51
|
Bosch X, Moreno P, Guerra-García M, Guasch N, López-Soto A. What is the relevance of an ambulatory quick diagnosis unit or inpatient admission for the diagnosis of pancreatic cancer? A retrospective study of 1004 patients. Medicine (Baltimore) 2020; 99:e19009. [PMID: 32176029 PMCID: PMC7440208 DOI: 10.1097/md.0000000000019009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Quick diagnosis units (QDU) have become an alternative hospital-based ambulatory medicine strategy to inpatient hospitalization for potentially serious illnesses in Spain. Whether diagnosis of pancreatic cancer is better accomplished by an ambulatory or inpatient approach is unknown. The main objective of this retrospective study was to examine and compare the diagnostic effectiveness of a QDU or inpatient setting in patients with pancreatic cancer.Patients with a diagnosis of pancreatic adenocarcinoma who had been referred to a university, tertiary hospital-based QDU or hospitalized between 2005 and 2018 were eligible. Presenting symptoms and signs, risk and prognostic factors, and time to diagnosis were compared. The costs incurred during the diagnostic assessment were analyzed with a microcosting method.A total of 1004 patients (508 QDU patients and 496 inpatients) were eligible. Admitted patients were more likely than QDU patients to have weight loss, asthenia, anorexia, abdominal pain, jaundice, and palpable hepatomegaly. Time to diagnosis of inpatients was similar to that of QDU patients (4.1 [0.8 vs 4.3 [0.6] days; P = .163). Inpatients were more likely than QDU patients to have a tumor on the head of the pancreas, a tumor size >2 cm, a more advanced nodal stage, and a poorer histological differentiation. No differences were observed in the proportion of metastatic and locally advanced disease and surgical resections. Microcosting revealed a cost of &OV0556;347.76 (48.69) per QDU patient and &OV0556;634.36 (80.56) per inpatient (P < .001).Diagnosis of pancreatic cancer is similarly achieved by an inpatient or QDU clinical approach, but the latter seems to be cost-effective. Because the high costs of hospitalization, an ambulatory diagnostic assessment may be preferable in these patients.
Collapse
Affiliation(s)
- Xavier Bosch
- Quick Diagnosis Unit, Adult Day Care Center, Hospital Clínic, University of Barcelona
| | - Pedro Moreno
- Department of Internal Medicine, Hospital Clínic, University of Barcelona
| | - Mar Guerra-García
- Adult Day Care Center, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Neus Guasch
- Adult Day Care Center, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Alfons López-Soto
- Department of Internal Medicine, Hospital Clínic, University of Barcelona
| |
Collapse
|
52
|
Dai WF, Beca J, Guo H, Isaranawatchai W, Schwartz D, Naipaul R, Arias J, Qiao Y, Gavura S, Redmond‐Misner R, Ismail Z, Barbera L, Chan K. Are population-based patient-reported outcomes associated with overall survival in patients with advanced pancreatic cancer? Cancer Med 2020; 9:215-224. [PMID: 31736256 PMCID: PMC6943146 DOI: 10.1002/cam4.2704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/27/2019] [Accepted: 10/22/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Advanced pancreatic cancer (APC) patients often have substantial symptom burden. In Ontario, patients routinely complete the Edmonton Symptom Assessment Scale (ESAS), which screens for nine symptoms (scale: 0-10), in cancer clinics. We explored the association between baseline patient-reported outcomes, via ESAS, and overall survival (OS). METHODS Advanced pancreatic cancer patients with ESAS records prior to receiving publicly funded drugs from November 2008 to March 2016 were retrospectively identified from Cancer Care Ontario's administrative databases. We examined three composite ESAS scores: total symptom distress score (TSDS: 9 symptoms), physical symptom score (PHS: 6/9 symptoms), and psychological symptom score (PSS: 2/9 symptoms); Composite scores greater than defined thresholds (TSDS ≥36, PHS ≥24, PSS ≥8) were considered as high symptom burden. Crude OS was assessed using Kaplan-Meier method. Hazard ratios (HRs) were assessed using multivariable Cox models. Analysis was repeated in a sub-cohort with Eastern Cooperative Oncology Group (ECOG) status and metastasis. RESULTS We identified 2199 APC patients (mean age 64 years, 55% male) with ESAS records prior to receiving chemotherapy. Crude median survival was 4.5 and 7.3 months for high and low TSDS, respectively. High TSDS was associated with lower OS (HR = 1.47, 95% CI: 1.33, 1.63). In the sub-cohort (n = 393) with ECOG status and metastasis, high TSDS was also associated with lower OS (HR = 1.34, 95% CI: 1.04, 1.73). Similar trends were observed for PHS and PSS. CONCLUSIONS Higher burden of patient-reported outcome was associated with reduced OS among APC patients. The effect was prominent after adjusting for ECOG status.
Collapse
Affiliation(s)
- Wei Fang Dai
- Cancer Care OntarioTorontoONCanada
- Canadian Centre for Applied Research in Cancer ControlTorontoONCanada
| | - Jaclyn Beca
- Cancer Care OntarioTorontoONCanada
- Canadian Centre for Applied Research in Cancer ControlTorontoONCanada
| | | | - Wanrudee Isaranawatchai
- Cancer Care OntarioTorontoONCanada
- Canadian Centre for Applied Research in Cancer ControlTorontoONCanada
- St Michael's HospitalTorontoONCanada
- Insitute of Health Policy, Management and EvaluationUniversity of TorontoTorontoONCanada
| | | | | | | | - Yao Qiao
- Cancer Care OntarioTorontoONCanada
| | | | | | | | - Lisa Barbera
- Cancer Care OntarioTorontoONCanada
- Tom Baker Cancer CentreCalgaryABCanada
- University of CalgaryCalgaryABCanada
| | - Kelvin Chan
- Cancer Care OntarioTorontoONCanada
- Canadian Centre for Applied Research in Cancer ControlTorontoONCanada
- Sunnybrook Health Sciences CentreTorontoONCanada
| |
Collapse
|
53
|
Macarulla Mercadé T, Chen LT, Li CP, Siveke JT, Cunningham D, Bodoky G, Blanc JF, Lee KH, Dean A, Belanger B, Wang-Gillam A. Liposomal Irinotecan + 5-FU/LV in Metastatic Pancreatic Cancer: Subgroup Analyses of Patient, Tumor, and Previous Treatment Characteristics in the Pivotal NAPOLI-1 Trial. Pancreas 2020; 49:62-75. [PMID: 31856081 PMCID: PMC6946097 DOI: 10.1097/mpa.0000000000001455] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/04/2019] [Indexed: 12/26/2022]
Abstract
OBJECTIVES The NAnoliPOsomaL Irinotecan (NAPOLI-1) study (NCT01494506) was the largest global phase 3 study in a post-gemcitabine metastatic pancreatic adenocarcinoma (mPAC) population (N = 417). The subanalyses reported here investigated the prognostic effect of tumor characteristics and disease stage, prior treatment characteristics, baseline patient characteristics on survival outcomes in NAPOLI-1, and whether liposomal irinotecan (nal-IRI) + 5-fluorouracil/leucovorin (5-FU/LV) benefited patients with mPAC across subgroups. METHODS Post hoc analyses were performed in the NAPOLI-1 population (4 across tumor characteristics and disease stage, 6 across prior treatment characteristics, and 4 across patient baseline characteristics). Survival outcomes were estimated by Kaplan-Meier analysis and patient safety data were evaluated. RESULTS Mortality and morbidity risk was lower on nal-IRI+5-FU/LV treatment across subgroups. Exceptions were patients who had received prior nonliposomal irinotecan and those who had undergone prior Whipple procedure (overall survival hazard ratio = 1.25 and 1.23, respectively). Decreased appetite, liver metastases, and number of measurable metastatic lesions seemed to be prognostic of survival in this population. Subgroup safety data were generally comparable with those in the overall NAPOLI-1 safety population. CONCLUSIONS A diverse population of patients with mPAC that progressed on gemcitabine-based therapy benefited from nal-IRI+5-FU/LV versus 5-FU/LV, potentially helping guide treatment decisions for challenging cases.
Collapse
Affiliation(s)
- Teresa Macarulla Mercadé
- From the Vall d'Hebron University Hospital (HUVH) and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes
- Department of Internal Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan
| | - Chung-Pin Li
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital
- National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Jens T. Siveke
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen
- German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - David Cunningham
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - György Bodoky
- Department of Oncology, Szent László Hospital, Budapest, Hungary
| | | | - Kyung-Hun Lee
- Department of Internal Medicine and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Andrew Dean
- St. John of God Hospital, Subiaco, Western Australia, Australia
| | | | | |
Collapse
|
54
|
Dandawate P, Ghosh C, Palaniyandi K, Paul S, Rawal S, Pradhan R, Sayed AAA, Choudhury S, Standing D, Subramaniam D, Padhye S, Gunewardena S, Thomas SM, O’ Neil M, Tawfik O, Welch DR, Jensen RA, Maliski S, Weir S, Iwakuma T, Anant S, Dhar A. The Histone Demethylase KDM3A, Increased in Human Pancreatic Tumors, Regulates Expression of DCLK1 and Promotes Tumorigenesis in Mice. Gastroenterology 2019; 157:1646-1659.e11. [PMID: 31442435 PMCID: PMC6878178 DOI: 10.1053/j.gastro.2019.08.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 07/31/2019] [Accepted: 08/08/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS The histone lysine demethylase 3A (KDM3A) demethylates H3K9me1 and H3K9Me2 to increase gene transcription and is upregulated in tumors, including pancreatic tumors. We investigated its activities in pancreatic cancer cell lines and its regulation of the gene encoding doublecortin calmodulin-like kinase 1 (DCLK1), a marker of cancer stem cells. METHODS We knocked down KDM3A in MiaPaCa-2 and S2-007 pancreatic cancer cell lines and overexpressed KDM3A in HPNE cells (human noncancerous pancreatic ductal cell line); we evaluated cell migration, invasion, and spheroid formation under hypoxic and normoxic conditions. Nude mice were given orthotopic injections of S2-007 cells, with or without (control) knockdown of KDM3A, and HPNE cells, with or without (control) overexpression of KDM3A; tumor growth was assessed. We analyzed pancreatic tumor tissues from mice and pancreatic cancer cell lines by immunohistochemistry and immunoblotting. We performed RNA-sequencing analysis of MiaPaCa-2 and S2-007 cells with knockdown of KDM3A and evaluated localization of DCLK1 and KDM3A by immunofluorescence. We analyzed the cancer genome atlas for levels of KDM3A and DCLK1 messenger RNA in human pancreatic ductal adenocarcinoma (PDAC) tissues and association with patient survival time. RESULTS Levels of KDM3A were increased in human pancreatic tumor tissues and cell lines, compared with adjacent nontumor pancreatic tissues, such as islet and acinar cells. Knockdown of KDM3A in S2-007 cells significantly reduced colony formation, invasion, migration, and spheroid formation, compared with control cells, and slowed growth of orthotopic tumors in mice. We identified KDM3A-binding sites in the DCLK1 promoter; S2-007 cells with knockdown of KDM3A had reduced levels of DCLK1. HPNE cells that overexpressed KDM3A formed foci and spheres in culture and formed tumors and metastases in mice, whereas control HPNE cells did not. Hypoxia induced sphere formation and increased levels of KDM3A in S2-007 cells and in HPNE cells that overexpressed DCLK1, but not control HPNE cells. Levels of KDM3A and DCLK1 messenger RNA were higher in human PDAC than nontumor pancreatic tissues and correlated with shorter survival times of patients. CONCLUSIONS We found human PDAC samples and pancreatic cancer cell lines to overexpress KDM3A. KDM3A increases expression of DCLK1, and levels of both proteins are increased in human PDAC samples. Knockdown of KDM3A in pancreatic cancer cell lines reduced their invasive and sphere-forming activities in culture and formation of orthotopic tumors in mice. Hypoxia increased expression of KDM3A in pancreatic cancer cells. Strategies to disrupt this pathway might be developed for treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Prasad Dandawate
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Chandrayee Ghosh
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Kanagaraj Palaniyandi
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Santanu Paul
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Sonia Rawal
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Rohan Pradhan
- Interdisciplinary Science and Technology Research Academy, Abeda Inamdar Senior College, Camp, Pune 411001, India
| | - Afreen Asif Ali Sayed
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Sonali Choudhury
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - David Standing
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Dharmalingam Subramaniam
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Subhash Padhye
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA.,Interdisciplinary Science and Technology Research Academy, Abeda Inamdar Senior College, Camp, Pune 411001, India
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Sufi M. Thomas
- Department of Otolaryngology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Moura O’ Neil
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Ossama Tawfik
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Danny R. Welch
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Roy A. Jensen
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Sally Maliski
- School of Nursing, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Scott Weir
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Tomoo Iwakuma
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Shrikant Anant
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas.
| | - Animesh Dhar
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
55
|
Sandhu V, Labori KJ, Borgida A, Lungu I, Bartlett J, Hafezi-Bakhtiari S, Denroche RE, Jang GH, Pasternack D, Mbaabali F, Watson M, Wilson J, Kure EH, Gallinger S, Haibe-Kains B. Meta-Analysis of 1,200 Transcriptomic Profiles Identifies a Prognostic Model for Pancreatic Ductal Adenocarcinoma. JCO Clin Cancer Inform 2019; 3:1-16. [DOI: 10.1200/cci.18.00102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE With a dismal 8% median 5-year overall survival, pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy. Only 10% to 20% of patients are eligible for surgery, and more than 50% of these patients will die within 1 year of surgery. Building a molecular predictor of early death would enable the selection of patients with PDAC who are at high risk. MATERIALS AND METHODS We developed the Pancreatic Cancer Overall Survival Predictor (PCOSP), a prognostic model built from a unique set of 89 PDAC tumors in which gene expression was profiled using both microarray and sequencing platforms. We used a meta-analysis framework that was based on the binary gene pair method to create gene expression barcodes that were robust to biases arising from heterogeneous profiling platforms and batch effects. Leveraging the largest compendium of PDAC transcriptomic data sets to date, we show that PCOSP is a robust single-sample predictor of early death—1 year or less—after surgery in a subset of 823 samples with available transcriptomics and survival data. RESULTS The PCOSP model was strongly and significantly prognostic, with a meta-estimate of the area under the receiver operating curve of 0.70 ( P = 2.6E−22) and d-index (robust hazard ratio) of 1.9 (range, 1.6 to 2.3; ( = 1.4E−04) for binary and survival predictions, respectively. The prognostic value of PCOSP was independent of clinicopathologic parameters and molecular subtypes. Over-representation analysis of the PCOSP 2,619 gene pairs—1,070 unique genes—unveiled pathways associated with Hedgehog signaling, epithelial–mesenchymal transition, and extracellular matrix signaling. CONCLUSION PCOSP could improve treatment decisions by identifying patients who will not benefit from standard surgery/chemotherapy but who may benefit from a more aggressive treatment approach or enrollment in a clinical trial.
