101
|
Abdulmawjood B, Roma-Rodrigues C, Fernandes AR, Baptista PV. Liquid biopsies in myeloid malignancies. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:1044-1061. [PMID: 35582281 PMCID: PMC9019201 DOI: 10.20517/cdr.2019.88] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022]
Abstract
Hematologic malignancies are the most common type of cancer affecting children and young adults, and encompass diseases, such as leukemia, lymphoma, and myeloma, all of which impact blood associated tissues such as the bone marrow, lymphatic system, and blood cells. Clinical diagnostics of these malignancies relies heavily on the use of bone marrow samples, which is painful, debilitating, and not free from risks for leukemia patients. Liquid biopsies are based on minimally invasive assessment of markers in the blood (and other fluids) and have the potential to improve the efficacy of diagnostic/therapeutic strategies in leukemia patients, providing a useful tool for the real time molecular profiling of patients. The most promising noninvasive biomarkers are circulating tumor cells, circulating tumor DNA, microRNAs, and exosomes. Herein, we discuss the role of assessing these circulating biomarkers for the understanding of tumor progression and metastasis, tumor progression dynamics through treatment and for follow-up.
Collapse
Affiliation(s)
- Bilal Abdulmawjood
- UCIBIO, Department of Life Sciences, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Campus Caparica, Caparica 2829-516, Portugal
| | - Catarina Roma-Rodrigues
- UCIBIO, Department of Life Sciences, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Campus Caparica, Caparica 2829-516, Portugal
| | - Alexandra R Fernandes
- UCIBIO, Department of Life Sciences, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Campus Caparica, Caparica 2829-516, Portugal
| | - Pedro V Baptista
- UCIBIO, Department of Life Sciences, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Campus Caparica, Caparica 2829-516, Portugal
| |
Collapse
|
102
|
Abstract
Pancreatic cancer (PC) is a leading cause of cancer-related death in developed countries, and since most patients have incurable disease at the time of diagnosis, developing a screening method for early detection is of high priority. Due to its metabolic importance, alterations in pancreatic functions may affect the composition of the gut microbiota, potentially yielding biomarkers for PC. However, the usefulness of these biomarkers may be limited if they are specific for advanced stages of disease, which may involve comorbidities such as biliary obstruction or diabetes. In this study we analyzed the fecal microbiota of 30 patients with pancreatic adenocarcinoma, 6 patients with pre-cancerous lesions, 13 healthy subjects and 16 with non-alcoholic fatty liver disease, using amplicon sequencing of the bacterial 16S rRNA gene. Fourteen bacterial features discriminated between PC and controls, and several were shared with findings from a recent Chinese cohort. A Random Forest model based on the microbiota classified PC and control samples with an AUC of 82.5%. However, inter-subject variability was high, and only a small part of the PC-associated microbial signals were also observed in patients with pre-cancerous pancreatic lesions, implying that microbiome-based early detection of such lesions will be challenging.
Collapse
|
103
|
Ren S, Zhang J, Chen J, Cui W, Zhao R, Qiu W, Duan S, Chen R, Chen X, Wang Z. Evaluation of Texture Analysis for the Differential Diagnosis of Mass-Forming Pancreatitis From Pancreatic Ductal Adenocarcinoma on Contrast-Enhanced CT Images. Front Oncol 2019; 9:1171. [PMID: 31750254 PMCID: PMC6848378 DOI: 10.3389/fonc.2019.01171] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/18/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose: To investigate the potential of computed tomography (CT) imaging features and texture analysis to differentiate between mass-forming pancreatitis (MFP) and pancreatic ductal adenocarcinoma (PDAC). Materials and Methods: Thirty patients with pathologically proved MFP and 79 patients with PDAC were included in this study. Clinical data and CT imaging features of the two lesions were evaluated. Texture features were extracted from arterial and portal phase CT images using commercially available software (AnalysisKit). Multivariate logistic regression analyses were used to identify relevant CT imaging and texture parameters to discriminate MFP from PDAC. Receiver operating characteristic curves were performed to determine the diagnostic performance of predictions. Results: MFP showed a larger size compared to PDAC (p = 0.009). Cystic degeneration, pancreatic ductal dilatation, vascular invasion, and pancreatic sinistral portal hypertension were more frequent and duct penetrating sign was less frequent in PDAC compared to MFP. Arterial CT attenuation, arterial, and portal enhancement ratios of MFP were higher than PDAC (p < 0.05). In multivariate analysis, arterial CT attenuation and pancreatic duct penetrating sign were independent predictors. Texture features in arterial phase including SurfaceArea, Percentile40, InverseDifferenceMoment_angle90_offset4, LongRunEmphasis_angle45_offset4, and uniformity were independent predictors. Texture features in portal phase including LongRunEmphasis_angle135_offset7, VoxelValueSum, LongRunEmphasis_angle135_offset4, and GLCMEntropy_angle45_offset1 were independent predictors. Areas under the curve of imaging feature-based, texture feature-based in arterial and portal phases, and the combined models were 0.84, 0.96, 0.93, and 0.98, respectively. Conclusions: CT texture analysis demonstrates great potential to differentiate MFP from PDAC.
Collapse
Affiliation(s)
- Shuai Ren
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Jingjing Zhang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Jingya Chen
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Wenjing Cui
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Rui Zhao
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Wenli Qiu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | | | - Rong Chen
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Xiao Chen
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Zhongqiu Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| |
Collapse
|
104
|
Rezaee M, Wang J, Razavi M, Ren G, Zheng F, Hussein A, Ullah M, Thakor AS. A Study Comparing the Effects of Targeted Intra-Arterial and Systemic Chemotherapy in an Orthotopic Mouse Model of Pancreatic Cancer. Sci Rep 2019; 9:15929. [PMID: 31685925 PMCID: PMC6828954 DOI: 10.1038/s41598-019-52490-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 10/18/2019] [Indexed: 02/06/2023] Open
Abstract
Systemic chemotherapy is the first line treatment for patients with unresectable pancreatic cancer, however, insufficient drug delivery to the pancreas is a major problem resulting in poor outcomes. We evaluated the therapeutic effects of targeted intra-arterial (IA) delivery of gemcitabine directly into the pancreas in an orthotopic mouse model of pancreatic cancer. Nude mice with orthotopic pancreatic tumors were randomly assigned into 3 groups receiving gemcitabine: systemic intravenous (IV) injection (low: 0.3 mg/kg and high: 100 mg/kg) and direct IA injection (0.3 mg/kg). Treatments were administered weekly for 2 weeks. IA treatment resulted in a significantly greater reduction in tumor growth compared to low IV treatment. To achieve a comparable reduction in tumor growth as seen with IA treatment, gemcitabine had to be given IV at over 300x the dose (high IV treatment) which was associated with some toxicity. After 2 weeks, tumor samples from animals treated with IA gemcitabine had significantly lower residual cancer cells, higher cellular necrosis and evidence of increased apoptosis when compared to animals treated with low IV gemcitabine. Our study shows targeted IA injection of gemcitabine directly into the pancreas, via its arterial blood supply, has a superior therapeutic effect in reducing tumor growth compared to the same concentration administered by conventional systemic injection.
Collapse
MESH Headings
- Administration, Intravenous
- Animals
- Antimetabolites, Antineoplastic/adverse effects
- Antimetabolites, Antineoplastic/therapeutic use
- Cell Line, Tumor
- Deoxycytidine/adverse effects
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/therapeutic use
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drug Administration Schedule
- Female
- Humans
- Infusions, Intra-Arterial
- Male
- Mice
- Mice, Nude
- Neoplasm, Residual
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Transplantation, Heterologous
- Gemcitabine
Collapse
Affiliation(s)
- Melika Rezaee
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
- Chicago Medical School, Rosalind Franklin University, North Chicago, Illinois, 60064, USA
| | - Jing Wang
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Mehdi Razavi
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Gang Ren
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Fengyan Zheng
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Ahmed Hussein
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA.
| |
Collapse
|
105
|
Cheng N, Du D, Wang X, Liu D, Xu W, Luo Y, Lin Y. Recent Advances in Biosensors for Detecting Cancer-Derived Exosomes. Trends Biotechnol 2019; 37:1236-1254. [DOI: 10.1016/j.tibtech.2019.04.008] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/14/2019] [Accepted: 04/17/2019] [Indexed: 02/07/2023]
|
106
|
Buscail E, Alix-Panabières C, Quincy P, Cauvin T, Chauvet A, Degrandi O, Caumont C, Verdon S, Lamrissi I, Moranvillier I, Buscail C, Marty M, Laurent C, Vendrely V, Moreau-Gaudry F, Bedel A, Dabernat S, Chiche L. High Clinical Value of Liquid Biopsy to Detect Circulating Tumor Cells and Tumor Exosomes in Pancreatic Ductal Adenocarcinoma Patients Eligible for Up-Front Surgery. Cancers (Basel) 2019; 11:cancers11111656. [PMID: 31717747 PMCID: PMC6895804 DOI: 10.3390/cancers11111656] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 02/08/2023] Open
Abstract
PURPOSE Expediting the diagnosis of pancreatic ductal adenocarcinoma (PDAC) would benefit care management, especially for the start of treatments requiring histological evidence. This study evaluated the combined diagnostic performance of circulating biomarkers obtained by peripheral and portal blood liquid biopsy in patients with resectable PDAC. EXPERIMENTAL DESIGN Liquid biopsies were performed in a prospective translational clinical trial (PANC-CTC #NCT03032913) including 22 patients with resectable PDAC and 28 noncancer controls from February to November 2017. Circulating tumor cells (CTCs) were detected using the CellSearch® method or after RosetteSep® enrichment combined with CRISPR/Cas9-improved KRAS mutant alleles quantification by droplet digital PCR. CD63 bead-coupled Glypican-1 (GPC1)-positive exosomes were quantified by flow cytometry. RESULTS Liquid biopsies were positive in 7/22 (32%), 13/22 (59%), and 14/22 (64%) patients with CellSearch® or RosetteSep®-based CTC detection or GPC1-positive exosomes, respectively, in peripheral and/or portal blood. Liquid biopsy performance was improved in portal blood only with CellSearch®, reaching 45% of PDAC identification (5/11) versus 10% (2/22) in peripheral blood. Importantly, combining CTC and GPC1-positive-exosome detection displayed 100% of sensitivity and 80% of specificity, with a negative predictive value of 100%. High levels of GPC1+-exosomes and/or CTC presence were significantly correlated with progression-free survival and with overall survival when CTC clusters were found. CONCLUSION This study is the first to evaluate combined CTC and exosome detection to diagnose resectable pancreatic cancers. Liquid biopsy combining several biomarkers could provide a rapid, reliable, noninvasive decision-making tool in early, potentially curable pancreatic cancer. Moreover, the prognostic value could select patients eligible for neoadjuvant treatment before surgery. This exploratory study deserves further validation.
Collapse
Affiliation(s)
- Etienne Buscail
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells, University Medical Centre of Montpellier, EA2415 Montpellier, France;
| | - Pascaline Quincy
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Thomas Cauvin
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Alexandre Chauvet
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Olivier Degrandi
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Charline Caumont
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Séverine Verdon
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
| | - Isabelle Lamrissi
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Isabelle Moranvillier
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Camille Buscail
- Nutritional Epidemiology Research Team (EREN), Paris 13 University, U1153 INSERM, U1125 Institut national de la recherche agronomique (INRA), Conservatoire national des arts et métiers (CNAM), Paris Cité Epidemiology and Statistics Research Center (CRESS), 93017 Bobigny, France;
| | - Marion Marty
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
| | - Christophe Laurent
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Véronique Vendrely
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - François Moreau-Gaudry
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Aurélie Bedel
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Sandrine Dabernat
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
- Correspondence: ; Tel.: +33-(0)5-5757-1374; Fax: +33-(0)5-5757-1374
| | - Laurence Chiche
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| |
Collapse
|
107
|
Liu P, Kong L, Jin H, Wu Y, Tan X, Song B. Differential secretome of pancreatic cancer cells in serum-containing conditioned medium reveals CCT8 as a new biomarker of pancreatic cancer invasion and metastasis. Cancer Cell Int 2019; 19:262. [PMID: 31632196 PMCID: PMC6788113 DOI: 10.1186/s12935-019-0980-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 09/27/2019] [Indexed: 12/22/2022] Open
Abstract
Background Pancreatic cancer is a malignancy with a very poor prognosis. The emergence of liquid biopsy is expected to achieve accurate early diagnosis through detection of tumor-derived secreted proteins in the blood. Early diagnosis and treatment of pancreatic cancer could help to improve prognosis. Methods The pretreatment approach of samples can have a major effect on downstream analysis. In this study, we used a pair of homologous pancreatic cancer cell supernatants with different capacities for invasion and metastasis to examine secreted proteins in the conditioned media without the removal of fetal bovine serum, namely through size exclusion chromatography combined with high-abundance protein affinity chromatography to enrich low-concentration protein, followed by mass spectrometry using triple dimethyl labeling. Identification of proteins was performed using an online public database and western blot. Results Mass spectrometry data revealed 77 proteins with quantitative properties, of which 12 proteins had over a 1.5-fold difference (in the supernatant of the highly invasive pancreatic cancer cell line PC-1.0, the expression of 8 proteins were increased and the expression of 4 proteins were decreased). Bioinformatics analysis results showed that CCT8, CTSL, SAA1, IGF2 are secreted via the exosome pathway. According to the literature, with the exception of CCT8, the other three proteins can be detected in blood samples of pancreatic cancer patients, and they can be used as prognostic markers. Western blot results were used to validate consistency with MS results. Conclusion This study found that CCT8 can be used as a liquid biopsy marker to assess the prognosis of pancreatic cancer patients.
