201
|
Fusaroli P, Kypraios D, Caletti G, Eloubeidi MA. Pancreatico-biliary endoscopic ultrasound: a systematic review of the levels of evidence, performance and outcomes. World J Gastroenterol 2012; 18:4243-4256. [PMID: 22969187 PMCID: PMC3436039 DOI: 10.3748/wjg.v18.i32.4243] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 08/01/2012] [Accepted: 08/03/2012] [Indexed: 02/06/2023] Open
Abstract
Our aim was to record pancreaticobiliary endoscopic ultrasound (EUS) literature of the past 3 decades and evaluate its role based on a critical appraisal of published studies according to levels of evidence (LE). Original research articles (randomized controlled trials, prospective and retrospective studies), meta-analyses, reviews and surveys pertinent to gastrointestinal EUS were included. All articles published until September 2011 were retrieved from PubMed and classified according to specific disease entities, anatomical subdivisions and therapeutic applications of EUS. The North of England evidence-based guidelines were used to determine LE. A total of 1089 pertinent articles were reviewed. Published research focused primarily on solid pancreatic neoplasms, followed by disorders of the extrahepatic biliary tree, pancreatic cystic lesions, therapeutic-interventional EUS, chronic and acute pancreatitis. A uniform observation in all six categories of articles was the predominance of LE III studies followed by LE IV, II b, II a, I b and I a, in descending order. EUS remains the most accurate method for detecting small (< 3 cm) pancreatic tumors, ampullary neoplasms and small (< 4 mm) bile duct stones, and the best test to define vascular invasion in pancreatic and peri-ampullary neoplasms. Detailed EUS imaging, along with biochemical and molecular cyst fluid analysis, improve the differentiation of pancreatic cysts and help predict their malignant potential. Early diagnosis of chronic pancreatitis appears feasible and reliable. Novel imaging techniques (contrast-enhanced EUS, elastography) seem promising for the evaluation of pancreatic cancer and autoimmune pancreatitis. Therapeutic applications currently involve pancreaticobiliary drainage and targeted fine needle injection-guided antitumor therapy. Despite the ongoing development of extra-corporeal imaging modalities, such as computed tomography, magnetic resonance imaging, and positron emission tomography, EUS still holds a leading role in the investigation of the pancreaticobiliary area. The major challenge of EUS evolution is its expanding therapeutic potential towards an effective and minimally invasive management of complex pancreaticobiliary disorders.
Collapse
|
202
|
Gold DV, Gaedcke J, Ghadimi BM, Goggins M, Hruban RH, Liu M, Newsome G, Goldenberg DM. PAM4 enzyme immunoassay alone and in combination with CA 19-9 for the detection of pancreatic adenocarcinoma. Cancer 2012; 119:522-8. [PMID: 22898932 DOI: 10.1002/cncr.27762] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 05/21/2012] [Accepted: 06/12/2012] [Indexed: 12/17/2022]
Abstract
BACKGROUND The monoclonal antibody PAM4 has high specificity for pancreatic ductal adenocarcinoma (PDAC), as well as its precursor lesions, but has not been found to be reactive with normal and benign pancreatic tissues. The objective of the current study was to evaluate a PAM4-based serum enzyme immunoassay alone and in combination with the carbohydrate antigen (CA) 19-9 assay for the detection of PDAC, with particular attention to early stage disease. METHODS Sera from patients with confirmed PDAC (N = 298), other cancers (N = 99), benign disease of the pancreas (N = 120), and healthy adults (N = 79) were evaluated by a specific enzyme immunoassay for the concentration of PAM4 and CA 19-9 antigen levels by blinded analyses. All tests for statistical significance were 2-sided. RESULTS The overall sensitivity for PAM4 detection of PDAC was 76%, with 64% of patients with stage I disease also identified. The detection rate was considerably higher (85%) for patients with advanced disease. The assay demonstrated high specificity compared with benign pancreatic disease (85%), with a positive likelihood ratio of 4.93. CA 19-9 provided an overall sensitivity of 77%, and was positive in 58% of patients with stage I disease; however, the specificity was significantly lower for CA 19-9 (68%), with a positive likelihood ratio of 2.85 (P = .026 compared with PAM4). It is important to note that a combined PAM4 and CA 19-9 biomarker serum assay demonstrated an improved sensitivity (84%) for the overall detection of PDAC without a significant loss of specificity (82%) compared with either arm alone. CONCLUSIONS The PAM4 enzyme immunoassay identified approximately two-thirds of patients with stage I PDAC with high discriminatory power with respect to benign, nonneoplastic pancreatic disease. These results provide a rationale for testing patient groups considered to be at high risk for PDAC with a combined PAM4 and CA 19-9 biomarker serum assay for the detection of early stage PDAC.
Collapse
Affiliation(s)
- David V Gold
- Center for Molecular Medicine and Immunology, Garden State Cancer Center, Morris Plains, NJ 07950, USA.
| | | | | | | | | | | | | | | |
Collapse
|
203
|
Suzuki R, Ohira H, Irisawa A, Bhutani MS. Pancreatic cancer: early detection, diagnosis, and screening. Clin J Gastroenterol 2012; 5:322-6. [DOI: 10.1007/s12328-012-0327-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2012] [Accepted: 07/22/2012] [Indexed: 12/18/2022]
|
204
|
Abstract
Pancreatic cancer is notoriously difficult to diagnose until a late stage when curative options are no longer available. Owing to its relatively low incidence and the lack of sensitivity of current diagnostic tool, screening of pancreatic cancer in the general population is not recommended. However, in high-risk individuals, especially those with well-described genetic syndromes and a strong family history of pancreatic cancer, screening can be carried out. Detection of a lesion of the diameter < 1 cm without lymph node involvements and subsequent removal of the tumor results in long-term cure of the cancer. Endoscopic ultrasound (EUS) is the only diagnostic tool that is able to detect such small lesions. EUS is often combined with endoscopic retrograde cholangiography to augment the diagnostic yield. The conundrum in clinical practice is to differentiate between a malignant and a benign lesion. Resection of the pancreas constitutes major surgery with a high morbidity and mortality. The need continues, therefore, to find even more accurate imaging modalities to diagnose small pancreatic cancers with confidence.
Collapse
Affiliation(s)
- Khean-Lee Goh
- Departments of Gastroenterology and Hepatology Hepatobiliary Surgery, University of Malaya, Kuala Lumpur, Malaysia.
| | | |
Collapse
|
205
|
Abstract
Familial pancreatic cancer (FPC) describes families with at least two first-degree relatives with confirmed exocrine pancreatic cancer that do not fulfil the criteria of other inherited tumour syndromes with increased risks of pancreatic cancer, such as Peutz-Jeghers syndrome, hereditary pancreatitis, and hereditary breast and ovarian cancer. The inheritance of FPC is mostly autosomal dominant and with a heterogeneous phenotype. The major gene defect is yet to be identified, although germline mutations in BRCA2, PALB2 and ATM are causative in some FPC families. Expert consensus conferences considered it appropriate to screen for pancreatic cancer in high-risk individuals using a multidisciplinary approach under research protocol conditions. However, neither biomarkers nor reliable imaging modalities for the detection of high-grade precursor lesions are yet available. Most screening programmes are currently based on findings from endoscopic ultrasonography and MRI, and data has demonstrated that precursor lesions of pancreatic cancer can be identified. No consensus exists regarding the age to initiate or stop screening and the optimal intervals for follow-up. Timing and extent of surgery as a treatment for FPC are debated. This Review focuses on the clinical phenotype of FPC, its histopathological characteristics, known underlying genetic changes and associated genetic counselling and screening.
Collapse
|
206
|
Segura PP, Ponce CG, Ramón Y Cajal T, Blanch RS, Aranda E. Hereditary pancreatic cancer: molecular bases and their application in diagnosis and clinical management: a guideline of the TTD group. Clin Transl Oncol 2012; 14:553-63. [PMID: 22855135 DOI: 10.1007/s12094-012-0840-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 04/19/2012] [Indexed: 12/16/2022]
Abstract
Pancreatic carcinoma (PC) represents the fourth leading cause of cancer death in Spain with a death rate of 2,400 males and 2,000 females per year. Poor outcome related to its silent nature and the lack of reliable secondary prevention measures translate into advanced-stage diagnosis, 75 % of deaths within the first year of diagnosis and 5-year survival rate of <5 %. Family history was first recognized as a risk factor for PC. Further population-based and case-control studies subsequently found that 7.8 % of patients with PC have a family history of the same tumor and individuals with a first-degree relative with PC have a 3.2-fold increased risk of developing PC. Overall, it is estimated that up to 10 % of PC have a familial component. However, known genetic syndromes account for <20 % of the observed familial aggregation of PC. We review the most important aspects in epidemiology, molecular biology and clinical management of familial PC.
Collapse
Affiliation(s)
- P Pérez Segura
- Medical Oncology, Clinical Hospital San Carlos, Madrid, Spain.
| | | | | | | | | |
Collapse
|
207
|
Breitkopf CR, Sinicrope PS, Rabe KG, Brockman TA, Patten CA, McWilliams RR, Ehlers S, Petersen GM. Factors influencing receptivity to future screening options for pancreatic cancer in those with and without pancreatic cancer family history. Hered Cancer Clin Pract 2012; 10:8. [PMID: 22738386 PMCID: PMC3410777 DOI: 10.1186/1897-4287-10-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 06/27/2012] [Indexed: 02/08/2023] Open
Abstract
Background Pancreatic cancer (PC) is considered the most lethal cancer and approximately 10% of PC is hereditary. The purpose of the study was to assess attitudes of at-risk family members with two or more relatives affected with pancreas cancer (PC) toward PC risk and future screening options. Methods At-risk family members and primary care controls were surveyed regarding perceived PC risk, PC worry/concern, attitude toward cancer screening, screening test accuracy, and intentions regarding PC screening via blood testing or more invasive endoscopic ultrasound (EUS). Results PC family members reported greater perceived risk of PC than controls (54% vs. 6%, respectively, p < 0.0001). PC family members also reported higher levels of PC worry/concern than controls (p < 0.0001), although 19% of PC family members indicated they were “not at all concerned” about getting PC. PC family members indicated greater acceptance of a false-negative result on a PC screening test relative to controls (12% vs. 8%, p = 0.02). Both groups reported high (>89%) receptivity to the potential PC screening options presented, though receptivity was greater among PC family members as compared to controls (p < 0.0001) for EUS. In multivariable analyses, degree of PC concern (p < 0.0001) was associated with intention to screen for PC by blood test and EUS, while perceived PC risk was associated with likelihood of undergoing EUS only (p < 0.0001). Conclusions Receptivity to screening options for PC appears high. Clinicians should address behavioral and genetic risk factors for PC and foster appropriate concern regarding PC risk among at-risk individuals.