Collapse
Affiliation(s)
- Vandana Sandhu
- University Health Network, Toronto, Ontario, Canada
- Oslo University Hospital, Institute for Cancer Research, Oslo, Norway
| | | | | | - Ilinca Lungu
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - John Bartlett
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | | | - Gun Ho Jang
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | | | - Matthew Watson
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Julie Wilson
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Elin H. Kure
- Oslo University Hospital, Institute for Cancer Research, Oslo, Norway
- University of South-Eastern Norway, Bø in Telemark, Norway
| | - Steven Gallinger
- University Health Network, Toronto, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Benjamin Haibe-Kains
- University Health Network, Toronto, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
56
|
Misiura M, Zińczuk J, Zaręba K, Kamińska D, Guzińska-Ustymowicz K, Pryczynicz A. Actin-Bundling Proteins (Actinin-4 and Fascin-1) are Involved in the Development of Pancreatic Intraepithelial Neoplasia (PanIN). Am J Med Sci 2019; 359:147-155. [PMID: 31889512 DOI: 10.1016/j.amjms.2019.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/09/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Fascin-1 and actinin-4 are involved in key processes of tumor cell adhesion, migration and metastasis. Actinin-4 plays an important role in promotion of cell proliferation, whereas fascin-1 regulates cellular motility. Its over-expression leads to the loss of cell adhesion and metastasis. The aim of our study was to assess fascin-1 and actinin-4 expression in normal pancreatic ducts and in pancreatic intraepithelial neoplasia (PanIN) - precursor lesion of pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS The study involved 70 patients treated surgically due to PDAC, cysts and pancreatitis, who had also been diagnosed with pancreatic intraepithelial neoplasia. Fascin-1 and actinin-4 expressions were evaluated using the immunohistochemistry method. RESULTS A statistically significant relationship was observed between the expression of fascin-1 and actinin-4 (cytoplasmic) and patients' age (P = 0.01, P = 0.002, respectively). The expression of fascin-1 and actinin-4 was associated with the diagnosis (P <0.001, P = 0.04, respectively). Statistical analysis revealed correlations of fascin-1 and actinin-4 expressions with the presence and grade of PanIN (P < 0.001, P = 0.002, respectively). The expression of these proteins was observed in each degree of PanIN and increased with the pancreatic intraepithelial neoplasia progression. CONCLUSIONS The expression of fascin-1 and actinin-4 is connected with the degree of PanIN advancement and depends on the type of the primary disease. Overexpression of these proteins may be linked to cytological and architectural abnormalities observed in advanced PanIN. Elevated expression of fascin-1 and actinin-4 indicates the role of these proteins in the progression from PanIN to PDAC.
Collapse
Affiliation(s)
| | | | - Konrad Zaręba
- 2nd Clinical Department of General and Gastroenterological Surgery, Medical University of Bialystok, Bialystok, Poland
| | - Dorota Kamińska
- Department of Laboratory Diagnostics, Independent Public Health Care Unit of the Provincial Hospital Jędrzej Śniadecki in Bialystok, Bialystok, Poland
| | | | - Anna Pryczynicz
- Department of General Pathomorphology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
57
|
Ferencz S, Reglodi D, Kaszas B, Bardosi A, Toth D, Vekony Z, Vicena V, Karadi O, Kelemen D. PACAP and PAC1 receptor expression in pancreatic ductal carcinoma. Oncol Lett 2019; 18:5725-5730. [PMID: 31788045 PMCID: PMC6865831 DOI: 10.3892/ol.2019.10971] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/12/2019] [Indexed: 12/26/2022] Open
Abstract
Pancreatic carcinoma is one of the most malignant diseases and is associated with a poor survival rate. Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuropeptide that acts on three different G protein-coupled receptors: the specific PAC1 and the VPAC1/2 that also bind vasoactive intestinal peptide. PACAP is widely distributed in the body and has diverse physiological effects. Among other things, it acts as a trophic factor and influences proliferation and differentiation of several different cells both under normal circumstances and tumourous transformation. Changes of PACAP and its receptors have been shown in various tumour types. However, it is not known whether PACAP and its specific receptor are altered in pancreatic cancer. Perioperative data of patients with pancreas carcinoma was investigated over a five-year period. Histological results showed Grade 2 or Grade 3 adenocarcinoma in most cases. PACAP and PAC1 receptor expression were investigated by immunohistochemistry. Staining intensity of PAC1 receptor was strong in normal tissues both in the exocrine and endocrine parts of the pancreas, the receptor staining was markedly weaker in the adenocarcinoma. PACAP immunostaining was weak in the exocrine part and very strong in the islets and nerve elements in non-tumourous tissues. The PACAP immunostaining almost disappeared in the adenocarcinoma samples. Based on these findings a decrease or lack of the PAC1 receptor/PACAP signalling might have an influence on tumour growth and/or differentiation.
Collapse
Affiliation(s)
- Sandor Ferencz
- Department of Surgery, University of Pécs, Medical School, Pécs 7622, Hungary
| | - Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pécs, Medical School, Pécs 7622, Hungary
| | - Balint Kaszas
- Department of Pathology, University of Pécs, Medical School, Pécs 7622, Hungary
| | - Attila Bardosi
- Center for Histology, Cytology and Molecular Diagnostics, and Proteopath GmbH, Trier 54296, Germany
| | - Denes Toth
- Department of Forensic Medicine, University of Pécs, Medical School, Pécs 7622, Hungary
| | - Zsofia Vekony
- Department of Surgery, University of Pécs, Medical School, Pécs 7622, Hungary
| | - Viktoria Vicena
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pécs, Medical School, Pécs 7622, Hungary
| | - Oszkar Karadi
- Department of Oncology, University of Pécs, Medical School, Pécs 7622, Hungary
| | - Dezso Kelemen
- Department of Surgery, University of Pécs, Medical School, Pécs 7622, Hungary
| |
Collapse
|
58
|
Zhang L, Huang P, Li Q, Wang D, Xu CX. miR-134-5p Promotes Stage I Lung Adenocarcinoma Metastasis and Chemoresistance by Targeting DAB2. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:627-637. [PMID: 31689617 PMCID: PMC6838973 DOI: 10.1016/j.omtn.2019.09.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/15/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023]
Abstract
Despite surgery and adjuvant therapy, early-stage lung adenocarcinoma (LUAD) treatment often fails due to local or metastatic recurrence. However, the mechanism is largely unknown. Here, we report that increased expression levels of miR-134-5p and decreased levels of disabled-2 (DAB2) were significantly correlated with recurrence in stage I LUAD patients. Our data show that miR-134-5p overexpression or DAB2 silencing strongly stimulated LUAD cell metastasis and chemoresistance. In contrast, inhibition of miR-134-5p or overexpression of DAB2 strongly suppressed LUAD cell metastasis and overcame the insensitivity of chemoresistant LUAD cells to chemotherapy. In addition, we demonstrated that DAB2 is a target of miR-134-5p and that miR-134-5p stimulates chemoresistance and metastasis through DAB2 in LUAD. Taken together, these findings suggest that miR-134-5p and its target gene DAB2 have potential as a biomarker for predicting recurrence in stage I LUAD patients. Additionally, miR-134-5p inhibition or DAB2 restoration may be a novel strategy for inhibiting LUAD metastasis and overcoming LUAD cell resistance to chemotherapy.
Collapse
Affiliation(s)
- Liang Zhang
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Ping Huang
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Qing Li
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Dong Wang
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China.
| | - Cheng-Xiong Xu
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China.
| |
Collapse
|
59
|
Winer LK, Dhar VK, Wima K, Morris MC, Lee TC, Shah SA, Ahmad SA, Patel SH. The Impact of Tumor Location on Resection and Survival for Pancreatic Ductal Adenocarcinoma. J Surg Res 2019; 239:60-66. [DOI: 10.1016/j.jss.2019.01.061] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 01/16/2019] [Accepted: 01/25/2019] [Indexed: 12/11/2022]
|
60
|
Fang C, Guo X, Lv X, Yin R, Lv X, Wang F, Zhao J, Bai Q, Yao X, Chen Y. Dysbindin promotes progression of pancreatic ductal adenocarcinoma via direct activation of PI3K. J Mol Cell Biol 2019; 9:504-515. [PMID: 29040676 DOI: 10.1093/jmcb/mjx043] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/05/2017] [Indexed: 12/26/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents a biggest challenge in clinic oncology due to its invasiveness and lack of targeted therapeutics. Our recent study showed that schizophrenia susceptibility factor dysbindin exhibited significant higher level in serum of PDAC patients. However, the functional relevance of dysbindin in PDAC is still unclear. Here, we show that dysbindin promotes tumor growth both in vitro and in vivo by accelerating the G1/S phase transition in cell cycle via PI3K/AKT signaling pathway. Mechanistically, dysbindin interacts with PI3K and stimulates the kinase activity of PI3K. Moreover, overexpression of dysbindin in PDAC is correlated with clinicopathological characteristics significantly, such as histological differentiation (P = 0.011) and tumor size (P = 0.007). Kaplan-Meier survival curves show that patients with high dysbindin expression exhibit poorer overall survival, compared to those with low dysbindin expression (P < 0.001). Multivariate analysis reveals that dysbindin is an independent prognostic factor for pancreatic ductal adenocarcinoma (P = 0.001). Thus, our findings reveal that dysbindin is a novel PI3K activator and promotes PDAC progression via stimulation of PI3K/AKT. Dysbindin therefore represents a potential target for prognosis and therapy of PDAC.
Collapse
Affiliation(s)
- Cheng Fang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xin Guo
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xing Lv
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ruozhe Yin
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaohui Lv
- Department of Gynecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fengsong Wang
- Department of Biology, School of Life Science, Anhui Medical University, Hefei, China
| | - Jun Zhao
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Quan Bai
- Institute of Modern Separation Science, College of Chemistry & Materials Science, Northwest University, Xi'an, China
| | - Xuebiao Yao
- Department of Hefei Laboratory for Physical Sciences at Microscale, School of Life Science, University of Science and Technology of China, Hefei, China
| | - Yong Chen
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
61
|
Giampieri R, Piva F, Occhipinti G, Bittoni A, Righetti A, Pagliaretta S, Murrone A, Bianchi F, Amantini C, Giulietti M, Ricci G, Principato G, Santoni G, Berardi R, Cascinu S. Clinical impact of different exosomes' protein expression in pancreatic ductal carcinoma patients treated with standard first line palliative chemotherapy. PLoS One 2019; 14:e0215990. [PMID: 31048929 PMCID: PMC6497273 DOI: 10.1371/journal.pone.0215990] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/11/2019] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma is associated to dismal prognosis despite the use of palliative chemotherapy, partly due to the lack of knowledge of biological processes underlying disease progression. Exosomes have been identified as biomarkers sources in different cancer types. Aim of the study was to analyse the contents of circulating exosomes in patients with pancreatic cancer who received palliative chemotherapy. PATIENTS AND METHODS Patients were submitted to blood sample collection before chemotherapy (T0) and after 3 months (T3). We quantified by an ELISA-based technique specific proteins of cancer-derived exosomes (CD44,CD44v6,EpCAM,CD9,CD81,Tspan8,Integrin α6,Integrin β4,CD24,CXCR4). We correlated the baseline levels of these factors and changes between T3 and T0 and survival outcomes. Survival analyses were performed by Kaplan-Meier method. Correlation was assessed by log-rank test and level of statistical significance was set at 0.05. Multivariate analysis was performed by logistic regression analysis. RESULTS Nineteen patients were enrolled. EpCAM T0 levels and increased EpCAM levels from T0 to T3 were those mostly associated with differences in survival. Patients having higher EpCAM had median progression free survival (PFS) of 3.18vs7.31 months (HR:2.82,95%CI:1.03-7.73,p = 0.01). Overall survival (OS) was shorter for patients having higher EpCAM (5.83vs16.45 months,HR:6.16,95%CI:1.93-19.58,p = 0.0001) and also response rates (RR) were worse (20%vs87%,p = 0.015). EpCAM increase during treatment was associated with better median PFS (2.88vs7.31 months,HR:0.24,95%CI:0.04-1.22,p = 0.003). OS was also better (8.75vs11.04 months, HR:0.77,95%CI:0.21-2.73,p = 0.66) and RR were 60%vs20% (p = 0.28). Among clinical factors that might determine changes on PFS and OS, only ECOG PS was associated to significantly worse PFS and OS (p = 0.0137and<0.001 respectively).Multivariate analysis confirmed EpCAM T0 levels and EpCAM T0/T3 changes as independent prognostic factors for PFS. CONCLUSIONS Pancreatic cancer patients exosomes express EpCAM, whose levels change during treatment. This represents a useful prognostic factor and also suggests that future treatment modalities who target EpCAM should be tested in pancreatic cancer patients selected by exosome EpCAM expression.
