
- Cancer Prevention and Control Program and
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - James R. Hébert
- Cancer Prevention and Control Program and
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Gloria M. Petersen
- *To whom correspondence should be addressed. Tel: +1 5075 381563; Fax: +1 5072 662478;
| |
Collapse
|
216
|
Li YG, Han BB, Li F, Yu JW, Dong ZF, Niu GM, Qing YW, Li JB, Wei M, Zhu W. High Glucose Induces Down-Regulated GRIM-19 Expression to Activate STAT3 Signaling and Promote Cell Proliferation in Cell Culture. PLoS One 2016; 11:e0153659. [PMID: 27101310 PMCID: PMC4839686 DOI: 10.1371/journal.pone.0153659] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 04/01/2016] [Indexed: 11/27/2022] Open
Abstract
Recent studies indicated that Gene Associated with Retinoid-IFN-Induced Mortality 19 (GRIM-19), a newly discovered mitochondria-related protein, can regulate mitochondrial function and modulate cell viability possibly via interacting with STAT3 signal. In the present study we sought to test: 1) whether GRIM-19 is involved in high glucose (HG) induced altered cell metabolism in both cancer and cardiac cells, 2) whether GRIM-19/STAT3 signaling pathway plays a role in HG induced biological effects, especially whether AMPK activity could be involved. Our data showed that HG enhanced cell proliferation of both HeLa and H9C2 cells, which was closely associated with down-regulated GRIM-19 expression and increased phosphorylated STAT3 level. We showed that GRIM-19 knock-down alone in normal glucose cultured cells can also result in an increase in phosphorylated STAT3 level and enhanced proliferation capability, whereas GRIM-19 over-expression can abolished HG induced STAT3 activation and enhanced cell proliferation. Importantly, both down-regulated or over-expression of GRIM-19 increased lactate production in both HeLa and H9C2 cells. The activated STAT3 was responsible for increased cell proliferation as either AG-490, an inhibitor of JAK2, or siRNA targeting STAT3 can attenuate cell proliferation increased by HG. In addition, HG increased lactate acid levels in HeLa cells, which was also observed when GRIM-19 was genetically manipulated. However, HG did not affect the lactate levels in H9C2 cells. Of note, over-expression of GRIM-19 and silencing of STAT3 both increased lactate production in H9C2 cells. As expected, HG resulted in significant decreases in phosphorylated AMPKα levels in H9C2 cells, but not in HeLa cells. Interestingy, activation of AMPKα by metformin was associated with a reversal of the suppressed GRIM-19 expression in H9C2 cells, the fold of changes in GRIM-19 expression by metformin were much less in HeLa cells. Metformin did not affect the phosphorylated STAT3 lelvels, however, decreased its levels in H9C2, especially in the setting of HG culture. Not like HG alone which resulted in no changes in lactate acid in H9C2 cells, metformin can increase lactate acid levels in H9C2 cells. Increased lactate induced by metformin was also observed in HeLa cells.
Collapse
Affiliation(s)
- Yong-Guang Li
- State Key Discipline Division, Heart Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Bei-Bei Han
- State Key Discipline Division, Heart Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Feng Li
- State Key Discipline Division, Heart Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Jian-Wu Yu
- Division of Cardiology, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang, People’s Republic of China
| | - Zhi-Feng Dong
- State Key Discipline Division, Heart Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Geng-Ming Niu
- State Key Discipline Division, Heart Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Yan-Wei Qing
- State Key Discipline Division, Heart Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Jing-Bo Li
- State Key Discipline Division, Heart Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Meng Wei
- State Key Discipline Division, Heart Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
- * E-mail: (WZ); (MW)
| | - Wei Zhu
- State Key Discipline Division, Heart Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
- Provincial Key Laboratory of Cardiovascular Research, Department of Cardiology, Zhejiang University School of Medicine 2nd Affiliated Hospital, Hangzhou, Zhejiang, People’s Republic of China
- * E-mail: (WZ); (MW)
| |
Collapse
|
217
|
Weledji EP, Enoworock G, Mokake M, Sinju M. How Grim is Pancreatic Cancer? Oncol Rev 2016; 10:294. [PMID: 27471581 PMCID: PMC4943093 DOI: 10.4081/oncol.2016.294] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/12/2016] [Accepted: 06/29/2016] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal carcinoma continues to be the most lethal malignancy with rising incidence. It is the fourth most common cause of cancer death in the western world due to its low treatment success rate. In addition, because of its rapid growth and silent course, diagnosis is often only established in the advanced stages. As one of the most aggressive malignancies, the treatment of this disease is a great challenge to clinicians. This paper reviewed the natural history of pancreatic cancer, the current clinical practice and the future in pancreatic cancer management.
Collapse
Affiliation(s)
| | | | - Martin Mokake
- Department of Surgery and Obstetrics and Gynaecology, University of Buea, Cameroon
| | - Motaze Sinju
- Department of Surgery and Obstetrics and Gynaecology, University of Buea, Cameroon
| |
Collapse
|
218
|
Laeseke PF, Chen R, Jeffrey RB, Brentnall TA, Willmann JK. Combining in Vitro Diagnostics with in Vivo Imaging for Earlier Detection of Pancreatic Ductal Adenocarcinoma: Challenges and Solutions. Radiology 2016; 277:644-61. [PMID: 26599925 DOI: 10.1148/radiol.2015141020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth-leading cause of cancer-related death in the United States and is associated with a dismal prognosis, particularly when diagnosed at an advanced stage. Overall survival is significantly improved if PDAC is detected at an early stage prior to the onset of symptoms. At present, there is no suitable screening strategy for the general population. Available diagnostic serum markers are not sensitive or specific enough, and clinically available imaging modalities are inadequate for visualizing early-stage lesions. In this article, the role of currently available blood biomarkers and imaging tests for the early detection of PDAC will be reviewed. Also, the emerging biomarkers and molecularly targeted imaging agents being developed to improve the specificity of current imaging modalities for PDAC will be discussed. A strategy incorporating blood biomarkers and molecularly targeted imaging agents could lead to improved screening and earlier detection of PDAC in the future. (©) RSNA, 2015.
Collapse
Affiliation(s)
- Paul F Laeseke
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621 (P.F.L., R.B.J., J.K.W.); and Department of Medicine, University of Washington, Seattle, Wash (R.C., T.A.B.)
| | - Ru Chen
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621 (P.F.L., R.B.J., J.K.W.); and Department of Medicine, University of Washington, Seattle, Wash (R.C., T.A.B.)
| | - R Brooke Jeffrey
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621 (P.F.L., R.B.J., J.K.W.); and Department of Medicine, University of Washington, Seattle, Wash (R.C., T.A.B.)
| | - Teresa A Brentnall
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621 (P.F.L., R.B.J., J.K.W.); and Department of Medicine, University of Washington, Seattle, Wash (R.C., T.A.B.)
| | - Jürgen K Willmann
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621 (P.F.L., R.B.J., J.K.W.); and Department of Medicine, University of Washington, Seattle, Wash (R.C., T.A.B.)
| |
Collapse
|
219
|
|
220
|
Ambe CM, Mahipal A, Fulp J, Chen L, Malafa MP. Effect of Metformin Use on Survival in Resectable Pancreatic Cancer: A Single-Institution Experience and Review of the Literature. PLoS One 2016; 11:e0151632. [PMID: 26967162 PMCID: PMC4788143 DOI: 10.1371/journal.pone.0151632] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 03/02/2016] [Indexed: 12/16/2022] Open
Abstract
Observational studies have demonstrated that metformin use in diabetic patients is associated with reduced cancer incidence and mortality. Here, we aimed to determine whether metformin use was associated with improved survival in patients with resected pancreatic cancer. All patients with diabetes who underwent resection for pancreatic adenocarcinoma between 12/1/1986 and 4/30/2013 at our institution were categorized by metformin use. Survival analysis was done using the Kaplan-Meier method, with log-rank test and Cox proportional hazards multivariable regression models. For analyses of our data and the only other published study, we used Meta-Analysis version 2.2. We identified 44 pancreatic cancer patients with diabetes who underwent resection of the primary tumor (19 with ongoing metformin use, 25 never used metformin). There were no significant differences in major clinical and demographic characteristics between metformin and non-metformin users. Metformin users had a better median survival than nonusers, but the difference was not statistically significant (35.3 versus 20.2 months; P = 0.3875). The estimated 2-, 3-, and 5-year survival rates for non-metformin users were 42%, 28%, and 14%, respectively. Metformin users fared better with corresponding rates of 68%, 34%, and 34%, respectively. In our literature review, which included 111 patients from the two studies (46 metformin users and 65 non-users), overall hazard ratio was 0.668 (95% CI 0.397–1.125), with P = 0.129. Metformin use was associated with improved survival outcomes in patients with resected pancreatic cancer, but the difference was not statistically significant. The potential benefit of metformin should be investigated in adequately powered prospective studies.