Collapse
Affiliation(s)
- Carmen Radecki Breitkopf
- Mayo Clinic College of Medicine, Department of Health Sciences Research, Charlton 6, 200 First Street SW, Rochester, MN, 55905, USA.
| | | | | | | | | | | | | | | |
Collapse
|
208
|
Grüner BM, Hahne H, Mazur PK, Trajkovic-Arsic M, Maier S, Esposito I, Kalideris E, Michalski CW, Kleeff J, Rauser S, Schmid RM, Küster B, Walch A, Siveke JT. MALDI imaging mass spectrometry for in situ proteomic analysis of preneoplastic lesions in pancreatic cancer. PLoS One 2012; 7:e39424. [PMID: 22761793 PMCID: PMC3383687 DOI: 10.1371/journal.pone.0039424] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Accepted: 05/20/2012] [Indexed: 01/07/2023] Open
Abstract
The identification of new biomarkers for preneoplastic pancreatic lesions (PanINs, IPMNs) and early pancreatic ductal adenocarcinoma (PDAC) is crucial due to the diseases high mortality rate upon late detection. To address this task we used the novel technique of matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) on genetically engineered mouse models (GEM) of pancreatic cancer. Various GEM were analyzed with MALDI IMS to investigate the peptide/protein-expression pattern of precursor lesions in comparison to normal pancreas and PDAC with cellular resolution. Statistical analysis revealed several discriminative m/z-species between normal and diseased tissue. Intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasm (IPMN) could be distinguished from normal pancreatic tissue and PDAC by 26 significant m/z-species. Among these m/z-species, we identified Albumin and Thymosin-beta 4 by liquid chromatography and tandem mass spectrometry (LC-MS/MS), which were further validated by immunohistochemistry, western blot, quantitative RT-PCR and ELISA in both murine and human tissue. Thymosin-beta 4 was found significantly increased in sera of mice with PanIN lesions. Upregulated PanIN expression of Albumin was accompanied by increased expression of liver-restricted genes suggesting a hepatic transdifferentiation program of preneoplastic cells. In conclusion we show that GEM of endogenous PDAC are a suitable model system for MALDI-IMS and subsequent LC-MS/MS analysis, allowing in situ analysis of small precursor lesions and identification of differentially expressed peptides and proteins.
Collapse
Affiliation(s)
- Barbara M. Grüner
- II. Medizinische Klinik, Technische Universität München, Munich, Germany
| | - Hannes Hahne
- Chair of Proteomics and Bioanalytics, Center of Life and Food Sciences, Technische Universität München, Munich, Germany
| | - Pawel K. Mazur
- II. Medizinische Klinik, Technische Universität München, Munich, Germany
| | | | - Stefan Maier
- Chair of Proteomics and Bioanalytics, Center of Life and Food Sciences, Technische Universität München, Munich, Germany
- Institute of Pathology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Germany
| | - Irene Esposito
- Institute of Pathology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Pathology, Technische Universität München, Munich, Germany
| | - Evdokia Kalideris
- II. Medizinische Klinik, Technische Universität München, Munich, Germany
| | | | - Jörg Kleeff
- Department of Surgery, Technische Universität München, Munich, Germany
| | - Sandra Rauser
- Institute of Pathology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Germany
| | - Roland M. Schmid
- II. Medizinische Klinik, Technische Universität München, Munich, Germany
| | - Bernhard Küster
- Chair of Proteomics and Bioanalytics, Center of Life and Food Sciences, Technische Universität München, Munich, Germany
- Center for Integrated Protein Science Munich, Munich, Germany
| | - Axel Walch
- Institute of Pathology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Germany
| | - Jens T. Siveke
- II. Medizinische Klinik, Technische Universität München, Munich, Germany
- * E-mail:
| |
Collapse
|
209
|
Olson SH, Kurtz RC. Epidemiology of pancreatic cancer and the role of family history. J Surg Oncol 2012; 107:1-7. [PMID: 22589078 DOI: 10.1002/jso.23149] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 04/17/2012] [Indexed: 12/20/2022]
Abstract
Pancreatic cancer is a lethal disease for which only a small number of risk factors have been identified. In addition to older age, male gender, and black race, risk factors include smoking, obesity, long-standing diabetes and pancreatitis, and heavy alcohol use; allergies such as hay fever are related to lowered risk. Several genetic syndromes increase risk of pancreatic cancer. Work on more common genetic variants promises to reveal more potentially important genetic associations.
Collapse
Affiliation(s)
- Sara H Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
| | | |
Collapse
|
210
|
Rebours V, Couvelard A, Peyroux JL, Sauvanet A, Hammel P, Ruszniewski P, Lévy P. Familial intraductal papillary mucinous neoplasms of the pancreas. Dig Liver Dis 2012; 44:442-6. [PMID: 21824831 DOI: 10.1016/j.dld.2011.07.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 06/24/2011] [Accepted: 07/02/2011] [Indexed: 12/11/2022]
Abstract
UNLABELLED The prevalence of intraductal papillary mucinous neoplasms in patients with a high risk of pancreatic adenocarcinoma was estimated to be 15%. However, a familial form of intraductal papillary mucinous neoplasms was never described. METHODS Three families (8 patients) with intraductal papillary mucinous neoplasms familial forms were described. Diagnosis was made according to radiological criteria and was confirmed by pathological data. Genetic predisposing factors of pancreatic cancer were searched for. RESULTS Symptoms related to intraductal papillary mucinous neoplasms were recurrent acute pancreatitis (n=3) or fortuitous discovery (n=5). Number of cystic lesions was ≤3 (n=4) or >3 (n=4). Intraductal papillary mucinous neoplasms involved branch ducts (n=7) or both main pancreatic duct and branch duct (n=1). Severe and moderate dysplasia was found on surgical specimens. No genetic alteration was found (BRCA2, p16 or CDKN2A genes). CONCLUSION A familial form of intraductal papillary mucinous neoplasms was found in three families. No pancreatic cancer was found in relatives but an attentive survey has to be proposed.
Collapse
Affiliation(s)
- Vinciane Rebours
- Pôle des Maladies de l'Appareil Digestif, Service de Gastroentérologie - Pancréatologie, Hôpital Beaujon, APHP, Université Denis Diderot-Paris VII, 100, boulevard du général Leclerc, 92118 Clichy, France.
| | | | | | | | | | | | | |
Collapse
|
211
|
Al-Sukhni W, Borgida A, Rothenmund H, Holter S, Semotiuk K, Grant R, Wilson S, Moore M, Narod S, Jhaveri K, Haider MA, Gallinger S. Screening for pancreatic cancer in a high-risk cohort: an eight-year experience. J Gastrointest Surg 2012; 16:771-83. [PMID: 22127781 DOI: 10.1007/s11605-011-1781-6] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 11/11/2011] [Indexed: 02/06/2023]
Abstract
BACKGROUND Pancreatic adenocarcinoma is the fourth leading cause of cancer death. METHODS A prospective cohort study was undertaken between 2003 and 2011 at a tertiary care centre in Toronto, Canada. Two hundred and sixty-two subjects were enrolled based on an elevated estimated lifetime risk for pancreatic cancer due to known genetic mutations and/or cancer family history. Subjects underwent annual magnetic resonance imaging, followed by additional investigations if abnormal findings were detected. Evidence of malignancy or suspicious macroscopic abnormalities prompted referral for surgical intervention. RESULTS Average length of follow-up was 4.2 years, during which 84/262 (32%) subjects demonstrated pancreatic abnormalities. Three participants developed pancreatic adenocarcinoma (one 1.5-cm tumor was resected but recurred, while the other two subjects developed metastatic cancer), and a fourth participant developed a pancreatic neuroendocrine tumor that was resected. Fifteen subjects had radiologic evidence of branch-duct intraductal papillary mucinous neoplasms, of which two underwent surgical resection. Sixty-five subjects had simple pancreatic cysts that have remained stable. CONCLUSION Magnetic resonance imaging can detect small pancreatic tumors and cystic lesions, but further improvement in sensitivity is needed. An understanding of the natural history of pre-invasive lesions in members of high-risk families is necessary for developing a more effective screening program.
Collapse
Affiliation(s)
- Wigdan Al-Sukhni
- Hepatobiliary/Pancreatic Surgical Oncology Program, Division of General Surgery, Department of Surgery, University Health Network, University of Toronto, Toronto, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Canto MI, Hruban RH, Fishman EK, Kamel IR, Schulick R, Zhang Z, Topazian M, Takahashi N, Fletcher J, Petersen G, Klein AP, Axilbund J, Griffin C, Syngal S, Saltzman JR, Mortele KJ, Lee J, Tamm E, Vikram R, Bhosale P, Margolis D, Farrell J, Goggins M, For the American Cancer of the Pancreas Screening (CAPS) Consortium. Frequent detection of pancreatic lesions in asymptomatic high-risk individuals. Gastroenterology 2012; 142:796-804; quiz e14-5. [PMID: 22245846 PMCID: PMC3321068 DOI: 10.1053/j.gastro.2012.01.005] [Citation(s) in RCA: 485] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 12/16/2011] [Accepted: 01/05/2012] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS The risk of pancreatic cancer is increased in patients with a strong family history of pancreatic cancer or a predisposing germline mutation. Screening can detect curable, noninvasive pancreatic neoplasms, but the optimal imaging approach is not known. We determined the baseline prevalence and characteristics of pancreatic abnormalities using 3 imaging tests to screen asymptomatic, high-risk individuals (HRIs). METHODS We screened 225 asymptomatic adult HRIs at 5 academic US medical centers once, using computed tomography (CT), magnetic resonance imaging (MRI), and endoscopic ultrasonography (EUS). We compared results in a blinded, independent fashion. RESULTS Ninety-two of 216 HRIs (42%) were found to have at least 1 pancreatic mass (84 cystic, 3 solid) or a dilated pancreatic duct (n = 5) by any of the imaging modalities. Fifty-one of the 84 HRIs with a cyst (60.7%) had multiple lesions, typically small (mean, 0.55 cm; range, 2-39 mm), in multiple locations. The prevalence of pancreatic lesions increased with age; they were detected in 14% of subjects younger than 50 years old, 34% of subjects 50-59 years old, and 53% of subjects 60-69 years old (P < .0001). CT, MRI, and EUS detected a pancreatic abnormality in 11%, 33.3%, and 42.6% of the HRIs, respectively. Among these abnormalities, proven or suspected neoplasms were identified in 85 HRIs (82 intraductal papillary mucinous neoplasms and 3 pancreatic endocrine tumors). Three of 5 HRIs who underwent pancreatic resection had high-grade dysplasia in less than 3 cm intraductal papillary mucinous neoplasms and in multiple intraepithelial neoplasias. CONCLUSIONS Screening of asymptomatic HRIs frequently detects small pancreatic cysts, including curable, noninvasive high-grade neoplasms. EUS and MRI detect pancreatic lesions better than CT.