Collapse
Affiliation(s)
- Riccardo Giampieri
- Oncologia Clinica c/o Università Politecnica delle Marche, Dipartimento Scienze Cliniche e Molecolari – Azienda Ospedaliera Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Francesco Piva
- Biologia e biochimica c/o Università Politecnica delle Marche, Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche – Azienda Ospedaliera Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Giulia Occhipinti
- Biologia e biochimica c/o Università Politecnica delle Marche, Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche – Azienda Ospedaliera Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Alessandro Bittoni
- Oncologia Clinica c/o Università Politecnica delle Marche, Dipartimento Scienze Cliniche e Molecolari – Azienda Ospedaliera Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Alessandra Righetti
- Biologia e biochimica c/o Università Politecnica delle Marche, Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche – Azienda Ospedaliera Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Silvia Pagliaretta
- Oncologia Clinica c/o Università Politecnica delle Marche, Dipartimento Scienze Cliniche e Molecolari – Azienda Ospedaliera Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Alberto Murrone
- Oncologia Clinica c/o Università Politecnica delle Marche, Dipartimento Scienze Cliniche e Molecolari – Azienda Ospedaliera Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Francesca Bianchi
- Oncologia Clinica c/o Università Politecnica delle Marche, Dipartimento Scienze Cliniche e Molecolari – Azienda Ospedaliera Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | | | | | - Giulia Ricci
- Oncologia Clinica c/o Università Politecnica delle Marche, Dipartimento Scienze Cliniche e Molecolari – Azienda Ospedaliera Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Giovanni Principato
- Biologia e biochimica c/o Università Politecnica delle Marche, Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche – Azienda Ospedaliera Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | | | - Rossana Berardi
- Oncologia Clinica c/o Università Politecnica delle Marche, Dipartimento Scienze Cliniche e Molecolari – Azienda Ospedaliera Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Stefano Cascinu
- Dipartimento Onco-ematologia Ospedale Universitario di Modena, Università di Modena e Reggio Emilia, Modena, Italy
| |
Collapse
|
62
|
Dai FQ, Li CR, Fan XQ, Tan L, Wang RT, Jin H. miR-150-5p Inhibits Non-Small-Cell Lung Cancer Metastasis and Recurrence by Targeting HMGA2 and β-Catenin Signaling. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 16:675-685. [PMID: 31121479 PMCID: PMC6529773 DOI: 10.1016/j.omtn.2019.04.017] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 01/23/2023]
Abstract
Dysregulated microRNAs (miRNAs) play crucial roles in the regulation of cancer stem cells (CSCs), and CSCs are closely associated with tumor initiation, metastasis, and recurrence. Here we found that miR-150-5p was significantly downregulated in CSCs of non-small-cell lung cancer (NSCLC) and its expression level was negatively correlated with disease progression and poor survival in patients with NSCLC. Inhibition of miR-150-5p increased the CSC population and sphere formation of NSCLC cells in vitro and stimulated NSCLC cell tumorigenicity and metastatic colonization in vivo. In contrast, miR-150-5p overexpression potently inhibited sphere-formed NSCLC cell tumor formation, metastatic colonization, and recurrence in xenograft models. Furthermore, we identified that miR-150-5p significantly inhibited wingless (Wnt)-β-catenin signaling by simultaneously targeting glycogen synthase kinase 3 beta interacting protein (GSKIP) and β-catenin in NSCLC cells. miR-150-5p also targeted high mobility group AT-hook 2 (HMGA2), another regulator of CSCs, and Wnt-β-catenin signaling. The restoration of HMGA2 and β-catenin blocked miR-150-5p overexpression-induced inhibition of CSC traits in NSCLC cells. These findings suggest that miR-150-5p functions as a CSC suppressor and that overexpression of miR-150-5p may be a novel strategy to inhibit CSC-induced metastasis and recurrence in NSCLC.
Collapse
Affiliation(s)
- Fu-Qiang Dai
- Department of Thoracic Surgery, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Cheng-Run Li
- Department of Thoracic Surgery, The General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - Xiao-Qing Fan
- Department of Thoracic Surgery, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Long Tan
- Department of Thoracic Surgery, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Ren-Tao Wang
- Department of Respiratory, The General Hospital of Chinese People's Liberation Army, Beijing 100853, China.
| | - Hua Jin
- Department of Thoracic Surgery, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China.
| |
Collapse
|
63
|
Overexpression of BUB1B, CCNA2, CDC20, and CDK1 in tumor tissues predicts poor survival in pancreatic ductal adenocarcinoma. Biosci Rep 2019; 39:BSR20182306. [PMID: 30765611 PMCID: PMC6390130 DOI: 10.1042/bsr20182306] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 01/29/2019] [Accepted: 02/04/2019] [Indexed: 01/28/2023] Open
Abstract
Overexpressed genes in tumors usually contributed to aggressiveness in pancreatic ductal adenocarcinoma (PDAC). Using Gene Expression Omnibus (GEO) profiles including GSE46234, GSE71989, and GSE107610, we detected overexpressed genes in tumors with R program, which were enriched by Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene ontology (GO), and Reactome pathway databases. Then, we performed a survival analysis of enriched genes based on TCGA profile. Our results revealed that high BUB1B, CCNA2, CDC20, and CDK1 expression in tumors was significantly associated with worse overall survival (OS) (Log rank P=0.00338, P=0.0447, P=0.00965, and P=0.00479, respectively), which was validated using a Kaplan–Meier plotter with a median cutoff (Log rank P=0.028, P=0.0035, P=0.039, and P=0.0033, respectively). Moreover, overexpression of BUB1B, CCNA2, CDC20, and CDK1 in tumor tissues was significantly associated with disease-free survival (DFS) in PDAC patients (Log rank P=0.00565, P=0.0357, P=0.00104, and P=0.00121, respectively). BUB1B, CCNA2, CDC20, and CDK1 were significantly overexpressed in deceased PDAC patients (all P<0.01) and in patients with recurrence/disease progression (all P<0.05). In addition, PDAC patients with neoplasms of histologic grade G3-4 had significantly higher BUB1B, CCNA2 and CDC20 levels (all P<0.05). In conclusion, the up-regulation of BUB1B, CCNA2, CDC20, CDK1, and WEE1 in tumor tissues are associated with worse OS and DFS in PDAC and is correlated with advanced tumor stage and tumor development.
Collapse
|
64
|
NAPOLI-1 phase 3 study of liposomal irinotecan in metastatic pancreatic cancer: Final overall survival analysis and characteristics of long-term survivors. Eur J Cancer 2019; 108:78-87. [PMID: 30654298 DOI: 10.1016/j.ejca.2018.12.007] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/06/2018] [Accepted: 12/11/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Liposomal irinotecan (nal-IRI) plus 5-fluorouracil and leucovorin (5-FU/LV) is approved for patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) previously treated with gemcitabine-based therapy. This approval was based on significantly improved median overall survival compared with 5-FU/LV alone (6.1 vs 4.2 months; hazard ratio [HR], 0.67) in the global phase 3 NAPOLI-1 trial. Here, we report the final survival analysis and baseline characteristics associated with long-term survivors (survival of ≥1 year) in the NAPOLI-1 trial. PATIENTS AND METHODS Patients with mPDAC were randomised to receive nal-IRI + 5-FU/LV (n = 117), nal-IRI (n = 151), or 5-FU/LV (n = 149) for the first 4 weeks of 6-week cycles. Baseline characteristics and efficacy in the overall population were compared with those in patients who survived ≥1 year. Through 16th November 2015, 382 overall survival events had occurred. RESULTS The overall survival advantage for nal-IRI+5-FU/LV vs 5-FU/LV was maintained from the original nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1) analysis (6.2 vs 4.2 months, respectively; HR, 0.75; 95% confidence interval: 0.57-0.99). Median progression-free survival, objective response rate and disease control rate also favoured nal-IRI+5-FU/LV therapy. Estimated one-year overall survival rates were 26% with nal-IRI+5-FU/LV and 16% with 5-FU/LV. Baseline characteristics associated with long-term survival in the nal-IRI+5-FU/LV arm were Karnofsky performance status ≥90, age ≤65 years, lower CA19-9 levels, neutrophil-to-lymphocyte ratio ≤5 and no liver metastases. No new safety concerns were detected. CONCLUSIONS The survival benefits of nal-IRI+5-FU/LV versus 5-FU/LV were maintained over an extended follow-up, and prognostic markers of survival ≥1 year were identified. CLINICAL TRIAL REGISTRATION NUMBER NCT01494506.
Collapse
|
65
|
DWI of Pancreatic Ductal Adenocarcinoma: A Pilot Study to Estimate the Correlation With Metastatic Disease Potential and Overall Survival. AJR Am J Roentgenol 2018; 212:323-331. [PMID: 30667305 DOI: 10.2214/ajr.18.20017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The purpose of this study is to analyze the relationship between the apparent diffusion coefficient (ADC) of pancreatic ductal adenocarcinoma (PDAC) and the presence or development of metastasis and overall survival (OS). MATERIALS AND METHODS Of 290 consecutive patients with histopathologically proven PDAC from January 2013 to December 2014, staging DWI was performed for 124 patients. Image quality was adequate in 112 studies. Sixty-five patients were treatment naïve, but 17 of the 65 were excluded because of the presence of other associated pancreatic pathologic abnormalities. Data for the remaining 48 patients (24 men and 24 women; median age, 65.5 years; interquartile range, 56-77 years) were obtained during a 4-year follow-up period (mean [± SD], 397 ± 415.1 days). The correlation between ADC and the presence or development of metastasis was assessed using descriptive statistics. OS was determined and mortality analysis was performed using Pearson correlation and Kaplan-Meier curves. RESULTS Of 48 patients, 10 had metastases at staging MRI, and 12 later developed metastatic disease. Among the latter, the mean time from staging MRI to metastasis was 258 ± 274.1 days. Most (86%) metastases were hepatic (n = 19). During the follow-up period, the remaining 26 patients (54%) never developed metastases. Patients with metastatic disease (n = 22) had significantly lower mean ADCs than did those without metastases (1.27 × 10-3 vs 1.43 × 10-3 mm2/s; p = 0.047). The ADC of PDAC had a positive correlation with survival: patients with PDAC with lower ADCs (< 1.36 × 10-3 mm2/s) had significantly worse 4-year OS rates than did patients with higher ADC values (p = 0.036). CONCLUSION Pretreatment ADC values of PDAC may be significantly lower in patients who have or will develop metastatic disease and may correlate with worse OS.
Collapse
|
66
|
Hang J, Wu L, Zhu L, Sun Z, Wang G, Pan J, Zheng S, Xu K, Du J, Jiang H. Prediction of overall survival for metastatic pancreatic cancer: Development and validation of a prognostic nomogram with data from open clinical trial and real-world study. Cancer Med 2018; 7:2974-2984. [PMID: 29856121 PMCID: PMC6051216 DOI: 10.1002/cam4.1573] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/29/2018] [Accepted: 05/01/2018] [Indexed: 12/20/2022] Open
Abstract
It is necessary to develop prognostic tools of metastatic pancreatic cancer (MPC) for optimizing therapeutic strategies. Thus, we tried to develop and validate a prognostic nomogram of MPC. Data from 3 clinical trials (NCT00844649, NCT01124786, and NCT00574275) and 133 Chinese MPC patients were used for analysis. The former 2 trials were taken as the training cohort while NCT00574275 was used as the validation cohort. In addition, 133 MPC patients treated in China were taken as the testing cohort. Cox regression model was used to investigate prognostic factors in the training cohort. With these factors, we established a nomogram and verified it by Harrell's concordance index (C‐index) and calibration plots. Furthermore, the nomogram was externally validated in the validation cohort and testing cohort. In the training cohort (n = 445), performance status, liver metastasis, Carbohydrate antigen 19‐9 (CA19‐9) log‐value, absolute neutrophil count (ANC), and albumin were independent prognostic factors for overall survival (OS). A nomogram was established with these factors to predict OS and survival probabilities. The nomogram showed an acceptable discrimination ability (C‐index: .683) and good calibration, and was further externally validated in the validation cohort (n = 273, C‐index: .699) and testing cohort (n = 133, C‐index: .653).The nomogram total points (NTP) had the potential to stratify patients into 3‐risk groups with median OS of 11.7, 7.0 and 3.7 months (P < .001), respectively. In conclusion, the prognostic nomogram with NTP can predict OS for patients with MPC with considerable accuracy.
Collapse
Affiliation(s)
- Junjie Hang
- Department of Oncology, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Lixia Wu
- Department of Oncology, Shanghai JingAn District ZhaBei Central Hospital, Shanghai, China
| | - Lina Zhu
- Department of Oncology, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Zhiqiang Sun
- Department of Oncology, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Ge Wang
- Department of Oncology, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Jingjing Pan
- Department of Oncology, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Suhua Zheng
- Department of Oncology, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Kequn Xu
- Department of Oncology, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Jiadi Du
- Center of Data Mining and Business Analytics, Rutgers Business School, Newark, NJ, USA
| | - Hua Jiang
- Department of Oncology, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
67
|
Löhr JM, Kordes M, Rutkowski W, Heuchel R, Gustafsson-Liljefors M, Russom A, Nilsson M. Overcoming diagnostic issues in precision treatment of pancreatic cancer. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2018. [DOI: 10.1080/23808993.2018.1476061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- J.-Matthias Löhr
- Department of Cancer Medicine, Division for Upper GI, Karolinska University Hospital, Stockholm, Sweden
- CLINTEC, Karolinska Institutet, Science for Life Laboratory, Stockholm, Sweden
| | - Maximilian Kordes
- Department of Cancer Medicine, Division for Upper GI, Karolinska University Hospital, Stockholm, Sweden
- CLINTEC, Karolinska Institutet, Science for Life Laboratory, Stockholm, Sweden
| | - Wiktor Rutkowski
- CLINTEC, Karolinska Institutet, Science for Life Laboratory, Stockholm, Sweden
| | - Rainer Heuchel
- CLINTEC, Karolinska Institutet, Science for Life Laboratory, Stockholm, Sweden
| | | | | | | |
Collapse
|
68
|
Kim MS, Jeon TJ, Park JY, Choi J, Shin WC, Park SE, Seo JY, Kim YM. Clinical Interpretation of Elevated CA 19-9 Levels in Obstructive Jaundice Following Benign and Malignant Pancreatobiliary Disease. THE KOREAN JOURNAL OF GASTROENTEROLOGY 2018; 70:96-102. [PMID: 28830135 DOI: 10.4166/kjg.2017.70.2.96] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background/Aims Elevated carbohydrate antigen (CA) 19-9 level may be unable to differentiate between benign and malignant pancreatobiliary disease with obstructive jaundice. The study aims to determine the clinical interpretation and the diagnostic value of CA 19-9 level in pancreatobiliary diseases with coexistent obstructive jaundice. Methods We retrospectively reviewed the data of 981 patients who underwent biliary drainage due to obstructive jaundice following pancreatobiliary disease at Sanggye Paik Hospital for 5 years. 114 patients with serial follow-up data for CA 19-9 level were included in this study (80 patients with malignancy and 34 patients with benign diseases). We compared the levels of CA 19-9 levels and the biochemical value before and after biliary drainage. Results The rate of CA 19-9 elevation (>37 U/mL) was significantly different between the benign group and the malignant group (59% vs. 90%, p=0.001). Despite the decrease in serum bilirubin after biliary drainage, CA 19-9 levels remained elevated in 12% of patients in the benign group and in 63% of patients in the malignant group (p<0.001). Finally, 12% of patients in the benign group turned out to have malignant disease. A receiver operating characteristic analysis provided a cut-off value of 38 U/mL for differentiating benign disease from malignant disease after biliary drainage (area under curve, 0.787; 95% confidence interval, 0.703 to 0.871; sensitivity, 62%; specificity, 88%). Conclusions This study suggested that we should consider the possibility of malignant causes if the CA 19-9 levels remain high or are more than 38 U/mL after resolution of biliary obstruction.