Collapse
Affiliation(s)
- Chenwi M. Ambe
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Amit Mahipal
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Jimmy Fulp
- Department of Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Lu Chen
- Department of Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Mokenge P. Malafa
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
- * E-mail:
| |
Collapse
|
221
|
Clinical importance of Familial Pancreatic Cancer Registry in Japan: a report from kick-off meeting at International Symposium on Pancreas Cancer 2012. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2016; 20:557-66. [PMID: 23604538 DOI: 10.1007/s00534-013-0611-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pancreatic cancer is still a highly lethal disease with a 5-year survival rate of approximately 5 %. Early detection offers one of the best hopes for improving survival. Previous cohort studies and case-control studies showed that 4-10 % of pancreatic cancers have a hereditary basis, and individuals with a family history have an increased risk of developing pancreatic and extra-pancreatic malignancies. Since individuals with a family history of pancreatic cancer and those with a known genetic syndrome that predisposes to pancreatic cancer will be the first to benefit from early detection tests as they become available, familial pancreatic cancer (FPC) registries have been established in the US and Europe, but not yet in Japan. Such registries form the basis for epidemiological studies, clinical trials, and basic research on familial pancreatic cancer. There is a need for FPC registries in Japan as cancer risk varies among different populations and discoveries made in Western populations may not translate to the Japanese population. These registries in Japan will align with ongoing international efforts and add to a better understanding of the natural history, risk factors, screening strategies, and responsible genes, for improving survival of this dismal disease.
Collapse
|
222
|
Blood expression of matrix metalloproteinases 8 and 9 and of their inducers S100A8 and S100A9 supports diagnosis and prognosis of PDAC-associated diabetes mellitus. Clin Chim Acta 2016; 456:24-30. [PMID: 26923392 DOI: 10.1016/j.cca.2016.02.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/24/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND Based on the knowledge that matrix metalloproteinases (MMPs) and S100A8/A9 synergistically work in causing PDAC-associated type 2 diabetes mellitus (T2DM), we verified whether tissue and blood MMP8, MMP9, S100A8 and S100A9 expression might help in distinguishing PDAC among diabetics. METHODS Relative quantification of MMP8, MMP9, S100A8 and S100A9 mRNA was performed in tissues obtained from 8 PDAC, 4 chronic pancreatitis (ChrPa), 4 non-PDAC tumors and in PBMCs obtained from 30 controls, 43 T2DM, 41 ChrPa, 91 PDAC and 33 pancreatic-biliary tract tumors. RESULTS T2DM was observed in PDAC (66%), in pancreatic-biliary tract tumors (64%) and in ChrPa (70%). In diabetics, with or without PDAC, MMP9 tissue expression was increased (p<0.05). Both MMPs increased in PDAC and MMP9 increased also in pancreatic-biliary tract tumors PBMCs. In diabetics, MMP9 was independently associated with PDAC (p=0.025), but failed to enhance CA 19-9 discriminant efficacy. A highly reduced S100A9 expression, found in 7 PDAC, was significantly correlated with a reduced overall survival (p=0.015). CONCLUSIONS An increased expression of tissue and blood MMP9 reflects the presence of PDAC-associated diabetes mellitus. This finding fits with the hypothesized role of MMPs as part of the complex network linking cancer to diabetes.
Collapse
|
223
|
De Souza A, Irfan K, Masud F, Saif MW. Diabetes Type 2 and Pancreatic Cancer: A History Unfolding. JOP : JOURNAL OF THE PANCREAS 2016; 17:144-148. [PMID: 29568247 PMCID: PMC5860818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Pancreatic Cancer is the fourth cause of cancer-related deaths in the United States. Up to 80% of pancreatic cancer patients present with either new-onset type 2 diabetes or impaired glucose tolerance at the time of diagnosis. Recent literature suggests that diabetes mellitus type 2 is a risk factor, a manifestation and a prognostic factor for pancreatic cancer. This article is intended to clarify the evidence about diabetes as a risk factor for pancreatic cancer.
Collapse
Affiliation(s)
- Andre De Souza
- Department of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
| | - Khawaja Irfan
- Department of Endocrinology and Metabolism, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Faisal Masud
- King Edward Medical University, Lahore, Pakistan
| | - Muhammad Wasif Saif
- Department of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
224
|
Azoulay L, Filion KB, Platt RW, Dahl M, Dormuth CR, Clemens KK, Durand M, Juurlink DN, Targownik LE, Turin TC, Paterson JM, Ernst P. Incretin based drugs and the risk of pancreatic cancer: international multicentre cohort study. BMJ 2016; 352:i581. [PMID: 26888382 PMCID: PMC4772785 DOI: 10.1136/bmj.i581] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To determine whether the use of incretin based drugs compared with sulfonylureas is associated with an increased risk of incident pancreatic cancer in people with type 2 diabetes. DESIGN Population based cohort. SETTING Large, international, multicentre study combining the health records from six participating sites in Canada, the United States, and the United Kingdom. PARTICIPANTS A cohort of 972,384 patients initiating antidiabetic drugs between 1 January 2007 and 30 June 2013, with follow-up until 30 June 2014. MAIN OUTCOME MEASURES Within each cohort we conducted nested case-control analyses, where incident cases of pancreatic cancer were matched with up to 20 controls on sex, age, cohort entry date, duration of treated diabetes, and duration of follow-up. Hazard ratios and 95% confidence intervals for incident pancreatic cancer were estimated, comparing use of incretin based drugs with use of sulfonylureas, with drug use lagged by one year for latency purposes. Secondary analyses assessed whether the risk varied by class (dipeptidyl peptidase-4 inhibitors and glucagon-like peptide-1 receptor agonists) or by duration of use (cumulative duration of use and time since treatment initiation). Site specific hazard ratios were pooled using random effects models. RESULTS During 2,024,441 person years of follow-up (median follow-up ranging from 1.3 to 2.8 years; maximum 8 years), 1221 patients were newly diagnosed as having pancreatic cancer (incidence rate 0.60 per 1000 person years). Compared with sulfonylureas, incretin based drugs were not associated with an increased risk of pancreatic cancer (pooled adjusted hazard ratio 1.02, 95% confidence interval 0.84 to 1.23). Similarly, the risk did not vary by class and evidence of a duration-response relation was lacking. CONCLUSIONS In this large, population based study the use of incretin based drugs was not associated with an increased risk of pancreatic cancer compared with sulfonylureas. Although this potential adverse drug reaction will need to be monitored long term owing to the latency of the cancer, these findings provide some reassurance on the safety of incretin based drugs.
Collapse
Affiliation(s)
- Laurent Azoulay
- Center for Clinical Epidemiology, Lady Davis Research Institute, Jewish General Hospital, Montreal, Quebec, H3T 1E2, Canada Department of Oncology, McGill University, Montreal, Canada
| | - Kristian B Filion
- Center for Clinical Epidemiology, Lady Davis Research Institute, Jewish General Hospital, Montreal, Quebec, H3T 1E2, Canada Department of Medicine, McGill University, Montreal, Canada
| | - Robert W Platt
- Departments of Pediatrics and of Epidemiology, Biostatistics, and Occupational Health, McGill University, and the Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Matthew Dahl
- Manitoba Centre for Health Policy, Department of Community Health Sciences, University of Manitoba, Winnipeg, Canada and Department of Family Medicine, McMaster University, Hamilton, Canada
| | - Colin R Dormuth
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, Vancouver, Canada
| | | | - Madeleine Durand
- Department of Internal Medicine, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| | | | - Laura E Targownik
- Manitoba Centre for Health Policy, Department of Community Health Sciences, University of Manitoba, Winnipeg, Canada and Department of Family Medicine, McMaster University, Hamilton, Canada Section of Gastroenterology, Division of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Tanvir C Turin
- Department of Family Medicine, University of Calgary, Calgary, Canada
| | - J Michael Paterson
- Institute for Clinical Evaluative Sciences, Toronto, Canada Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto
| | - Pierre Ernst
- Center for Clinical Epidemiology, Lady Davis Research Institute, Jewish General Hospital, Montreal, Quebec, H3T 1E2, Canada Department of Medicine, McGill University, Montreal, Canada
| |
Collapse
|
225
|
Kang M, Qin W, Buya M, Dong X, Zheng W, Lu W, Chen J, Guo Q, Wu Y. VNN1, a potential biomarker for pancreatic cancer-associated new-onset diabetes, aggravates paraneoplastic islet dysfunction by increasing oxidative stress. Cancer Lett 2016; 373:241-50. [PMID: 26845448 DOI: 10.1016/j.canlet.2015.12.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 12/11/2015] [Accepted: 12/25/2015] [Indexed: 12/28/2022]
Abstract
In our previous clinical microarray analysis, we were the first to report on Vanin-1 (VNN1) as a novel clinically derived biomarker of pancreatic cancer-associated new-onset diabetes (PCAND). The functional mechanisms of VNN1 in the pathogenesis of PCAND, however, are not completely understood. In the present study, we further extend our previous clinical study to include laboratory research. The functions and mechanisms of neoplastic overexpressed VNN1 in PCAND have been explored using a co-culture model. Furthermore, the serum concentrations and discrimination power of downstream molecules of VNN1 were tested in a PCAND cohort. Pancreatic ductal adenocarcinoma (PDA) overexpressed VNN1 further aggravates paraneoplastic islet dysfunction; decreases in GSH/PPAR-γ concentrations and increases in ROS/cysteamine might be primary cause of this effect. Clinical serum analyses revealed that the expression profiles of these molecules were aberrant in the PCAND group. Our results further demonstrated that PCAND is a type of paraneoplastic diabetes. As the only clinically derived biomarker for PCAND screening available today, the biological role of VNN1 in triggering oxidative stress within the pancreatic microenvironment is important. The molecules downstream of VNN1 are also potential biomarkers for PCAND screening.