Collapse
Affiliation(s)
- Peng Liu
- 11st Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang, 110004 China
| | - Lingming Kong
- 11st Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang, 110004 China
| | - Haoyi Jin
- 11st Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang, 110004 China
| | - Yunhao Wu
- 11st Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang, 110004 China
| | - Xiaodong Tan
- 11st Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang, 110004 China
| | - Bing Song
- 11st Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang, 110004 China.,2Cardiff Institute of Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, CF14 4XY UK
| |
Collapse
|
108
|
Zhou B, Wu D, Liu H, Du LT, Wang YS, Xu JW, Qiu FB, Hu SY, Zhan HX. Obesity and pancreatic cancer: An update of epidemiological evidence and molecular mechanisms. Pancreatology 2019; 19:941-950. [PMID: 31447281 DOI: 10.1016/j.pan.2019.08.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 07/04/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022]
Abstract
Despite advances in therapy and achievements in translational research, pancreatic cancer (PC) remains an invariably fatal malignancy. Risk factors that affect the incidence of PC include diabetes, smoking, obesity, chronic pancreatitis, and diet. The growing worldwide obesity epidemic is associated with an increased risk of the most common cancers, including PC. Chronic inflammation, hormonal effects, circulating adipokines, and adipocyte-mediated inflammatory and immunosuppressive microenvironment are involved in the association of obesity with PC. Herein, we systematically review the epidemiology of PC and the biological mechanisms that may account for this association. Included in this review is a discussion of adipokine-mediated inflammation, lipid metabolism, and the interactions of adipocytes with cancer cells. We consider the influence of bariatric surgery on the risk of PC risk as well as potential molecular targets of therapy. Our review leads us to conclude that targeting adipose tissue to achieve weight loss may represent a new therapeutic strategy for preventing and treating PC.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China; Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China
| | - Dong Wu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Han Liu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Lu-Tao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, 250033, China; Tumor Marker Detection Engineering Laboratory of Shandong Province, Jinan, Shandong Province, 250033, China
| | - Yun-Shan Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, 250033, China; Tumor Marker Detection Engineering Laboratory of Shandong Province, Jinan, Shandong Province, 250033, China
| | - Jian-Wei Xu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Fa-Bo Qiu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China; Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China
| | - San-Yuan Hu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Han-Xiang Zhan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China.
| |
Collapse
|
109
|
Pasquale V, Dugnani E, Liberati D, Marra P, Citro A, Canu T, Policardi M, Valla L, Esposito A, Piemonti L. Glucose metabolism during tumorigenesis in the genetic mouse model of pancreatic cancer. Acta Diabetol 2019; 56:1013-1022. [PMID: 30989379 DOI: 10.1007/s00592-019-01335-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 03/28/2019] [Indexed: 12/11/2022]
Abstract
AIM More than 40% of pancreatic ductal adenocarcinoma (PDAC) patients have glucose intolerance or diabetes. The association has led to two hypotheses: PDAC causes diabetes or diabetes shares risk factors for the development of PDAC. In order to elucidate the relationship between diabetes and PDAC, we investigated the glucose metabolism during tumorigenesis in the LSL-KrasG12D/+; LSL-Trp53R172H/+; and Pdx-1-Cre (KPC) mouse, a genetically engineered model of PDAC. METHODS Male and female KPCs have been fed with standard diet (SD) or high-fat diet (HFD). The imaging-based 4-class tumor staging was used to follow pancreatic cancer development. Not fasting glycemia, 4-h fasting glycemia, insulin, C-peptide, glucose tolerance after OGTT and abdominal fat volume were measured during tumorigenesis. RESULTS PDAC development did not lead to an overt diabetic phenotype or to any alterations in glucose tolerance in KPC fed with SD. Consumption of HFD induced higher body weight/abdominal fat volume and worsened glucose homeostasis both in control CRE mice and only in early tumorigenesis stages of the KPC mice, excluding that the cancer development itself acts as a trigger for the onset of dysmetabolic features. CONCLUSION Our data demonstrate that carcinogenesis in KPC mice is not associated with paraneoplastic diabetes.
Collapse
Affiliation(s)
- Valentina Pasquale
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Erica Dugnani
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Daniela Liberati
- Division of Genetics and Cell biology, Genomic Unit for the diagnosis of human pathologies, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Paolo Marra
- Department of Radiology, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Antonio Citro
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Tamara Canu
- Department of Radiology, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Martina Policardi
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Libera Valla
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Antonio Esposito
- Department of Radiology, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
110
|
Buscail E, Chauvet A, Quincy P, Degrandi O, Buscail C, Lamrissi I, Moranvillier I, Caumont C, Verdon S, Brisson A, Marty M, Chiche L, Laurent C, Vendrely V, Moreau-Gaudry F, Bedel A, Dabernat S. CD63-GPC1-Positive Exosomes Coupled with CA19-9 Offer Good Diagnostic Potential for Resectable Pancreatic Ductal Adenocarcinoma. Transl Oncol 2019; 12:1395-1403. [PMID: 31400579 PMCID: PMC6699195 DOI: 10.1016/j.tranon.2019.07.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 12/13/2022] Open
Abstract
Tumor-released extracellular vesicles (EVs) contain tumor-specific cargo distinguishing them from healthy EVs, and making them eligible as circulating biomarkers. Glypican 1 (GPC1)-positive exosome relevance as liquid biopsy elements is still debated. We carried out a prospective study to quantify GPC1-positive exosomes in sera from pancreatic ductal adenocarcinoma (PDAC) patients undergoing up-front surgery, as compared to controls including patients without cancer history and patients displaying pancreatic preneoplasic lesions. Sera were enriched in EVs, and exosomes were pulled down with anti-CD63 coupled magnetic beads. GPC1-positive bead percentages determined by flow cytometry were significantly higher in PDAC than in the control group. Diagnosis accuracy reached 78% (sensitivity 64% and specificity 90%), when results from peripheral and portal blood were combined. In association with echo-guided-ultrasound-fine-needle-aspiration (EUS-FNA) negative predictive value was 80% as compared to 33% for EUS-FNA only. This approach is clinically relevant as a companion test to the already available diagnostic tools, since patients with GPC1-positive exosomes in peripheral blood showed decreased tumor free survival.
Collapse
Affiliation(s)
- Etienne Buscail
- INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France; Université de Bordeaux, Bordeaux, France
| | - Alexandre Chauvet
- INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France; Université de Bordeaux, Bordeaux, France
| | - Pascaline Quincy
- INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France; Université de Bordeaux, Bordeaux, France
| | - Olivier Degrandi
- INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France; Université de Bordeaux, Bordeaux, France
| | - Camille Buscail
- Nutritional Epidemiology Research Team (EREN), Paris 13 University, U1153 INSERM, U1125 INRA, CNAM, CRESS) Bobigny, France
| | - Isabelle Lamrissi
- INSERM U1035, Bordeaux, France; Université de Bordeaux, Bordeaux, France
| | | | - Charline Caumont
- CHU de Bordeaux, Bordeaux, France; Université de Bordeaux, Bordeaux, France
| | | | - Alain Brisson
- Université de Bordeaux, Bordeaux, France; UMR-5248, CNRS, Talence, France
| | | | - Laurence Chiche
- INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France; Université de Bordeaux, Bordeaux, France
| | - Christophe Laurent
- INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France; Université de Bordeaux, Bordeaux, France
| | - Veronique Vendrely
- INSERM U1035, Bordeaux, France; Université de Bordeaux, Bordeaux, France
| | - François Moreau-Gaudry
- INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France; Université de Bordeaux, Bordeaux, France
| | - Aurelie Bedel
- INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France; Université de Bordeaux, Bordeaux, France
| | - Sandrine Dabernat
- INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France; Université de Bordeaux, Bordeaux, France.
| |
Collapse
|
111
|
Buscail E, Chiche L, Laurent C, Vendrely V, Denost Q, Denis J, Thumerel M, Lacorte JM, Bedel A, Moreau-Gaudry F, Dabernat S, Alix-Panabières C. Tumor-proximal liquid biopsy to improve diagnostic and prognostic performances of circulating tumor cells. Mol Oncol 2019; 13:1811-1826. [PMID: 31216108 PMCID: PMC6717761 DOI: 10.1002/1878-0261.12534] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/04/2019] [Accepted: 06/17/2019] [Indexed: 12/11/2022] Open
Abstract
Circulating tumor cell (CTC) detection and numeration are becoming part of the common clinical practice, especially for breast, colon, and prostate cancer. However, their paucity in peripheral blood samples is an obstacle for their identification. Several groups have tried to improve CTC recovery rate by developing highly sensitive cellular and molecular detection methods. However, CTCs are still difficult to detect in peripheral blood. Therefore, their recovery rate could be increased by obtaining blood samples from vessels close to the drainage territories of the invaded organ, when the anatomical situation is favorable. This approach has been tested mostly during tumor resection surgery, when the vessels nearest to the tumor are easily accessible. Moreover, radiological (including echo‐guided based and endovascular techniques) and/or endoscopic routes could be utilized to obtain CTC samples close to the tumor in a less invasive way than conventional biopsies. The purpose of this article is to summarize the available knowledge on CTC recovery from blood samples collected close to the tumor (i.e., in vessels located in the drainage area of the primary tumor or metastases). The relevance of such an approach for diagnostic and prognostic evaluations will be discussed, particularly for pancreatic ductal adenocarcinoma, colorectal adenocarcinoma, hepatocellular carcinoma, and non‐small‐cell lung cancer.
Collapse
Affiliation(s)
- Etienne Buscail
- INSERM U1035, Bordeaux, France.,CHU de Bordeaux, France.,Université de Bordeaux, France
| | - Laurence Chiche
- INSERM U1035, Bordeaux, France.,CHU de Bordeaux, France.,Université de Bordeaux, France
| | - Christophe Laurent
- INSERM U1035, Bordeaux, France.,CHU de Bordeaux, France.,Université de Bordeaux, France
| | - Véronique Vendrely
- INSERM U1035, Bordeaux, France.,CHU de Bordeaux, France.,Université de Bordeaux, France
| | | | - Jérôme Denis
- Laboratory of Rare Human Circulating Cells, University Medical Centre of Montpellier, France
| | | | - Jean-Marc Lacorte
- Laboratory of Rare Human Circulating Cells, University Medical Centre of Montpellier, France
| | - Aurélie Bedel
- INSERM U1035, Bordeaux, France.,CHU de Bordeaux, France.,Université de Bordeaux, France
| | | | - Sandrine Dabernat
- INSERM U1035, Bordeaux, France.,CHU de Bordeaux, France.,Université de Bordeaux, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells, University Medical Centre of Montpellier, France.,Service de Biochimie Endocrinienne et Oncologie, Hôpital Pitié Salpêtrière Assistance Publique Hôpitaux de Paris, France
| |
Collapse
|
112
|
Duan B, Hu X, Fan M, Xiong X, Han L, Wang Z, Tong D, Liu L, Wang X, Li W, Yang J, Huang C. RNA-Binding Motif Protein 6 is a Candidate Serum Biomarker for Pancreatic Cancer. Proteomics Clin Appl 2019; 13:e1900048. [PMID: 31207145 DOI: 10.1002/prca.201900048] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/26/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Early diagnosis is crucial to improve outcomes for pancreatic cancer patients (PC). The present study is designed to identify differently expressed peptides involved in PC as potential biomarkers. EXPERIMENTAL DESIGN The serum proteome of 22 PC patients, 12 pancreatitis patients (PP), and 45 healthy controls (HC) are analyzed using magnetic bead-based weak cation exchange (MB-WCX) and matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS). Next, a supervised neural network (SNN) algorithm model is established by ClinProTools and the candidate biomarker identified using liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS). Finally, the candidate biomarker is validated in tissue samples. RESULTS The SNN algorithm model discriminates PC from HC with 92.97% sensitivity and 94.55% specificity. Seventy-six differentially expressed peptides are identified, seven of which are significantly different among PC, PP, and HC (p < 0.05). Only one peak (m/z: 1466.99) tends to be upregulated in samples from HC, PP, and PC, which is identified as region of RNA-binding motif protein 6 (RBM6). In subsequent tissue analysis, it is verified that RBM6 expression is significantly higher in PC tissues than paracancerous tissue. CONCLUSIONS AND CLINICAL RELEVANCE The results indicate that RBM6 might serve as a candidate diagnostic biomarker for PC. CLINICAL RELEVANCE Methods used in this study could generate serum peptidome profiles of PC, PP, and HC, and present an approach to identify potential biomarkers for diagnosis of this malignancy.