Collapse
Affiliation(s)
- Min Seong Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Tae Joo Jeon
- Division of Gastroenterology, Department of Internal Medicine, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Ji Young Park
- Division of Gastroenterology, Department of Internal Medicine, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Jeongmin Choi
- Division of Gastroenterology, Department of Internal Medicine, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Won Chang Shin
- Division of Gastroenterology, Department of Internal Medicine, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Seong Eun Park
- Division of Gastroenterology, Department of Internal Medicine, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Ji Young Seo
- Division of Gastroenterology, Department of Internal Medicine, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Young Moon Kim
- Division of Gastroenterology, Department of Internal Medicine, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| |
Collapse
|
69
|
Murthy D, Attri KS, Singh PK. Phosphoinositide 3-Kinase Signaling Pathway in Pancreatic Ductal Adenocarcinoma Progression, Pathogenesis, and Therapeutics. Front Physiol 2018; 9:335. [PMID: 29670543 PMCID: PMC5893816 DOI: 10.3389/fphys.2018.00335] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/19/2018] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy characterized by its sudden manifestation, rapid progression, poor prognosis, and limited therapeutic options. Genetic alterations in key signaling pathways found in early pancreatic lesions are pivotal for the development and progression of pancreatic intraepithelial neoplastic lesions into invasive carcinomas. More than 90% of PDAC tumors harbor driver mutations in K-Ras that activate various downstream effector-signaling pathways, including the phosphoinositide-3-kinase (PI3K) pathway. The PI3K pathway also responds to stimuli from various growth factor receptors present on the cancer cell surface that, in turn, modulate downstream signaling cascades. Thus, the inositide signaling acts as a central node in the complex cellular signaling networks to impact cancer cell growth, motility, metabolism, and survival. Also, recent publications highlight the importance of PI3K signaling in stromal cells, whereby PI3K signaling modifies the tumor microenvironment to dictate disease outcome. The high incidence of mutations in the PI3K signaling cascade, accompanied by activation of parallel signaling pathways, makes PI3K a promising candidate for drug therapy. In this review, we describe the role of PI3K signaling in pancreatic cancer development and progression. We also discuss the crosstalk between PI3K and other major cellular signaling cascades, and potential therapeutic opportunities for targeting pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Divya Murthy
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kuldeep S Attri
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Pankaj K Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
70
|
Garcia-Cremades M, Pitou C, Iversen PW, Troconiz IF. Predicting tumour growth and its impact on survival in gemcitabine-treated patients with advanced pancreatic cancer. Eur J Pharm Sci 2018; 115:296-303. [PMID: 29366960 DOI: 10.1016/j.ejps.2018.01.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 12/17/2022]
Abstract
The aim of this evaluation was to characterize the impact of the tumour size (TS) effects driven by the anticancer drug gemcitabine on overall survival (OS) in patients with advanced pancreatic cancer by building and validating a predictive semi-mechanistic joint TS-OS model. TS and OS data were obtained from one phase II and one phase III study where gemcitabine was administered (1000-1250 mg/kg over 30-60 min i.v infusion) as single agent to patients (n = 285) with advanced pancreatic cancer. Drug exposure, TS and OS were linked using the population approach with NONMEM 7.3. Pancreatic tumour progression was characterized by exponential growth (doubling time = 67 weeks), and tumour response to treatment was described as a function of the weekly area under the gemcitabine triphosphate concentration vs time curve (AUC), including treatment-related resistance development. The typical predicted percentage of tumour growth inhibition with respect to no treatment was 22.3% at the end of 6 chemotherapy cycles. Emerging resistance elicited a 57% decrease in drug effects during the 6th chemotherapy cycle. Predicted TS profile was identified as main prognostic factor of OS, with tumours responders' profiles improving median OS by 30 weeks compared to stable-disease TS profiles. Results of NCT00574275 trial were predicted using this modelling framework, thereby validating the approach as a prediction tool in clinical development. Our analyses show that despite the advanced stage of the disease in this patient population, the modelling framework herein can be used to predict the likelihood of treatment success using early clinical data.
Collapse
Affiliation(s)
- Maria Garcia-Cremades
- Pharmacometrics and Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), University of Navarra, Pamplona, Spain.
| | - Celine Pitou
- Global Pharmacokinetic/Pharmacodynamics and Pharmacometrics, Eli Lilly and Company Windlesham, Surrey, United Kingdom.
| | - Philip W Iversen
- Lilly Research laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA.
| | - Iñaki F Troconiz
- Pharmacometrics and Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), University of Navarra, Pamplona, Spain.
| |
Collapse
|
71
|
Nishida S, Ishikawa T, Egawa S, Koido S, Yanagimoto H, Ishii J, Kanno Y, Kokura S, Yasuda H, Oba MS, Sato M, Morimoto S, Fujiki F, Eguchi H, Nagano H, Kumanogoh A, Unno M, Kon M, Shimada H, Ito K, Homma S, Oka Y, Morita S, Sugiyama H. Combination Gemcitabine and WT1 Peptide Vaccination Improves Progression-Free Survival in Advanced Pancreatic Ductal Adenocarcinoma: A Phase II Randomized Study. Cancer Immunol Res 2018; 6:320-331. [PMID: 29358173 DOI: 10.1158/2326-6066.cir-17-0386] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/17/2017] [Accepted: 01/09/2018] [Indexed: 11/16/2022]
Abstract
We investigated the efficacy of a Wilms' tumor gene 1 (WT1) vaccine combined with gemcitabine (GEMWT1) and compared it with gemcitabine (GEM) monotherapy for advanced pancreatic ductal adenocarcinoma (PDAC) in a randomized phase II study. We randomly assigned HLA-A*02:01- or HLA-A*24:02-positive patients with advanced PDAC to receive GEMWT1 or GEM. We assessed WT1-specific immune responses via delayed-type hypersensitivity (DTH) to the WT1 peptide and a tetramer assay to detect WT1-specific cytotoxic T lymphocytes (WT1-CTL). Of 91 patients enrolled, 85 were evaluable (GEMWT1: n = 42; GEM: n = 43). GEMWT1 prolonged progression-free survival [PFS; hazard ratio (HR), 0.66; P = 0.084] and improved overall survival rate at 1 year (1-year OS%; GEMWT1: 35.7%; GEM: 20.9%). However, the difference in OS was not significant (HR: 0.82; P = 0.363). These effects were particularly evident in metastatic PDAC (PFS: HR 0.51, P = 0.0017; 1-year OS%: GEMWT1 27.3%; GEM 11.8%). The combination was well tolerated, with no unexpected serious adverse events. In patients with metastatic PDAC, PFS in the DTH-positive GEMWT1 group was significantly prolonged, with a better HR of 0.27 compared with the GEM group, whereas PFS in the DTH-negative GEMWT1 group was similar to that in the GEM group (HR 0.86; P = 0.001). DTH positivity was associated with an increase in WT1-CTLs induced by the WT1 vaccine. GEM plus the WT1 vaccine prolonged PFS and may improve 1-year OS% in advanced PDAC. These clinical effects were associated with the induction of WT1-specific immune responses. Cancer Immunol Res; 6(3); 320-31. ©2018 AACR.
Collapse
Affiliation(s)
- Sumiyuki Nishida
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Takeshi Ishikawa
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shinichi Egawa
- Division of International Cooperation for Disaster Medicine, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - Shigeo Koido
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Jun Ishii
- Division of General and Gastroenterological Surgery, Department of Surgery, Toho University Faculty of Medicine, Tokyo, Japan
| | - Yoshihide Kanno
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan
| | - Satoshi Kokura
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroaki Yasuda
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mari Saito Oba
- Department of Biostatistics, Yokohama City University, Yokohama, Japan
| | - Maho Sato
- Department of Biostatistics, Yokohama City University, Yokohama, Japan
| | - Soyoko Morimoto
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Fumihiro Fujiki
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan.,Department of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masanori Kon
- Department of Surgery, Kansai Medical University, Hirakata, Japan
| | - Hideaki Shimada
- Division of General and Gastroenterological Surgery, Department of Surgery, Toho University Faculty of Medicine, Tokyo, Japan
| | - Kei Ito
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan
| | - Sadamu Homma
- Division of Oncology, Research Center for Medical Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshihiro Oka
- Department of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
72
|
Chakraborty A, Dorsett KA, Trummell HQ, Yang ES, Oliver PG, Bonner JA, Buchsbaum DJ, Bellis SL. ST6Gal-I sialyltransferase promotes chemoresistance in pancreatic ductal adenocarcinoma by abrogating gemcitabine-mediated DNA damage. J Biol Chem 2018; 293:984-994. [PMID: 29191829 PMCID: PMC5777269 DOI: 10.1074/jbc.m117.808584] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/14/2017] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with a poor prognosis. Gemcitabine, as a single agent or in combination therapy, remains the frontline chemotherapy despite its limited efficacy due to de novo or acquired chemoresistance. There is an acute need to decipher mechanisms underlying chemoresistance and identify new targets to improve patient outcomes. Here, we report a novel role for the ST6Gal-I sialyltransferase in gemcitabine resistance. Utilizing MiaPaCa-2 and BxPC-3 PDAC cells, we found that knockdown (KD) of ST6Gal-I expression, as well as removal of surface α2-6 sialic acids by neuraminidase, enhances gemcitabine-mediated cell death assessed via clonogenic assays and cleaved caspase 3 expression. Additionally, KD of ST6Gal-I potentiates gemcitabine-induced DNA damage as measured by comet assays and quantification of γH2AX foci. ST6Gal-I KD also alters mRNA expression of key gemcitabine metabolic genes, RRM1, RRM2, hENT1, and DCK, leading to an increased gemcitabine sensitivity ratio, an indicator of gemcitabine toxicity. Gemcitabine-resistant MiaPaCa-2 cells display higher ST6Gal-I levels than treatment-naïve cells along with a reduced gemcitabine sensitivity ratio, suggesting that chronic chemotherapy selects for clonal variants with more abundant ST6Gal-I. Finally, we examined Suit2 PDAC cells and Suit2 derivatives with enhanced metastatic potential. Intriguingly, three metastatic and chemoresistant subclones, S2-CP9, S2-LM7AA, and S2-013, exhibit up-regulated ST6Gal-I relative to parental Suit2 cells. ST6Gal-I KD in S2-013 cells increases gemcitabine-mediated DNA damage, indicating that suppressing ST6Gal-I activity sensitizes inherently resistant cells to gemcitabine. Together, these findings place ST6Gal-I as a critical player in imparting gemcitabine resistance and as a potential target to restore PDAC chemoresponse.