Collapse
Affiliation(s)
- Muxing Kang
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China; Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Wenjie Qin
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China; Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Miranbieke Buya
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Xin Dong
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Wen Zheng
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Wenjie Lu
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Jian Chen
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Qingqu Guo
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Yulian Wu
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China; Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China.
| |
Collapse
|
226
|
Risk Factors for Early-Onset and Very-Early-Onset Pancreatic Adenocarcinoma: A Pancreatic Cancer Case-Control Consortium (PanC4) Analysis. Pancreas 2016; 45:311-6. [PMID: 26646264 PMCID: PMC4710562 DOI: 10.1097/mpa.0000000000000392] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES While pancreatic cancer (PC) most often affects older adults, to date, there has been no comprehensive assessment of risk factors among PC patients younger than 60 years. METHODS We defined early-onset PC (EOPC) and very-early-onset PC (VEOPC) as diagnosis of PC in patients younger than 60 and 45 years, respectively. We pooled data from 8 case-control studies, including 1954 patients with EOPC and 3278 age- and sex-matched control subjects. Logistic regression analysis was performed to identify associations with EOPC and VEOPC. RESULTS Family history of PC, diabetes mellitus, smoking, obesity, and pancreatitis were associated with EOPC. Alcohol use equal to or greater than 26 g daily also was associated with increased risk of EOPC (odds ratio, 1.49; 95% confidence interval, 1.21-1.84), and there appeared to be a dose- and age-dependent effect of alcohol on risk. The point estimate for risk of VEOPC was an odds ratio of 2.18 (95% confidence interval, 1.17-4.09). CONCLUSIONS The established risk factors for PC, including smoking, diabetes, family history of PC, and obesity, also apply to EOPC. Alcohol intake appeared to have an age-dependent effect; the strongest association was with VEOPC.
Collapse
|
227
|
Liang D, Shi S, Xu J, Zhang B, Qin Y, Ji S, Xu W, Liu J, Liu L, Liu C, Long J, Ni Q, Yu X. New insights into perineural invasion of pancreatic cancer: More than pain. Biochim Biophys Acta Rev Cancer 2016; 1865:111-22. [PMID: 26794395 DOI: 10.1016/j.bbcan.2016.01.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/26/2015] [Accepted: 01/11/2016] [Indexed: 01/01/2023]
Abstract
Pancreatic cancer is one of the most malignant human tumors. Perineural invasion, whereby a cancer cell invades the perineural spaces surrounding nerves, is acknowledged as a gradual contributor to cancer aggressiveness. Furthermore, perineural invasion is considered one of the root causes of the recurrence and metastasis observed after pancreatic resection, and it is also an independent predictor of prognosis. Advanced research has demonstrated that the neural microenvironment is closely associated with perineural invasion in pancreatic cancer. Therapy targeting the molecular mechanism of perineural invasion may enable the durable clinical treatment of this formidable disease. This review provides an overview of the present status of perineural invasion, the relevant molecular mechanisms of perineural invasion, pain and hyperglycemia associated with perineural invasion in pancreatic cancer, and the targeted therapeutics based on these studies.
Collapse
Affiliation(s)
- Dingkong Liang
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Si Shi
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yi Qin
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wenyan Xu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jiang Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Liang Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Chen Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jiang Long
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Quanxing Ni
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
228
|
De Souza A, Khawaja KI, Masud F, Saif MW. Metformin and pancreatic cancer: Is there a role? Cancer Chemother Pharmacol 2016; 77:235-42. [PMID: 26740120 DOI: 10.1007/s00280-015-2948-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/11/2015] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is the fourth leading cause of cancer-related deaths in the USA, with a 5-year survival rate of 6 %. Anti-hyperglycemic treatments for type 2 diabetes mellitus that induce hyperinsulinemia (i.e., sulfonylureas) are thought to increase cancer risk, whereas treatments that lower insulin resistance and hyperinsulinemia (i.e., metformin) are considered cancer prevention strategies. Metformin is a cornerstone in the treatment of diabetes mellitus type 2. Retrospective studies have shown a survival benefit in diabetic patients with many solid tumors including pancreatic cancer that have been treated with metformin compared with patients treated with insulin or sulfonylureas. Metformin influences various cellular pathways, including activation of the LKB1/AMPK pathway, inhibition of cell division, promotion of apoptosis and autophagy, down-regulation of circulating insulin, and activation of the immune system. Ongoing research is redefining our understanding about how metformin modulates the molecular pathways implicated in pancreatic cancer. The authors review the topic critically and also give their opinion. Further studies investigating the effect of metformin in combination with chemotherapy, targeted agents, or radiation therapy are undergoing. In addition, the role of metabolic and other biomarkers is needed.
Collapse
Affiliation(s)
- Andre De Souza
- Section of GI Cancers and Experimental Therapeutics, Department of Hematology and Oncology, Tufts University School of Medicine and Tufts Cancer Center, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA
| | - Khadija Irfan Khawaja
- Department of Endocrinology and Metabolism, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Faisal Masud
- King Edward Medical University, Lahore, Pakistan
| | - Muhammad Wasif Saif
- Section of GI Cancers and Experimental Therapeutics, Department of Hematology and Oncology, Tufts University School of Medicine and Tufts Cancer Center, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA.
| |
Collapse
|
229
|
Illés D, Terzin V, Holzinger G, Kosár K, Róka R, Zsóri G, Ábrahám G, Czakó L. New-onset type 2 diabetes mellitus--A high-risk group suitable for the screening of pancreatic cancer? Pancreatology 2015; 16:266-71. [PMID: 26777407 DOI: 10.1016/j.pan.2015.12.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 11/05/2015] [Accepted: 12/04/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Type 2 diabetes mellitus is widely considered to be associated with pancreatic cancer. OBJECTIVE To determine the incidence of pancreatic cancer in new-onset type 2 diabetic patients by measuring the serum level of CA 19-9 and performing abdominal ultrasonography (US). PATIENTS AND METHODS Consecutive type 2 diabetic patients in whom diabetes was diagnosed within 36 months were included in this prospective study. Serum CA 19-9 measurement and US were performed in all patients. If any of two was positive, abdominal computer tomography (CT) was carried out. Endoscopic ultrasound-guided fine needle aspiration or direct surgical referral was performed on patients with CT-identified lesions. RESULTS A total of 115 patients were enrolled. CA 19-9 was elevated in 10 patients but pancreatic cancer diagnosed in neither of them. Pancreatic cancer was revealed by morphological means in three patients without elevated CA 19-9 level. The sensitivity, specificity, positive-, negative predictive values and validity were 0%, 90.4%, 0%, 97.9% and 87.9% for CA 19-9, 66.7%, 100%, 100%, 99% and 99% for US, respectively. The value of the Standardized Incidence Ratio for pancreatic cancer in new-onset type-2 diabetic patients was 198.6 (95% CI = 6.25-46.9). CONCLUSIONS The prevalence of pancreatic cancer in patients with new-onset type-2 diabetes is significantly higher than that in the general population and screening is beneficial for detecting PaC in this patient population. CA 19-9 and US is not reliable screening modality for pancreatic cancer screening in this population.