Collapse
Affiliation(s)
- Baojun Duan
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China.,Department of Medical Oncology of Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Xiaoyan Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Meiyang Fan
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Xiaofan Xiong
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Lin Han
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Dongdong Tong
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Liying Liu
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Xiaofei Wang
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Wensheng Li
- Department of Pathology of Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Juan Yang
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Chen Huang
- Key Laboratory of Environment and Disease-Related Gene, Ministry of Education/Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Health Science Center, Xi'an, 710061, Shaanxi, China
| |
Collapse
|
113
|
Yang W, Liu H, Duan B, Xu X, Carmody D, Luo S, Walsh KM, Abbruzzese JL, Zhang X, Chen X, Wei Q. Three novel genetic variants in NRF2 signaling pathway genes are associated with pancreatic cancer risk. Cancer Sci 2019; 110:2022-2032. [PMID: 30972876 PMCID: PMC6550126 DOI: 10.1111/cas.14017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/06/2019] [Accepted: 04/07/2019] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PanC) is one of the most lethal solid malignancies, and metastatic PanC is often present at the time of diagnosis. Although several high- and low-penetrance genes have been implicated in PanC, their roles in carcinogenesis remain only partially elucidated. Because the nuclear factor erythroid2-related factor2 (NRF2) signaling pathway is involved in human cancers, we hypothesize that genetic variants in NRF2 pathway genes are associated with PanC risk. To test this hypothesis, we assessed associations between 31 583 common single nucleotide polymorphisms (SNP) in 164 NRF2-related genes and PanC risk using three published genome-wide association study (GWAS) datasets, which included 8474 cases and 6944 controls of European descent. We also carried out expression quantitative trait loci (eQTL) analysis to assess the genotype-phenotype correlation of the identified significant SNP using publicly available data in the 1000 Genomes Project. We found that three novel SNP (ie, rs3124761, rs17458086 and rs1630747) were significantly associated with PanC risk (P = 5.17 × 10-7 , 5.61 × 10-4 and 5.52 × 10-4 , respectively). Combined analysis using the number of unfavorable genotypes (NUG) of these three SNP suggested that carriers of two to three NUG had an increased risk of PanC (P < 0.0001), compared with those carrying zero to one NUG. Furthermore, eQTL analysis showed that both rs3124761 T and rs17458086 C alleles were associated with increased mRNA expression levels of SLC2A6 and SLC2A13, respectively (P < 0.05). In conclusion, genetic variants in NRF2 pathway genes could play a role in susceptibility to PanC, and further functional exploration of the underlying molecular mechanisms is warranted.
Collapse
Affiliation(s)
- Wenjun Yang
- Key Laboratory of Fertility Preservation and MaintenanceSchool of Basic Medicine and the General HospitalNingxia Medical UniversityYinchuanChina
- Cancer Research ProgramJulius L. Chambers Biomedical Biotechnology Research InstituteNorth Carolina Central UniversityDurham
| | - Hongliang Liu
- Duke Cancer InstituteDuke University Medical CenterDurham
- Department of Population Health SciencesDuke University School of MedicineDurham
| | - Bensong Duan
- Duke Cancer InstituteDuke University Medical CenterDurham
- Department of Population Health SciencesDuke University School of MedicineDurham
| | - Xinyuan Xu
- Duke Cancer InstituteDuke University Medical CenterDurham
- Department of Population Health SciencesDuke University School of MedicineDurham
| | - Dennis Carmody
- Duke Cancer InstituteDuke University Medical CenterDurham
- Department of Population Health SciencesDuke University School of MedicineDurham
| | - Sheng Luo
- Department of Biostatistics and BioinformaticsDuke University School of MedicineDurham
| | - Kyle M. Walsh
- Duke Cancer InstituteDuke University Medical CenterDurham
- Department of NeurosurgeryDuke University School of MedicineDurham
| | - James L. Abbruzzese
- Duke Cancer InstituteDuke University Medical CenterDurham
- Department of Medicine, Population Health SciencesDuke University School of MedicineDurham
| | - Xuefeng Zhang
- Duke Cancer InstituteDuke University Medical CenterDurham
- Department of PathologyDuke University School of MedicineDurham
| | - Xiaoxin Chen
- Cancer Research ProgramJulius L. Chambers Biomedical Biotechnology Research InstituteNorth Carolina Central UniversityDurham
| | - Qingyi Wei
- Duke Cancer InstituteDuke University Medical CenterDurham
- Department of Population Health SciencesDuke University School of MedicineDurham
- Department of Medicine, Population Health SciencesDuke University School of MedicineDurham
| |
Collapse
|
114
|
Primary and Secondary Prevention of Pancreatic Cancer. CURR EPIDEMIOL REP 2019. [DOI: 10.1007/s40471-019-00189-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
115
|
BRM transcriptionally regulates miR-302a-3p to target SOCS5/STAT3 signaling axis to potentiate pancreatic cancer metastasis. Cancer Lett 2019; 449:215-225. [DOI: 10.1016/j.canlet.2019.02.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/03/2019] [Accepted: 02/14/2019] [Indexed: 12/11/2022]
|
116
|
Yoshida A, Kitayama Y, Kiguchi K, Yamada T, Akasaka H, Sasaki R, Takeuchi T. Gold Nanoparticle-Incorporated Molecularly Imprinted Microgels as Radiation Sensitizers in Pancreatic Cancer. ACS APPLIED BIO MATERIALS 2019; 2:1177-1183. [DOI: 10.1021/acsabm.8b00766] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Aoi Yoshida
- Graduate School of Engineering, Kobe University, 1-1, Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Yukiya Kitayama
- Graduate School of Engineering, Kobe University, 1-1, Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
- Medical Device Fabrication Engineering Center, Graduate School of Engineering, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Kentaro Kiguchi
- Graduate School of Engineering, Kobe University, 1-1, Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Takuya Yamada
- Graduate School of Engineering, Kobe University, 1-1, Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Hiroaki Akasaka
- Division of Radiation Oncology, Kobe University Hospital, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Ryohei Sasaki
- Division of Radiation Oncology, Kobe University Hospital, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
- Medical Device Fabrication Engineering Center, Graduate School of Engineering, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Toshifumi Takeuchi
- Graduate School of Engineering, Kobe University, 1-1, Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
- Medical Device Fabrication Engineering Center, Graduate School of Engineering, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| |
Collapse
|
117
|
Pang Y, Holmes MV, Chen Z, Kartsonaki C. A review of lifestyle, metabolic risk factors, and blood-based biomarkers for early diagnosis of pancreatic ductal adenocarcinoma. J Gastroenterol Hepatol 2019; 34:330-345. [PMID: 30550622 PMCID: PMC6378598 DOI: 10.1111/jgh.14576] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/09/2018] [Accepted: 12/12/2018] [Indexed: 12/28/2022]
Abstract
We aimed to review the epidemiologic literature examining lifestyle and metabolic risk factors, and blood-based biomarkers including multi-omics (genomics, proteomics, and metabolomics) and to discuss how these predictive markers can inform early diagnosis of pancreatic ductal adenocarcinoma (PDAC). A search of the PubMed database was conducted in June 2018 to review epidemiologic studies of (i) lifestyle and metabolic risk factors for PDAC, genome-wide association studies, and risk prediction models incorporating these factors and (ii) blood-based biomarkers for PDAC (conventional diagnostic markers, metabolomics, and proteomics). Prospective cohort studies have reported at least 20 possible risk factors for PDAC, including smoking, heavy alcohol drinking, adiposity, diabetes, and pancreatitis, but the relative risks and population attributable fractions of individual risk factors are small (mostly < 10%). High-throughput technologies have continued to yield promising genetic, metabolic, and protein biomarkers in addition to conventional biomarkers such as carbohydrate antigen 19-9. Nonetheless, most studies have utilized a hospital-based case-control design, and the diagnostic accuracy is low in studies that collected pre-diagnostic samples. Risk prediction models incorporating lifestyle and metabolic factors as well as other clinical parameters have shown good discrimination and calibration. Combination of traditional risk factors, genomics, and blood-based biomarkers can help identify high-risk populations and inform clinical decisions. Multi-omics investigations can provide valuable insights into disease etiology, but prospective cohort studies that collect pre-diagnostic samples and validation in independent studies are warranted.
Collapse
Affiliation(s)
- Yuanjie Pang
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Michael V Holmes
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- National Institute for Health Research Oxford Biomedical Research CentreOxford University HospitalOxfordUK
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Christiana Kartsonaki
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| |
Collapse
|
118
|
Zhou X, Wang X, Duan J, Sun W, Chen Z, Li Q, Ou Z, Jiang G, Ren X, Liu S. HBXIP protein overexpression predicts the poor prognosis of pancreatic ductal adenocarcinomas. Pathol Res Pract 2019; 215:343-346. [PMID: 30583814 DOI: 10.1016/j.prp.2018.12.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/23/2018] [Accepted: 12/11/2018] [Indexed: 01/17/2023]
Abstract
BACKGROUND Hepatitis B virus X-interacting protein (HBXIP) is associated with a variety of tumors. The purpose of this study was to investigate the clinicopathological significance of HBXIP expression in pancreatic ductal adenocarcinoma (PDAC) and to explore its potential as a biomarker for PDAC. METHODS Immunohistochemical (IHC) staining was performed on 126 PDAC tissues, 36 paraneoplastic tissues and 22 normal pancreatic tissues. The relationship between high levels of HBXIP expression and pathological features of PDAC patients was evaluated by chi-squared values. RESULTS The positive rate of HBXIP protein in PDAC tissues was 85.7% (108/126), which was significantly higher than that of adjacent pancreatic tissue (41.7%, 15/36) and normal pancreas (18.2%, 4/22). In addition, strong positive expression of HBXIP was associated with tumor size, positive lymph node metastasis, clinical stage and 80-month overall survival. Patient's age, gender, degree of differentiation, Ki-67 expression index, and calcification were, however, not associated with high levels of HBXIP expression. CONCLUSIONS We present association between HBXIP expression and the pathological features of patients with PDAC.