Collapse
Affiliation(s)
- Asmi Chakraborty
- From the Departments of Cell Developmental and Integrative Biology and
| | - Kaitlyn A Dorsett
- From the Departments of Cell Developmental and Integrative Biology and
| | - Hoa Q Trummell
- Radiation Oncology, University of Alabama, Birmingham, Alabama 35294
| | - Eddy S Yang
- Radiation Oncology, University of Alabama, Birmingham, Alabama 35294
| | - Patsy G Oliver
- Radiation Oncology, University of Alabama, Birmingham, Alabama 35294
| | - James A Bonner
- Radiation Oncology, University of Alabama, Birmingham, Alabama 35294
| | | | - Susan L Bellis
- From the Departments of Cell Developmental and Integrative Biology and
| |
Collapse
|
73
|
Campa D, Pastore M, Capurso G, Hackert T, Di Leo M, Izbicki JR, Khaw KT, Gioffreda D, Kupcinskas J, Pasquali C, Macinga P, Kaaks R, Stigliano S, Peeters PH, Key TJ, Talar-Wojnarowska R, Vodicka P, Valente R, Vashist YK, Salvia R, Papaconstantinou I, Shimizu Y, Valsuani C, Zambon CF, Gazouli M, Valantiene I, Niesen W, Mohelnikova-Duchonova B, Hara K, Soucek P, Malecka-Panas E, Bueno-de-Mesquita HBA, Johnson T, Brenner H, Tavano F, Fogar P, Ito H, Sperti C, Butterbach K, Latiano A, Andriulli A, Cavestro GM, Busch ORC, Dijk F, Greenhalf W, Matsuo K, Lombardo C, Strobel O, König AK, Cuk K, Strothmann H, Katzke V, Cantore M, Mambrini A, Oliverius M, Pezzilli R, Landi S, Canzian F. Do pancreatic cancer and chronic pancreatitis share the same genetic risk factors? A PANcreatic Disease ReseArch (PANDoRA) consortium investigation. Int J Cancer 2018; 142:290-296. [PMID: 28913878 DOI: 10.1002/ijc.31047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/29/2017] [Accepted: 07/17/2017] [Indexed: 02/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a very aggressive tumor with a five-year survival of less than 6%. Chronic pancreatitis (CP), an inflammatory process in of the pancreas, is a strong risk factor for PDAC. Several genetic polymorphisms have been discovered as susceptibility loci for both CP and PDAC. Since CP and PDAC share a consistent number of epidemiologic risk factors, the aim of this study was to investigate whether specific CP risk loci also contribute to PDAC susceptibility. We selected five common SNPs (rs11988997, rs379742, rs10273639, rs2995271 and rs12688220) that were identified as susceptibility markers for CP and analyzed them in 2,914 PDAC cases, 356 CP cases and 5,596 controls retrospectively collected in the context of the international PANDoRA consortium. We found a weak association between the minor allele of the PRSS1-PRSS2-rs10273639 and an increased risk of developing PDAC (ORhomozygous = 1.19, 95% CI 1.02-1.38, p = 0.023). Additionally all the SNPs confirmed statistically significant associations with risk of developing CP, the strongest being PRSS1-PRSS2-rs10273639 (ORheterozygous = 0.51, 95% CI 0.39-0.67, p = 1.10 × 10-6 ) and MORC4-rs 12837024 (ORhomozygous = 2.07 (1.55-2.77, ptrend = 0.7 × 10-11 ). Taken together, the results from our study do not support variants rs11988997, rs379742, rs10273639, rs2995271 and rs12688220 as strong predictors of PDAC risk, but further support the role of these SNPs in CP susceptibility. Our study suggests that CP and PDAC probably do not share genetic susceptibility, at least in terms of high frequency variants.
Collapse
Affiliation(s)
- Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - Manuela Pastore
- Department of Biology, University of Pisa, Pisa, Italy
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gabriele Capurso
- Digestive and Liver Disease Unit, S. Andrea Hospital 'Sapienza' University of Rome, Rome, Italy
| | - Thilo Hackert
- Department of General Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Milena Di Leo
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, San Raffaele Scientific Institute, Milan, Italy
| | - Jakob R Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kay-Tee Khaw
- Clinical Gerontology Unit, Addenbrooke's Hospital, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Domenica Gioffreda
- Division of Gastroenterology and Research Laboratory, Department of Surgery, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Italy
| | - Juozas Kupcinskas
- Department of Gastroenterology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Claudio Pasquali
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
| | - Peter Macinga
- Institute of Experimental Medicine, Czech Academy of Sciences and Institute of Clinical and Experimental Medicine, Prague, Czech Republic
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Serena Stigliano
- Digestive and Liver Disease Unit, S. Andrea Hospital 'Sapienza' University of Rome, Rome, Italy
| | - Petra H Peeters
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, United Kingdom
| | - Timothy J Key
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | | | - Pavel Vodicka
- Institute of Experimental Medicine, Czech Academy of Sciences and Institute of Clinical and Experimental Medicine, Prague, Czech Republic
| | - Roberto Valente
- Digestive and Liver Disease Unit, S. Andrea Hospital 'Sapienza' University of Rome, Rome, Italy
| | - Yogesh K Vashist
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Visceral Surgery, Kantonsspital Aarau AG, Aarau, Switzerland
| | - Roberto Salvia
- Department of Surgery, Pancreas Institute, University and Hospital Trust of Verona, Verona, Italy
| | - Ioannis Papaconstantinou
- Second Department of Surgery, Aretaieion Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Yasuhiro Shimizu
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Chiara Valsuani
- Oncological Department, Azienda USL Toscana Nord Ovest, Oncological Unit of Massa Carrara, Carrara, Massa and Carrara, Italy
| | | | - Maria Gazouli
- Department of Basic Medical Sciences, Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Irena Valantiene
- Department of Gastroenterology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Willem Niesen
- Department of General Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Kazuo Hara
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Pavel Soucek
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Ewa Malecka-Panas
- Department of Digestive Tract Diseases, Medical University of Lodz, Lodz, Poland
| | - H B As Bueno-de-Mesquita
- Department for Determinants of Chronic Diseases (DCD), National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, St Mary's Campus, London, United Kingdom
- Department of Social & Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Theron Johnson
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Herman Brenner
- Division of Clinical Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ), and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Francesca Tavano
- Division of Gastroenterology and Research Laboratory, Department of Surgery, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Italy
| | - Paola Fogar
- Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy
| | - Hidemi Ito
- Division of Molecular and Clinical Epidemiology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Cosimo Sperti
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
| | - Katja Butterbach
- Division of Clinical Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anna Latiano
- Division of Gastroenterology and Research Laboratory, Department of Surgery, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Italy
| | - Angelo Andriulli
- Division of Gastroenterology and Research Laboratory, Department of Surgery, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Italy
| | - Giulia Martina Cavestro
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, San Raffaele Scientific Institute, Milan, Italy
| | - Olivier R C Busch
- Department of Surgery, Academic Medical Centre, Amsterdam, the Netherlands
| | - Frederike Dijk
- Department of Pathology, Academic Medical Centre, Amsterdam, the Netherlands
| | - William Greenhalf
- Institute for Health Research, Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Liverpool, United Kingdom
| | - Keitaro Matsuo
- Division of Molecular and Clinical Epidemiology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Carlo Lombardo
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, University of Pisa, Pisa, Italy
| | - Oliver Strobel
- Department of General Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Anna-Katharina König
- Department of General Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Katarina Cuk
- Division of Clinical Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hendrik Strothmann
- Department of General Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Verena Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Maurizio Cantore
- Oncological Department, Azienda USL Toscana Nord Ovest, Oncological Unit of Massa Carrara, Carrara, Massa and Carrara, Italy
| | - Andrea Mambrini
- Oncological Department, Azienda USL Toscana Nord Ovest, Oncological Unit of Massa Carrara, Carrara, Massa and Carrara, Italy
| | - Martin Oliverius
- Transplant Surgery Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Raffaele Pezzilli
- Pancreas Unit, Department of Digestive Diseases and Internal Medicine Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | - Stefano Landi
- Department of Biology, University of Pisa, Pisa, Italy
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
74
|
Vogl TJ, Mohamed SA, Albrecht MH, Gruber-Roh T, Lin H, Nour Eldin NEA, Bednarova I, Naguib NN, Panahi B. Transarterial chemoembolization in pancreatic adenocarcinoma with liver metastases: MR-based tumor response evaluation, apparent diffusion coefficient (ADC) patterns, and survival rates. Pancreatology 2018; 18:94-99. [PMID: 29221632 DOI: 10.1016/j.pan.2017.11.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 11/26/2017] [Accepted: 11/28/2017] [Indexed: 12/11/2022]
Abstract
PURPOSE To retrospectively investigate the effectiveness of triple drug combination transarterial chemoembolization (TACE) on local tumor response and survival in patients with liver metastases from pancreatic cancer. Also, this study will evaluate the variances in response regarding the number of metastases, assess the correlation between tumor response and the changes in the apparent diffusion coefficients (ADC) in diffusion weighted (DW) MRI. MATERIALS AND METHODS One hundred and twelve patients (58 men and 54 women; mean age 57) with malignant liver metastases from pancreatic adenocarcinoma underwent at least one session of TACE with a chemotherapeutic combination of mitomycin C, cisplatin, and gemcitabine. A size-based evaluation of tumor response (response evaluation criteria in solid tumors (RECIST)) was conducted, along with ADC values, and survival indices as related to treatment pattern. RESULTS Four weeks following the end of the treatment, 78.26% of patients showed stable disease and 11.59% showed partial response. The median survival time was 19 months and for the stable disease group, 26 months. Low pretreatment ADC values showed no significant correlation to poor response to treatment (r = 0.347,p = 0.146). CONCLUSION The triple drug TACE technique showed improvements in median survival times in patients with hepatic metastases from pancreatic carcinoma and helped control disease progression, whereas the number of hepatic lesions was not a statistically significant factor in patients' response to TACE. The data suggest that pre-treatment ADC values in DW-MRI have no statistical correlation with tumor response.
Collapse
Affiliation(s)
- Thomas J Vogl
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Frankfurt, Germany
| | - Sherif A Mohamed
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Frankfurt, Germany.
| | - Moritz H Albrecht
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Frankfurt, Germany; Medical University of South Carolina, Department of Radiology and Radiological Science, Charleston SC, USA
| | - Tatjana Gruber-Roh
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Frankfurt, Germany
| | - Han Lin
- Medical University of South Carolina, Department of Radiology and Radiological Science, Charleston SC, USA
| | - Nour Eldin A Nour Eldin
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Frankfurt, Germany
| | - Iliana Bednarova
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Frankfurt, Germany; Institute of Diagnostic Radiology, Department of Medical and Biological Science, Udine, Italy
| | - Nagy N Naguib
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Frankfurt, Germany
| | - Bita Panahi
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Frankfurt, Germany
| |
Collapse
|
75
|
Dando I, Pacchiana R, Pozza ED, Cataldo I, Bruno S, Conti P, Cordani M, Grimaldi A, Butera G, Caraglia M, Scarpa A, Palmieri M, Donadelli M. UCP2 inhibition induces ROS/Akt/mTOR axis: Role of GAPDH nuclear translocation in genipin/everolimus anticancer synergism. Free Radic Biol Med 2017; 113:176-189. [PMID: 28962872 DOI: 10.1016/j.freeradbiomed.2017.09.022] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/25/2017] [Indexed: 12/20/2022]
Abstract
Several studies indicate that mitochondrial uncoupling protein 2 (UCP2) plays a pivotal role in cancer development by decreasing reactive oxygen species (ROS) produced by mitochondrial metabolism and by sustaining chemoresistance to a plethora of anticancer drugs. Here, we demonstrate that inhibition of UCP2 triggers Akt/mTOR pathway in a ROS-dependent mechanism in pancreatic adenocarcinoma cells. This event reduces the antiproliferative outcome of UCP2 inhibition by genipin, creating the conditions for the synergistic counteraction of cancer cell growth with the mTOR inhibitor everolimus. Inhibition of pancreatic adenocarcinoma cell growth and induction of apoptosis by genipin and everolimus treatment are functionally related to nuclear translocation of the cytosolic glycolytic enzyme glyceraldehyde 3-phosphate dehydrogenase (GAPDH). The synthetic compound (S)-benzyl-2-amino-2-(S)-3-bromo-4,5-dihydroisoxazol-5-yl-acetate (AXP3009), which binds GAPDH at its redox-sensitive Cys152, restores cell viability affected by the combined treatment with genipin and everolimus, suggesting a role for ROS production in the nuclear translocation of GAPDH. Caspase-mediated apoptosis by genipin and everolimus is further potentiated by the autophagy inhibitor 3-methyladenine revealing a protective role for Beclin1-mediated autophagy induced by the treatment. Mice xenograft of pancreatic adenocarcinoma further confirmed the antiproliferative outcome of drug combination without toxic effects for animals. Tumor masses from mice injected with UCP2 and mTOR inhibitors revealed a strong reduction in tumor volume and number of mitosis associated with a marked GAPDH nuclear positivity. Altogether, these results reveal novel mechanisms through which UCP2 promotes cancer cell proliferation and support the combined inhibition of UCP2 and of Akt/mTOR pathway as a novel therapeutic strategy in the treatment of pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Ilaria Dando
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy.
| | - Raffaella Pacchiana
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Elisa Dalla Pozza
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Ivana Cataldo
- Applied Research on Cancer Centre (ARC-Net) and Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Stefano Bruno
- Food and Dug Department, University of Parma, Parma, Italy
| | - Paola Conti
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Marco Cordani
- Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| | - Anna Grimaldi
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Giovanna Butera
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Aldo Scarpa
- Applied Research on Cancer Centre (ARC-Net) and Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Marta Palmieri
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Massimo Donadelli
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy.
| |
Collapse
|
76
|
Jiao F, Hu H, Han T, Zhuo M, Yuan C, Yang H, Wang L, Wang L. Aberrant expression of nuclear HDAC3 and cytoplasmic CDH1 predict a poor prognosis for patients with pancreatic cancer. Oncotarget 2017; 7:16505-16. [PMID: 26918727 PMCID: PMC4941331 DOI: 10.18632/oncotarget.7663] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 02/11/2016] [Indexed: 12/02/2022] Open
Abstract
Previous studies showed that aberrant CDH1 or/and HDAC3 localization is essential for the progression of some human cancers. Here, we investigate the prognostic significance of aberrant CDH1 and HDAC3 localization in 84 pancreatic cancer patients. Our results show that increases in both membrane and cytoplasmic CDH1 correlate with lymph node metastasis (P = 0.026 and P < 0.001, respectively) and clinical stage (P = 0.020 and P < 0.001, respectively). Increased nuclear HDAC3 correlates with lymph node metastasis (P < 0.001) and advanced clinical stage (P < 0.001), but increased cytoplasmic HDAC3 does not (P > 0.05). Multivariate analysis showed that nuclear HDAC3 and cytoplasmic CDH1 (P = 0.001 and P = 0.010, respectively), as well as tumor differentiation (P = 0.009) are independent prognostic factors. Most importantly, patients with high co-expression of nuclear HDAC3 and cytoplasmic CDH1 had shorter survival times (P < 0.001), more frequent lymph node metastasis (P < 0.001), and advanced clinical stage (P < 0.001). Our studies provide convincing evidence that nuclear HDAC3 and cytoplasmic CDH1 have independent prognostic value in pancreatic cancer and provide novel targets for prognostic therapeutics.