Collapse
Affiliation(s)
- Dóra Illés
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Viktória Terzin
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Gábor Holzinger
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Klára Kosár
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Richárd Róka
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Gábor Zsóri
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - György Ábrahám
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - László Czakó
- First Department of Medicine, University of Szeged, Szeged, Hungary.
| |
Collapse
|
230
|
Incio J, Suboj P, Chin SM, Vardam-Kaur T, Liu H, Hato T, Babykutty S, Chen I, Deshpande V, Jain RK, Fukumura D. Metformin Reduces Desmoplasia in Pancreatic Cancer by Reprogramming Stellate Cells and Tumor-Associated Macrophages. PLoS One 2015; 10:e0141392. [PMID: 26641266 PMCID: PMC4671732 DOI: 10.1371/journal.pone.0141392] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 10/06/2015] [Indexed: 02/06/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is a highly desmoplastic tumor with a dismal prognosis for most patients. Fibrosis and inflammation are hallmarks of tumor desmoplasia. We have previously demonstrated that preventing the activation of pancreatic stellate cells (PSCs) and alleviating desmoplasia are beneficial strategies in treating PDAC. Metformin is a widely used glucose-lowering drug. It is also frequently prescribed to diabetic pancreatic cancer patients and has been shown to associate with a better outcome. However, the underlying mechanisms of this benefit remain unclear. Metformin has been found to modulate the activity of stellate cells in other disease settings. In this study, we examine the effect of metformin on PSC activity, fibrosis and inflammation in PDACs. Methods/Results In overweight, diabetic PDAC patients and pre-clinical mouse models, treatment with metformin reduced levels of tumor extracellular matrix (ECM) components, in particular hyaluronan (HA). In vitro, we found that metformin reduced TGF-ß signaling and the production of HA and collagen-I in cultured PSCs. Furthermore, we found that metformin alleviates tumor inflammation by reducing the expression of inflammatory cytokines including IL-1β as well as infiltration and M2 polarization of tumor-associated macrophages (TAMs) in vitro and in vivo. These effects on macrophages in vitro appear to be associated with a modulation of the AMPK/STAT3 pathway by metformin. Finally, we found in our preclinical models that the alleviation of desmoplasia by metformin was associated with a reduction in ECM remodeling, epithelial-to-mesenchymal transition (EMT) and ultimately systemic metastasis. Conclusion Metformin alleviates the fibro-inflammatory microenvironment in obese/diabetic individuals with pancreatic cancer by reprogramming PSCs and TAMs, which correlates with reduced disease progression. Metformin should be tested/explored as part of the treatment strategy in overweight diabetic PDAC patients.
Collapse
Affiliation(s)
- Joao Incio
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Internal Medicine, Hospital S. Joao, Porto, Portugal
- I3S, Institute for Innovation and Research in Heath, Metabolism, Nutrition and Endocrinology group, Biochemistry Department, Faculty of Medicine, Porto University, Porto, Portugal
| | - Priya Suboj
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Botany and Biotechnology, St. Xaviers College, Thumba, Trivandrum, Kerala, India
| | - Shan M. Chin
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Trupti Vardam-Kaur
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hao Liu
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Program of Biology and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Tai Hato
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Suboj Babykutty
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Zoology, Mar Ivanios College, Nalanchira, Trivandrum, Kerala, India
| | - Ivy Chen
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
| | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rakesh K. Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (RKJ); (DF)
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (RKJ); (DF)
| |
Collapse
|
231
|
Wang X, Zhang H, Wang T, Lau WY, Wang X, Sun J, Yuan Z, Zhang Y. The concept and controversy of retroperitoneal nerve dissection in pancreatic head carcinoma (Review). Int J Oncol 2015; 47:2017-27. [PMID: 26458369 DOI: 10.3892/ijo.2015.3190] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 09/15/2015] [Indexed: 01/29/2023] Open
Abstract
Pancreatic head cancer is a common but the most lethal cancer of the human digestive system. It is invasive, resulting in early infiltration of adjacent structures and lymph node and distant metastases. Its biological characteristics of neurotropic growth lead to early neural invasion (NI) which is an independent prognostic factor of survival for pancreatic cancer. Radical surgical resection remains the only form of curative treatment. The extent of surgical resection and whether extended resection results in better long-term survival have been controversial. Studies have reported that peripancreatic plexus invasion is a frequent cause of pancreatic cancer recurrence and death. The relationship between cancer microenvironment and nerve cells, and whether the peripancreatic nerve plexus nearby needs to be resected require further studies. The present review aims to discuss the role of peripancreatic nerve and its implications in pancreatic head cancer resection.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Hongwei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Taihong Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Wan Yee Lau
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, P.R. China
| | - Xin Wang
- Department of Oncology, Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Jingfeng Sun
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Zhenhua Yuan
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Yewei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
232
|
Lu Y, García Rodríguez LA, Malgerud L, González-Pérez A, Martín-Pérez M, Lagergren J, Bexelius TS. New-onset type 2 diabetes, elevated HbA1c, anti-diabetic medications, and risk of pancreatic cancer. Br J Cancer 2015; 113:1607-14. [PMID: 26575601 PMCID: PMC4705881 DOI: 10.1038/bjc.2015.353] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/04/2015] [Accepted: 09/14/2015] [Indexed: 02/06/2023] Open
Abstract
Background: Associations between type 2 diabetes, anti-diabetic medications and pancreatic cancer are controversial. This study aims to clarify such associations with new-onset type 2 diabetes and repeated measurements of glycated haemoglobin (HbA1c) levels. Methods: A nested case–control study was initiated from the Health Improvement Network (THIN) in UK from 1996 to 2010. Information of pancreatic cancer cases was retrieved electronically from the medical records and manually validated. Control subjects were randomly selected and frequency-matched to the cases on sex, age, and calendar years. Multivariable unconditional logistic regression was performed to estimate odds ratios (OR) and 95% confidence intervals (CI), and adjusted for potential confounders. Results: Among 1 574 768 person-years of follow-up, 529 pancreatic cancer cases and 5000 controls were identified. Type 2 diabetes, or changed HbA1c levels (rather than HbA1c levels at diabetes diagnosis) in diabetes patients (⩾4 mmol mol−1 compared with <0 mmol mol−1) were followed by an increased OR of pancreatic cancer (OR, 2.16, 95% CI 1.72–2.72 and OR, 5.06, 95% CI 1.52–16.87, respectively). Among the anti-diabetic medications in diabetes patients, the OR for insulin users was 25.57 (95% CI 11.55–56.60), sulphonylureas 2.22 (95% CI 1.13, 4.40), and metformin users 1.46 (95% CI 0.85–2.52), compared with no use of any anti-diabetic medications. Conclusions: New-onset type 2 diabetes and, particularly, diabetes with rising HbA1c seem to be independent risk factors for pancreatic cancer. The relation between different anti-diabetic medications and pancreatic cancer seems to vary in strength, with the highest risk among users of insulin.
Collapse
Affiliation(s)
- Yunxia Lu
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 76, Sweden.,Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
| | | | - Linnéa Malgerud
- Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm 171 77, Sweden
| | | | - Mar Martín-Pérez
- Centro Español de Investigación Farmacoepidemiológica, Madrid 28004, Spain
| | - Jesper Lagergren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 76, Sweden.,Division of Cancer Studies, King's College London, London WC2R 2LS, UK
| | - Tomas S Bexelius
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 76, Sweden.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm 171 77, Sweden
| |
Collapse
|
233
|
Advances in Biomedical Imaging, Bioengineering, and Related Technologies for the Development of Biomarkers of Pancreatic Disease: Summary of a National Institute of Diabetes and Digestive and Kidney Diseases and National Institute of Biomedical Imaging and Bioengineering Workshop. Pancreas 2015; 44:1185-94. [PMID: 26465948 PMCID: PMC4608388 DOI: 10.1097/mpa.0000000000000552] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A workshop sponsored by the National Institute of Diabetes and Digestive and Kidney Diseases and the National Institute of Biomedical Imaging and Bioengineering focused on research gaps and opportunities in the development of new biomarkers of pancreatic disease. The session was held on July 22, 2015, and structured into 6 sessions: 1) Introduction and Overview; 2) Keynote Address; 3) New Approaches to the Diagnosis of Chronic Pancreatitis; 4) Biomarkers of Pain and Inflammation; 5) New Approaches to the Detection of Pancreatic Cancer; and 6) Shed Exosomes, Shed Cells, and Shed Proteins. Recent advances in the fields of pancreatic imaging, functional markers of pancreatic disease, proteomics, molecular and cellular imaging, and detection of circulating cancer cells and exosomes were reviewed. Knowledge gaps and research needs were highlighted. The development of new methods for the noninvasive determination of pancreatic pathology; the use of cellular markers of pancreatic function, inflammation, pain, and malignancy; and the refinement of methods to identify cells and cellular constituents of pancreatic cancer were discussed. The further refinement of sophisticated technical methods and the need for clinical studies to validate these new approaches in large-scale studies of patients at risk for the development of pancreatic disease were repeatedly emphasized.