Collapse
Affiliation(s)
- Xingzhi Zhou
- Department of Pathology, Medical College, Dalian University, Dalian 116622, Liaoning, China; Department of Biology, Life Science and Technology College, Dalian University, Dalian 116622, Liaoning, China
| | - Xuanyu Wang
- Department of Pathology, Medical College, Dalian University, Dalian 116622, Liaoning, China
| | - Jiahong Duan
- Department of Pathology, Medical College, Dalian University, Dalian 116622, Liaoning, China
| | - Wenxin Sun
- Department of Pathology, Medical College, Dalian University, Dalian 116622, Liaoning, China
| | - Zhuo Chen
- Department of Pathology, Medical College, Dalian University, Dalian 116622, Liaoning, China
| | - Qiulan Li
- Department of Pathology, Medical College, Dalian University, Dalian 116622, Liaoning, China
| | - Zitong Ou
- Department of Pathology, Medical College, Dalian University, Dalian 116622, Liaoning, China
| | - Ge Jiang
- Department of Biology, Life Science and Technology College, Dalian University, Dalian 116622, Liaoning, China
| | - Xin Ren
- Department of Pathology, Medical College, Dalian University, Dalian 116622, Liaoning, China.
| | - Shuangping Liu
- Department of clinical laboratory, Xinhua Hospital Affiliated to Dalian University, Dalian, 116021, China; Department of Pathology, Medical College, Dalian University, Dalian 116622, Liaoning, China.
| |
Collapse
|
119
|
Sattar Z, Ali S, Hussain I, Sattar F, Hussain S, Ahmad S. Diagnosis of pancreatic cancer. THERANOSTIC APPROACH FOR PANCREATIC CANCER 2019:51-68. [DOI: 10.1016/b978-0-12-819457-7.00002-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
120
|
Cao J, Li J, Sun L, Qin T, Xiao Y, Chen K, Qian W, Duan W, Lei J, Ma J, Ma Q, Han L. Hypoxia-driven paracrine osteopontin/integrin αvβ3 signaling promotes pancreatic cancer cell epithelial-mesenchymal transition and cancer stem cell-like properties by modulating forkhead box protein M1. Mol Oncol 2018; 13:228-245. [PMID: 30367545 PMCID: PMC6360359 DOI: 10.1002/1878-0261.12399] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/05/2018] [Accepted: 09/19/2018] [Indexed: 12/30/2022] Open
Abstract
Pancreatic stellate cells (PSCs), a key component of the tumor microenvironment, contribute to tumor invasion, metastasis, and chemoresistance. Osteopontin (OPN), a phosphorylated glycoprotein, is overexpressed in pancreatic cancer. However, OPN expression in PSCs and its potential roles in tumor–stroma interactions remain unclear. The present study first showed that OPN is highly expressed and secreted in activated PSCs driven by hypoxia, and this process is in a ROS‐dependent manner; in addition, OPN was shown to be involved in the PSC‐induced epithelial–mesenchymal transition (EMT) and cancer stem cell (CSC)‐like properties of pancreatic cancer cells (PCCs). Mechanistically, OPN from activated PSCs interacts with the transmembrane receptor integrin αvβ3 on PCCs to upregulate forkhead box protein M1 (FOXM1) expression and induce malignant phenotypes of PCCs. Moreover, the Akt and Erk pathways participate in OPN/integrin αvβ3 axis‐induced FOXM1 expression of PCCs. Our further analysis showed that OPN and FOXM1 are significantly upregulated in pancreatic cancer tissues and are associated with poor clinical outcome, indicating that OPN and FOXM1 might be considered as diagnostic and prognostic biomarkers for patients with pancreatic cancer. In conclusion, we show here for the first time that OPN promotes the EMT and CSC‐like properties of PCCs by activating the integrin αvβ3‐Akt/Erk‐FOXM1 cascade in a paracrine manner, suggesting that targeting the tumor microenvironment represents a promising therapeutic strategy in pancreatic cancer.
Collapse
Affiliation(s)
- Junyu Cao
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Jie Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Liankang Sun
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Tao Qin
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Ying Xiao
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Ke Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Weikun Qian
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Jianjun Lei
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Jiguang Ma
- Department of Anesthesiology, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Liang Han
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| |
Collapse
|
121
|
The 150 most important questions in cancer research and clinical oncology series: questions 94-101 : Edited by Cancer Communications. Cancer Commun (Lond) 2018; 38:69. [PMID: 30477575 PMCID: PMC6257962 DOI: 10.1186/s40880-018-0341-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
Since the beginning of 2017, Cancer Communications (former title: Chinese Journal of Cancer) has published a series of important questions regarding cancer research and clinical oncology, to provide an enhanced stimulus for cancer research, and to accelerate collaborations between institutions and investigators. In this edition, the following 8 valuable questions are presented. Question 94. The origin of tumors: time for a new paradigm? Question 95. How can we accelerate the identification of biomarkers for the early detection of pancreatic ductal adenocarcinoma? Question 96. Can we improve the treatment outcomes of metastatic pancreatic ductal adenocarcinoma through precision medicine guided by a combination of the genetic and proteomic information of the tumor? Question 97. What are the parameters that determine a competent immune system that gives a complete response to cancers after immune induction? Question 98. Is high local concentration of metformin essential for its anti-cancer activity? Question 99. How can we monitor the emergence of cancer cells anywhere in the body through plasma testing? Question 100. Can phytochemicals be more specific and efficient at targeting P-glycoproteins to overcome multi-drug resistance in cancer cells? Question 101. Is cell migration a selectable trait in the natural evolution of carcinoma?
Collapse
|
122
|
McGuigan A, Kelly P, Turkington RC, Jones C, Coleman HG, McCain RS. Pancreatic cancer: A review of clinical diagnosis, epidemiology, treatment and outcomes. World J Gastroenterol 2018; 24:4846-4861. [PMID: 30487695 PMCID: PMC6250924 DOI: 10.3748/wjg.v24.i43.4846] [Citation(s) in RCA: 1187] [Impact Index Per Article: 169.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 10/19/2018] [Accepted: 10/27/2018] [Indexed: 02/06/2023] Open
Abstract
This review aims to outline the most up-to-date knowledge of pancreatic adenocarcinoma risk, diagnostics, treatment and outcomes, while identifying gaps that aim to stimulate further research in this understudied malignancy. Pancreatic adenocarcinoma is a lethal condition with a rising incidence, predicted to become the second leading cause of cancer death in some regions. It often presents at an advanced stage, which contributes to poor five-year survival rates of 2%-9%, ranking firmly last amongst all cancer sites in terms of prognostic outcomes for patients. Better understanding of the risk factors and symptoms associated with this disease is essential to inform both health professionals and the general population of potential preventive and/or early detection measures. The identification of high-risk patients who could benefit from screening to detect pre-malignant conditions such as pancreatic intraepithelial neoplasia, intraductal papillary mucinous neoplasms and mucinous cystic neoplasms is urgently required, however an acceptable screening test has yet to be identified. The management of pancreatic adenocarcinoma is evolving, with the introduction of new surgical techniques and medical therapies such as laparoscopic techniques and neo-adjuvant chemoradiotherapy, however this has only led to modest improvements in outcomes. The identification of novel biomarkers is desirable to move towards a precision medicine era, where pancreatic cancer therapy can be tailored to the individual patient, while unnecessary treatments that have negative consequences on quality of life could be prevented for others. Research efforts must also focus on the development of new agents and delivery systems. Overall, considerable progress is required to reduce the burden associated with pancreatic cancer. Recent, renewed efforts to fund large consortia and research into pancreatic adenocarcinoma are welcomed, but further streams will be necessary to facilitate the momentum needed to bring breakthroughs seen for other cancer sites.
Collapse
Affiliation(s)
- Andrew McGuigan
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast BT9 7AE, United Kingdom
| | - Paul Kelly
- Department of Pathology, Royal Victoria Hospital, Belfast BT12 6BA, United Kingdom
| | - Richard C Turkington
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast BT9 7AE, United Kingdom
| | - Claire Jones
- Department of Hepatobiliary Surgery, Mater Hospital, Belfast BT14 6AB, United Kingdom
| | - Helen G Coleman
- Centre for Public Health, Queen’s University Belfast, Belfast BT12 6BJ, United Kingdom
| | - R Stephen McCain
- Department of Hepatobiliary Surgery, Mater Hospital, Belfast BT14 6AB, United Kingdom
- Centre for Public Health, Queen’s University Belfast, Belfast BT12 6BJ, United Kingdom
| |
Collapse
|
123
|
Cao J, Ma J, Sun L, Li J, Qin T, Zhou C, Cheng L, Chen K, Qian W, Duan W, Wang F, Wu E, Wang Z, Ma Q, Han L. Targeting glypican-4 overcomes 5-FU resistance and attenuates stem cell-like properties via suppression of Wnt/β-catenin pathway in pancreatic cancer cells. J Cell Biochem 2018; 119:9498-9512. [PMID: 30010221 DOI: 10.1002/jcb.27266] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/12/2018] [Accepted: 06/22/2018] [Indexed: 12/12/2022]
Abstract
The existences of cancer stem cells in patients with pancreatic cancer are considered as pivotal factors contributing to chemoresistance and disease relapse. Glypican-4 (GPC4) is one of the members of the glypicans family, which underlies human congenital malformations and multiple diseases. However, its potential biological function in pancreatic cancer still remains elusive. In this study, we are the first to demonstrate that GPC4 was involved in 5-fluorouracil (5-FU) resistance and pancreatic cancer stemness through comprehensive bioinformatical analysis. Functional experiments showed that knockdown of GPC4 sensitized pancreatic cancer cells to 5-FU and attenuated stem cell-like properties. In terms of mechanism research, knockdown of GPC4 suppressed the activation of Wnt/β-catenin pathway and its downstream targets. Furthermore, the expression of GPC4 was significantly upregulated in pancreatic cancer tissues compared with normal tissues and remarkably correlated with patients' overall survival according to the data derived from the Cancer Genome Atlas database. Taken together, our results suggest that GPC4 is a key regulator in chemoresistance and pancreatic cancer stemness. Thus, targeting GPC4 may serve as a promising strategy for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Junyu Cao
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jiguang Ma
- Department of Anesthesiology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Liankang Sun
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jie Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Tao Qin
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Cancan Zhou
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Liang Cheng
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Ke Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Weikun Qian
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Fengfei Wang
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas
- Neuroscience Institute, Baylor Scott & White Health, Temple, Texas
- Department of Surgery, Texas A & M University College of Medicine, College Station, Texas
- Department of Neurology, Baylor Scott & White Health, Temple, Texas
| | - Erxi Wu
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas
- Neuroscience Institute, Baylor Scott & White Health, Temple, Texas
- Department of Surgery, Texas A & M University College of Medicine, College Station, Texas
- Department of Pharmaceutical Sciences, Texas A & M University College of Pharmacy, College Station, Texas
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Liang Han
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
124
|
Samandari M, Julia MG, Rice A, Chronopoulos A, Del Rio Hernandez AE. Liquid biopsies for management of pancreatic cancer. Transl Res 2018; 201:98-127. [PMID: 30118658 DOI: 10.1016/j.trsl.2018.07.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/17/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is one of the main causes of cancer-related deaths worldwide. It is asymptomatic at an early stage, and most diagnosis occurs when the disease is already at a late stage, by which time the tumor is nonresectable. In order to increase the overall survival of patients with pancreatic cancer, as well as to decrease the cancer burden, it is necessary to perform early diagnosis, prognosis stratifications and cancer monitoring using accurate, minimally invasive, and cost-effective methods. Liquid biopsies seek to detect tumor-associated biomarkers in a variety of extractable body fluids and can help to monitor treatment response and disease progression, and even predict patient outcome. In patients with pancreatic cancer, tumor-derived materials, primarily circulating tumor DNA, circulating tumor cells and exosomes, are being studied for inclusion in the management of the disease. This review focuses on describing the biology of these biomarkers, methods for their enrichment and detection, as well as their potential for clinical application. Moreover, we discuss the future direction of liquid biopsies and introduce how they can be exploited toward point of care personalized medicine for the management of pancreatic cancer.
Collapse
Affiliation(s)
- Mohamadmahdi Samandari
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - María Gil Julia
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Antonios Chronopoulos
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Armando E Del Rio Hernandez
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom.
| |
Collapse
|
125
|
Benzel J, Fendrich V. Familial Pancreatic Cancer. Oncol Res Treat 2018; 41:611-618. [PMID: 30269130 DOI: 10.1159/000493473] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/04/2018] [Indexed: 12/13/2022]
Abstract
Familial pancreatic cancer accounts for 10% of all patients with pancreatic cancer. Because the 5-year survival rate of pancreatic cancer is only 7%, screening programs for high-risk individuals are essential and might be advantageous. Pancreatic ductal adenocarcinoma mostly shows symptoms at an advanced state and treatment is not efficient enough to cure most patients. People with hereditary tumor syndromes or their affected relatives can also be included in such screening programs. Besides the collection of data to investigate the background of the disease, these screening programs aim to diagnose and treat precursor lesions so that more dangerous, invasive lesions are prevented. These precursor lesions can be pancreatic intraepithelial neoplasia, intraductal papillary mucinous neoplasm, and mucinous cystic neoplasm. This review summarizes the latest knowledge of pancreatic screening programs, shows the procedure of pancreatic cancer screening, and gives an overview of current guidelines.