Collapse
Affiliation(s)
- Feng Jiao
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai 201620, China
| | - Hai Hu
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Ting Han
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai 201620, China
| | - Meng Zhuo
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Cuncun Yuan
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Haiyan Yang
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Lei Wang
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai 201620, China
| | - Liwei Wang
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai 201620, China
| |
Collapse
|
77
|
Xia D, Li X, Niu Q, Liu X, Xu W, Ma C, Gu H, Liu Z, Shi L, Tian X, Chen X, Zhang Y. MicroRNA-185 suppresses pancreatic cell proliferation by targeting transcriptional coactivator with PDZ-binding motif in pancreatic cancer. Exp Ther Med 2017; 15:657-666. [PMID: 29399068 PMCID: PMC5772449 DOI: 10.3892/etm.2017.5447] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 04/28/2017] [Indexed: 12/12/2022] Open
Abstract
The aim of the present study was to compare the expression of transcriptional coactivator with the PDZ-binding motif (TAZ) in pancreatic cancer (PC) patients, and to investigate the regulation mechanisms of TAZ in the proliferation of PC. PC tissues and matched peritumoral tissues, pancreatic juice and serum were collected from PC patients who underwent pancreatectomy between June 2012 and December 2015 at the Affiliated Hospital of Qingdao University (Qingdao, China). Pancreatic juice and serum were collected from patients with chronic pancreatitis as a control. The levels of taz mRNA expression in the samples were examined by reverse-transcription quantitative polymerase chain reaction, and the protein expression of TAZ was assessed by western blot analysis and ELISA. MicroRNAs (miRNAs) that regulate TAZ expression were also predicted by bioinformatics analysis and validated by dual luciferase reporter and rescue assays. In addition, the proliferation of PC cells was evaluated after transfection with TAZ small interfering RNA (siRNA) or its upstream miRNA agomir. Expression of TAZ was significantly increased in the PC tissues, pancreatic juice and serum of PC patients at the mRNA and protein levels compared with controls (P<0.05). Furthermore, TAZ was predicted and verified to be a target of miRNA (miR)-185, and miR-185 and TAZ were inversely expressed in samples from PC patients (P<0.05). In addition, TAZ siRNA or agomiR-185 transfection significantly inhibited human pancreatic adenocarcinoma cell proliferation (P<0.05). However, overexpression of TAZ in the agomiR-185 group rescued the inhibition (P<0.05). Finally, the expression of TAZ effector proteins, namely ankyrin repeat domain-containing protein and cysteine-rich 61, were upregulated in PC tissues (P<0.05), but repressed following transfection of PC cells with agomiR-185 (P<0.05). Thus, miR-185 may regulate the proliferation of PC by targeting TAZ, making it a promising diagnostic marker for PC.
Collapse
Affiliation(s)
- Di Xia
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xiaoyu Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Qinghui Niu
- Department of Infectious Disease, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xishuang Liu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Wanqun Xu
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Chengtai Ma
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Huali Gu
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Zhenfang Liu
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Lei Shi
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xintao Tian
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xiaoxue Chen
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yubao Zhang
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
78
|
Rees DO, Crick PJ, Jenkins GJ, Wang Y, Griffiths WJ, Brown TH, Al-Sarireh B. Comparison of the composition of bile acids in bile of patients with adenocarcinoma of the pancreas and benign disease. J Steroid Biochem Mol Biol 2017; 174:290-295. [PMID: 29031685 PMCID: PMC5668629 DOI: 10.1016/j.jsbmb.2017.10.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 09/15/2017] [Accepted: 10/10/2017] [Indexed: 12/20/2022]
Abstract
Bile acids have been implicated in the development of gastrointestinal malignancies. Both the specific nature of individual bile acids and their concentration appear key factors in the carcinogenic potency of bile. Using liquid chromatography mass spectrometry (LC-MS) we performed quantitative profiling of bile extracted directly from the common bile duct in 30 patients (15 patients with pancreatic cancer and 15 patients with benign disease). Separation and detection of bile acids was performed using a 1.7μm particle size reversed-phase C18 LC column at a flow rate of 200μL/min with negative electrospray ionization MS. A significant difference (p=0.018) was seen in the concentration of unconjugated cholic acid in the malignant group (0.643mmol/L) compared to the benign group (0.022mmol/L), with an overall significant difference (p=0.04) seen in the level of total unconjugated bile acids in the malignant group (1.816mmol/L) compared to the benign group (0.069mmol/L). This finding may offer the possibility of both understanding the biology of cancer development in the pancreas, as well as offering a potential diagnostic avenue to explore. However, a larger study is necessary to confirm the alterations in bile acid profiles reported here and explore factors such as diet and microbial populations on the bile acid profiles of these patient groups.
Collapse
Affiliation(s)
- David O Rees
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK; Department of Surgery, Morriston Hospital, Swansea, SA6 6NL, UK
| | - Peter J Crick
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK
| | - Gareth J Jenkins
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK
| | - Yuqin Wang
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK
| | - William J Griffiths
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, UK.
| | - Tim H Brown
- Department of Surgery, Morriston Hospital, Swansea, SA6 6NL, UK
| | | |
Collapse
|
79
|
Effects of Rab27A and Rab27B on Invasion, Proliferation, Apoptosis, and Chemoresistance in Human Pancreatic Cancer Cells. Pancreas 2017; 46:1173-1179. [PMID: 28902788 DOI: 10.1097/mpa.0000000000000910] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Rab family members are key regulatory factors that function as molecular switches in multiple phases of vesicular trafficking. Our previous study demonstrated that Rab27A and Rab27B overexpression may predict a poor outcome of pancreatic ductal adenocarcinoma. The purpose of this study was to investigate the role of Rab27A and Rab27B in the progression of pancreatic cancer. METHODS We down-regulated Rab27A and Rab27B expression in pancreatic cancer cell lines. The regulatory effects of knockdown Rab27A and Rab27B on pancreatic cancer cell were measured by cisplatin assay, invasion assay, proliferation assay, and Western blot assay. RESULTS Rab27A and Rab27B down-regulation enhances sensitivity to cisplatin and induces apoptosis in ASPC-1 and PANC-1 cells. In addition, down-regulation of Rab27A reduced the invasive and proliferative ability of ASPC-1 cells, and Rab27B knockdown significantly prevented cancer invasion and proliferation in PANC-1 cells. CONCLUSIONS Our findings provide evidence that Rab27A and Rab27B play significant roles in cell invasion, proliferation, and apoptosis, as well as in chemotherapy resistance.
Collapse
|
80
|
Ishii N, Iwata Y, Nishikawa H, Enomoto H, Aizawa N, Ishii A, Miyamoto Y, Yuri Y, Hasegawa K, Nakano C, Nishimura T, Yoh K, Sakai Y, Ikeda N, Takashima T, Takata R, Iijima H, Nishiguchi S. Effect of pretreatment psoas muscle mass on survival for patients with unresectable pancreatic cancer undergoing systemic chemotherapy. Oncol Lett 2017; 14:6059-6065. [PMID: 29113246 DOI: 10.3892/ol.2017.6952] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/04/2017] [Indexed: 12/18/2022] Open
Abstract
To the best of our knowledge, there are few previous studies that have investigated the effect of decreased skeletal muscle mass (DSMM) on survival in patients with unresectable advanced pancreatic cancer (APC) who are undergoing systemic chemotherapy. Thus, the present study aimed to investigate the impact of DSMM, as determined by the psoas muscle index (PMI) following computed tomography and prior to systemic chemotherapy, on the outcomes of patients with unresectable APC (n=61). The primary endpoint used was the overall survival (OS) rate. The OS rates in the PMI-High group (exceeds the median PMI value in each gender) were retrospectively compared with those in the PMI-Low group (below the median PMI value in each gender), and factors associated with OS were investigated using univariate and multivariate analyses. The study cohort included 31 male and 30 female patients with a median age of 72 years, 13 of whom were stage IVA, and 48 were stage IVB. The median PMI in males was 4.3 cm2/m2 (range, 1.6-8.2 cm2/m2), while that in females was 2.3 cm2/m2 (range, 0.7-6.1 cm2/m2). The proportion of patients with performance status 0 in the PMI-High group was significantly high, compared with that in the PMI-Low group [83.3% (25/30) vs. 58.1% (18/31); P=0.0486]. Body mass index in the PMI-High group was significantly higher compared with that in the PMI-Low group (P=0.0154). The 1-year cumulative survival rate was 43.3% in the PMI-High group and 12.9% in the PMI-Low group (P=0.0027). Following multivariate analysis, PMI (P=0.0036), prothrombin time (P=0.0044) and carbohydrate antigen 19-9 (P=0.0451) were identified to be significant predictors of OS. In conclusion, DSMM, as determined by the PMI, could be a significant predictor of prognosis in patients with unresectable APC who are receiving systemic chemotherapy.
Collapse
Affiliation(s)
- Noriko Ishii
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Yoshinori Iwata
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Hiroki Nishikawa
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Hirayuki Enomoto
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Nobuhiro Aizawa
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Akio Ishii
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Yuho Miyamoto
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Yukihisa Yuri
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Kunihiro Hasegawa
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Chikage Nakano
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Takashi Nishimura
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Kazunori Yoh
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Yoshiyuki Sakai
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Naoto Ikeda
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Tomoyuki Takashima
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Ryo Takata
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Hiroko Iijima
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| | - Shuhei Nishiguchi
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiyashi, Hyogo 663-8501, Japan
| |
Collapse
|
81
|
Validation of Lymphocyte-to-Monocyte Ratio as a Prognostic Factor in Advanced Pancreatic Cancer: An East Asian Cohort Study of 2 Countries. Pancreas 2017; 46:1011-1017. [PMID: 28787331 DOI: 10.1097/mpa.0000000000000891] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Although the prognostic value of lymphocyte-to-monocyte ratio (LMR) has been recently demonstrated in solid tumors, little is known of its impact on advanced pancreatic cancer (APC). This study evaluates and validates the cutoff value of LMR for predicting palliative chemotherapy outcome using a transnational cohort of APC patients. METHODS A total of 405 APC patients receiving first-line palliative chemotherapy were retrospectively reviewed. Of these, 153 patients were from Shanghai General Hospital (training set) and 252 patients were from Kyoto University Hospital (validation set). The optimal cutoff value of LMR was determined by a generating receiver operating characteristic curve for the training set. The association between LMR and survival was evaluated using log-rank tests, and a Cox regression model was used to validate the independent prognostic significance of LMR in APC patients. RESULTS The optimal cutoff value of LMR was 2.8. Overall survival was significantly longer in patients with LMR of 2.8 or greater than those with LMR of less than 2.8 (P < 0.001). Cox regression analysis showed that LMR was an independent prognostic factor. The impact of LMR was widely observed in all subgroups except the performance status 2 subgroup. CONCLUSIONS Lymphocyte-to-monocyte ratio may be considered as a promising prognostic marker for APC patients receiving palliative chemotherapy.
Collapse
|
82
|
Mehta KY, Wu HJ, Menon SS, Fallah Y, Zhong X, Rizk N, Unger K, Mapstone M, Fiandaca MS, Federoff HJ, Cheema AK. Metabolomic biomarkers of pancreatic cancer: a meta-analysis study. Oncotarget 2017; 8:68899-68915. [PMID: 28978166 PMCID: PMC5620306 DOI: 10.18632/oncotarget.20324] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 08/04/2017] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer (PC) is an aggressive disease with high mortality rates, however, there is no blood test for early detection and diagnosis of this disease. Several research groups have reported on metabolomics based clinical investigations to identify biomarkers of PC, however there is a lack of a centralized metabolite biomarker repository that can be used for meta-analysis and biomarker validation. Furthermore, since the incidence of PC is associated with metabolic syndrome and Type 2 diabetes mellitus (T2DM), there is a need to uncouple these common metabolic dysregulations that may otherwise diminish the clinical utility of metabolomic biosignatures. Here, we attempted to externally replicate proposed metabolite biomarkers of PC reported by several other groups in an independent group of PC subjects. Our study design included a T2DM cohort that was used as a non-cancer control and a separate cohort diagnosed with colorectal cancer (CRC), as a cancer disease control to eliminate possible generic biomarkers of cancer. We used targeted mass spectrometry for quantitation of literature-curated metabolite markers and identified a biomarker panel that discriminates between normal controls (NC) and PC patients with high accuracy. Further evaluation of our model with CRC, however, showed a drop in specificity for the PC biomarker panel. Taken together, our study underscores the need for a more robust study design for cancer biomarker studies so as to maximize the translational value and clinical implementation.
Collapse
Affiliation(s)
- Khyati Y Mehta
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Hung-Jen Wu
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Smrithi S Menon
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Yassi Fallah
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Xiaogang Zhong
- Department of Biostatistics Bioinformatics and Biomathematics, Georgetown University, Washington, DC, United States of America
| | - Nasser Rizk
- Department of Health Sciences, Qatar University, Doha, Qatar
| | - Keith Unger
- Lombardi Comprehensive Cancer Center, Med-Star Georgetown University Hospital, Washington, DC, United States of America
| | - Mark Mapstone
- Department of Neurology, University of California, Irvine, CA, United States of America
| | - Massimo S Fiandaca
- Department of Neurology, University of California, Irvine, CA, United States of America.,Department of Neurological Surgery, University of California, Irvine, CA, United States of America
| | - Howard J Federoff
- Department of Neurology, University of California, Irvine, CA, United States of America
| | - Amrita K Cheema
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, United States of America
| |
Collapse
|
83
|
Sinn M, Bahra M, Liersch T, Gellert K, Messmann H, Bechstein W, Waldschmidt D, Jacobasch L, Wilhelm M, Rau BM, Grützmann R, Weinmann A, Maschmeyer G, Pelzer U, Stieler JM, Striefler JK, Ghadimi M, Bischoff S, Dörken B, Oettle H, Riess H. CONKO-005: Adjuvant Chemotherapy With Gemcitabine Plus Erlotinib Versus Gemcitabine Alone in Patients After R0 Resection of Pancreatic Cancer: A Multicenter Randomized Phase III Trial. J Clin Oncol 2017; 35:3330-3337. [PMID: 28817370 DOI: 10.1200/jco.2017.72.6463] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Purpose Gemcitabine is standard of care in the adjuvant treatment of resectable pancreatic ductal adenocarcinoma (PDAC). The epidermal growth factor receptor tyrosine kinase inhibitor erlotinib in combination with gemcitabine has shown efficacy in the treatment of advanced PDAC and was considered to improve survival in patients with primarily resectable PDAC after R0 resection. Patients and Methods In an open-label, multicenter trial, patients were randomly assigned to one of two study arms: gemcitabine 1,000 mg/m2 days 1, 8, 15, every 4 weeks plus erlotinib 100 mg once per day (GemErlo) or gemcitabine (Gem) alone for six cycles. The primary end point of the study was to improve disease-free survival (DFS) from 14 to 18 months by adding erlotinib to gemcitabine. Results In all, 436 patients were randomly assigned at 57 study centers between April 2008 and July 2013. A total of 361 instances (83%) of disease recurrence were observed after a median follow-up of 54 months. Median treatment duration was 22 weeks in both arms. There was no difference in median DFS (GemErlo 11.4 months; Gem 11.4 months) or median overall survival (GemErlo 24.5 months; Gem 26.5 months). There was a trend toward long-term survival in favor of GemErlo (estimated survival after 1, 2, and 5 years for GemErlo was 77%, 53%, and 25% v 79%, 54%, and 20% for Gem, respectively). The occurrence or the grade of rash was not associated with a better survival in the GemErlo arm. Conclusion To the best of our knowledge, CONKO-005 is the first study to investigate the combination of chemotherapy and a targeted therapy in the adjuvant treatment of PDAC. GemErlo for 24 weeks did not improve DFS or overall survival over Gem.