Collapse
|
234
|
Wan X, Lei Y, Li Z, Wang J, Chen Z, McNutt M, Lin D, Zhao C, Jiang C, Li J, Pu Q, Su M, Wang Y, Gu J. Pancreatic Expression of Immunoglobulin G in Human Pancreatic Cancer and Associated Diabetes. Pancreas 2015; 44:1304-13. [PMID: 26390427 DOI: 10.1097/mpa.0000000000000544] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES The prognosis of pancreatic cancer (PC) is poor and the pathogenesis of PC-associated diabetes is unknown. We investigated the possible expression of immunoglobulin G (IgG) in human pancreatic carcinomas and adjacent pancreatic islets to gain a better understanding of these diseases. METHODS We employed immunohistochemistry, Western Blot, real-time polymerase chain reaction, and in situ hybridization to examine IgG expression in PC tissues and adjacent islets with and without cancer-associated diabetes. The IgG mRNA and IgG synthesizing-related enzymes were examined in PC cell lines. The IgG expression and secretion were downregulated with specific small interfering RNA and antibody to IgG followed by flow cytometry to assess its effect on apoptosis of cultured PC cells. RESULTS The expression of IgG was detected in pancreatic carcinoma and adjacent islets. Small interfering RNA and antibody treatments induced apoptosis in PC cell lines. In the carcinoma tissue, the levels of IgG expression varied depending on the stages of the cancers with more malignant cancers expressing more IgG (P < 0.05). The IgG levels in cancer cells were also increased when the patients had diabetes or hyperglycemia (P < 0.05). In addition, the extent of IgG expression in the seemingly normal islet cells adjacent to the tumor varied in relation to the grade of cancer differentiation and distance to the cancer nests. CONCLUSIONS (1) Immunoglobulin G was locally produced by PC cells and adjacent islet cells. (2) Immunoglobulin G may promote tumor growth by inhibiting cancer cell apoptosis. (3) Locally produced IgG might play a role in PC-associated diabetes.
Collapse
Affiliation(s)
- Xia Wan
- From the *Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, Guangdong; †Department of Pathology, School of Basic Medical Sciences, Peking (Beijing) University, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Shi W, Qiu W, Wang W, Zhou X, Zhong X, Tian G, Deng A. Osteoprotegerin is up-regulated in pancreatic cancers and correlates with cancer-associated new-onset diabetes. Biosci Trends 2015; 8:322-6. [PMID: 25641178 DOI: 10.5582/bst.2014.01092] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
New-onset diabetes might help to yield biomarkers for the early diagnosis of pancreatic cancer (PaC). In this study, we computationally predicted and experimentally validated osteoprotegerin (OPG) being associated with pancreatic cancer related new-onset diabetes. We first performed a meta-analysis on microarray datasets to search for genes specifically highly expressed in PaC, and then filtered for cytokines involved in islet dysfunction. The expression of OPG in PaC and normal pancreas were validated by immunohistochemistry. Serum OPG levels in healthy controls, non-cancerous diabetes and PaC patients with or without diabetes were detected by enzyme-linked immunosorbent assay (ELISA). In silico assay found that OPG up-regulated in PaC tissues in comparison to normal pancreas. Immunohistochemical data further confirmed that OPG was overexpressed in PaC samples. Furthermore, increased expression of OPG in PaC tissues correlated to the occurrence of new-onset diabetes, and adversely affected the patients' overall survival in both univariate and multivariate analysis. In addition, the serum levels of OPG were significantly higher in pancreatic cancer patients with new-onset diabetes than other groups including pancreatic patients without diabetes, new-onset type 2 diabetes and healthy controls. In conclusion, there is a close association between OPG and pancreatic cancer related new-onset diabetes, and OPG might serve as a potential biomarker for the early diagnosis of pancreatic cancer from populations with new-onset diabetes.
Collapse
Affiliation(s)
- Wanchun Shi
- Department of Endocrinology, Huzhou Central Hospital
| | | | | | | | | | | | | |
Collapse
|
236
|
Borazanci E, Millis SZ, Korn R, Han H, Whatcott CJ, Gatalica Z, Barrett MT, Cridebring D, Hoff DDV. Adenosquamous carcinoma of the pancreas: Molecular characterization of 23 patients along with a literature review. World J Gastrointest Oncol 2015; 7:132-140. [PMID: 26380056 PMCID: PMC4569590 DOI: 10.4251/wjgo.v7.i9.132] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 05/22/2015] [Accepted: 07/14/2015] [Indexed: 02/05/2023] Open
Abstract
Adenosquamous carcinoma of the pancreas (ASCP) is a rare entity. Like adenocarcinoma of the pancreas, overall survival is poor. Characteristics of ASCP include central tumor necrosis, along with osteoclasts and hypercalcemia. Various theories exist as to why this histological subtype exists, as normal pancreas tissue has no benign squamous epithelium. Due to the rarity of this disease, limited molecular analysis has been performed, and those reports indicate unique molecular features of ASCP. In this paper, we characterize 23 patients diagnosed with ASCP through molecular profiling using immunohistochemistry staining, fluorescent in situ hybridization, chromogenic in situ hybridization, and gene sequencing, Additionally, we provide a comprehensive literature review of what is known to date of ASCP. Molecular characterization revealed overexpression in MRP1 (80%), MGMT (79%), TOP2A (75), RRM1 (42%), TOPO1 (42%), PTEN (45%), CMET (40%), and C-KIT (10%) among others. One hundred percent of samples tested were positive for KRAS mutations. This analysis shows heretofore unsuspected leads to be considered for treatments of this rare type of exocrine pancreas cancer. Molecular profiling may be appropriate to provide maximum information regarding the patient’s tumor. Further work should be pursued to better characterize this disease.
Collapse
|
237
|
Ehehalt F, Sturm D, Rösler M, Distler M, Weitz J, Kersting S, Ludwig B, Schwanebeck U, Saeger HD, Solimena M, Grützmann R. Blood Glucose Homeostasis in the Course of Partial Pancreatectomy--Evidence for Surgically Reversible Diabetes Induced by Cholestasis. PLoS One 2015; 10:e0134140. [PMID: 26248027 PMCID: PMC4527702 DOI: 10.1371/journal.pone.0134140] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 06/27/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND AIM Partial pancreatic resection is accompanied not only by a reduction in the islet cell mass but also by a variety of other factors that are likely to interfere with glucose metabolism. The aim of this work was to characterize the patient dynamics of blood glucose homeostasis during the course of partial pancreatic resection and to specify the associated clinico-pathological variables. METHODS In total, 84 individuals undergoing elective partial pancreatic resection were consecutively recruited into this observational trial. The individuals were assigned based on their fasting glucose or oral glucose tolerance testing results into one of the following groups: (I) deteriorated, (II) stable or (III) improved glucose homeostasis three months after surgery. Co-variables associated with blood glucose dynamics were identified. RESULTS Of the 84 participants, 25 (30%) displayed a normal oGTT, 17 (20%) showed impaired glucose tolerance, and 10 (12%) exhibited pathological glucose tolerance. Elevated fasting glucose was present in 32 (38%) individuals before partial pancreatic resection. Three months after partial pancreatic resection, 14 (17%) patients deteriorated, 16 (19%) improved, and 54 (64%) retained stable glucose homeostasis. Stability and improvement was associated with tumor resection and postoperative normalization of recently diagnosed glucose dysregulation, preoperatively elevated tumor markers and markers for common bile duct obstruction, acute pancreatitis and liver cell damage. Improvement was linked to preoperatively elevated insulin resistance, which normalized after resection and was accompanied by a decrease in fasting- and glucose-stimulated insulin secretion. CONCLUSIONS Surgically reversible blood glucose dysregulation diagnosed concomitantly with a (peri-) pancreatic tumor appears secondary to compromised liver function due to tumor compression of the common bile duct and the subsequent increase in insulin resistance. It can be categorized as "cholestasis-induced diabetes" and thereby distinguished from other forms of hyperglycemic disorders.
Collapse
Affiliation(s)
- Florian Ehehalt
- Department of GI, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Dorothée Sturm
- Department of GI, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Germany
| | - Manuela Rösler
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Marius Distler
- Department of GI, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Jürgen Weitz
- Department of GI, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Stephan Kersting
- Department of GI, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Barbara Ludwig
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Department of Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Uta Schwanebeck
- Coordination Center for Clinical Trials, TU Dresden, Germany
| | - Hans-Detlev Saeger
- Department of GI, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Michele Solimena
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- * E-mail: (RG); (MS)
| | - Robert Grützmann
- Department of GI, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Germany
- * E-mail: (RG); (MS)
| |
Collapse
|
238
|
Lu C, Xu CF, Wan XY, Zhu HT, Yu CH, Li YM. Screening for pancreatic cancer in familial high-risk individuals: A systematic review. World J Gastroenterol 2015; 21:8678-8686. [PMID: 26229410 PMCID: PMC4515849 DOI: 10.3748/wjg.v21.i28.8678] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 04/03/2015] [Accepted: 06/16/2015] [Indexed: 02/07/2023] Open
Abstract
AIM: To analyze the benefits and harms of pancreatic cancer screening in familial high-risk individuals (HRIs).