Collapse
|
126
|
Dankner R, Freedman LS, Gerstein HC, Roth J, Keinan-Boker L. Newly diagnosed type 2 diabetes may serve as a potential marker for pancreatic cancer. Diabetes Metab Res Rev 2018; 34:e3018. [PMID: 29673046 DOI: 10.1002/dmrr.3018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 01/22/2023]
Abstract
Pancreatic cancer has an extremely highly case fatality. Diabetes is a well-established strong risk factor for pancreatic cancer. Compared with a nondiabetic population, we previously reported a 15- and 14-fold greater risk for detecting pancreatic cancer during the first year after diagnosing diabetes in adult women and men, respectively, which dropped during the second year to 5.4-fold and 3.5-fold, respectively, and stabilized around 3-fold for the rest of the 11-year follow-up in our historical cohort. The population attributable risk during the 11-year period was 13.3% and 14.1% in prevalent diabetic women and men, respectively. This means that one out of about every 8 patients diagnosed with pancreatic cancer has been previously diagnosed with diabetes. The globally high prevalence of diabetes and the aggravating implications of a delayed pancreatic cancer diagnosis call for newly-onset diabetes to be considered a potential marker for an underlying pancreatic cancer and addressed accordingly.
Collapse
Affiliation(s)
- Rachel Dankner
- The Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, School of Public Health, Department of Epidemiology and Preventive Medicine, Tel Aviv University, Tel Aviv, Israel
- Patient Oriented Research, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Laurence S Freedman
- The Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, School of Public Health, Department of Epidemiology and Preventive Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hertzel C Gerstein
- Department of Medicine and Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada
| | - Jesse Roth
- Laboratory of Diabetes and Diabetes-Related Disorders, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
- Hofstra Northwell School of Medicine, Hempstead, NY, USA
- Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA
| | - Lital Keinan-Boker
- The Israel Center for Disease Control, Israel Ministry of Health, Jerusalem, Israel
- School of Public Health, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
127
|
Zhang Y, Huang S, Li P, Chen Q, Li Y, Zhou Y, Wang L, Kang M, Zhang B, Yang B, Dong X, Wu Y. Pancreatic cancer-derived exosomes suppress the production of GIP and GLP-1 from STC-1 cells in vitro by down-regulating the PCSK1/3. Cancer Lett 2018; 431:190-200. [PMID: 29800682 DOI: 10.1016/j.canlet.2018.05.027] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/25/2018] [Accepted: 05/17/2018] [Indexed: 01/08/2023]
Abstract
One hallmark of pancreatic cancer (PC) is the high prevalence of pancreatic cancer-associated diabetes mellitus (PC-DM), but the mechanisms remain to be elucidated. Patients with PC who are diagnosed with new-onset diabetes/prediabetes have recently been shown to display significantly lower levels of glucose-dependent insulinotropic peptide (GIP) secreted mainly by enteroendocrine cells. We hypothesized that PC-derived exosomes are responsible for the decreased levels of incretins in patients with PC-DM. In this study, exosomes were successfully isolated from PANC-1, MIA PaCa-2 and SW620 cells and characterized. Only the exosomes from MIA PaCa-2 cells (Exo-Mia) reduce the production of GIP and glucagon-like peptide-1 (GLP-1) from STC-1 cells in vitro in a concentration- and time-dependent manner. Moreover, Exo-Mia increased the levels of the Gip and proglucagon mRNAs and decreased the expression of proprotein convertase subtilisin/kexin type 1/3 (PCSK1/3), which is responsible for the post-translational processing of Gip and proglucagon. Furthermore, differentially expressed exosomal miRNAs (miR-6796-3p, miR-6763-5p, miR-4750-3p and miR-197-3p) were identified and considered to be responsible for the inhibitory effects on GIP and GLP-1 production. To further determine the approach of cancer-derived exosomes reaching enteroendocrine cells, we analyzed the uptake and distribution of exosomes in animal model. It was observed that exosomes infused into the intestinal cavity were more easily internalized by the intestinal epithelium than exosomes injected into blood. In conclusion, pancreatic cancer-derived exosomes (Exo-Mia) suppress the synthesis of GIP and GLP-1 from STC-1 cells in vitro by down-regulating the PCSK1/3. Moreover, it may be the pancreatic juice that transport cancer-derived exosomes to target cells (K and L cells) in the gut.
Collapse
Affiliation(s)
- Yuefeng Zhang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Shifei Huang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Pengping Li
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Qing Chen
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Yongzhou Li
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Yizhao Zhou
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Lantian Wang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Muxing Kang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Bo Zhang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Bin Yang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Xin Dong
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Department of Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, PR China
| | - Yulian Wu
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
128
|
Jiao Y, Jin D, Jiang F, Liu J, Qu L, Ni W, Liu Z, Lu C, Ni R, Zhu J, Xiao M. Characterization and proteomic profiling of pancreatic cancer‐derived serum exosomes. J Cell Biochem 2018; 120:988-999. [PMID: 30160795 DOI: 10.1002/jcb.27465] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/25/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Yu‐J. Jiao
- Department of Gastroenterology Affiliated Hospital of Nantong University Nantong Jiangsu China
- Medical College, Nantong University Nantong China
| | - Dan‐D. Jin
- Department of Gastroenterology Affiliated Hospital of Nantong University Nantong Jiangsu China
- Medical College, Nantong University Nantong China
| | - Feng Jiang
- Department of Gastroenterology Affiliated Hospital of Nantong University Nantong Jiangsu China
| | - Jin‐X. Liu
- Department of Gastroenterology Affiliated Hospital of Nantong University Nantong Jiangsu China
| | - Li‐S. Qu
- Department of Gastroenterology Affiliated Hospital of Nantong University Nantong Jiangsu China
| | - Wen‐K. Ni
- Department of Gastroenterology Affiliated Hospital of Nantong University Nantong Jiangsu China
| | - Zhao‐X. Liu
- Department of Gastroenterology Affiliated Hospital of Nantong University Nantong Jiangsu China
| | - Cui‐H. Lu
- Department of Gastroenterology Affiliated Hospital of Nantong University Nantong Jiangsu China
| | - Run‐Z. Ni
- Department of Gastroenterology Affiliated Hospital of Nantong University Nantong Jiangsu China
| | - Jing Zhu
- Department of Gastroenterology Affiliated Hospital of Nantong University Nantong Jiangsu China
| | - Ming‐B. Xiao
- Department of Gastroenterology Affiliated Hospital of Nantong University Nantong Jiangsu China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University Nantong Jiangsu China
| |
Collapse
|
129
|
Kunovsky L, Tesarikova P, Kala Z, Kroupa R, Kysela P, Dolina J, Trna J. The Use of Biomarkers in Early Diagnostics of Pancreatic Cancer. Can J Gastroenterol Hepatol 2018; 2018:5389820. [PMID: 30186820 PMCID: PMC6112218 DOI: 10.1155/2018/5389820] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/27/2018] [Accepted: 08/06/2018] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid malignancies with increasing incidence. The poor prognosis is due to the aggressive nature of the tumor, late detection, and the resistance to chemotherapy and radiotherapy. A radical surgery procedure is the only treatment that has been shown to improve the 5-year survival rate to 20-25%. However, the majority of patients (80-85%) are diagnosed with locally advanced or metastatic disease and just 15-20% patients are diagnosed in an early stage allowing them to undergo the potentially curative surgical resection. The early detection of PDAC without the use of invasive methods is challenging and discovery of a cost-effective biomarker with high specificity and sensitivity could significantly improve the treatment and survival in these patients. In this review, we summarize current and newly examined biomarkers in early PDAC detection.
Collapse
Affiliation(s)
- Lumir Kunovsky
- Department of Gastroenterology, University Hospital Brno Bohunice, Faculty of Medicine, Masaryk University, Czech Republic
- Department of Surgery, University Hospital Brno Bohunice, Faculty of Medicine, Masaryk University, Czech Republic
| | - Pavla Tesarikova
- Department of Internal Medicine, Hospital Boskovice, Czech Republic
| | - Zdenek Kala
- Department of Surgery, University Hospital Brno Bohunice, Faculty of Medicine, Masaryk University, Czech Republic
| | - Radek Kroupa
- Department of Gastroenterology, University Hospital Brno Bohunice, Faculty of Medicine, Masaryk University, Czech Republic
| | - Petr Kysela
- Department of Surgery, University Hospital Brno Bohunice, Faculty of Medicine, Masaryk University, Czech Republic
| | - Jiri Dolina
- Department of Gastroenterology, University Hospital Brno Bohunice, Faculty of Medicine, Masaryk University, Czech Republic
| | - Jan Trna
- Department of Gastroenterology, University Hospital Brno Bohunice, Faculty of Medicine, Masaryk University, Czech Republic
- Department of Internal Medicine, Hospital Boskovice, Czech Republic
| |
Collapse
|
130
|
Giulietti M, Righetti A, Principato G, Piva F. LncRNA co-expression network analysis reveals novel biomarkers for pancreatic cancer. Carcinogenesis 2018; 39:1016-1025. [PMID: 29796634 DOI: 10.1093/carcin/bgy069] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/30/2018] [Accepted: 05/21/2018] [Indexed: 12/19/2022] Open
Abstract
High mortality and low survival rates for pancreatic ductal adenocarcinoma (PDAC) mainly result from the delay in diagnosis and treatment. Therefore, there is an urgent need to identify early PDAC biomarkers and new therapeutic targets. In this study, we applied a commonly used systems biology approach, the weighted gene co-expression network analysis (WGCNA), on lncRNA expression data. Eleven lncRNAs, namely A2M-AS1, DLEU2, LINC01133, LINC00675, MIR155HG, SLC25A25-AS1, LINC01857, LOC642852 (LINC00205), ITGB2-AS1, TSPOAP1-AS1 and PSMB8-AS1 have been identified and validated on an independent PDAC expression dataset. Furthermore, we characterized them by functional and pathway enrichment analysis and identified which lncRNAs showed differential expression, differential promoter methylation levels and copy number alterations between normal and PDAC samples. Finally, we also performed a survival analysis and identified A2M-AS1, LINC01133, LINC00205 and TSPOAP1-AS1 as prognostic biomarkers for PDAC. Interestingly, although only a few cancer-associated lncRNAs have been functionally characterized, LINC00675 and LINC01133 lncRNAs have already been demonstrated to be involved in PDAC development and progression. Therefore, our results provide new potential diagnostic/prognostic biomarkers and therapeutic targets for PDAC that deserve to be further investigated. Moreover, these lncRNAs may improve the understanding about molecular pathogenesis of PDAC.
Collapse
Affiliation(s)
- Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Alessandra Righetti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Giovanni Principato
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
131
|
Xu JW, Wang L, Cheng YG, Zhang GY, Hu SY, Zhou B, Zhan HX. Immunotherapy for pancreatic cancer: A long and hopeful journey. Cancer Lett 2018; 425:143-151. [PMID: 29605510 DOI: 10.1016/j.canlet.2018.03.040] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/28/2018] [Accepted: 03/26/2018] [Indexed: 12/11/2022]
Abstract
Multiple therapeutic strategies have been developed to treat pancreatic cancer. However, the outcomes of these approaches are disappointing. Due to deeper understandings of the pivotal roles of the immune system in pancreatic cancer tumorigenesis and progression, novel therapeutic strategies based on immune cells and the tumor microenvironment are being investigated. Some of these approaches, such as checkpoint inhibitors, chimeric antigen receptor T-cell therapy, and BiTE antibodies, have achieved exciting outcomes in preclinical and clinical trials. The current review describes the roles of immune cells and the immunosuppressive microenvironment in the development of pancreatic cancer, as well as the preclinical and clinical outcomes and benefits of recent immunotherapeutic approaches, which may help us further disclose the mechanisms of pancreatic cancer progression and the dialectical views of feasibility and effectiveness of immunotherapy in treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Jian-Wei Xu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Lei Wang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Yu-Gang Cheng
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Guang-Yong Zhang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - San-Yuan Hu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Bin Zhou
- Department of Hepatopancreatobiliary Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China.
| | - Han-Xiang Zhan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China.
| |
Collapse
|
132
|
Llop E, Guerrero PE, Duran A, Barrabés S, Massaguer A, Ferri MJ, Albiol-Quer M, de Llorens R, Peracaula R. Glycoprotein biomarkers for the detection of pancreatic ductal adenocarcinoma. World J Gastroenterol 2018; 24:2537-2554. [PMID: 29962812 PMCID: PMC6021768 DOI: 10.3748/wjg.v24.i24.2537] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/04/2018] [Accepted: 06/09/2018] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PaC) shows a clear tendency to increase in the next years and therefore represents an important health and social challenge. Currently, there is an important need to find biomarkers for PaC early detection because the existing ones are not useful for that purpose. Recent studies have indicated that there is a large window of time for PaC early detection, which opens the possibility to find early biomarkers that could greatly improve the dismal prognosis of this tumor. The present manuscript reviews the state of the art of the existing PaC biomarkers. It focuses on the anomalous glycosylation process and its role in PaC. Glycan structures of glycoconjugates such as glycoproteins are modified in tumors and these modifications can be detected in biological fluids of the cancer patients. Several studies have found serum glycoproteins with altered glycan chains in PaC patients, but they have not shown enough specificity for PaC. To find more specific cancer glycoproteins we propose to analyze the glycan moieties of a battery of glycoproteins that have been reported to increase in PaC tissues and that can also be found in serum. The combination of these new candidate glycoproteins with their aberrant glycosylation together with the existing biomarkers could result in a panel, which would expect to give better results as a new tool for early diagnosis of PaC and to monitor the disease.