Collapse
Affiliation(s)
- Marcia Irene Canto
- Department of Medicine (Division of Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland 21205, USA.
| | - Ralph H. Hruban
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Elliot K. Fishman
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Ihab R. Kamel
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Richard Schulick
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Zhe Zhang
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Mark Topazian
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Mayo Clinic
| | - Naoki Takahashi
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Mayo Clinic
| | - Joel Fletcher
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Mayo Clinic
| | - Gloria Petersen
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Mayo Clinic
| | - Alison P. Klein
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Jennifer Axilbund
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Constance Griffin
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Sapna Syngal
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Dana Farber Cancer Institute,Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Brigham and Women's Hospital
| | - John R. Saltzman
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Brigham and Women's Hospital
| | - Koenraad J. Mortele
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Brigham and Women's Hospital
| | - Jeffrey Lee
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, MD Anderson Cancer Center
| | - Eric Tamm
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, MD Anderson Cancer Center
| | - Raghunandan Vikram
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, MD Anderson Cancer Center
| | - Priya Bhosale
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, MD Anderson Cancer Center
| | - Daniel Margolis
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, University of California Los Angeles
| | - James Farrell
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, University of California Los Angeles
| | - Michael Goggins
- Department of Medicine (Gastroenterology), Epidemiology, Oncology, Biostatistics, Radiology, Anesthesia, Surgery, and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | | |
Collapse
|
213
|
Lüttges J. [What's new? The 2010 WHO classification for tumours of the pancreas]. DER PATHOLOGE 2012; 32 Suppl 2:332-6. [PMID: 21915659 DOI: 10.1007/s00292-011-1515-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The new WHO classification of tumours of the pancreas logically includes both exocrine and neuroendocrine neoplasms in one volume, thus differing from all previous editions. Ductal adenocarcinoma is still the most frequent and clinically the most relevant malignant tumour. Its subtypes and variants are described in detail, as are mixed tumours. Other ductal tumours [mucinous cystic neoplasms (MCN) and intraductal papillary mucinous neoplasms (IPNM)] are classified as neoplasms with various grades of dysplasia up to invasive carcinoma. A new subtype of IPNM, intraductal tubulopapillary neoplasm (ITPN), has been characterized and newly added to the IPMN group. Serous and acinar tumours are classified as neoplasms with varying grades of dysplasia. Solid pseudopapillary neoplasm (SPN) is regarded as malignant (low grade) as a matter of principle because of its inherent potential to metastasize. Neuroendocrine neoplasms are characterized as G1 or G2 neuroendocrine tumours (NET) and neuroendocrine carcinomas (NEC, highly malignant). Syndromatic NETs are described and named according to their hormone expression pattern. The problems of staging when applying either the TNM or AJCC/UICC (American Joint Committee on Cancer/Union Internationale Contre le Cancer) classifications, which apply equally to endocrine and exocrine tumors, are discussed.
Collapse
Affiliation(s)
- J Lüttges
- Institut für Pathologie, Klinikum Saarbrücken, Winterberg 1, 66119, Saarbrücken, Deutschland.
| |
Collapse
|
214
|
Clinicopathological characteristics and molecular analyses of multifocal intraductal papillary mucinous neoplasms of the pancreas. Ann Surg 2012; 255:326-33. [PMID: 22167000 DOI: 10.1097/sla.0b013e3182378a18] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To examine the clinicopathologic features and clonal relationship of multifocal intraductal papillary mucinous neoplasms (IPMNs) of the pancreas. BACKGROUND Intraductal papillary mucinous neoplasms are increasingly diagnosed cystic precursor lesions of pancreatic cancer. Intraductal papillary mucinous neoplasms can be multifocal and a potential cause of recurrence after partial pancreatectomy. METHODS Thirty four patients with histologically documented multifocal IPMNs were collected and their clinicopathologic features catalogued. In addition, thirty multifocal IPMNs arising in 13 patients from 3 hospitals were subjected to laser microdissection followed by KRAS pyrosequencing and loss of heterozygosity (LOH) analysis on chromosomes 6q and 17p. Finally, we sought to assess the clonal relationships among multifocal IPMNs. RESULTS We identified 34 patients with histologically documented multifocal IPMNs. Synchronous IPMNs were present in 29 patients (85%), whereas 5 (15%) developed clinically significant metachronous IPMNs. Six patients (18%) had a history of familial pancreatic cancer. A majority of multifocal IPMNs (86% synchronous, 100% metachronous) were composed of branch duct lesions, and typically demonstrated a gastric-foveolar subtype epithelium with low or intermediate grades of dysplasia. Three synchronous IPMNs (10%) had an associated invasive cancer. Molecular analysis of multiple IPMNs from 13 patients demonstrated nonoverlapping KRAS gene mutations in 8 patients (62%) and discordant LOH profiles in 7 patients (54%); independent genetic alterations were established in 9 of the 13 patients (69%). CONCLUSIONS The majority of multifocal IPMNs arise independently and exhibit a gastric-foveolar subtype, with low to intermediate dysplasia. These findings underscore the importance of life-long follow-up after resection for an IPMN.
Collapse
|
215
|
Abstract
Accumulating data indicate that clinically available abdominal imaging tests such as EUS and MRI/MRCP can detect asymptomatic precursor benign (IPMN, PanIN) and invasive malignant pancreatic neoplasms, such as ductal adenocarcinoma, in individuals with an inherited predisposition. These asymptomatic FPCs detected have been more likely to be resectable, compared to symptomatic tumors. The most challenging part of screening high-risk individuals is the selection of individuals with high-grade precursor neoplasms for preventive treatment (ie, surgical resection before development of invasive cancer). Ongoing and future research should focus on formulating and validating a model for FPC risk and neoplastic progression using patient characteristics, imaging, and biomarkers. The comparative cost and effectiveness of various approaches for screening and surveillance of high-risk individuals also deserves study. For now, screening is best performed in high-risk individuals within the research protocols in academic centers with multidisciplinary teams with expertise in genetics, gastroenterology, radiology, surgery, and pathology.
Collapse
|
216
|
Fusaroli P, Kypraios D, Eloubeidi MA, Caletti G. Levels of evidence in endoscopic ultrasonography: a systematic review. Dig Dis Sci 2012; 57:602-609. [PMID: 22057240 DOI: 10.1007/s10620-011-1961-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 10/20/2011] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS A prolific trend currently designates endoscopic ultrasonography (EUS) literature. We aimed to record all EUS-studies published during the past decade and evaluate them in terms of scientific quality, creating a stratification based on levels of evidence (LE). METHODS All articles on EUS published between January 2001 and December 2010 were retrieved using a Pubmed and Cochrane Library search. Inclusion criteria were: original research papers (randomized controlled trials-RCTs, prospective and retrospective studies), meta-analyses, reviews and surveys pertinent to gastrointestinal EUS. Levels of evidence (LE) were assessed using the North of England evidence-based guidelines. RESULTS Overall, 1,832 eligible articles were reviewed. The majority (46.1%) of reports comprised retrospective descriptive studies (LE III). Expert reviews and committee reports (LE IV) accounted for 28.9%, well-designed quasi-experimental studies (LE IIb) for 20.1%, RCTs (LE Ib) for 2.4%, prospective controlled trials (LE IIa) for 1.4%, and meta-analyses (LE Ia) for 1.1% of the total. High LE (Ia-Ib) were assigned to loco-regional staging of luminal gastrointestinal cancers; mediastinal staging of lung cancer; diagnostic work-up of solid pancreatic tumors, suspected biliary obstruction and choledocholithiasis; celiac plexus neurolysis; and pancreatic pseudocysts drainage. Intermediate to low LE (IIa-IV) were assigned to submucosal tumors, pancreatic cysts, chronic pancreatitis and novel therapeutic applications (pancreato-biliary drainage, tumor ablation). CONCLUSIONS Diagnostic and staging EUS has matured and has proven its clinical impact on patient management. Therapeutic or interventional EUS is still evolving and more quality research and data are needed to establish EUS as the best next intervention to perform once firm evidence is available.
Collapse
Affiliation(s)
- Pietro Fusaroli
- Department of Clinical Medicine, GI Unit, University of Bologna/Hospital of Imola, Bologna, Italy.
| | | | | | | |
Collapse
|
217
|
Abstract
PURPOSE MicroRNA (miRNA) alterations are likely to contribute to the development of pancreatic cancer and may serve as markers for the early detection of pancreatic neoplasia. EXPERIMENTAL DESIGN To identify the miRNA alterations that arise during the development of pancreatic cancer, we determined the levels of 735 miRNAs in 34 pancreatic intraepithelial neoplasias (PanIN) and 15 normal pancreatic duct samples isolated by laser capture microdissection using TaqMan miRNA microarrays. Differential expression of selected miRNAs was confirmed by FISH analysis and by quantitative real-time reverse transcription PCR (qRT-PCR) analysis of selected candidate miRNAs in an independent set of PanIN and normal duct samples. RESULTS We identified 107 aberrantly expressed miRNAs in different PanIN grades compared with normal pancreatic duct samples and 35 aberrantly expressed miRNAs in PanIN-3 lesions compared with normal pancreatic duct samples. These differentially expressed miRNAs included those that have been previously identified as differentially expressed in pancreatic ductal adenocarcinomas (PDAC; including miR-21, miR-200a/b/c, miR-216a/b, miR-217, miR-146a, miR-155, miR-182, miR-196b, miR-203, miR-222, miR-338-3p, miR-486-3p, etc.) as well as miRNAs not previously described as differentially expressed in these lesions (miR-125b, miR-296-5p, miR-183*, miR-603, miR-625/*, miR-708, etc.). miR-196b was the most selectively differentially expressed miRNA in PanIN-3 lesions. CONCLUSIONS Many miRNAs undergo aberrant expression in PanIN lesions and are likely to be important in the development of PDAC. The miRNAs, such as miR-196b, whose expression is limited to PanIN-3 lesions or pancreatic cancers could be useful as diagnostic markers.