METHODS: Studies were identified by searching PubMed, EBSCO, ClinicalTrials.gov and the Cochrane database from database inception to June 2014. We also obtained papers from the reference lists of pertinent studies and systematic reviews. English-language trials and observational studies were searched. The key words used as search terms were “screening” and “surveillance”. Cost-effectiveness, diagnostic rate, survival rate, mortality and adverse events were the outcomes of interest. Age, sex, lifestyle and other confounding factors were also considered. However, anticipating only a few of these studies, we also included observational studies with or without control groups. We also included studies concerning the anxiety associated with pancreatic cancer risk and other psychological changes in familial HRIs. We extracted details on study design, objectives, population characteristics, inclusion criteria, year of enrollment, method of screening, adjusted and unadjusted mortality, cost-effectiveness and adverse events from the included studies. Studies were assessed using the Reporting of Observational studies in Epidemiology (STROBE) checklist.
RESULTS: Sixteen studies on pancreatic cancer screening were included. Five studies included control groups, nine were observational studies without control groups, and the other two studies investigated the worry associated with pancreatic cancer risk. We found that pancreatic cancer screening resulted in a high curative resection rate (60% vs 25%, P = 0.011), longer median survival time (14.5 mo vs 4 mo, P < 0.001), and higher 3-year survival rate (20% vs 15.0%, P = 0.624). We also found that familial HRIs had a higher diagnostic rate of pancreatic tumors than controls (34% vs 7.2%, P < 0.001). In patients who underwent regular physical examinations, more stage I pancreatic cancers were observed (19% vs 2.6%, P = 0.001). In addition, endoscopic ultrasonography, which was the main means of detection, diagnosed 64.3% of pancreatic cancers. In comparison, endoscopic retrograde cannulation of the pancreas, magnetic resonance imaging, and computed tomography diagnosed 28.6%, 42.9%, and 21.4%, respectively. For mass lesions, instant surgery was recommended because of the beneficial effects of post-operative chemotherapy. However, in patients with intraductal papillary mucinous neoplasms, we did not find a significant difference in outcome between surgery and follow-up without treatment. Moreover, pancreatic cancer screening in familial HRIs had a greater perceived risk of pancreatic cancer (P < 0.0001), higher levels of anxiety regarding pancreatic cancer (P < 0.0001), and increased economic burden.
CONCLUSION: Pancreatic cancer screening in familial HRIs is associated with a higher detection rate and longer survival, although screening may influence psychological function and increase the economic burden.
Collapse
|
239
|
Lin QJ, Yang F, Jin C, Fu DL. Current status and progress of pancreatic cancer in China. World J Gastroenterol 2015; 21:7988-8003. [PMID: 26185370 PMCID: PMC4499341 DOI: 10.3748/wjg.v21.i26.7988] [Citation(s) in RCA: 201] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/31/2015] [Accepted: 04/17/2015] [Indexed: 02/06/2023] Open
Abstract
Cancer is currently one of the most important public health problems in the world. Pancreatic cancer is a fatal disease with poor prognosis. As in most other countries, the health burden of pancreatic cancer in China is increasing, with annual mortality rates almost equal to incidence rates. The increasing trend of pancreatic cancer incidence is more significant in the rural areas than in the urban areas. Annual diagnoses and deaths of pancreatic cancer in China are now beyond the number of cases in the United States. GLOBOCAN 2012 estimates that cases in China account for 19.45% (65727/337872) of all newly diagnosed pancreatic cancer and 19.27% (63662/330391) of all deaths from pancreatic cancer worldwide. The population’s growing socioeconomic status contributes to the rapid increase of China’s proportional contribution to global rates. Here, we present an overview of control programs for pancreatic cancer in China focusing on prevention, early diagnosis and treatment. In addition, we describe key epidemiological, demographic, and socioeconomic differences between China and developed countries. Facts including no nationwide screening program for pancreatic cancer, delay in early detection resulting in a late stage at presentation, lack of awareness of pancreatic cancer in the Chinese population, and low investment compared with other cancer types by government have led to backwardness in China’s pancreatic cancer diagnosis and treatment. Finally, we suggest measures to improve health outcomes of pancreatic cancer patients in China.
Collapse
|
240
|
Sadr-Azodi O, Gudbjörnsdottir S, Ljung R. Pattern of increasing HbA1c levels in patients with diabetes mellitus before clinical detection of pancreatic cancer - a population-based nationwide case-control study. Acta Oncol 2015; 54:986-92. [PMID: 25734801 DOI: 10.3109/0284186x.2015.1006402] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Diabetes mellitus is a risk factor for pancreatic cancer. Impaired insulin resistance might precede the clinical detection of this cancer by several years. METHODS This was a nested case-control population-based study assessing the pattern of glycated hemoglobin (HbA1c) change before clinical detection of pancreatic cancer in a population of individuals with diabetes mellitus. All patients registered in the Swedish National Diabetes Register with a prescription of an anti-diabetic drug between 2005 and 2011 were identified. For each case of pancreatic cancer, 10 controls were randomly selected, matched for age, sex, and factors related to diabetes mellitus. Multivariable conditional logistic regression was used to calculate odds ratios (ORs) and 95% confidence intervals (CIs) for the association between HbA1c and pancreatic cancer. RESULTS In total, 391 cases and 3910 matched controls were identified. The risk of pancreatic cancer was increased more than two-fold in individuals with the highest HbA1c quartile compared with the lowest (OR 1.96, 95% CI 1.40-2.75). The risk of pancreatic cancer remained elevated when comparing the highest HbA1c quartile measured within five years from the clinical detection of pancreatic cancer to the lowest HbA1c quartile (p-value for trend < 0.05). No association was found between HbA1c and pancreatic cancer if HbA1c was measured > 5 years before the clinical detection of pancreatic cancer. CONCLUSIONS The pattern of increasing HbA1c in patients with diabetes mellitus preceded the clinical detection of pancreatic cancer by up to five years. These findings indicate that there is a lead time of several years during which the development of pancreatic cancer might be detectable through screening in patients with diabetes mellitus.
Collapse
Affiliation(s)
- Omid Sadr-Azodi
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Centre for Clinical Research Sörmland, Uppsala University, Eskilstuna, Sweden
| | - Soffia Gudbjörnsdottir
- National Diabetes Register, Registercentrum VGR, Gothenburg, Sweden
- Department of Medicine, Sahlgrenska University Hospital, University of Gothenburg, Sweden
| | - Rickard Ljung
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
241
|
Wu JM, Ho TW, Kuo TC, Yang CY, Lai HS, Chiang PY, Hsieh SH, Lai F, Tien YW. Glycemic Change After Pancreaticoduodenectomy: A Population-Based Study. Medicine (Baltimore) 2015; 94:e1109. [PMID: 26166104 PMCID: PMC4504605 DOI: 10.1097/md.0000000000001109] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/19/2015] [Accepted: 06/07/2015] [Indexed: 12/13/2022] Open
Abstract
The purpose of this population-based study was to determine the change of glucose metabolism in patients undergoing pancreaticoduodenectomy (PD).We conducted a nationwide cohort study using data from Taiwan's National Health Insurance Research Database collected between 2000 and 2010. Our sample included 861 subjects with type 2 diabetes mellitus (DM) and 3914 subjects without DM.Of 861 subjects with type 2 diabetes, 174 patients (20.2%) experienced resolution of their diabetes after PD, including patients with pancreatic ductal adenocarcinoma (PDAC) (20.5%), and non-PDAC (20.1%). Using a multiple logistic regression model, we found that subjects with comorbid chronic pancreatitis (odds ratio, 0.356; 95% CI, 0.167-0.759; P = 0.007) and use of insulin (odds ratio, 0.265; 95% CI, 0.171-0.412; P < 0.001) had significantly lower rates of resolution of diabetes. In the 3914 subjects without diabetes, the only statistically significant comorbidity contributing to pancreatogenic diabetes was chronic pancreatitis (odds ratio, 1.446; 95% CI, 1.146-1.823; P = 0.002).Subjects with comorbid chronic pancreatitis and use of insulin had lower rates of resolution of DM after PD. In subjects without diabetes, chronic pancreatitis contributed significantly to the development of pancreatogenic DM.