Collapse
Affiliation(s)
- Esther Llop
- Department of Biology, Biochemistry and Molecular Biology Unit, University of Girona, Girona 17003, Spain
- Biomedical Research Institute of Girona (IdIBGi). Parc Hospitalari Martí i Julià-Edifici M2, Salt 17190, Spain
| | - Pedro E Guerrero
- Department of Biology, Biochemistry and Molecular Biology Unit, University of Girona, Girona 17003, Spain
- Biomedical Research Institute of Girona (IdIBGi). Parc Hospitalari Martí i Julià-Edifici M2, Salt 17190, Spain
| | - Adrià Duran
- Department of Biology, Biochemistry and Molecular Biology Unit, University of Girona, Girona 17003, Spain
- Biomedical Research Institute of Girona (IdIBGi). Parc Hospitalari Martí i Julià-Edifici M2, Salt 17190, Spain
| | - Sílvia Barrabés
- Department of Biology, Biochemistry and Molecular Biology Unit, University of Girona, Girona 17003, Spain
- Biomedical Research Institute of Girona (IdIBGi). Parc Hospitalari Martí i Julià-Edifici M2, Salt 17190, Spain
| | - Anna Massaguer
- Department of Biology, Biochemistry and Molecular Biology Unit, University of Girona, Girona 17003, Spain
- Biomedical Research Institute of Girona (IdIBGi). Parc Hospitalari Martí i Julià-Edifici M2, Salt 17190, Spain
| | - María José Ferri
- Department of Biology, Biochemistry and Molecular Biology Unit, University of Girona, Girona 17003, Spain
- Biomedical Research Institute of Girona (IdIBGi). Parc Hospitalari Martí i Julià-Edifici M2, Salt 17190, Spain
- Clinic Laboratory, University Hospital Dr Josep Trueta, Girona 17007, Spain
| | - Maite Albiol-Quer
- Department of Surgery, Hepato-biliary and Pancreatic Surgery Unit, University Hospital Dr Josep Trueta, Girona 17007, Spain
| | - Rafael de Llorens
- Department of Biology, Biochemistry and Molecular Biology Unit, University of Girona, Girona 17003, Spain
- Biomedical Research Institute of Girona (IdIBGi). Parc Hospitalari Martí i Julià-Edifici M2, Salt 17190, Spain
| | - Rosa Peracaula
- Department of Biology, Biochemistry and Molecular Biology Unit, University of Girona, Girona 17003, Spain
- Biomedical Research Institute of Girona (IdIBGi). Parc Hospitalari Martí i Julià-Edifici M2, Salt 17190, Spain
| |
Collapse
|
133
|
Liu B, Yang H, Taher L, Denz A, Grützmann R, Pilarsky C, Weber GF. Identification of Prognostic Biomarkers by Combined mRNA and miRNA Expression Microarray Analysis in Pancreatic Cancer. Transl Oncol 2018; 11:700-714. [PMID: 29631214 PMCID: PMC6154866 DOI: 10.1016/j.tranon.2018.03.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/07/2018] [Accepted: 03/12/2018] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer is the fourth leading cause for cancer-related death, and early diagnosis is one key to improve the survival rate of this disease. Molecular biomarkers are an important method for diagnostic use in pancreatic cancer. We used data from three mRNA microarray datasets and a microRNA dataset (GSE16515, GSE15471, GSE28735, and GSE41372) to identify potential key genes. Differentially expressed genes (DEGs) and microRNAs (DEMs) were identified. Functional, pathway enrichment, and protein-protein interaction analyses were performed on common DEGs across all datasets. The target genes of the DEMs were identified. DEMs targets that were also DEGs were further scrutinized using overall survival analysis. A total of 236 DEGs and 21 DEMs were identified. There were a total of four DEGs (ECT2, NR5A2, NRP2, and TGFBI), which were also predicted target genes of DEMs. Overall survival analysis showed that high expression levels of three of these genes (ECT2, NRP2, and TGFBI) were associated with poor overall survival for pancreatic cancer patients. The basic expression of DEGs in pancreas stood lower level in various organ tissues. The expression of ECT2 and NRP2 was higher in different pancreatic cancer cell lines than normal pancreas cell line. Knockout of ECT2 by Crispr Cas9 gene editing system decreased proliferation and migration ability in pancreatic cancer cell line MiaPaCa2. In conclusion, we think that data mining method can do well in biomarker screening, and ECT2 and NRP2 can play as potential biomarker or therapy target by Crispr Cas9 in pancreatic cancer.
Collapse
Affiliation(s)
- Bin Liu
- Department of Surgery, Universitätsklinikum Erlangen, Krankenhausstraße 12, Erlangen, Germany
| | - Hai Yang
- Department of Surgery, Universitätsklinikum Erlangen, Krankenhausstraße 12, Erlangen, Germany
| | - Leila Taher
- Division of Bioinformatics, Department of Biology, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Axel Denz
- Department of Surgery, Universitätsklinikum Erlangen, Krankenhausstraße 12, Erlangen, Germany
| | - Robert Grützmann
- Department of Surgery, Universitätsklinikum Erlangen, Krankenhausstraße 12, Erlangen, Germany
| | - Christian Pilarsky
- Department of Surgery, Universitätsklinikum Erlangen, Krankenhausstraße 12, Erlangen, Germany.
| | - Georg F Weber
- Department of Surgery, Universitätsklinikum Erlangen, Krankenhausstraße 12, Erlangen, Germany
| |
Collapse
|
134
|
Huang X, Fan C, Zhu H, Le W, Cui S, Chen X, Li W, Zhang F, Huang Y, Sh D, Cui Z, Shao C, Chen B. Glypican-1-antibody-conjugated Gd-Au nanoclusters for FI/MRI dual-modal targeted detection of pancreatic cancer. Int J Nanomedicine 2018; 13:2585-2599. [PMID: 29750031 PMCID: PMC5933399 DOI: 10.2147/ijn.s158559] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Introduction Pancreatic cancer (PC) has a poor prognosis with high mortality, due to the lack of effective early diagnostic and prognostic tools. Materials and methods In order to target and diagnose PC, we developed a dual-modal imaging probe using Glypican-1 (GPC-1) antibody conjugated with Gd–Au nanoclusters (NCs; Gd-Au-NC-GPC-1). GPC-1 is a type of cell surface heparan sulfate proteoglycan, which is often highly expressed in PC. The probe was successfully prepared with a hydrodynamic diameter ranging from 13.5 to 24.4 nm. Results Spectral characteristics showed absorption at 280 nm and prominent emission at 650 nm. Confocal microscopic imaging showed effective detection of GPC-1 highly expressed PC cells by Gd-Au-NC-GPC-1, which was consistent with flow cytometry results. In vitro relaxivity characterization demonstrated that the r1 value of the probe was 17.722 s−1 mM−1 Gd, which was almost 4 times higher compared with that of Gd-diethylenetriaminepentacetate (DTPA; r1 value =4.6 s−1 mM−1 Gd). Gd-Au-NC-GPC-1 exhibited similar magnetic resonance (MR) signals when compared to Gd-DTPA even at lower Gd concentrations. Much higher MR signals were registered in PC cells (COLO-357) compared with normal cells (293T). Furthermore, Gd-Au-NC-GPC-1 could effectively detect PC cells in vivo by dual-modal fluorescence imaging/magnetic resonance imaging (FI/MRI) at 30 minutes postinjection. In addition, Gd-Au-NC-GPC-1 did not show significant biotoxicity to normal cells at tested concentrations both in vitro and in vivo. Conclusion Gd-Au-NC-GPC-1 has demonstrated to be a promising dual-modal FI/MRI contrast agent for targeted diagnosis of PC.
Collapse
Affiliation(s)
- Xin Huang
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China
| | - Chengqi Fan
- Radiology Department of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Huanhuan Zhu
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China
| | - Wenjun Le
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China
| | - Shaobin Cui
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China
| | - Xin Chen
- Department of Thyroid Surgery, The First Bethune Hospital of Jilin University, Jilin, China
| | - Wei Li
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, China
| | - Fulei Zhang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, China
| | - Yong Huang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, China
| | - Donglu Sh
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China.,The Materials Science & Engineering Program, Department of Mechanical & Materials Engineering, College of Engineering & Applied Science, University of Cincinnati, OH, USA
| | - Zheng Cui
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China.,Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Chengwei Shao
- Radiology Department of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Bingdi Chen
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
135
|
Armstrong EA, Beal EW, Chakedis J, Paredes AZ, Moris D, Pawlik TM, Schmidt CR, Dillhoff ME. Exosomes in Pancreatic Cancer: from Early Detection to Treatment. J Gastrointest Surg 2018; 22:737-750. [PMID: 29423813 DOI: 10.1007/s11605-018-3693-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/12/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Pancreatic cancer (PC) remains one of the most fatal forms of cancer worldwide with incidence nearly equal to mortality. This is often attributed to the fact that diagnosis is often not made until later disease stages when treatment proves difficult. Efforts have been made to reduce the mortality of PC through improvements in early screening techniques and treatments of late-stage disease. Exosomes, small extracellular vesicles involved in cellular communication, have shown promise in helping understand PC disease biology. METHODS In this review, we discuss current studies of the role of exosomes in PC physiology, and their potential use as diagnostic and treatment tools. RESULTS Exosomes have a role in diagnosing pancreatic cancer and in understanding tumor biology including migration, proliferation, chemoresistance, immunosuppression, cachexia and diabetes, and have a potential role in therapy for pancreatic cancer. CONCLUSIONS Exosomal analysis is beneficial in demonstrating mechanisms behind PC growth and metastasis, immunosuppression, drug resistance, and paraneoplastic conditions. Furthermore, the use of exosomes can be beneficial in detecting early-stage PC and exosomes have potential applications as therapeutic targets.