Collapse
Affiliation(s)
- Jun Yu
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Ang Li
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Seung-Mo Hong
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Ralph H. Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Michael Goggins
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| |
Collapse
|
218
|
Abstract
Pancreatic cancer is the fourth leading cause of cancer death in both men and women in the United States. However, it has the poorest prognosis of any major tumor type, with a 5-yr survival rate of approximately 5%. Cigarette smoking, increased body mass index, heavy alcohol consumption, and a diagnosis of diabetes mellitus have all been demonstrated to increase risk of pancreatic cancer. A family history of pancreatic cancer has also been associated with increased risk suggesting inherited genetic factors also play an important role, with approximately 5-10% of pancreatic cancer patients reporting family history of pancreatic cancer. While the genetic basis for the majority of the familial clustering of pancreatic cancer remains unclear, several important pancreatic cancer genes have been identified. These consist of high penetrance genes including BRCA2 or PALB2, to more common genetic variation associated with a modest increase risk of pancreatic cancer such as genetic variation at the ABO blood group locus. Recent advances in genotyping and genetic sequencing have accelerated the rate at which novel pancreatic cancer susceptibility genes have been identified with several genes identified within the past few years. This review addresses our current understanding of the familial aggregation of pancreatic cancer, established pancreatic cancer susceptablity genes and how this knowledge informs risk assessment and screening for high-risk families.
Collapse
Affiliation(s)
- Alison P Klein
- Department of Oncology and Pathology, Sol Goldman Pancreatic Cancer Research Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| |
Collapse
|
219
|
Czakó L, Szepes Z, Szepes A. [Diagnostic endoscopic ultrasonography in the gastrointestinal tract]. Orv Hetil 2012; 153:93-101. [PMID: 22236415 DOI: 10.1556/oh.2012.29288] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Endoscopic ultrasonography is a relatively new technology that combines the use of flexible fiberoptic endoscopes with high-resolution ultrasound technology. It proved to be highly accurate and useful in the staging of gastrointestinal malignancies, as well as in characterizing the nature of subepithelial lesions and disorders of the pancreaticobiliary system. In this overview the variety of current evidence based diagnostic options of endoscopic ultrasonography are discussed.
Collapse
Affiliation(s)
- László Czakó
- Általános Orvostudományi Kar I. Belgyógyászati Klinika Szeged Pf. 427 6701.
| | | | | |
Collapse
|
220
|
Sakorafas GH, Tsiotos GG, Korkolis D, Smyrniotis V. Individuals at high-risk for pancreatic cancer development: management options and the role of surgery. Surg Oncol 2012; 21:e49-58. [PMID: 22244849 DOI: 10.1016/j.suronc.2011.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 12/09/2011] [Accepted: 12/13/2011] [Indexed: 12/19/2022]
Abstract
Pancreatic cancer (PC) is a highly lethal disease. Despite advances regarding the safety and long-term results of pancreatectomies, early diagnosis remains the only hope for cure. This necessitates the implementation of an intensive screening program (based mainly on modern imaging), which - given the incidence of PC - is not cost effective for the general population. However, this screening program is recommended for individuals at high-risk for PC development. Indications for screening include the following three clinical settings: hereditary cancer predisposition syndromes associated with PC, hereditary pancreatitis and familial pancreatic cancer syndrome. The aim of this strategy is to identify pre-invasive (precursor) lesions, which are curable. Surgery is recommended in the presence of recognizable lesion on imaging lesions. Partial (anatomic) pancreatectomy - depending on the location of the suspicious lesion - is the most widely accepted type of surgical intervention in this setting; occasionally, however, total pancreatectomy may be required, in carefully selected patients. Despite that experience still remains limited, there is evidence that this aggressive strategy allows early detection of neoplastic lesions, thereby improving the effectiveness of surgery and prognosis.
Collapse
Affiliation(s)
- George H Sakorafas
- Department of Surgery, Athens University, Medical School, Arkadias 19-21, Athens, Greece.
| | | | | | | |
Collapse
|
221
|
Aggarwal G, Rabe KG, Petersen GM, Chari ST. New-onset diabetes in pancreatic cancer: a study in the primary care setting. Pancreatology 2012; 12:156-61. [PMID: 22487526 PMCID: PMC4348043 DOI: 10.1016/j.pan.2012.02.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Accepted: 02/03/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND Onset of diabetes mellitus (DM) is often first noted by primary care physicians. New-onset DM (duration <36 months before PaC diagnosis) can be a harbinger of pancreatic cancer (PaC). However, its clinical significance remains unclear. METHODS To determine the prevalence, onset, and delay in diagnosis of DM in PaC patients in the primary care setting, we retrospectively reviewed the records of consecutive patients followed in Mayo Clinic's primary care clinics (at least one visit in the preceding 2 years) from 1995 to 2009 who were eventually diagnosed with PaC. Onset of DM was the first date the fasting blood glucose was ≥126 mg/dl. RESULTS Of the 111 PaC patients (59 male, median age 74 years), 52 (47%) had DM of whom 30 (58%) had new-onset DM. Among the 30 with new-onset DM, 24 (80%) were asymptomatic (no cancer-specific symptoms), at DM onset. In these 24 patients, median duration of DM prior to PaC diagnosis was 6.5 (0.5-35) months, and median delay between onset and physician diagnosis of DM was 2.5 (0.25-14) months, which decreased from 8.8 (3.5-14) months in patients with DM onset between 1995 and 1999 to 0 (0-2) months, in patients with DM onset between 2004 and 2009. However, the proportion of patients with undiagnosed DM (~33%) remained unchanged. CONCLUSIONS Diabetes is very common (~50%) in PaC. In over a fifth of PaC, the onset of DM occurs when the cancer is asymptomatic, providing a potential window-of-opportunity to diagnose early PaC. However, nearly a third of new-onset DM in PaC remains undiagnosed.
Collapse
Affiliation(s)
- Gaurav Aggarwal
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Kari G. Rabe
- Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Gloria M. Petersen
- Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Suresh T. Chari
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA, Corresponding author. Fax: +1 507 284 5486. (S.T. Chari)
| |
Collapse
|
222
|
Intraductal papillary mucinous neoplasms: does a family history of pancreatic cancer matter? Pancreatology 2012; 12:358-63. [PMID: 22898638 PMCID: PMC3806100 DOI: 10.1016/j.pan.2012.05.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 05/29/2012] [Accepted: 05/30/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND/OBJECTIVES The purpose of this study is to compare surgically resected intraductal papillary mucinous neoplasms (IPMNs) in patients with and without a family history of pancreatic cancer to gain insight into differences that may suggest the need for differential management. METHODS A retrospective review of patients who underwent resection of an IPMN at the Massachusetts General Hospital (1990-2011) was conducted. Three hundred and twenty-four patients of whom 45 (13.9%) had a family history of pancreatic cancer were identified. Patients with (PFH) and without (NFH) a family history of pancreatic cancer were compared. RESULTS There were no differences in demographic characteristics between groups. Extra-pancreatic malignancies diagnosed prior to the IPMN were more common in those with a PFH (35.6% vs 20.1%, p = 0.03). There were no differences in IPMN characteristics between groups including no difference in the presence of invasive disease (p = 0.55). Concurrent pancreatic ductal adenocarcinomas were more common in those with a PFH (11.1% vs 2.9%, p = 0.02). The survival in the PFH group was marginally lower than the NFH group, a difference found to be attributable to the higher prevalence of extra-pancreatic malignancies. CONCLUSION Characteristics of surgically resected IPMNs are not different between patients with and without a family history of pancreatic cancer. Most importantly, the incidence of invasive disease is not different, suggesting that these lesions may not be more aggressive when they occur in the presence of a family history of pancreatic cancer.
Collapse
|
223
|
Abstract
The evaluation, management, and follow-up of patients with chronic pancreatitis (CP) can be simple, but it can also be complex, so having a good referral network of subspecialists experienced in this field is essential. Identifying the cause of CP requires a systematic review of the many potential causes when the cause is not obvious. The identification of patients with autoimmune CP is particularly important because treatment with steroids may be effective. Alterations in pain or other symptoms in patients with CP should not be attributed to worsening disease before evaluations for complications including malignancy are done.
Collapse
Affiliation(s)
- John Affronti
- Division of Gastroenterology, Hepatology and Nutrition, Stritch School of Medicine, Loyola University of Chicago, 2160 South First Avenue, Maywood, IL 60153, USA.
| |
Collapse
|
224
|
Hong SM, Omura N, Vincent A, Li A, Knight S, Yu J, Hruban RH, Goggins M. Genome-wide CpG island profiling of intraductal papillary mucinous neoplasms of the pancreas. Clin Cancer Res 2011; 18:700-12. [PMID: 22173550 DOI: 10.1158/1078-0432.ccr-11-1718] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Intraductal papillary mucinous neoplasms (IPMN) are precursors to infiltrating pancreatic ductal adenocarcinomas. Widespread epigenetic alterations are characteristic of many cancers, yet few studies have systematically analyzed epigenetic alterations of neoplastic precursors. Our goal was to conduct genome-wide CpG island methylation profiling to identify aberrantly methylated loci in IPMNs. EXPERIMENTAL DESIGN We compared the CpG island methylation profiles of six IPMNs to normal primary pancreatic duct samples using methylation CpG island amplification (MCA) and Agilent CpG island microarray (MCAM) analysis. When selected 13 genes identified as differentially methylated by MCAM for methylation-specific PCR (MSP) analysis in an independent set of IPMNs and normal pancreas samples and conducted expression analysis of selected genes. RESULTS We identified 2,259 loci as differentially methylated in at least one of six IPMNs including 245 genes hypermethylated in IPMNs with high-grade dysplasia compared with normal pancreatic duct samples. Eleven of 13 genes evaluated by MSP were more commonly methylated in 61 IPMNs than in 43 normal pancreas samples. Several genes (BNIP3, PTCHD2, SOX17, NXPH1, EBF3) were significantly more likely to be methylated in IPMNs with high-grade than with low-grade dysplasia. One gene, SOX17, showed loss of protein expression by immunohistochemistry in 22% (19 of 88) of IPMNs. The most specific marker, BNIP3, was not methylated in any IPMNs with low-grade dysplasia or in normal pancreas samples. CONCLUSIONS IPMNs undergo extensive aberrant CpG island hypermethylation. The detection of genes selectively methylated in high-grade IPMNs such as BNIP3 may have use in the clinical evaluation of IPMNs.