Collapse
Affiliation(s)
- Marianne Sinn
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Marcus Bahra
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Torsten Liersch
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Klaus Gellert
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Helmut Messmann
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Wolf Bechstein
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Dirk Waldschmidt
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Lutz Jacobasch
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Martin Wilhelm
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Bettina M Rau
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Robert Grützmann
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Arndt Weinmann
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Georg Maschmeyer
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Uwe Pelzer
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Jens M Stieler
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Jana K Striefler
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Michael Ghadimi
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Sven Bischoff
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Bernd Dörken
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Helmut Oettle
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| | - Hanno Riess
- Marianne Sinn, Marcus Bahra, Uwe Pelzer, Jens M. Stieler, Jana K. Striefler, Sven Bischoff, Bernd Dörken, and Hanno Riess, Charité-Universitätsmedizin Berlin; Klaus Gellert, Sana Klinikum Lichtenberg, Berlin; Torsten Liersch and Michael Ghadimi, Universitätsmedizin Göttingen, Göttingen; Helmut Messmann, Klinikum Augsburg, Augsburg; Wolf Bechstein, Universitätsklinikum Frankfurt, Frankfurt; Dirk Waldschmidt, Universitätsklinikum Köln, Köln; Lutz Jacobasch, Clinical Center, Dresden; Martin Wilhelm, Paracelsus Medical University, Nürnberg; Bettina M. Rau, Universitätsmedizin Rostock, and Municipal Hospital of Neumarkt, Rostock; Robert Grützmann, Universitätsklinikum Carl Gustav Carus, Dresden, and Universitätsklinikum Erlangen, Erlangen; Arndt Weinmann, Klinikum der Johannes Gutenberg-Universität, Mainz; Georg Maschmeyer, Ernst von Bergmann Klinikum, Potsdam; and Helmut Oettle, Clinical Center, Friedrichshafen, Germany
| |
Collapse
|
84
|
Niu Q, Li X, Xia D, Jiang Y, Tian Z, Bian C, Zhang C, Liu P, Zhang F, Yang Y, Wang G. MicroRNA-186 affects the proliferation of tumor cells via yes-associated protein 1 in the occurrence and development of pancreatic cancer. Exp Ther Med 2017; 14:2094-2100. [PMID: 28962129 PMCID: PMC5609192 DOI: 10.3892/etm.2017.4770] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 03/31/2017] [Indexed: 12/12/2022] Open
Abstract
The present study aimed to determine the expression of microRNA (miRNA or miR)-186 in tumor tissues and peripheral blood of patients with pancreatic cancer (PC), as well as its mechanism of regulation. A total of 65 patients with PC who underwent surgery between June 2013 and October 2015 were included. In addition, 59 healthy subjects were recruited as controls. Reverse transcription-quantitative polymerase chain reaction was used to measure the expression of mRNA and miRNA. Western blotting and enzyme-linked immunosorbent assay were used to determine protein expression. Bioinformatics was employed for the prediction of the target gene of miR-186, whereas dual luciferase reporter assay was performed to identify whether miR-186 directly bound to YAP1 mRNA. Human pulmonary aortic endothelial cells (HPACs) were transfected with ago-miR-186. YAP1 expression in HPACs was silenced by siRNA. MTT assay was used to evaluate the viability of HPACs. YAP1 mRNA and protein expression levels were elevated in PC. In addition, expression levels of miR-186 in PC were downregulated. miR-186 regulated the expression of YAP1 by binding with the 3'-untranslated region of YAP1. Elevated expression of miR-186 inhibited the proliferation of HPACs by downregulating the expression of YAP1. Decreased expression of YAP1 by siRNA reduced the viability of HPACs. The present study demonstrates that YAP1 is upregulated in the tumor tissues and blood of PC patients, and this may be associated with the downregulation of miR-186. In addition, miR-186 may affect the occurrence and development of PC by controlling the proliferation of PC cells via YAP1.
Collapse
Affiliation(s)
- Qinghui Niu
- Department of Infection, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xiaoyu Li
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Di Xia
- Department of Emergency, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yueping Jiang
- Department of Infection, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Zibin Tian
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Cheng Bian
- Department of Infection, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Cuiping Zhang
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Pei Liu
- Department of Infection, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Fengjuan Zhang
- Department of Infection, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yuling Yang
- Department of Infection, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Guanglan Wang
- Department of Infection, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
85
|
Wang SN, Luo S, Liu C, Piao Z, Gou W, Wang Y, Guan W, Li Q, Zou H, Yang ZZ, Wang D, Wang Y, Xu M, Jin H, Xu CX. miR-491 Inhibits Osteosarcoma Lung Metastasis and Chemoresistance by Targeting αB-crystallin. Mol Ther 2017. [PMID: 28648665 DOI: 10.1016/j.ymthe.2017.05.018] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Dysregulated microRNAs (miRNAs) play an important role in osteosarcoma (OS) progression. In the present study, we investigate the clinical significance of serum miR-491 level and the potential role of miR-491 in OS lung metastasis and chemoresistance. Clinical data show that the level of miR-491 was decreased in serum from OS patients compared with healthy control subjects, and that a decreased serum miR-491 level is correlated with increased metastasis, poor chemoresponse, and lower survival rate in OS patients. In vitro and in vivo experiments show that overexpression of miR-491 suppresses OS cell lung metastasis, whereas it enhances cisplatin (CDDP)-induced tumor growth inhibition and apoptosis. In contrast, inhibition of miR-491 stimulates OS cell lung metastasis and suppresses CDDP-induced tumor growth inhibition and apoptosis. Furthermore, we demonstrate that miR-491 exerts its role by directly targeting αB-crystallin (CRYAB) in OS. Our findings suggest that serum level of miR-491 has potential as a biomarker for predicting OS progression and prognosis of OS patients. Additionally, restoration of miR-491 may be a novel strategy for inhibiting OS lung metastasis and overcoming OS cell resistance to chemotherapy.
Collapse
Affiliation(s)
- Shu-Nan Wang
- Department of Radiology, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Song Luo
- Department of Orthopaedics, The General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - Chang Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Zhenghao Piao
- Department of Basic Medical Science, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China
| | - Wenlong Gou
- Department of Orthopaedics, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Yun Wang
- Department of Pathology, The General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - Wei Guan
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Qing Li
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Hua Zou
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Zhen-Zhou Yang
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Dong Wang
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Yan Wang
- Department of Orthopaedics, The General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - Meng Xu
- Department of Orthopaedics, The General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - Hua Jin
- Department of Thoracic Surgery, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China; Department of Pharmaceutical Science, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA.
| | - Cheng-Xiong Xu
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China.
| |
Collapse
|
86
|
Loosen SH, Neumann UP, Trautwein C, Roderburg C, Luedde T. Current and future biomarkers for pancreatic adenocarcinoma. Tumour Biol 2017; 39:1010428317692231. [DOI: 10.1177/1010428317692231] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Sven H Loosen
- Department of Gastroenterology, Digestive Diseases and Intensive Care Medicine (Department of Medicine III), Division of GI Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Ulf P Neumann
- Department of Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Christian Trautwein
- Department of Gastroenterology, Digestive Diseases and Intensive Care Medicine (Department of Medicine III), Division of GI Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Christoph Roderburg
- Department of Gastroenterology, Digestive Diseases and Intensive Care Medicine (Department of Medicine III), Division of GI Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Tom Luedde
- Department of Gastroenterology, Digestive Diseases and Intensive Care Medicine (Department of Medicine III), Division of GI Oncology, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
87
|
Pretreatment C-reactive protein to albumin ratio for predicting overall survival in advanced pancreatic cancer patients. Sci Rep 2017; 7:2993. [PMID: 28592881 PMCID: PMC5462766 DOI: 10.1038/s41598-017-03153-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 04/25/2017] [Indexed: 02/06/2023] Open
Abstract
Although previous studies demonstrated that elevated C-reactive protein to albumin ratio (CAR) predicted poor prognosis in various solid tumors, little was known about the prognostic value of CAR in patients with advanced pancreatic cancer (APC). The aim of the present study was to assess CAR as one independent prognostic factor in predicting overall survival (OS) in APC patients who had received palliative chemotherapy. Data of 142 APC patients who received palliative chemotherapy between 2009 and 2014 were retrospectively documented. We classified the patients into two groups based on the optimal cutoff value of CAR identified by generating receiver operating characteristics (ROC) curve. The clinicopathological parameters were compared between two CAR groups. Pearson correlation test showed that the level of C-reactive protein (CRP) was inversely correlated with albumin (r = −0.387; P < 0.001). Kaplan-Meier analysis demonstrated overall survival (OS) was significantly longer in CAR < 0.156 group than CAR ≥ 0.156 group (11.2 vs 5.9 months, P < 0.001). CAR was an independent prognostic factor for OS in the Cox regression model (HR, 1.623; 95% CI, 1.093–2.410; P = 0.016). Furthermore, the discrimination ability of CAR (AUC = 0.648, P = 0.025) was slightly higher than that of other inflammation-based factors. Therefore, pretreatment CAR could be an independent prognostic biomarker for APC patients.
Collapse
|
88
|
Maruyama T, Tomofuji T, Machida T, Kato H, Tsutsumi K, Uchida D, Takaki A, Yoneda T, Miyai H, Mizuno H, Ekuni D, Okada H, Morita M. Association between periodontitis and prognosis of pancreatobiliary tract cancer: A pilot study. Mol Clin Oncol 2017; 6:683-687. [PMID: 28515921 PMCID: PMC5431377 DOI: 10.3892/mco.2017.1220] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 03/10/2017] [Indexed: 12/16/2022] Open
Abstract
Several studies have indicated that periodontitis is a risk factor for cancer. However, the association between periodontitis and the prognosis of pancreatobiliary tract cancer remains unclear. The aim of this pilot study was to investigate the association between periodontitis and prognosis of pancreatobiliary tract cancer. A total of 22 patients diagnosed with pancreatobiliary tract cancer were analyzed. Oral health status, including severity of periodontitis, general health status and biochemical serum markers were evaluated. The Kaplan-Meier method and Cox proportional hazards model were used to assess factors affecting the prognosis of pancreatobiliary tract cancer. The Kaplan-Meier analysis demonstrated that low body mass index, high concentration of serum C-reactive protein (CRP) and severe periodontitis were significant prognostic factors for survival rate. The Cox proportional hazards model revealed that serum carbohydrate antigen 19-9 concentration [hazard ratio (HR)=1.002; 95% confidence interval (CI): 1.000–1.004] and serum CRP concentration (HR=2.57; 95% CI: 1.15–5.74) were significantly associated with the prognosis of pancreatobiliary tract cancer. In addition, cancer patients with severe periodontitis had higher serum CRP concentrations compared with those without severe periodontitis. Therefore, severe periodontitis indirectly affected the prognosis of pancreatobiliary tract cancer through promoting systemic inflammation.
Collapse
Affiliation(s)
- Takayuki Maruyama
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama 700-8558, Japan
| | - Takaaki Tomofuji
- Department of Community Oral Health, Asahi University School of Dentistry, Mizuho, Gifu 501-0296, Japan
| | - Tatsuya Machida
- Department of Preventive Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hironari Kato
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Koichiro Tsutsumi
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Daisuke Uchida
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Akinobu Takaki
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Toshiki Yoneda
- Department of Preventive Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hisataka Miyai
- Department of Preventive Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hirofumi Mizuno
- Department of Preventive Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Daisuke Ekuni
- Department of Preventive Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hiroyuki Okada
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Manabu Morita
- Department of Preventive Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
89
|
Conrad TOF, Genzel M, Cvetkovic N, Wulkow N, Leichtle A, Vybiral J, Kutyniok G, Schütte C. Sparse Proteomics Analysis - a compressed sensing-based approach for feature selection and classification of high-dimensional proteomics mass spectrometry data. BMC Bioinformatics 2017; 18:160. [PMID: 28274197 PMCID: PMC5343371 DOI: 10.1186/s12859-017-1565-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 02/24/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND High-throughput proteomics techniques, such as mass spectrometry (MS)-based approaches, produce very high-dimensional data-sets. In a clinical setting one is often interested in how mass spectra differ between patients of different classes, for example spectra from healthy patients vs. spectra from patients having a particular disease. Machine learning algorithms are needed to (a) identify these discriminating features and (b) classify unknown spectra based on this feature set. Since the acquired data is usually noisy, the algorithms should be robust against noise and outliers, while the identified feature set should be as small as possible. RESULTS We present a new algorithm, Sparse Proteomics Analysis (SPA), based on the theory of compressed sensing that allows us to identify a minimal discriminating set of features from mass spectrometry data-sets. We show (1) how our method performs on artificial and real-world data-sets, (2) that its performance is competitive with standard (and widely used) algorithms for analyzing proteomics data, and (3) that it is robust against random and systematic noise. We further demonstrate the applicability of our algorithm to two previously published clinical data-sets.