Collapse
Affiliation(s)
- Emily A Armstrong
- The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Eliza W Beal
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA.
| | - Jeffery Chakedis
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Anghela Z Paredes
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Demetrios Moris
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Carl R Schmidt
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Mary E Dillhoff
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| |
Collapse
|
136
|
Habartová L, Bunganič B, Tatarkovič M, Zavoral M, Vondroušová J, Syslová K, Setnička V. Chiroptical spectroscopy and metabolomics for blood-based sensing of pancreatic cancer. Chirality 2018; 30:581-591. [DOI: 10.1002/chir.22834] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/23/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Lucie Habartová
- Department of Analytical Chemistry; University of Chemistry and Technology Prague; Prague 6 Czech Republic
| | - Bohuš Bunganič
- Department of Internal Medicine, First Faculty of Medicine, Military University Hospital; Charles University; Prague 6 Czech Republic
| | - Michal Tatarkovič
- Department of Analytical Chemistry; University of Chemistry and Technology Prague; Prague 6 Czech Republic
| | - Miroslav Zavoral
- Department of Internal Medicine, First Faculty of Medicine, Military University Hospital; Charles University; Prague 6 Czech Republic
| | - Jana Vondroušová
- Department of Organic Technology; University of Chemistry and Technology Prague; Prague 6 Czech Republic
| | - Kamila Syslová
- Department of Organic Technology; University of Chemistry and Technology Prague; Prague 6 Czech Republic
| | - Vladimír Setnička
- Department of Analytical Chemistry; University of Chemistry and Technology Prague; Prague 6 Czech Republic
| |
Collapse
|
137
|
Wu ST, Williams CD, Grover PA, Moore LJ, Mukherjee P. Early detection of pancreatic cancer in mouse models using a novel antibody, TAB004. PLoS One 2018; 13:e0193260. [PMID: 29462213 PMCID: PMC5819830 DOI: 10.1371/journal.pone.0193260] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 02/07/2018] [Indexed: 01/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is the fourth-leading cause of cancer death in the United States with a 5-year overall survival rate of 8% for all stages combined. But this decreases to 3% for the majority of patients that present with stage IV PDA at time of diagnosis. The lack of distinct early symptoms for PDA is one of the primary reasons for the late diagnosis. Common symptoms like weight loss, abdominal and back pains, and jaundice are often mistaken for symptoms of other issues and do not appear until the cancer has progressed to a late stage. Thus the development of novel imaging platforms for PDA is crucial for the early detection of the disease. MUC1 is a tumor-associated antigen (tMUC1) expressed on 80% of PDA. The goal of this study was to determine the targeting and detection capabilities of a tMUC1 specific antibody, TAB004. TAB004 antibody conjugated to a near infrared fluorescent probe was injected intraperitoneally into immune competent orthotopic and spontaneous models of PDA. Results show that fluorophore conjugated TAB004 specifically targets a) 1 week old small tumor in the pancreas in an orthotopic PDA model and b) very early pre-neoplastic lesions (PanIN lesions) that develop in the spontaneous PDA model before progression to adenocarcinoma. Thus, TAB004 is a promising antibody to deliver imaging agents directly to the pancreatic tumor microenvironment, significantly affecting early detection of PDA.
Collapse
Affiliation(s)
- Shu-ta Wu
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
| | - Chandra D. Williams
- Department of Animal Laboratory Resources, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
| | - Priyanka A. Grover
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
| | - Laura J. Moore
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
| | - Pinku Mukherjee
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
138
|
Elevated Polyamines in Saliva of Pancreatic Cancer. Cancers (Basel) 2018; 10:cancers10020043. [PMID: 29401744 PMCID: PMC5836075 DOI: 10.3390/cancers10020043] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 01/26/2018] [Accepted: 02/02/2018] [Indexed: 12/13/2022] Open
Abstract
Detection of pancreatic cancer (PC) at a resectable stage is still difficult because of the lack of accurate detection tests. The development of accurate biomarkers in low or non-invasive biofluids is essential to enable frequent tests, which would help increase the opportunity of PC detection in early stages. Polyamines have been reported as possible biomarkers in urine and saliva samples in various cancers. Here, we analyzed salivary metabolites, including polyamines, using capillary electrophoresis-mass spectrometry. Salivary samples were collected from patients with PC (n = 39), those with chronic pancreatitis (CP, n = 14), and controls (C, n = 26). Polyamines, such as spermine, N₁-acetylspermidine, and N₁-acetylspermine, showed a significant difference between patients with PC and those with C, and the combination of four metabolites including N₁-acetylspermidine showed high accuracy in discriminating PC from the other two groups. These data show the potential of saliva as a source for tests screening for PC.
Collapse
|
139
|
Chirlaque MD, Salmerón D, Galceran J, Ameijide A, Mateos A, Torrella A, Jiménez R, Larrañaga N, Marcos-Gragera R, Ardanaz E, Sant M, Minicozzi P, Navarro C, Sánchez MJ. Cancer survival in adult patients in Spain. Results from nine population-based cancer registries. Clin Transl Oncol 2018; 20:201-211. [PMID: 28718071 DOI: 10.1007/s12094-017-1710-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 06/20/2017] [Indexed: 01/24/2023]
Abstract
INTRODUCTION With the aim of providing cancer control indicators, this work presents cancer survival in adult (≥15 years) patients in Spain diagnosed during the period 2000-2007 from Spanish cancer registries participating in the EUROCARE project. METHODS Cancer cases from nine Spanish population-based cancer registries were included and analysed as a whole. All primary malignant neoplasms diagnosed in adult patients were eligible for the analysis. Cancer patients were followed until 31 December 2008. For each type of cancer, 1-, 3- and 5-year observed and relative survival were estimated by sex, age and years from diagnosis. Furthermore, age-standardized 5-year relative survival for the period 2000-2007 has been compared with that of the period 1995-1999. RESULTS Skin melanoma (84.6 95% CI 83.0-86.2), prostate (84.6% 95% CI 83.6-85.6) and thyroid (84.2% CI 95% 82.0-86.6) cancers showed the highest 5-year relative survival, whereas the worst prognosis was observed in pancreatic (6% 95% CI 5.1-7.0) and oesophageal (9.4% 95% CI 7.9-11.1) cancers. Overall, survival is higher in women (58.0%) than in men (48.9%). The absolute difference in relative survival between 2000-2007 and 1995-1999 was positive for all cancers as a whole (+4.8% in men, +1.6% in women) and for most types of tumours. Survival increased significantly for chronic myeloid leukaemia, non-Hodgkin's lymphoma and rectum cancer in both sexes, and for acute lymphoid leukaemia, prostate, liver and colon cancers in men and Hodgkin's lymphoma and breast cancer in women. Survival patterns by age were similar in Europe and Spain. A decline in survival by age was observed in all tumours, being more pronounced for ovarian, corpus uteri, prostate and urinary bladder and less for head and neck and rectum cancers. CONCLUSION High variability and differences have been observed in survival among adults in Spain according to the type of cancer diagnosed, from above 84% to below 10%, reflecting high heterogeneity. The differences in prognosis by age, sex and period of diagnosis reveal opportunities for improving cancer care in Spain.
Collapse
Affiliation(s)
- M D Chirlaque
- Department of Epidemiology, Regional Health Authority, IMIB-Arrixaca, Murcia University, Ronda de Levante, 11, 30008, Murcia, Spain.
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain.
| | - D Salmerón
- Department of Epidemiology, Regional Health Authority, IMIB-Arrixaca, Murcia University, Ronda de Levante, 11, 30008, Murcia, Spain
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - J Galceran
- Tarragona Cancer Registry, Foundation Society for Cancer Research and Prevention, Pere Virgili Health Research Institute, Reus, Spain
| | - A Ameijide
- Tarragona Cancer Registry, Foundation Society for Cancer Research and Prevention, Pere Virgili Health Research Institute, Reus, Spain
| | - A Mateos
- Albacete Cancer Registry, Health and Social Welfare Authority, Castile-La Mancha, Spain
| | - A Torrella
- Castellón Cancer Registry, Public Health Directorate, Valencian Government, Castellón, Spain
| | - R Jiménez
- Cuenca Cancer Registry, Health and Social Welfare Authority, Castile-La Mancha, Spain
| | - N Larrañaga
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Basque Country Cancer Registry, Basque Country Regional Authority, Vitoria-Gasteiz, Spain
| | - R Marcos-Gragera
- Epidemiology Unit and Girona Cancer Registry (UERCG), Oncology Coordination Plan, Department of Health, Autonomous Government of Catalonia, Catalan Institute of Oncology (ICO), Girona Biomedical Institute (IDIBGI), University of Girona, Girona, Spain
| | - E Ardanaz
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Navarre Cancer Registry, Navarre Public Health Institute, Pamplona, Spain
| | - M Sant
- Analytical Epidemiology and Health Impact Unit, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - P Minicozzi
- Analytical Epidemiology and Health Impact Unit, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - C Navarro
- Department of Epidemiology, Regional Health Authority, IMIB-Arrixaca, Murcia University, Ronda de Levante, 11, 30008, Murcia, Spain
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - M J Sánchez
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Granada Cancer Registry, Andalusian School of Public Health, Instituto de Investigación Biosanitaria ibs.GRANADA, Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
| |
Collapse
|
140
|
Nordgård O, Tjensvoll K, Gilje B, Søreide K. Circulating tumour cells and DNA as liquid biopsies in gastrointestinal cancer. Br J Surg 2018; 105:e110-e120. [DOI: 10.1002/bjs.10782] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/02/2017] [Accepted: 11/09/2017] [Indexed: 12/15/2022]
Abstract
Abstract
Background
Blood is the most extensively studied body fluid and, because it contains circulating tumour cells (CTCs) and circulating tumour-derived cell-free DNA (ctDNA), it may represent a liquid biopsy for cancer. Methods for enrichment and detection of CTCs and ctDNA, their clinical applications and future opportunities in gastrointestinal cancers were the focus of this review.
Methods
The PubMed database was searched for literature up to 24 June 2017, with a focus on the past 10 years. Identified articles were further scrutinized for relevant references. Articles were those in English relating to colorectal, gastric and pancreatic cancer.
Results
Both CTCs and ctDNA are in low abundance compared with other cellular components of blood, but effective enrichment and highly sensitive techniques are available for their detection. Potential clinical applications of these liquid biopsies include screening, prognostic stratification, therapy administration, monitoring of treatment effect or resistance, and surveillance. Liquid biopsies provide opportunities to reduce the need for invasive tissue sampling, especially in the context of intratumoral heterogeneity and the need for tumour genotyping.
Conclusion
Liquid biopsies have applications in gastrointestinal cancers to improve clinical decision-making.
Collapse
Affiliation(s)
- O Nordgård
- Department of Haematology and Oncology, Stavanger University Hospital, Stavanger, Norway
- Department of Mathematics and Natural Science, University of Stavanger, Stavanger, Norway
| | - K Tjensvoll
- Department of Haematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - B Gilje
- Department of Haematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - K Søreide
- Department of Gastrointestinal Surgery, Stavanger University Hospital, Stavanger, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Clinical Surgery, Royal Infirmary of Edinburgh and University of Edinburgh, Edinburgh, UK
| |
Collapse
|
141
|
Moutinho-Ribeiro P, Macedo G, Melo SA. Pancreatic Cancer Diagnosis and Management: Has the Time Come to Prick the Bubble? Front Endocrinol (Lausanne) 2018; 9:779. [PMID: 30671023 PMCID: PMC6331408 DOI: 10.3389/fendo.2018.00779] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/11/2018] [Indexed: 02/01/2023] Open
Abstract
Pancreatic cancer (PC) is associated with poor prognosis and very dismal survival rates. The most effective possibility of cure is tumor resection, which is only possible in about 15% of patients diagnosed at early stages of disease progression. Recent whole-genome sequencing studies pointed genetic alterations in 12 core signaling pathways in PC. These observations hint at the possibility that the initial mutation in PC might appear nearly 20 years before any symptoms occur, suggesting that a large window of opportunity may exist for early detection. Biomarkers with the potential to identify pre-neoplastic disease or very early stages of cancer are of great promise to improve patient survival. The concept of liquid biopsy refers to a minimally invasive sampling and analysis of liquid biomarkers that can be isolated from body fluids, primarily blood, urine and saliva. A myriad of circulating molecules may be useful as tumor markers, including cell-free DNA (cfDNA), cell-free RNA (cfRNA), circulating tumor cells (CTC), circulating tumor proteins, and extracellular vesicles, more specifically exosomes. In this review, we discuss with more detail the potential role of exosomes in several aspects related to PC, from initiation to tumor progression and its applicability in early detection and treatment. Exosomes are small circulating extracellular vesicles of 50-150 nm in diameter released from the plasma membrane by almost all cells and exhibit some advantages over other biomarkers. Exosomes are central players of intercellular communication and they have been implicated in a series of biological process, including tumorigenesis, migration and metastasis. Several exosomal microRNAs and proteins have been observed to distinguish PC from benign pancreatic diseases and healthy controls. Besides their possible role in diagnosis, understanding exosomes functions in cancer has clarified the importance of microenvironment in PC progression as well as its influence in proliferation, metastasis and resistance to chemotherapy. Increasing knowledge on cancer exosomes provides valuable insights on new therapeutic targets and can potentially open new strategies to treat this disease. Continuous research is needed to ascertain the reliability of using exosomes and their content as potential biomarkers, so that, hopefully, in the near future, they will provide the opportunity for early diagnosis, treatment intervention and increase survival of PC patients.