Collapse
Affiliation(s)
- Seung-Mo Hong
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland 21231, USA
| | | | | | | | | | | | | | | |
Collapse
|
225
|
Beane JD, House MG, Coté GA, DeWitt JM, Al-Haddad M, LeBlanc JK, McHenry L, Sherman S, Schmidt CM, Zyromski NJ, Nakeeb A, Pitt HA, Lillemoe KD. Outcomes after preoperative endoscopic ultrasonography and biopsy in patients undergoing distal pancreatectomy. Surgery 2011; 150:844-53. [PMID: 22000199 DOI: 10.1016/j.surg.2011.07.068] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 07/22/2011] [Indexed: 12/26/2022]
Abstract
BACKGROUND This retrospective cohort study analyzes the potential risks associated with preoperative fine needle aspiration (FNA) biopsy guided by endoscopic ultrasonography (EUS) in patients undergoing distal pancreatectomy. METHODS Excluding 204 patients with acute or chronic pancreatitis and those with previous pancreatic resections, 230 consecutive patients with primary pancreatic neoplasms underwent elective distal pancreatectomy between 2002 and 2009. The most common indications were adenocarcinoma (28%), intraductal papillary mucinous neoplasm (IPMN; 20%), and endocrine neoplasms (17%). Two-way statistical comparisons were performed between patients who did (EUS(+)) or did not (EUS(-)) undergo preoperative EUS-FNA. RESULTS Distal pancreatectomy was performed open in 118 patients (56%) and laparoscopically in 102 patients (44%). No differences were observed in age, sex, American Society of Anesthesiologists class, operative time, or blood loss between the EUS(+) (n = 179) and EUS(-) (n = 51) groups. Splenectomy was performed in 162 patients (70%) and was more common in the EUS(+) group. With the exception of adenocarcinoma (n = 57 [32%] EUS(+) vs n = 6 [12%] EUS(-); P < .01), the final pathologic diagnosis did not differ significantly between the EUS groups. Postoperative complications were more common in the EUS(+) patients with cystic neoplasms (43% vs 16% EUS(-); P = .04). EUS-FNA caused pancreatitis in 2 patients preoperatively. No differences in overall or recurrence-free survival were noted between cancer patients in the EUS groups. Patterns of tumor recurrence were not associated with EUS-FNA. CONCLUSION Preoperative EUS-FNA is not associated with adverse perioperative or long-term outcomes in patients undergoing distal pancreatectomy for solid neoplasms of the pancreas. The potentially detrimental long-term impact of preoperative EUS-FNA in patients with resectable pancreatic adenocarcinoma was not observed, but will require additional study.
Collapse
Affiliation(s)
- Joal D Beane
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Harinck F, Nagtegaal T, Kluijt I, Aalfs C, Smets E, Poley JW, Wagner A, van Hooft J, Fockens P, Bruno M, Bleiker EMA. Feasibility of a pancreatic cancer surveillance program from a psychological point of view. Genet Med 2011; 13:1015-24. [DOI: 10.1097/gim.0b013e31822934f5] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
227
|
Yu J, Li A, Hong SM, Hruban RH, Goggins M. MicroRNA alterations of pancreatic intraepithelial neoplasias. Clin Cancer Res 2011. [PMID: 22114139 DOI: 10.1158/1078-0432.ccr- 11-2347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE MicroRNA (miRNA) alterations are likely to contribute to the development of pancreatic cancer and may serve as markers for the early detection of pancreatic neoplasia. EXPERIMENTAL DESIGN To identify the miRNA alterations that arise during the development of pancreatic cancer, we determined the levels of 735 miRNAs in 34 pancreatic intraepithelial neoplasias (PanIN) and 15 normal pancreatic duct samples isolated by laser capture microdissection using TaqMan miRNA microarrays. Differential expression of selected miRNAs was confirmed by FISH analysis and by quantitative real-time reverse transcription PCR (qRT-PCR) analysis of selected candidate miRNAs in an independent set of PanIN and normal duct samples. RESULTS We identified 107 aberrantly expressed miRNAs in different PanIN grades compared with normal pancreatic duct samples and 35 aberrantly expressed miRNAs in PanIN-3 lesions compared with normal pancreatic duct samples. These differentially expressed miRNAs included those that have been previously identified as differentially expressed in pancreatic ductal adenocarcinomas (PDAC; including miR-21, miR-200a/b/c, miR-216a/b, miR-217, miR-146a, miR-155, miR-182, miR-196b, miR-203, miR-222, miR-338-3p, miR-486-3p, etc.) as well as miRNAs not previously described as differentially expressed in these lesions (miR-125b, miR-296-5p, miR-183*, miR-603, miR-625/*, miR-708, etc.). miR-196b was the most selectively differentially expressed miRNA in PanIN-3 lesions. CONCLUSIONS Many miRNAs undergo aberrant expression in PanIN lesions and are likely to be important in the development of PDAC. The miRNAs, such as miR-196b, whose expression is limited to PanIN-3 lesions or pancreatic cancers could be useful as diagnostic markers.
Collapse
Affiliation(s)
- Jun Yu
- Department of Pathology, Oncology, and Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | | | | | | | | |
Collapse
|
228
|
Yu J, Li A, Hong SM, Hruban RH, Goggins M. MicroRNA alterations of pancreatic intraepithelial neoplasias. Clin Cancer Res 2011. [PMID: 22114139 DOI: 10.1158/1073-0432.ccr-1-2347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE MicroRNA (miRNA) alterations are likely to contribute to the development of pancreatic cancer and may serve as markers for the early detection of pancreatic neoplasia. EXPERIMENTAL DESIGN To identify the miRNA alterations that arise during the development of pancreatic cancer, we determined the levels of 735 miRNAs in 34 pancreatic intraepithelial neoplasias (PanIN) and 15 normal pancreatic duct samples isolated by laser capture microdissection using TaqMan miRNA microarrays. Differential expression of selected miRNAs was confirmed by FISH analysis and by quantitative real-time reverse transcription PCR (qRT-PCR) analysis of selected candidate miRNAs in an independent set of PanIN and normal duct samples. RESULTS We identified 107 aberrantly expressed miRNAs in different PanIN grades compared with normal pancreatic duct samples and 35 aberrantly expressed miRNAs in PanIN-3 lesions compared with normal pancreatic duct samples. These differentially expressed miRNAs included those that have been previously identified as differentially expressed in pancreatic ductal adenocarcinomas (PDAC; including miR-21, miR-200a/b/c, miR-216a/b, miR-217, miR-146a, miR-155, miR-182, miR-196b, miR-203, miR-222, miR-338-3p, miR-486-3p, etc.) as well as miRNAs not previously described as differentially expressed in these lesions (miR-125b, miR-296-5p, miR-183*, miR-603, miR-625/*, miR-708, etc.). miR-196b was the most selectively differentially expressed miRNA in PanIN-3 lesions. CONCLUSIONS Many miRNAs undergo aberrant expression in PanIN lesions and are likely to be important in the development of PDAC. The miRNAs, such as miR-196b, whose expression is limited to PanIN-3 lesions or pancreatic cancers could be useful as diagnostic markers.
Collapse
Affiliation(s)
- Jun Yu
- Department of Pathology, Oncology, and Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | | | | | | | | |
Collapse
|
229
|
Familial pancreatic cancer and hereditary syndromes: screening strategy for high-risk individuals. J Gastroenterol 2011; 46:1249-59. [PMID: 21847571 DOI: 10.1007/s00535-011-0457-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 07/25/2011] [Indexed: 02/04/2023]
Abstract
Globally, and almost evenly across nations, a familial disposition can be found in 4-10% of patients with pancreatic cancer (PC). A family history of PC is a risk for this disease and the risk level changes in correlation with the number of affected relatives. Several hereditary syndromes with potential germline mutation also have a high risk for PC; however, little is yet known regarding the genes responsible for familial pancreatic cancer (FPC). Characteristics of FPC cases are similar to those of other familial tumors, including younger onset than in sporadic cases and an ethnic difference (Ashkenazi Jewish > other Caucasian). Other risks resemble those of sporadic cases and include smoking and diabetes mellitus. People with several genetic syndromes, including Peutz-Jeghers syndrome, hereditary pancreatitis, breast-ovarian cancer syndrome, hereditary nonpolyposis colorectal cancer, and familial adenomatous polyposis also have an increased risk of PC. In many countries, but not yet in Japan, screening of these high-risk individuals is now ongoing for the detection of early PC under established familial pancreatic cancer registries. In addition to the ordinary risk factors, such as smoking, diabetes, pancreatitis, cysts, duct ectasia, and intraductal papillary mucinous neoplasm (IPMN), individuals with a family history of PC and hereditary syndromes are expected to be entered into the screening protocol.