Collapse
Affiliation(s)
- Jin-Ming Wu
- From the Department of Surgery (J-MW, T-CK, C-YY, H-SL, Y-WT); Department of Nursing, National Taiwan University Hospital and National Taiwan University College of Medicine (P-YC, S-HH); and Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan, ROC (J-MW, T-WH, FL)
| | | | | | | | | | | | | | | | | |
Collapse
|
242
|
Salvatore T, Marfella R, Rizzo MR, Sasso FC. Pancreatic cancer and diabetes: A two-way relationship in the perspective of diabetologist. Int J Surg 2015; 21 Suppl 1:S72-7. [PMID: 26123386 DOI: 10.1016/j.ijsu.2015.06.063] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 03/25/2015] [Accepted: 04/10/2015] [Indexed: 12/31/2022]
Abstract
Diabetes is a risk factor for pancreatic cancer as roughly half of all patients with pancreatic cancer are found to have diabetes at time of diagnosis. Moreover, an around 2-fold risk of pancreatic malignancy in diabetic patients has even be recently resulted from two meta-analysis. Actually, there is a bidirectional association between the two entities that implies a complex and reverse causality. In fact, while the risk for pancreatic cancer is modestly but significantly increased in patients with long-standing diabetes, recent-onset diabetes appears to be very frequently associated with pancreatic malignancy. Therefore, diabetes could serve as an excellent clue for early detection of pancreatic cancer. Moreover, recent epidemiological findings support the hypothesis that chronic exposure to hyperglycemia, higher insulin concentrations, and insulin resistance may be responsible for the enhanced risk of developing pancreatic cancer. Epidemiological data suggest that the type of anti-diabetic therapy may affect the risk of developing pancreatic cancer. In particular, metformin has been shown to reduce the risk of pancreatic cancer, as well as several other malignancies. On the other hand, some hypoglycemic agents could determine an increase of pancreatic cancer risk. These last findings were not confirmed. Finally, pancreatic cancer necessitates of a multidisciplinary management, primarily including surgeons and oncologists. In this context, the diabetologist plays an important role, given that his actions may influence the prevention and early diagnosis of pancreatic cancer, the perioperative complications associated to glycemic derangement, as well as the proper treatment of postpancreactomy diabetes.
Collapse
Affiliation(s)
- Teresa Salvatore
- Depart. of Internal and Experimental Medicine "Magrassi - Lanzara", Second University of Naples, Italy.
| | - Raffaele Marfella
- Depart. of Medicine, Surgery, Neurology, Metabolism and Geriatrics, Second University of Naples, Italy.
| | - Maria Rosaria Rizzo
- Depart. of Medicine, Surgery, Neurology, Metabolism and Geriatrics, Second University of Naples, Italy.
| | - Ferdinando Carlo Sasso
- Depart. of Internal and Experimental Medicine "Magrassi - Lanzara", Second University of Naples, Italy.
| |
Collapse
|
243
|
Fang L, Kojima K, Zhou L, Crossman DK, Mobley JA, Grams J. Analysis of the Human Proteome in Subcutaneous and Visceral Fat Depots in Diabetic and Non-diabetic Patients with Morbid Obesity. ACTA ACUST UNITED AC 2015; 8:133-141. [PMID: 26472921 PMCID: PMC4603752 DOI: 10.4172/jpb.1000361] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
No longer regarded as simply a storage depot, fat is a dynamic organ acting locally and systemically to modulate energy homeostasis, glucose sensitivity, insulin resistance, and inflammatory pathways. Here, mass spectrometry was used to survey the proteome of patient matched subcutaneous fat and visceral fat in 20 diabetic vs 22 nondiabetic patients with morbid obesity. A similar number of proteins (~600) were identified in each tissue type. When stratified by diabetic status, 19 and 41 proteins were found to be differentially abundant in subcutaneous fat and omentum, respectively. These proteins represent pathways known to be involved in metabolism. Five of these proteins were differentially abundant in both fat depots: moesin, 78 kDa glucose-regulated protein, protein cordon-bleu, zinc finger protein 611, and cytochrome c oxidase subunit 6B1. Three proteins, decorin, cytochrome c oxidase subunit 6B1, and 78 kDa glucose-regulated protein, were further tested for validation by western blot analysis. Investigation of the proteins reported here is expected to expand on the current knowledge of adipose tissue driven biochemistry in diabetes and obesity, with the ultimate goal of identifying clinical targets for the development of novel therapeutic interventions in the treatment of type 2 diabetes mellitus. To our knowledge, this study is the first to survey the global proteome derived from each subcutaneous and visceral adipose tissue obtained from the same patient in the clinical setting of morbid obesity, with and without diabetes. It is also the largest study of diabetic vs nondiabetic patients with 42 patients surveyed.
Collapse
Affiliation(s)
- Lingling Fang
- Ningbo Lihuili Hospital; Ningbo, Zhejiang, China ; Department of Surgery, University of Alabama at Birmingham; Birmingham, AL, USA
| | - Kyoko Kojima
- Comprehensive Cancer Center, University of Alabama at Birmingham; Birmingham, AL, USA
| | - Lihua Zhou
- Department of Surgery, University of Alabama at Birmingham; Birmingham, AL, USA
| | - David K Crossman
- Heflin Center for Genomic Science, University of Alabama at Birmingham; Birmingham, AL, USA ; Department of Genetics, University of Alabama at Birmingham; Birmingham, AL, USA
| | - James A Mobley
- Department of Surgery, University of Alabama at Birmingham; Birmingham, AL, USA ; Comprehensive Cancer Center, University of Alabama at Birmingham; Birmingham, AL, USA ; Heflin Center for Genomic Science, University of Alabama at Birmingham; Birmingham, AL, USA
| | - Jayleen Grams
- Department of Surgery, University of Alabama at Birmingham; Birmingham, AL, USA ; Department of Surgery, Birmingham Veterans Administration Medical Center, Birmingham, AL, USA
| |
Collapse
|
244
|
Li J, Ma J, Han L, Xu Q, Lei J, Duan W, Li W, Wang F, Wu E, Ma Q, Huo X. Hyperglycemic tumor microenvironment induces perineural invasion in pancreatic cancer. Cancer Biol Ther 2015; 16:912-21. [PMID: 25946624 DOI: 10.1080/15384047.2015.1040952] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Glucose intolerance and frank diabetes mellitus (DM) can increase the risk of cancer death for pancreatic cancer (PanCa). However, the mechanism by which these factors influence cancer deaths is not clear. In this study, we established a model system to mimic the pancreatic tumor microenvironment in patients with DM to examine the biological behavior of PanCa cells and nerves in cell culture and in animals. Our in vitro studies demonstrated that hyperglycemia promoted the proliferation and invasion of PanCa cell lines and upregulated the expression of nerve growth factor in these cells. Also, the migration of Schwann cells (SCs) was inhibited by hyperglycemia and neurites exerted pathological regeneration. Furthermore, the interaction between the PanCa cells and nerves was enhanced in the tumor microenvironment. We further showed that hyperglycemia promoted the perineural invasion (PNI) of PanCa in vivo. These data suggest that DM worsens the prognosis of PanCa because of aggravated PNI. Thus, our study illustrates a novel mechanism by which hyperglycemia decreases survival in patients with PanCa.
Collapse
Affiliation(s)
- Junhui Li
- a Department of Hepatobiliary Surgery ; First Affiliated Hospital of Medical College; Xi'an Jiaotong University ; Xi'an , China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Pan S, Brentnall TA, Chen R. Proteomics analysis of bodily fluids in pancreatic cancer. Proteomics 2015; 15:2705-15. [PMID: 25780901 DOI: 10.1002/pmic.201400476] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 02/06/2015] [Accepted: 03/13/2015] [Indexed: 12/12/2022]
Abstract
Proteomics study of pancreatic cancer using bodily fluids emphasizes biomarker discovery and clinical application, presenting unique prospect and challenges. Depending on the physiological nature of the bodily fluid and its proximity to pancreatic cancer, the proteomes of bodily fluids, such as pancreatic juice, pancreatic cyst fluid, blood, bile, and urine, can be substantially different in terms of protein constitution and the dynamic range of protein concentration. Thus, a comprehensive discovery and specific detection of cancer-associated proteins within these varied fluids is a complex task, requiring rigorous experiment design and a concerted approach. While major challenges still remain, fluid proteomics studies in pancreatic cancer to date have provided a wealth of information in revealing proteome alterations associated with pancreatic cancer in various bodily fluids.
Collapse
Affiliation(s)
- Sheng Pan
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Ru Chen
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
246
|
Javeed N, Sagar G, Dutta SK, Smyrk TC, Lau JS, Bhattacharya S, Truty M, Petersen GM, Kaufman RJ, Chari ST, Mukhopadhyay D. Pancreatic Cancer-Derived Exosomes Cause Paraneoplastic β-cell Dysfunction. Clin Cancer Res 2015; 21:1722-33. [PMID: 25355928 PMCID: PMC4383684 DOI: 10.1158/1078-0432.ccr-14-2022] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/11/2014] [Indexed: 12/14/2022]
Abstract
PURPOSE Pancreatic cancer frequently causes diabetes. We recently proposed adrenomedullin as a candidate mediator of pancreatic β-cell dysfunction in pancreatic cancer. How pancreatic cancer-derived adrenomedullin reaches β cells remote from the cancer to induce β-cell dysfunction is unknown. We tested a novel hypothesis that pancreatic cancer sheds adrenomedullin-containing exosomes into circulation, which are transported to β cells and impair insulin secretion. EXPERIMENTAL METHODS We characterized exosomes from conditioned media of pancreatic cancer cell lines (n = 5) and portal/peripheral venous blood of patients with pancreatic cancer (n = 20). Western blot analysis showed the presence of adrenomedullin in pancreatic cancer-exosomes. We determined the effect of adrenomedullin-containing pancreatic cancer exosomes on insulin secretion from INS-1 β cells and human islets, and demonstrated the mechanism of exosome internalization into β cells. We studied the interaction between β-cell adrenomedullin receptors and adrenomedullin present in pancreatic cancer-exosomes. In addition, the effect of adrenomedullin on endoplasmic reticulum (ER) stress response genes and reactive oxygen/nitrogen species generation in β cells was shown. RESULTS Exosomes were found to be the predominant extracellular vesicles secreted by pancreatic cancer into culture media and patient plasma. Pancreatic cancer-exosomes contained adrenomedullin and CA19-9, readily entered β cells through caveolin-mediated endocytosis or macropinocytosis, and inhibited insulin secretion. Adrenomedullin in pancreatic cancer exosomes interacted with its receptor on β cells. Adrenomedullin receptor blockade abrogated the inhibitory effect of exosomes on insulin secretion. β cells exposed to adrenomedullin or pancreatic cancer exosomes showed upregulation of ER stress genes and increased reactive oxygen/nitrogen species. CONCLUSIONS Pancreatic cancer causes paraneoplastic β-cell dysfunction by shedding adrenomedullin(+)/CA19-9(+) exosomes into circulation that inhibit insulin secretion, likely through adrenomedullin-induced ER stress and failure of the unfolded protein response.