Collapse
Affiliation(s)
- Pedro Moutinho-Ribeiro
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal
- Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Guilherme Macedo
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal
- Faculty of Medicine of the University of Porto, Porto, Portugal
- *Correspondence: Guilherme Macedo
| | - Sónia A. Melo
- Faculty of Medicine of the University of Porto, Porto, Portugal
- Institute for Research Innovation in Health (i3S), Porto, Portugal
- Institute of Pathology and Molecular Immunology of the University of Porto, Porto, Portugal
- Sónia A. Melo
| |
Collapse
|
142
|
Kanno A, Masamune A, Hanada K, Maguchi H, Shimizu Y, Ueki T, Hasebe O, Ohtsuka T, Nakamura M, Takenaka M, Kitano M, Kikuyama M, Gabata T, Yoshida K, Sasaki T, Serikawa M, Furukawa T, Yanagisawa A, Shimosegawa T. Multicenter study of early pancreatic cancer in Japan. Pancreatology 2018; 18:61-67. [PMID: 29170051 DOI: 10.1016/j.pan.2017.11.007] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 10/21/2017] [Accepted: 11/16/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES The diagnosis of early-stage pancreatic ductal adenocarcinoma (PDAC) is still challenging. We conducted a multicenter study to clarify the clinical features of early-stage PDAC in Japan. METHODS We collected patients with stage 0 and stage I PDAC according to the sixth edition of the Japanese Classification of Pancreatic Carcinoma. We retrospectively analyzed the clinical profiles including opportunities for medical examination, imaging modalities and findings, methods of cytological diagnosis, and prognosis according to the stages at diagnosis. RESULTS Two hundred cases with Stage 0 and stage I PDAC were reported from 14 institutions, which accounted for approximately 0.7% and 3% of all PDAC cases, respectively. Overall, 20% of the early-stage PDAC cases were symptomatic. Indirect imaging findings such as dilatation of the main pancreatic duct were useful to detect early-stage PDAC. In particular, local fatty changes may be specific to early-stage PDAC. For preoperative pathologic diagnosis, cytology during endoscopic retrograde cholangiopancreatography was more commonly applied than endoscopic ultrasound fine-needle aspiration. Although the overall prognosis was favorable, new PDAC lesions developed in the remnant pancreas in 11.5% cases. CONCLUSIONS This multicenter study revealed several key points concerning the diagnosis and management of early-stage PDAC, including screening of asymptomatic cases, importance of indirect imaging findings, application of cytology during endoscopic retrograde cholangiopancreatography, and the risk of carcinogenesis in the remnant pancreas.
Collapse
Affiliation(s)
- Atsushi Kanno
- Division of Gastroenterology, Tohoku University, Graduate School of Medicine, Japan.
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University, Graduate School of Medicine, Japan
| | - Keiji Hanada
- Department of Gastroenterology, JA Onomichi General Hospital, Japan
| | | | - Yasuhiro Shimizu
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Japan
| | - Toshiharu Ueki
- Department of Gastroenterology, Fukuoka University Chikushi Hospital, Japan
| | - Osamu Hasebe
- Department of Gastroenterology, Nagano Municipal Hospital, Japan
| | - Takao Ohtsuka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Mamoru Takenaka
- Department of Gastroenterology and Hepatology, Kindai University, Faculty of Medicine, Japan
| | - Masayuki Kitano
- Second Department of Internal Medicine, Wakayama Medical University, School of Medicine, Japan
| | | | | | - Koji Yoshida
- Department of Interventional Bilio-Pancreatology, Kawasaki Medical School, Japan
| | - Tamito Sasaki
- Department of Gastroenterology, Hiroshima Prefectural Hospital, Japan
| | - Masahiro Serikawa
- Department of Gastroenterology and Metabolism, Hiroshima University Hospital, Japan
| | - Toru Furukawa
- Institute for Integrated Medical Sciences, Tokyo Women's Medical University, Japan; Department of Histopathology, Tohoku University, Graduate School of Medicine, Japan
| | - Akio Yanagisawa
- Department of Pathology, Kyoto Prefectural University of Medicine, Japan
| | - Tooru Shimosegawa
- Division of Gastroenterology, Tohoku University, Graduate School of Medicine, Japan
| |
Collapse
|
143
|
Boulaiz H, Ramos MC, Griñán-Lisón C, García-Rubiño ME, Vicente F, Marchal JA. What's new in the diagnosis of pancreatic cancer: a patent review (2011-present). Expert Opin Ther Pat 2017; 27:1319-1328. [PMID: 28929814 DOI: 10.1080/13543776.2017.1379991] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Pancreatic cancer (PC) is the fourth leading cause of cancer-related death in the US with a 5-year survival rate of about 5%. Most patients have advanced metastatic disease mainly due to the lack of an effective early detection, and an extremely poor prognosis. Advancing in the fight against PC requires developing novel observable biomarkers at preclinical stages for early detection. Areas covered: This manuscript is an overview of different PC diagnostic modalities and the latest innovations made to enhance early PC detection through the patents published from 2011 to 2017. It also comments on the ongoing clinical trials and highlights the main challenges to be addressed in the future. Expert opinion: At present, real efforts are being made to identify new specific biomarkers with a potential clinical applicability, and to develop new devices that integrate several biomarkers in order to be more sensitive and specific for the early detection of PC. Although many biomarkers have been patented recently, they will not reach the clinic until they have been validated by clinical trials. We believe that the high-throughput screening of '-omic' technologies to detect tumor-specific molecular alterations can lead to an enhanced understanding of the disease mechanisms and the discovery of new clinical diagnostic biomarkers.
Collapse
Affiliation(s)
- Houria Boulaiz
- a Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research , University of Granada , Granada , Spain .,b Biosanitary Institute of Granada (ibs.GRANADA) , University Hospitals of Granada-University of Granada , Granada , Spain
| | - María C Ramos
- c Fundación MEDINA , Parque Tecnológico de la Salud , Granada , Spain
| | - Carmen Griñán-Lisón
- a Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research , University of Granada , Granada , Spain .,b Biosanitary Institute of Granada (ibs.GRANADA) , University Hospitals of Granada-University of Granada , Granada , Spain
| | - Maria E García-Rubiño
- d Department of Inorganic Chemistry, Faculty of Pharmacy , University of Granada , Granada , Spain
| | - Francisca Vicente
- c Fundación MEDINA , Parque Tecnológico de la Salud , Granada , Spain
| | - Juan Antonio Marchal
- a Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research , University of Granada , Granada , Spain .,b Biosanitary Institute of Granada (ibs.GRANADA) , University Hospitals of Granada-University of Granada , Granada , Spain
| |
Collapse
|
144
|
Liu X, Zhou X, Xu H, He Z, Shi X, Wu S. SLC34A2 Regulates the Proliferation, Migration, and Invasion of Human Osteosarcoma Cells Through PTEN/PI3K/AKT Signaling. DNA Cell Biol 2017; 36:775-780. [PMID: 28777670 DOI: 10.1089/dna.2017.3750] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Osteosarcoma (OS) is a bone malignancy with high incidence. The underlying molecular mechanisms that are associated with the development of OS need further investigation. In this study, we showed that SLC34A2, a member of the solute carrier gene family, was significantly downregulated in OS patients and cell lines. Overexpression of SLC34A2 inhibited the proliferation, migration, and invasion of OS cells. Mechanistically, we found that SLC34A2 interacted with PTEN, and inactivated the PI3K/AKT signaling pathway. Collectively, our results demonstrated that SLC34A2 plays important roles in regulating the cancer cell growth of OS. The downregulation of SLC34A2 in OS patients suggested that it might be a promising target in the diagnosis and therapy of OS.
Collapse
Affiliation(s)
- Xiaozhou Liu
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University , Nanjing China
| | - Xing Zhou
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University , Nanjing China
| | - Haidong Xu
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University , Nanjing China
| | - Zhiwei He
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University , Nanjing China
| | - Xin Shi
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University , Nanjing China
| | - Sujia Wu
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University , Nanjing China
| |
Collapse
|
145
|
Xu J, Li L, Chen N, She Y, Wang S, Liu N, Xiao X. Endonuclease IV based competitive DNA probe assay for differentiation of low-abundance point mutations by discriminating stable single-base mismatches. Chem Commun (Camb) 2017; 53:9422-9425. [PMID: 28792020 DOI: 10.1039/c7cc04816e] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We disclosed the unique discrimination property of Endo IV toward stable single-base mismatches located at the second nucleotide 3' to the AP site. Coupled with thermodynamic differentiation and competitive blocker strands, a highly sensitive and specific detection system was established with discrimination factors of 510-1079 for G:X mismatches and LODs of 0.003-0.005% for KRAS G12A, KRAS G12V and KRAS G12S mutations.
Collapse
Affiliation(s)
- Jiaju Xu
- Family Planning Research Institute/Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
146
|
Makler A, Narayanan R. Mining Exosomal Genes for Pancreatic Cancer Targets. Cancer Genomics Proteomics 2017; 14:161-172. [PMID: 28446531 PMCID: PMC5420817 DOI: 10.21873/cgp.20028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/03/2017] [Accepted: 04/05/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Exosomes, cell-derived vesicles encompassing lipids, DNA, proteins coding genes and noncoding RNAs (ncRNAs) are present in diverse body fluids. They offer novel biomarker and drug therapy potential for diverse diseases, including cancer. MATERIALS AND METHODS Using gene ontology, exosomal genes database and GeneCards metadata analysis tools, a database of cancer-associated protein coding genes and ncRNAs (n=2,777) was established. Variant analysis, expression profiling and pathway mapping were used to identify putative pancreatic cancer exosomal gene candidates. RESULTS Five hundred and seventy-five protein-coding genes, 26 RNA genes and one pseudogene directly associated with pancreatic cancer were identified in the study. Nine open reading frames (ORFs) encompassing enzymes, apoptosis and transcriptional regulators, and secreted factors and five cDNAs (enzymes) emerged from the analysis. Among the ncRNA class, 26 microRNAs (miRs), one pseudogene, one long noncoding RNA (LNC) and one antisense gene were identified. Furthermore, 22 exosome-associated protein-coding targets (a cytokine, enzymes, membrane glycoproteins, receptors, and a transporter) emerged as putative leads for pancreatic cancer therapy. Seven of these protein-coding targets are FDA-approved and the drugs-based on these could provide repurposing opportunities for pancreatic cancer. CONCLUSION The database of exosomal genes established in this study provides a framework for developing novel biomarkers and drug therapy targets for pancreatic cancer.
Collapse
Affiliation(s)
- Amy Makler
- Department of Biological Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, U.S.A
| | - Ramaswamy Narayanan
- Department of Biological Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, U.S.A.
| |
Collapse
|
147
|
Khan MAA, Azim S, Zubair H, Bhardwaj A, Patel GK, Khushman M, Singh S, Singh AP. Molecular Drivers of Pancreatic Cancer Pathogenesis: Looking Inward to Move Forward. Int J Mol Sci 2017; 18:ijms18040779. [PMID: 28383487 PMCID: PMC5412363 DOI: 10.3390/ijms18040779] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) continues to rank among the most lethal cancers. The consistent increase in incidence and mortality has made it the seventh leading cause of cancer-associated deaths globally and the third in the United States. The biggest challenge in combating PC is our insufficient understanding of the molecular mechanism(s) underlying its complex biology. Studies during the last several years have helped identify several putative factors and events, both genetic and epigenetic, as well as some deregulated signaling pathways, with implications in PC onset and progression. In this review article, we make an effort to summarize our current understanding of molecular and cellular events involved in the pathogenesis of pancreatic malignancy. Specifically, we provide up-to-date information on the genetic and epigenetic changes that occur during the initiation and progression of PC and their functional involvement in the pathogenic processes. We also discuss the impact of the tumor microenvironment on the molecular landscape of PC and its role in aggressive disease progression. It is envisioned that a better understanding of these molecular factors and the mechanisms of their actions can help unravel novel diagnostic and prognostic biomarkers and can also be exploited for future targeted therapies.
Collapse
Affiliation(s)
- Mohammad Aslam Aslam Khan
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| | - Shafquat Azim
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| | - Haseeb Zubair
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| | - Arun Bhardwaj
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| | - Girijesh Kumar Patel
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| | - Moh'd Khushman
- Departments of Interdisciplinary Clinical Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| | - Seema Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA.
| | - Ajay Pratap Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA.
| |
Collapse
|