Collapse
|
230
|
Lubezky N, Ben-Haim M, Lahat G, Marmor S, Solar I, Brazowski E, Nackache R, Klausner JM. Intraductal papillary mucinous neoplasm of the pancreas: associated cancers, family history, genetic predisposition? Surgery 2011; 151:70-5. [PMID: 21975290 DOI: 10.1016/j.surg.2011.06.036] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2010] [Accepted: 06/16/2011] [Indexed: 12/17/2022]
Abstract
BACKGROUND High rates of extrapancreatic malignancies (EPM) have been observed in patients with intraductal papillary mucinous neoplasm (IPMN). IPMN in patients with familial pancreatic cancer have also been reported. Our purpose was to evaluate the association of IPMN with EPM, malignancies in family members, and germline BRCA1 and BRCA2 mutations. METHODS Using retrospective analysis on prospectively collected data from 82 patients with IPMN and direct contact for familial cancer history, data were compared with those of 150 patients with pancreatic ductal adenocarcinoma (PDAC). The common germline mutations in the BRCA1 and BRCA2 genes were evaluated on available IPMN patients. RESULTS EPM rates were greater in IPMN than PDAC patients (P = .002). Malignancies in first-degree relatives, specifically pancreatic cancer, were more common among IPMN than PDAC patients (P = .028). IPMN patients with EPM had high rates of relatives with colorectal cancer (31%). Two of the 51 genetically tested patients (4%) were BRCA2 mutation carriers, and both had first-degree relatives with pancreatic cancer. One patient fulfilled the Amsterdam criteria for hereditary nonpolyposis colon cancer; however, the neoplasm was microsatellite stable. CONCLUSION Our results demonstrated high rates of EPM among IPMN patients. There was an increased rate of cancer in families of IPMN patients, specifically pancreatic cancer. A high rate of colorectal cancer in families of IPMN patients who have EPM was also observed. These findings suggest a genetic component in the pathogenesis of IPMN. Possible genetic changes include BRCA2 mutations, which are found in 25% of IPMN patients with a family history of pancreatic cancer.
Collapse
Affiliation(s)
- Nir Lubezky
- Department of Surgery B, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | | | | | | | | | | | | | | |
Collapse
|
231
|
Schneider R, Slater EP, Sina M, Habbe N, Fendrich V, Matthäi E, Langer P, Bartsch DK. German national case collection for familial pancreatic cancer (FaPaCa): ten years experience. Fam Cancer 2011. [PMID: 21207249 DOI: 10.1007/s10689-010 -9414-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Familial pancreatic cancer (FPC) is a rare hereditary tumor syndrome. The 10-years experience of the national case collection for familial pancreatic cancer of Germany (FaPaCa) is reported. Since 1999 FaPaCa has collected families with at least two first-degree relatives with confirmed pancreatic cancer (PC), who did not fulfill the criteria of other hereditary tumor syndromes. Histopathological verification of tumor diagnoses, and genetic counseling were prerequisites for enrollment of families in FaPaCa. 94 of 452 evaluated families fulfilled the criteria for partaking in FaPaCa. PC represented the sole tumor entity in 38 (40%) families. In 56 families additional tumor types occurred, including breast cancer (n = 28), colon cancer (n = 20) and lung cancer (n = 11). In 70 (74%) families the pattern of inheritance was consistent with an autosomal dominant trait. Compared to the preceding generation, a younger age of onset was observed in the offspring of PC patients (median: 57 vs. 69 years), indicating anticipation. Mutation analyses of BRCA2, PALB2, CDKN2a, RNASEL, STK11, NOD2, CHEK2 and PALLD, revealed deleterious causative germline mutations of BRCA2 and PALB2 in 2 of 70 (3%) and 2 of 41 (4.9%) German FPC families, respectively. Prospective PC screening with EUS, MRI and MRCP detected precancerous lesions (IPMN, multifocal PanIN2/3) or carcinoma in 5.5% (4 of 72) to 12.5% (9 of 72) of individuals at risk, depending on histological verification. Appropriate inclusion of families at high risk for PC in registries, such as FaPaCa, provides a unique and excellent tool to gain clinical and genetic knowledge of FPC. Focused research projects can be conducted most efficiently, when data of different FPC registries are combined.
Collapse
Affiliation(s)
- Ralph Schneider
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Baldingerstrasse, 35043 Marburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
232
|
Schneider R, Slater EP, Sina M, Habbe N, Fendrich V, Matthäi E, Langer P, Bartsch DK. German national case collection for familial pancreatic cancer (FaPaCa): ten years experience. Fam Cancer 2011; 10:323-30. [PMID: 21207249 DOI: 10.1007/s10689-010-9414-x] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Familial pancreatic cancer (FPC) is a rare hereditary tumor syndrome. The 10-years experience of the national case collection for familial pancreatic cancer of Germany (FaPaCa) is reported. Since 1999 FaPaCa has collected families with at least two first-degree relatives with confirmed pancreatic cancer (PC), who did not fulfill the criteria of other hereditary tumor syndromes. Histopathological verification of tumor diagnoses, and genetic counseling were prerequisites for enrollment of families in FaPaCa. 94 of 452 evaluated families fulfilled the criteria for partaking in FaPaCa. PC represented the sole tumor entity in 38 (40%) families. In 56 families additional tumor types occurred, including breast cancer (n = 28), colon cancer (n = 20) and lung cancer (n = 11). In 70 (74%) families the pattern of inheritance was consistent with an autosomal dominant trait. Compared to the preceding generation, a younger age of onset was observed in the offspring of PC patients (median: 57 vs. 69 years), indicating anticipation. Mutation analyses of BRCA2, PALB2, CDKN2a, RNASEL, STK11, NOD2, CHEK2 and PALLD, revealed deleterious causative germline mutations of BRCA2 and PALB2 in 2 of 70 (3%) and 2 of 41 (4.9%) German FPC families, respectively. Prospective PC screening with EUS, MRI and MRCP detected precancerous lesions (IPMN, multifocal PanIN2/3) or carcinoma in 5.5% (4 of 72) to 12.5% (9 of 72) of individuals at risk, depending on histological verification. Appropriate inclusion of families at high risk for PC in registries, such as FaPaCa, provides a unique and excellent tool to gain clinical and genetic knowledge of FPC. Focused research projects can be conducted most efficiently, when data of different FPC registries are combined.
Collapse
Affiliation(s)
- Ralph Schneider
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Baldingerstrasse, 35043 Marburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
233
|
Kisiel JB, Yab TC, Taylor WR, Chari ST, Petersen GM, Mahoney DW, Ahlquist DA. Stool DNA testing for the detection of pancreatic cancer: assessment of methylation marker candidates. Cancer 2011. [PMID: 22083596 DOI: 10.1002/cncr/26558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Pancreatic cancer (PanC) presents at late stage with high mortality. Effective early detection methods are needed. Aberrantly methylated genes are unexplored as markers for noninvasive detection by stool testing. The authors aimed to select discriminant methylated genes and to assess accuracy of these and mutant KRAS in stool to detect PanC. METHODS Nine target genes were assayed by real-time methylation-specific polymerase chain reaction (MSP) in bisulfite-treated DNA from microdissected frozen specimens of 24 PanC cases and 30 normal colon controls. Archived stools from 58 PanC cases and 65 controls matched on sex, age, and smoking were analyzed. Target genes from fecal supernatants were enriched by hybrid capture, bisulfite-treated, and assayed by MSP. KRAS mutations were assayed using the QuARTS technique. RESULTS Areas under the receiver operating characteristics curves (AUCs) for tissue BMP3, NDRG4, EYA4, UCHL1, MDFI, Vimentin, CNTNAP2, SFRP2, and TFPI2 were 0.90, 0.79, 0.78, 0.78, 0.77, 0.77, 0.69, 0.67, and 0.66, respectively. The top 4 markers and mutant KRAS were evaluated in stool. BMP3 was the most discriminant methylation marker in stool. At 90% specificity, methylated BMP3 alone detected 51% of PanCs, mutant KRAS detected 50%, and combination detected 67%. AUCs for methylated BMP3, mutant KRAS, and combination in stool were 0.73, 0.75, and 0.85, respectively. CONCLUSIONS This study demonstrates that stool assay of a methylated gene marker can detect PanC. Among candidate methylated markers discriminant in tissue, BMP3 alone performed well in stool. Combining methylated BMP3 and mutant KRAS increased stool detection over either marker alone.
Collapse
Affiliation(s)
- John B Kisiel
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | |
Collapse
|
234
|
Kisiel JB, Yab TC, Taylor WR, Chari ST, Petersen GM, Mahoney DW, Ahlquist DA. Stool DNA testing for the detection of pancreatic cancer: assessment of methylation marker candidates. Cancer 2011; 118:2623-31. [PMID: 22083596 DOI: 10.1002/cncr.26558] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 08/10/2011] [Indexed: 12/21/2022]
Abstract
BACKGROUND Pancreatic cancer (PanC) presents at late stage with high mortality. Effective early detection methods are needed. Aberrantly methylated genes are unexplored as markers for noninvasive detection by stool testing. The authors aimed to select discriminant methylated genes and to assess accuracy of these and mutant KRAS in stool to detect PanC. METHODS Nine target genes were assayed by real-time methylation-specific polymerase chain reaction (MSP) in bisulfite-treated DNA from microdissected frozen specimens of 24 PanC cases and 30 normal colon controls. Archived stools from 58 PanC cases and 65 controls matched on sex, age, and smoking were analyzed. Target genes from fecal supernatants were enriched by hybrid capture, bisulfite-treated, and assayed by MSP. KRAS mutations were assayed using the QuARTS technique. RESULTS Areas under the receiver operating characteristics curves (AUCs) for tissue BMP3, NDRG4, EYA4, UCHL1, MDFI, Vimentin, CNTNAP2, SFRP2, and TFPI2 were 0.90, 0.79, 0.78, 0.78, 0.77, 0.77, 0.69, 0.67, and 0.66, respectively. The top 4 markers and mutant KRAS were evaluated in stool. BMP3 was the most discriminant methylation marker in stool. At 90% specificity, methylated BMP3 alone detected 51% of PanCs, mutant KRAS detected 50%, and combination detected 67%. AUCs for methylated BMP3, mutant KRAS, and combination in stool were 0.73, 0.75, and 0.85, respectively. CONCLUSIONS This study demonstrates that stool assay of a methylated gene marker can detect PanC. Among candidate methylated markers discriminant in tissue, BMP3 alone performed well in stool. Combining methylated BMP3 and mutant KRAS increased stool detection over either marker alone.