Collapse
Affiliation(s)
- Naureen Javeed
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Gunisha Sagar
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Shamit K Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Thomas C Smyrk
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Julie S Lau
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Mark Truty
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Randal J Kaufman
- Degenerative Disease Research Program, Sanford Burnham Medical Research Institute, La Jolla, California
| | - Suresh T Chari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| | | |
Collapse
|
247
|
Gao W, Zhou Y, Li Q, Zhou Q, Tan L, Song Y, Zhao X, Yu M, Zheng S, Ye H, Zeng B, Lin Q, Zhou J, Liu Y, Huang H, Zhang H, Hu X, Li Z, Dai X, Chen R. Analysis of global gene expression profiles suggests a role of acute inflammation in type 3C diabetes mellitus caused by pancreatic ductal adenocarcinoma. Diabetologia 2015; 58:835-44. [PMID: 25636208 DOI: 10.1007/s00125-014-3481-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 12/05/2014] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Pancreatic ductal adenocarcinoma (PDAC) can cause type 3C diabetes, known as PDAC-associated diabetes mellitus (PDAC-DM), but the mechanism is unknown. This study aimed to reveal the mechanism. METHODS PDAC lesions from patients with or without PDAC-DM (n = 4 in each group) were individually profiled for 23,512 mRNAs with microarrays. Bioinformatic analysis and in vivo and in vitro assays were then conducted. RESULTS We determined that 2,778 genes were differentially expressed; over-representation of ten genes was validated with quantitative RT-PCR. The analysis of gene ontology showed that the differentially expressed secretory genes were related mainly to inflammation. High levels of a marker of inflammation (C-reactive protein [CRP]) and an inflammatory mediator (TNF super-family member 13 [TNFSF13]) were found in the serum of patients with PDAC-DM. After surgical resection of PDAC lesions, CRP and TNFSF13 levels significantly decreased (p < 0.01). Furthermore, we found that the levels of TNFSF13 in PDAC lesions and TNFSF13 and CRP in serum were significantly correlated with the diabetic status of patients with PDAC-DM (p < 0.01). Assays in vivo showed that after exposure to an inhibitor of inflammation (celecoxib), the fasting blood glucose level in the mouse model of PDAC-DM dramatically decreased from 6.9 ± 0.1 to 5.6 ± 0.1 mmol/l in 2-4 days (p < 0.01). CONCLUSIONS/INTERPRETATION We found that acute inflammation was involved in the pathogenesis of PDAC-DM. We contend that acute inflammation is a potential target for the diagnosis and treatment of PDAC-DM.
Collapse
MESH Headings
- Aged
- Animals
- Anti-Inflammatory Agents/pharmacology
- Blood Glucose/metabolism
- Carcinoma, Pancreatic Ductal/complications
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/genetics
- Case-Control Studies
- Cells, Cultured
- Computational Biology
- Diabetes Mellitus/blood
- Diabetes Mellitus/diagnosis
- Diabetes Mellitus/genetics
- Diabetes Mellitus/prevention & control
- Disease Models, Animal
- Female
- Gene Expression Profiling/methods
- Genetic Association Studies
- Genetic Markers
- Humans
- Inflammation Mediators/blood
- Male
- Mice, Inbred BALB C
- Middle Aged
- Oligonucleotide Array Sequence Analysis
- Pancreatic Neoplasms/complications
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatitis, Chronic/blood
- Pancreatitis, Chronic/diagnosis
- Pancreatitis, Chronic/genetics
- Pancreatitis, Chronic/prevention & control
- Polymerase Chain Reaction
- Reproducibility of Results
Collapse
Affiliation(s)
- Wenchao Gao
- Department of Hepatopancreatobiliary Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, No. 107 Yanjiang Road, 510120, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Choi Y, Kim TY, Oh DY, Lee KH, Han SW, Im SA, Kim TY, Bang YJ. The Impact of Diabetes Mellitus and Metformin Treatment on Survival of Patients with Advanced Pancreatic Cancer Undergoing Chemotherapy. Cancer Res Treat 2015; 48:171-9. [PMID: 25779362 PMCID: PMC4720092 DOI: 10.4143/crt.2014.292] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 02/02/2015] [Indexed: 12/21/2022] Open
Abstract
PURPOSE A causal relationship between diabetes mellitus (DM) and pancreatic cancer is well established. However, in patients with advanced pancreatic cancer (APC) who receive palliative chemotherapy, the impact of DM on the prognosis of APC is unclear. MATERIALS AND METHODS We retrospectively enrolled APC patients who received palliative chemotherapy between 2003 and 2010. The patients were stratified according to the status of DM, in accordance with 2010 DM criteria (American Heart Association/American Diabetes Association). DM at least 2 years' duration prior to diagnosis of APC was defined as remote-onset DM (vs. recent-onset). RESULTS Of the 349 APC patients, 183 (52.4%) had DM. Among the patients with DM, 160 patients had DM at the time of diagnosis of APC (remote-onset, 87; recent-onset, 73) and the remaining 23 patients developed DM during treatment of APC. Ultimately, 73.2% of patients (134/183) with DM received antidiabetic medication, including metformin (56 patients, 41.8%), sulfonylurea (62, 45.5%), and insulin (43, 32.1%). In multivariate analysis, cancer extent (hazard ratio [HR], 1.792; 95% confidence interval [CI], 1.313 to 2.445; p < 0.001) showed association with decreased overall survival (OS), whereas a diagnosis of DM (HR, 0.788; 95% CI, 0.615 to 1.009; p=0.059) conferred positive tendency on the OS. Metformin treatment itself conferred better OS in comparison within DM patients (HR 0.693; 95% CI, 0.492 to 0.977; p=0.036) and even in all APC patients (adjusted HR, 0.697; 95% CI, 0.491 to 1.990; p=0.044). CONCLUSION For APC patients receiving palliative chemotherapy, metformin treatment is associated with longer OS. Patients with DM tend to survive longer than those without DM.
Collapse
Affiliation(s)
- Younak Choi
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Tae-Yong Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea ; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea ; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea ; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sae-Won Han
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea ; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea ; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Tae-You Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea ; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yung-Jue Bang
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea ; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
249
|
Pulmonary Embolism and Stroke as the Initial Manifestation of Advanced Metastatic Pancreatic Adenocarcinoma. J Gastrointest Cancer 2015; 46:182-4. [DOI: 10.1007/s12029-015-9697-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
250
|
Mayers JR, Vander Heiden MG. Famine versus feast: understanding the metabolism of tumors in vivo. Trends Biochem Sci 2015; 40:130-40. [PMID: 25639751 PMCID: PMC4340757 DOI: 10.1016/j.tibs.2015.01.004] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/07/2015] [Accepted: 01/08/2015] [Indexed: 12/20/2022]
Abstract
To fuel unregulated proliferation, cancer cells alter metabolism to support macromolecule biosynthesis. Cell culture studies have revealed how different oncogenic mutations and nutrients impact metabolism. Glucose and glutamine are the primary fuels used in vitro; however, recent studies have suggested that utilization of other amino acids as well as lipids and protein can also be important to cancer cells. Early investigations of tumor metabolism are translating these findings to the biology of whole tumors and suggest that additional complexity exists beyond nutrient availability alone in vivo. Whole-body metabolism and tumor heterogeneity also influence the metabolism of tumor cells, and successful targeting of metabolism for cancer therapy will require an understanding of tumor metabolism in vivo.
Collapse
Affiliation(s)
- Jared R Mayers
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard University, Cambridge, MA, USA.
| |
Collapse
|