Collapse
Affiliation(s)
- Tim O. F. Conrad
- Department of Mathematics, Freie Universität Berlin, Arnimallee 6, Berlin, Germany
- Zuse Institute Berlin, Takustr. 7, Berlin, Germany
| | - Martin Genzel
- Department of Mathematics, Technische Universität Berlin, Düsternbrooker Weg 20, Berlin, Germany
| | - Nada Cvetkovic
- Department of Mathematics, Freie Universität Berlin, Arnimallee 6, Berlin, Germany
| | - Niklas Wulkow
- Department of Mathematics, Freie Universität Berlin, Arnimallee 6, Berlin, Germany
| | - Alexander Leichtle
- Center of Laboratory Medicine, Inselspital - Bern University Hospital, Düsternbrooker Weg 20, Bern, 24105 Switzerland
| | - Jan Vybiral
- Department of Mathematical Analysis, Charles University, Düsternbrooker Weg 20, Prague, Czech Republic
| | - Gitta Kutyniok
- Department of Mathematics, Technische Universität Berlin, Düsternbrooker Weg 20, Berlin, Germany
| | - Christof Schütte
- Department of Mathematics, Freie Universität Berlin, Arnimallee 6, Berlin, Germany
- Zuse Institute Berlin, Takustr. 7, Berlin, Germany
| |
Collapse
|
90
|
Cao J, Fu Z, Gao L, Wang X, Cheng S, Wang X, Ren H. Evaluation of serum D-dimer, fibrinogen, and CA19-9 for postoperative monitoring and survival prediction in resectable pancreatic carcinoma. World J Surg Oncol 2017; 15:48. [PMID: 28219450 PMCID: PMC5319056 DOI: 10.1186/s12957-017-1104-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 01/18/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND We sought to investigate the efficacy of serum D-dimer, fibrinogen, and CA19-9 for postoperative monitoring and prediction of survival in patients with resectable pancreatic carcinoma (PC). METHODS One hundred and nineteen patients with resectable PC were enrolled. Serum D-dimer, fibrinogen, and CA19-9 values were analyzed before surgery and at the stages of relapse-free and progression disease. RESULTS D-dimer, fibrinogen, and CA19-9 were significantly higher at the active stage of PC than those at the relapse-free stage [1059.2 (1690.1) ng/ml vs 485.18 (289.84) ng/ml, (3.71 ± 0.83) g/l vs (2.75 ± 0.52) g/l, 207.2 (681.8) U/ml vs 24.5 (30) U/ml, respectively, p < 0.01]. Patients with elevated preoperative D-dimer had significantly shorter overall survival (18.9 ± 1.9 months vs 29.2 ± 2.6 months, p < 0.01) and progression-free survival (10.6 ± 1.2 months vs 20.4 ± 2.4 months, p < 0.01) than did those with low D-dimer. The correlation between CA19-9 values and survival depended on the threshold value of CA19-9: when the threshold value was 37 U/ml, there was no correlation between CA19-9 and survival; when the threshold value was 253.8 U/ml (median CA19-9 for the enrolled patients), patients with elevated preoperative CA19-9 had significantly shorter overall survival (19.9 ± 2. 1 months vs 29.0 ± 2. 7 months) and progression-free survival (11.5 ± 1.5 months vs 21.0 ± 2. 6 months) than did the patients with low CA19-9 (p < 0.01); when the threshold value was 1000 U/ml, the overall survival was 15.5 ± 2.3 months vs 28.0 ± 2.0 months and the progression-free survival 8.9 ± 1.9 months vs 19.1 ± 1.9 months (p < 0.01). There was no correlation between fibrinogen and overall survival (25.8 ± 2.1 months vs 21.2 ± 2.9 months; p = 0.096) and progression-free survival (17.8 ± 2.1 months vs 12.7 ± 1.7 months; p = 0.168). CONCLUSIONS For postoperative monitoring of patients with resectable PC, D-dimer, fibrinogen, and CA19-9 may be used as markers for monitoring disease relapse, but only preoperative D-dimer could predict survival.
Collapse
Affiliation(s)
- Junli Cao
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 People’s Republic of China
| | - Zhanzhao Fu
- Department of Oncology, First Hospital of Qinhuangdao, Hebei, 066000 People’s Republic of China
| | - Liming Gao
- Department of Oncology, First Hospital of Qinhuangdao, Hebei, 066000 People’s Republic of China
| | - Xin Wang
- Department of Intensive Medicine, Traditional Chinese Medicine Hospital of Qinhuangdao, Hebei, 066000 People’s Republic of China
| | - Shaohui Cheng
- Department of Oncology, First Hospital of Qinhuangdao, Hebei, 066000 People’s Republic of China
| | - Xiuchao Wang
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 People’s Republic of China
| | - He Ren
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 People’s Republic of China
| |
Collapse
|
91
|
Affiliation(s)
- Diana Behrens
- EPO - Experimental Pharmacology and Oncology GmbH - GmbH, Robert-Roessle-Str. 10, 13125 Berlin, Germany.
| | - Wolfgang Walther
- Experimental and Clinical Research Center (ECRC), Charité, University Medicine, Berlin; Max-Delbrueck-Center for Molecular Medicine, Robert-Roessle-Str. 10, 13125 Berlin, Germany
| | - Iduna Fichtner
- EPO - Experimental Pharmacology and Oncology GmbH - GmbH, Robert-Roessle-Str. 10, 13125 Berlin, Germany
| |
Collapse
|
92
|
Zinovyeva MV, Kuzmich AI, Monastyrskaya GS, Sverdlov ED. The role of FOXA subfamily factors in embryonic development and carcinogenesis of the pancreas. MOLECULAR GENETICS MICROBIOLOGY AND VIROLOGY 2017. [DOI: 10.3103/s0891416816030113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
93
|
Hastie E, Cataldi M, Moerdyk MJ, Felt SA, Steuerwald N, Grdzelishvili VZ. Novel biomarkers of resistance of pancreatic cancer cells to oncolytic vesicular stomatitis virus. Oncotarget 2016; 7:61601-61618. [PMID: 27533247 PMCID: PMC5308675 DOI: 10.18632/oncotarget.11202] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 07/27/2016] [Indexed: 02/04/2023] Open
Abstract
Vesicular stomatitis virus (VSV) based recombinant viruses (such as VSV-ΔM51) are effective oncolytic viruses (OVs) against a majority of pancreatic ductal adenocarcinoma (PDAC) cell lines. However, some PDAC cell lines are highly resistant to VSV-ΔM51. We recently showed that treatment of VSV-resistant PDAC cells with ruxolitinib (JAK1/2 inhibitor) or TPCA-1 (IKK-β inhibitor) breaks their resistance to VSV-ΔM51. Here we compared the global effect of ruxolitinib or TPCA-1 treatment on cellular gene expression in PDAC cell lines highly resistant to VSV-ΔM51. Our study identified a distinct subset of 22 interferon-stimulated genes (ISGs) downregulated by both ruxolitinib and TPCA-1. Further RNA and protein analyses demonstrated that 4 of these genes (MX1, EPSTI1, XAF1, and GBP1) are constitutively co-expressed in VSV-resistant, but not in VSV-permissive PDACs, thus serving as potential biomarkers to predict OV therapy success. Moreover, shRNA-mediated knockdown of one of such ISG, MX1, showed a positive effect on VSV-ΔM51 replication in resistant PDAC cells, suggesting that at least some of the identified ISGs contribute to resistance of PDACs to VSV-ΔM51. As certain oncogene and tumor suppressor gene variants are often associated with increased tropism of OVs to cancer cells, we also analyzed genomic DNA in a set of PDAC cell lines for frequently occurring cancer associated mutations. While no clear correlation was found between such mutations and resistance of PDACs to VSV-ΔM51, the analysis generated valuable genotypic data for future studies.
Collapse
Affiliation(s)
- Eric Hastie
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Marcela Cataldi
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Megan J. Moerdyk
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Sébastien A. Felt
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Nury Steuerwald
- Cannon Research Center, Carolinas Healthcare System, Charlotte, NC, USA
| | - Valery Z. Grdzelishvili
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| |
Collapse
|
94
|
Wang Y, Kumar S, Rachagani S, Sajja BR, Xie Y, Hang Y, Jain M, Li J, Boska MD, Batra SK, Oupický D. Polyplex-mediated inhibition of chemokine receptor CXCR4 and chromatin-remodeling enzyme NCOA3 impedes pancreatic cancer progression and metastasis. Biomaterials 2016; 101:108-120. [PMID: 27267632 PMCID: PMC4921319 DOI: 10.1016/j.biomaterials.2016.05.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/22/2016] [Accepted: 05/24/2016] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer (PC) is one of the most aggressive malignancies due to intense desmoplasia, extreme hypoxia and inherent chemoresistance. Studies have implicated the expression of chemokine receptor CXCR4 and nuclear receptor co-activator-3 (NCOA3) in the development of desmoplasia and metastatic spread of PC. Using a series of polymeric CXCR4 antagonists (PCX), we optimized formulation of PCX/siNCOA3 polyplexes to simultaneously target CXCR4 and NCOA3 in PC. Cholesterol-modified PCX showed maximum CXCR4 antagonism, NCOA3 silencing and inhibition of PC cell migration in vitro. The optimized PCX/siNCOA3 polyplexes were used in evaluating antitumor and antimetastatic activity in orthotopic mouse model of metastatic PC. The polyplexes displayed significant inhibition of primary tumor growth, which was accompanied by a decrease in tumor necrosis and increased tumor perfusion. The polyplexes also showed significant antimetastatic effect and effective suppression of metastasis to distant organs. Overall, dual-function PCX/siNCOA3 polyplexes can effectively regulate tumor microenvironment to decrease progression and dissemination of PC.
Collapse
Affiliation(s)
- Yan Wang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Nebraska, United States
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Nebraska, United States
| | | | - Ying Xie
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
| | - Yu Hang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Nebraska, United States
| | - Jing Li
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
| | - Michael D. Boska
- Department of Radiology, University of Nebraska Medical Center, Nebraska, United States
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Nebraska, United States
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
95
|
Mukherjee I, Powell B, Parianos M, Downs D, Ross SB. Available technologies and clinical applications of targeted chemotherapy in pancreatic cancer. Cancer Genet 2016; 209:582-591. [PMID: 27613576 DOI: 10.1016/j.cancergen.2016.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 08/01/2016] [Indexed: 02/08/2023]
Abstract
The incidence of pancreatic cancer, the fourth leading cause of cancer death in United States, is increasing worldwide. Even though the cure rate has doubled in 40 years, it is abysmally poor at 6-7%. As surgical resection remains the only curative treatment and less than 20% of the newly diagnosed cancers are resectable, the major burden of disease management lies in early diagnosis, good prognostication, and proper neo-adjuvant and/or adjuvant therapy. With advancing technologies and their ease of availability, researchers have better tools to understand pancreatic cancer. In the post-genetic era, proteomic, phosphoproteomic, metabolomic, and more have brought us to a multi-omics era. These newer avenues bring promises of better screening modalities, less invasive diagnostics and monitoring, subtyping of pancreatic cancer, and fine tuning the treatment modalities not only to the right stage of tumor but also to the right tumor biology. As the multitudes of technologies are generating extensive amounts of incongruous data, they are giving clinicians a lot of non-actionable information. In this paper, we wish to encompass the newer technologies, sub-classifications, and future treatment modalities in personalized care of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Indraneil Mukherjee
- Southeastern Center for Digestive Disorders and Pancreatic Cancer, Florida Hospital Tampa, Tampa, FL, USA.
| | - Brett Powell
- Southeastern Center for Digestive Disorders and Pancreatic Cancer, Florida Hospital Tampa, Tampa, FL, USA
| | - Mary Parianos
- Southeastern Center for Digestive Disorders and Pancreatic Cancer, Florida Hospital Tampa, Tampa, FL, USA
| | - Darrell Downs
- Southeastern Center for Digestive Disorders and Pancreatic Cancer, Florida Hospital Tampa, Tampa, FL, USA
| | - Sharona B Ross
- Southeastern Center for Digestive Disorders and Pancreatic Cancer, Florida Hospital Tampa, Tampa, FL, USA
| |
Collapse
|
96
|
Mar-Aguilar F, Rodríguez-Padilla C, Reséndez-Pérez D. Web-based tools for microRNAs involved in human cancer. Oncol Lett 2016; 11:3563-3570. [PMID: 27284356 DOI: 10.3892/ol.2016.4446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/10/2016] [Indexed: 12/18/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) are a family of small, endogenous and evolutionarily-conserved non-coding RNAs that are involved in the regulation of several cellular and functional processes. miRNAs can act as oncogenes or tumor suppressors in all types of cancer, and could be used as prognostic and diagnostic biomarkers. Databases and computational algorithms are behind the majority of the research performed on miRNAs. These tools assemble and curate the relevant information on miRNAs and present it in a user-friendly manner. The current review presents 14 online databases that address every aspect of miRNA cancer research. Certain databases focus on miRNAs and a particular type of cancer, while others analyze the behavior of miRNAs in different malignancies at the same time. Additional databases allow researchers to search for mutations in miRNAs or their targets, and to review the naming history of a particular miRNA. All these databases are open-access, and are a valuable tool for those researchers working with these molecules, particularly those who lack access to an advanced computational infrastructure.
Collapse
Affiliation(s)
- Fermín Mar-Aguilar
- Departamento de Biología Celular y Genética, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León 66451, México
| | - Cristina Rodríguez-Padilla
- Departamento de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León 66451, México
| | - Diana Reséndez-Pérez
- Departamento de Biología Celular y Genética, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León 66451, México; Departamento de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León 66451, México
| |
Collapse
|