Collapse
Affiliation(s)
- John B Kisiel
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | |
Collapse
|
235
|
de Wilde RF, Ottenhof NA, Jansen M, Morsink FHM, de Leng WWJ, Offerhaus GJA, Brosens LAA. Analysis of LKB1 mutations and other molecular alterations in pancreatic acinar cell carcinoma. Mod Pathol 2011; 24:1229-36. [PMID: 21572398 DOI: 10.1038/modpathol.2011.83] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Acinar cell carcinoma is a rare non-ductal neoplasm of the pancreas with poorly defined molecular genetic features. Recently, biallelic inactivation of LKB1 was described in an acinar cell carcinoma of a Peutz-Jeghers patient carrying a heterozygous germline LKB1 mutation, and inhibition of mTOR signaling resulted in partial remission of the tumor. To explore the potential of mTOR inhibitors in sporadic acinar cell carcinoma, the LKB1 gene was investigated in five sporadic acinar cell carcinomas by sequence analysis, methylation analysis and mRNA expression. In addition, microsatellite instability and methylation of a number of tumor suppressor genes were investigated and KRAS, TP53, CDKN1A, SMAD4 and CTNNB1 were studied by mutation analysis and immunohistochemistry. No mutations, deletions or promoter hypermethylation of LKB1 were found in any of the sporadic acinar cell carcinomas, and mRNA expression of LKB1 was not altered. Amplifications at chromosome 20q and 19p were found in 100 and 80% of the cases, respectively. In addition, hypermethylation of one or more tumor suppressor genes was found in 80% of cases. One case harbored a TP53 mutation, and expression of SMAD4 and CTNNB1 was altered in one case each. No KRAS mutations or microsatellite instability were found. To conclude, no evidence for a role for LKB1 in tumorigenesis of sporadic pancreatic acinar cell carcinoma was found. However, copy number variations and hypermethylation were found in a majority of cases. Molecular pathways involved in acinar cell carcinoma-tumorigenesis differ from those involved in ductal pancreatic neoplasms. Further studies are needed to increase our understanding of molecular pathogenesis of acinar cell carcinoma, which may eventually result in development of new therapeutic targets.
Collapse
Affiliation(s)
- Roeland F de Wilde
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
236
|
Abstract
Substantial progress has been made in our understanding of the biology of pancreatic cancer, and advances in patients' management have also taken place. Evidence is beginning to show that screening first-degree relatives of individuals with several family members affected by pancreatic cancer can identify non-invasive precursors of this malignant disease. The incidence of and number of deaths caused by pancreatic tumours have been gradually rising, even as incidence and mortality of other common cancers have been declining. Despite developments in detection and management of pancreatic cancer, only about 4% of patients will live 5 years after diagnosis. Survival is better for those with malignant disease localised to the pancreas, because surgical resection at present offers the only chance of cure. Unfortunately, 80-85% of patients present with advanced unresectable disease. Furthermore, pancreatic cancer responds poorly to most chemotherapeutic agents. Hence, we need to understand the biological mechanisms that contribute to development and progression of pancreatic tumours. In this Seminar we will discuss the most common and deadly form of pancreatic cancer, pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Audrey Vincent
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD 21231, USA
| | | | | | | | | |
Collapse
|
237
|
Stratégies de dépistage de l’adénocarcinome pancréatique chez les patients à haut risque. Bull Cancer 2011; 98:827-36. [DOI: 10.1684/bdc.2011.1396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
238
|
Zubarik R, Gordon SR, Lidofsky SD, Anderson SR, Pipas JM, Badger G, Ganguly E, Vecchio J. Screening for pancreatic cancer in a high-risk population with serum CA 19-9 and targeted EUS: a feasibility study. Gastrointest Endosc 2011; 74:87-95. [PMID: 21704809 DOI: 10.1016/j.gie.2011.03.1235] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 03/21/2011] [Indexed: 02/07/2023]
Abstract
BACKGROUND Earlier detection of pancreatic adenocarcinoma is needed. OBJECTIVE To determine whether early pancreatic neoplasia can be detected in a high-risk population by using CA 19-9 followed by targeted EUS. DESIGN Prospective cohort study. SETTING Two academic medical centers. PATIENTS Eligible patients met age criteria and had at least 1 first-degree relative with pancreatic adenocarcinoma. INTERVENTIONS A serum CA 19-9 was performed on all patients. EUS was performed if the CA 19-9 level was elevated. FNA of identified lesions was performed. Patients with pancreatic cancer detected by using this screening protocol were compared with patients presenting off-protocol for staging data. Medicare reimbursement rates were used to derive cost data. MAIN OUTCOME MEASUREMENTS Detection of early pancreatic neoplasia. RESULTS A total of 546 patients were enrolled. CA 19-9 was elevated in 27 patients (4.9%, 95% CI, 3.2%-7.1%). Neoplastic or malignant findings were detected in 5 patients (0.9%, 95% CI, 0.3%-2.1%), and pancreatic adenocarcinoma in 1 patient (0.2%, 95% CI, 0.005%-1.02%). The patient with pancreatic cancer detected as part of this protocol was 1 of 2 patients presenting to the University of Vermont with stage 1 cancer. The cost to detect 1 pancreatic neoplasia was $8431. The cost to detect 1 pancreatic adenocarcinoma was $41,133. LIMITATIONS The sample size is adequate only to demonstrate the feasibility of this approach. CONCLUSIONS Potentially curative pancreatic adenocarcinoma can be identified with this screening protocol. Stage 1 pancreatic cancer is more likely to be detected by using this screening protocol than by using standard means of detection.
Collapse
Affiliation(s)
- Richard Zubarik
- Division of Gastroenterology, University of Vermont, Burlington, VT, USA.
| | | | | | | | | | | | | | | |
Collapse
|
239
|
Vincent A, Omura N, Hong SM, Jaffe A, Eshleman J, Goggins M. Genome-wide analysis of promoter methylation associated with gene expression profile in pancreatic adenocarcinoma. Clin Cancer Res 2011; 17:4341-54. [PMID: 21610144 PMCID: PMC3131423 DOI: 10.1158/1078-0432.ccr-10-3431] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE The goal of this study was to comprehensively identify CpG island methylation alterations between pancreatic cancers and normal pancreata and their associated gene expression alterations. EXPERIMENTAL DESIGN We employed methylated CpG island amplification followed by CpG island microarray, a method previously validated for its accuracy and reproducibility, to analyze the methylation profile of 27,800 CpG islands covering 21 MB of the human genome in nine pairs of pancreatic cancer versus normal pancreatic epithelial tissues and in three matched pairs of pancreatic cancer versus lymphoid tissues from the same individual. RESULTS This analysis identified 1,658 known loci that were commonly differentially methylated in pancreatic cancer compared with normal pancreas. By integrating the pancreatic DNA methylation status with the gene expression profiles of the same samples before and after treatment with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine, and the histone deacetylase inhibitor, trichostatin A, we identified dozens of aberrantly methylated and differentially expressed genes in pancreatic cancers including a more comprehensive list of hypermethylated and silenced genes that have not been previously described as targets for aberrant methylation in cancer. CONCLUSION We expected that the identification of aberrantly hypermethylated and silenced genes will have diagnostic, prognostic, and therapeutic applications.
Collapse
Affiliation(s)
- Audrey Vincent
- Department of Pathology, the Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
| | - Noriyuki Omura
- Department of Pathology, the Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
| | - Seung-Mo Hong
- Department of Pathology, the Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew Jaffe
- Bloomberg School of Public Health, the Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
| | - James Eshleman
- Department of Pathology, the Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
- Department of Oncology, the Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
| | - Michael Goggins
- Department of Pathology, the Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
- Department of Oncology, the Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
- Department of Medicine, the Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
240
|
Naito Y, Okabe Y, Nagayama M, Nishinakagawa T, Taira T, Kawahara A, Hattori S, Machida K, Ishida Y, Kaji R, Mikagi K, Kinoshita H, Yasumoto M, Akiba J, Kage M, Nakashima M, Ohshima K, Yano H. Accuracy of differential diagnosis for pancreatic cancer is improved in the combination of RCAS1 and CEA measurements and cytology in pancreatic juice. Med Mol Morphol 2011; 44:86-92. [PMID: 21717311 DOI: 10.1007/s00795-010-0511-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Accepted: 04/06/2010] [Indexed: 12/25/2022]
Abstract
Improvement of diagnostic accuracy for pancreatic cancer in pancreatic disease patients was investigated by examining the combination of three diagnostic methods, i.e., measurements of RCAS1 and CEA levels in pancreatic juice and pancreatic juice cytology. Pancreatic juice was collected from 12 pancreatic cancer (PC) and 26 non-PC patients. RCAS1 and CEA levels were measured by using ELISA. RCAS1 expression on surgically resected tissue was immunohistochemically examined for 2 PC patients. By setting the cutoff level of RCAS1 at 10 U/ml and that of CEA at 18.5 μg/ml, sensitivity of RCAS1 was 42% and that of CEA was 50%. On the other hand, sensitivity and specificity increased from 42% and 85% of RCAS1 alone to 75% and 85% in the examination of RCAS1 + CEA + cytology, and the false-negative rate was also reduced to 25% in this combination. Immunohistochemically, a patient with a high RCAS1 level in pancreatic juice had numerous RCAS1-positive tumor cells in the pancreatic juice. We concluded that RCAS1 and CEA measurements together with cytology in pancreatic juice would be a useful combination method for making a differential diagnosis of PC from non-PC.
Collapse
Affiliation(s)
- Yoshiki Naito
- Department of Pathology, Kurume University School of Medicine, 67 Asahimachi, Kurume, Fukuoka, 830-0011, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
241
|
In vivo diagnosis of murine pancreatic intraepithelial neoplasia and early-stage pancreatic cancer by molecular imaging. Proc Natl Acad Sci U S A 2011; 108:9945-50. [PMID: 21628592 DOI: 10.1073/pnas.1100890108] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a fatal disease with poor patient outcome often resulting from late diagnosis in advanced stages. To date methods to diagnose early-stage PDAC are limited and in vivo detection of pancreatic intraepithelial neoplasia (PanIN), a preinvasive precursor of PDAC, is impossible. Using a cathepsin-activatable near-infrared probe in combination with flexible confocal fluorescence lasermicroscopy (CFL) in a genetically defined mouse model of PDAC we were able to detect and grade murine PanIN lesions in real time in vivo. Our diagnostic approach is highly sensitive and specific and proved superior to clinically established fluorescein-enhanced imaging. Translation of this endoscopic technique into the clinic should tremendously improve detection of pancreatic neoplasia, thus reforming management of patients at risk for PDAC.
Collapse
|
242
|
Abstract
Pancreatic intraepithelial neoplasias (PanINs) are microscopic lesions of the pancreas. Traditionally viewed as a benign metaplasia of small ducts, evidence suggests that PanINs are neoplastic and that some PanINs progress to invasive ductal adenocarcinoma. The primary diagnostic challenge is distinguishing PanINs from other lesions, including invasive ductal adenocarcinoma, intraductal papillary mucinous neoplasm, and cancerization of benign ducts. PanINs are the most common of the pancreatic cancer precursor lesions, yet they remain poorly understood and are so small that they are almost clinically undetectable. Further study is required to define the role of PanINs in the carcinogenesis and early detection of pancreatic cancer.
Collapse
|
243
|
Vincent A, Herman J, Schulick R, Hruban RH, Goggins M. Pancreatic cancer. LANCET (LONDON, ENGLAND) 2011. [PMID: 21620466 DOI: 10.1016/so140-6736(10)62307-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Substantial progress has been made in our understanding of the biology of pancreatic cancer, and advances in patients' management have also taken place. Evidence is beginning to show that screening first-degree relatives of individuals with several family members affected by pancreatic cancer can identify non-invasive precursors of this malignant disease. The incidence of and number of deaths caused by pancreatic tumours have been gradually rising, even as incidence and mortality of other common cancers have been declining. Despite developments in detection and management of pancreatic cancer, only about 4% of patients will live 5 years after diagnosis. Survival is better for those with malignant disease localised to the pancreas, because surgical resection at present offers the only chance of cure. Unfortunately, 80-85% of patients present with advanced unresectable disease. Furthermore, pancreatic cancer responds poorly to most chemotherapeutic agents. Hence, we need to understand the biological mechanisms that contribute to development and progression of pancreatic tumours. In this Seminar we will discuss the most common and deadly form of pancreatic cancer, pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Audrey Vincent
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Johns Hopkins University, Baltimore, MD 21231, USA
| | | | | | | | | |
Collapse
|
244
|
Kuroki-Suzuki S, Kuroki Y, Nasu K, Nagashima C, Machida M, Muramatsu Y, Moriyama N. Pancreatic cancer screening employing noncontrast magnetic resonance imaging combined with ultrasonography. Jpn J Radiol 2011; 29:265-71. [PMID: 21607840 DOI: 10.1007/s11604-010-0554-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Accepted: 12/16/2010] [Indexed: 01/06/2023]
Abstract
PURPOSE We have conducted an initial evaluation on the potential of combining noncontrast magnetic resonance imaging (MRI) and ultrasonography (US) to screen for pancreatic cancer. MATERIALS AND METHODS An independent ethics committee approved this study. A total of 2511 patients who underwent US were enrolled. Among them, noncontrast MRI was performed in patients in whom the entire pancreas was difficult to depict or in those with US-suspected pancreatic lesions. In total, using 1.5-T MRI, T1- and T2-weighted imaging, magnetic resonance cholangiopancreatography, and diffusion-weighted imaging, we acquired a variety of images. The efficacy of US and MRI in screening for pancreatic lesions, including pancreatic cancer, was evaluated. RESULTS Of 2511 patients, 184 underwent MRI, and the pancreas was demonstrated in all of them. Among the 2511, five pancreatic cancers were detected by MRI combined with US (detection rate 0.20%). Of the five pancreatic cancers, three were detected by US (detection rate 0.12%) and two by MRI. Four of the five pancreatic cancers were resectable. CONCLUSION By combining noncontrast MRI with US, pancreatic cancer can be detected with high accuracy. Other pancreatic lesions that require follow-up, including intraductal papillary mucinous neoplasms, can also be detected. Thus, pancreatic cancer screening with a combination of US and MRI is suggested.
Collapse
Affiliation(s)
- Seiko Kuroki-Suzuki
- Research Center for Cancer Prevention and Screening, National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | | | | | | | | | | | | |
Collapse
|
245
|
Stoita A, Penman ID, Williams DB. Review of screening for pancreatic cancer in high risk individuals. World J Gastroenterol 2011; 17:2365-71. [PMID: 21633635 PMCID: PMC3103788 DOI: 10.3748/wjg.v17.i19.2365] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 12/17/2010] [Accepted: 12/24/2010] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is difficult to diagnose at an early stage and is associated with a very poor survival. Ten percent of pancreatic cancers result from genetic susceptibility and/or familial aggregation. Individuals from families with multiple affected first-degree relatives and those with a known cancer-causing genetic mutation have been shown to be at much higher risk of developing pancreatic cancer. Recent efforts have focused on detecting disease at an earlier stage to improve survival in these high-risk groups. This article reviews high-risk groups, screening methods, and current screening programs and their results.
Collapse
|
246
|
Abstract
The concept of pancreatic cancer prevention through surveillance of high-risk patients has come of age. It is cost-effective to provide surveillance of patients who have a lifetime risk of pancreatic cancer that is ≥16%. Studies are currently ongoing that contribute to our understanding of the imaging methodologies that are best suited for surveillance and the best algorithm for the clinical management of patients who are at risk of this highly lethal disease. Long-term outcomes analyses will help shed light on the best management of these patients, as well as a better understanding of the natural history of familial pancreatic cancer.
Collapse
|
247
|
Ludwig E, Olson SH, Bayuga S, Simon J, Schattner MA, Gerdes H, Allen PJ, Jarnagin WR, Kurtz RC. Feasibility and yield of screening in relatives from familial pancreatic cancer families. Am J Gastroenterol 2011; 106:946-54. [PMID: 21468009 PMCID: PMC3683863 DOI: 10.1038/ajg.2011.65] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Pancreatic adenocarcinoma is a lethal disease. Over 80% of patients are found to have metastatic disease at the time of diagnosis. Strategies to improve disease-specific outcome include identification and early detection of precursor lesions or early cancers in high-risk groups. In this study, we investigate whether screening at-risk relatives of familial pancreatic cancer (FPC) patients is safe and has significant yield. METHODS We enrolled 309 asymptomatic at-risk relatives into our Familial Pancreatic Tumor Registry (FPTR) and offered them screening with magnetic resonance cholangiopancreaticogram (MRCP) followed by endoscopic ultrasound (EUS) with fine needle aspiration if indicated. Relatives with findings were referred for surgical evaluation. RESULTS As of 1 August 2009, 109 relatives had completed at least one cycle of screening. Abnormal radiographic findings were present on initial screening in 18/109 patients (16.5%), 15 of whom underwent EUS. A significant abnormality was confirmed in 9 of 15 patients, 6 of whom ultimately had surgery for an overall diagnostic yield of 8.3% (9/109). Yield was greatest in relatives >65 years old (35%, 6/17) when compared with relatives 55-65 years (3%, 1/31) and relatives <55 years (3%, 2/61). CONCLUSIONS Screening at-risk relatives from FPC families has a significant diagnostic yield, particularly in relatives >65 years of age, confirming prior studies. MRCP as initial screening modality is safe and effective.
Collapse
Affiliation(s)
- Emmy Ludwig
- Gastroenterology and Nutrition Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Shin SS, Armao DM, Burke LM, Kim HJ, Skrzynia C, Otey CA, Semelka RC. Comparison of the incidence of pancreatic abnormalities between high risk and control patients: Prospective pilot study with 3 tesla MR imaging. J Magn Reson Imaging 2011; 33:1080-5. [DOI: 10.1002/jmri.22551] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
249
|
Abstract
BACKGROUND Pancreatic cancer remains a devastating disease with a 5-year survival rate of less than 5%. Recent advances in diagnostic methods and therapeutic approaches have increased the possibility of improving the existing poor prognosis. DATA SOURCES English-language articles reporting early diagnosis and therapy of pancreatic cancer were searched from the MEDLINE and PubMed databases, Chinese-language articles were from CHKD (China Hospital Knowledge Database). RESULT The current literature about pancreatic cancer was reviewed from three aspects: statistics, screening and early detection, and therapy. CONCLUSIONS Early detection and screening of pancreatic cancer currently should be limited to high risk patients. Surgical resection is the only curative approach available, with some recent improvement in outcomes. Gemcitabine has been a standard treatment during the last decade. Gemcitabine-based combination treatment, especially combined with newer molecular targeted agents, is promising. The rationale for radiotherapy is controversial, but with the recent development of modern radiation delivery techniques, radiotherapy should be intensified. Patients with borderline pancreatic cancer could benefit from neoadjuvant therapy but more evidence is needed and the best neoadjuvant regimen is still to be determined.
Collapse
Affiliation(s)
- Qiang Xu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medial Sciences, Beijing 100730, China
| | | | | |
Collapse
|
250
|
Maheu C, Vodermaier A, Rothenmund H, Gallinger S, Ardiles P, Semotiuk K, Holter S, Thayalan S, Esplen MJ. Pancreatic cancer risk counselling and screening: impact on perceived risk and psychological functioning. Fam Cancer 2011; 9:617-24. [PMID: 20623197 DOI: 10.1007/s10689-010-9354-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Individuals at increased risk for pancreatic cancer who undergo screening can experience psychological and emotional distress. The objective of this study is to determine whether individuals participating in a pancreatic cancer screening program experience disruptions in risk perception, cancer-related anxiety or emotional distress. A pretestposttest design was used to examine perceived risk and psychological functioning of individuals participating in a pancreatic cancer screening protocol. The screening protocol includes genetic counselling, transcutaneous abdominal ultrasound, magnetic resonance imaging, and blood collection and eligible participants included individuals with a family history of pancreatic cancer or BRCA2 mutation carriers. At baseline, participants (n = 198) showed low to moderate levels of risk perception, pancreatic cancer-related anxiety, and general distress. Participants with familial pancreatic cancer (FPC) (n = 131) endorsed higher risk perception of pancreatic cancer than the BRCA2 carriers (n = 67) (perceived lifetime risk 42 vs. 15%), but did not differ on cancer worry or general distress prior to the first study appointment. From baseline to 3 months follow-up, no significant time or time by group interactions emerged on risk perception or general distress, but cancer worry decreased over time for the FPC group regardless of the number of affected relatives. Our findings indicate that participation in a pancreatic cancer screening program does not lead to a significant increase in risk perception, cancer worry, or general distress and that participants with high baseline levels of risk perception and distress may benefit from a more comprehensive risk assessment and psychological support.
Collapse
|