1
|
Granata V, Fusco R, Setola SV, Galdiero R, Maggialetti N, Silvestro L, De Bellis M, Di Girolamo E, Grazzini G, Chiti G, Brunese MC, Belli A, Patrone R, Palaia R, Avallone A, Petrillo A, Izzo F. Risk Assessment and Pancreatic Cancer: Diagnostic Management and Artificial Intelligence. Cancers (Basel) 2023; 15:351. [PMID: 36672301 PMCID: PMC9857317 DOI: 10.3390/cancers15020351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Pancreatic cancer (PC) is one of the deadliest cancers, and it is responsible for a number of deaths almost equal to its incidence. The high mortality rate is correlated with several explanations; the main one is the late disease stage at which the majority of patients are diagnosed. Since surgical resection has been recognised as the only curative treatment, a PC diagnosis at the initial stage is believed the main tool to improve survival. Therefore, patient stratification according to familial and genetic risk and the creation of screening protocol by using minimally invasive diagnostic tools would be appropriate. Pancreatic cystic neoplasms (PCNs) are subsets of lesions which deserve special management to avoid overtreatment. The current PC screening programs are based on the annual employment of magnetic resonance imaging with cholangiopancreatography sequences (MR/MRCP) and/or endoscopic ultrasonography (EUS). For patients unfit for MRI, computed tomography (CT) could be proposed, although CT results in lower detection rates, compared to MRI, for small lesions. The actual major limit is the incapacity to detect and characterize the pancreatic intraepithelial neoplasia (PanIN) by EUS and MR/MRCP. The possibility of utilizing artificial intelligence models to evaluate higher-risk patients could favour the diagnosis of these entities, although more data are needed to support the real utility of these applications in the field of screening. For these motives, it would be appropriate to realize screening programs in research settings.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, 41012 Napoli, Italy
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via della Signora 2, 20122 Milan, Italy
| | - Sergio Venanzio Setola
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Roberta Galdiero
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Nicola Maggialetti
- Department of Medical Science, Neuroscience and Sensory Organs (DSMBNOS), University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Lucrezia Silvestro
- Division of Clinical Experimental Oncology Abdomen, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Mario De Bellis
- Division of Gastroenterology and Digestive Endoscopy, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Elena Di Girolamo
- Division of Gastroenterology and Digestive Endoscopy, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Giulia Grazzini
- Department of Emergency Radiology, University Hospital Careggi, Largo Brambilla 3, 50134 Florence, Italy
| | - Giuditta Chiti
- Department of Emergency Radiology, University Hospital Careggi, Largo Brambilla 3, 50134 Florence, Italy
| | - Maria Chiara Brunese
- Diagnostic Imaging Section, Department of Medical and Surgical Sciences & Neurosciences, University of Molise, 86100 Campobasso, Italy
| | - Andrea Belli
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Renato Patrone
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Raffaele Palaia
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Antonio Avallone
- Division of Clinical Experimental Oncology Abdomen, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Antonella Petrillo
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Francesco Izzo
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| |
Collapse
|
2
|
Huang C, Simeone DM, Luk L, Hecht EM, Khatri G, Kambadakone A, Chandarana H, Ream JM, Everett JN, Guimaraes A, Liau J, Dasyam AK, Harmath C, Megibow AJ. Standardization of MRI Screening and Reporting in Individuals With Elevated Risk of Pancreatic Ductal Adenocarcinoma: Consensus Statement of the PRECEDE Consortium. AJR Am J Roentgenol 2022; 219:903-914. [PMID: 35856454 DOI: 10.2214/ajr.22.27859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies, with a dismal survival rate. Screening the general population for early detection of PDAC is not recommended, but because early detection improves survival, high-risk individuals, defined as those meeting criteria based on a family history of PDAC and/or the presence of known pathogenic germline variant genes with PDAC risk, are recommended to undergo screening with MRI and/or endoscopic ultrasound at regular intervals. The Pancreatic Cancer Early Detection (PRECEDE) Consortium was formed in 2018 and is composed of gastroenterologists, geneticists, pancreatic surgeons, radiologists, statisticians, and researchers from 40 sites in North America, Europe, and Asia. The overarching goal of the PRECEDE Consortium is to facilitate earlier diagnosis of PDAC for high-risk individuals to increase survival of the disease. A standardized MRI protocol and reporting template are needed to enhance the quality of screening examinations, improve consistency of clinical management, and facilitate multiinstitutional research. We present a consensus statement to standardize MRI screening and reporting for individuals with elevated risk of pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | | | - Gaurav Khatri
- University of Texas Southwestern Medical Center, Dallas, TX
| | | | | | | | | | | | - Joy Liau
- University of California at San Diego, La Jolla, CA
| | - Anil K Dasyam
- University of Pittsburgh Medical Center, Pittsburgh, PA
| | | | | |
Collapse
|
3
|
Li W, Wang J, Li Y, Yue Q, Cui M, Liu J. KRAS Mutations in Peripheral Blood (with or without CA19-9) for Differential Diagnosis of Pancreatic Cancer and Chronic Pancreatitis: a Systematic Review and Meta-analysis. Indian J Surg 2022. [DOI: 10.1007/s12262-022-03475-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
4
|
Zhang W, Wang L, Li D, Campbell DH, Walsh BJ, Packer NH, Dong Q, Wang E, Wang Y. Phenotypic profiling of pancreatic ductal adenocarcinoma plasma-derived small extracellular vesicles for cancer diagnosis and cancer stage prediction: a proof-of-concept study. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2022; 14:2255-2265. [PMID: 35612592 DOI: 10.1039/d2ay00536k] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Circulating pancreatic ductal adenocarcinoma (PDAC) derived small extracellular vesicles (sEVs) are nano-sized membranous vesicles secreted from PDAC cells and released into surrounding body fluids, such as blood. The use of plasma-derived sEVs for cancer diagnosis is particularly appealing in biomedical research because the sEVs reflect some key features (e.g. genetic and phenotypic status) related to the organs from which they originate. For example, the surface membrane proteins and their expression level on sEVs were reported to be related to the presence and progression of PDAC. However, difficulty in sEVs isolation and lack of ultrasensitive assays for simultaneous analysis of multiple protein biomarkers on patient plasma-derived sEVs hinder their application in the clinic. In our previous study, we have demonstrated the application of magnetic beads (MBs) and surface-enhanced Raman scattering (SERS) assay for phenotypic analysis of cancer cells-derived sEVs using different cell lines. To further demonstrate the clinical application of the proposed assay, we have profiled the sEVs' phenotypes (relative expression of biomarker Glypican 1, EpCAM and CD44V6) of healthy donors and PDAC patients to enable simultaneous detection of multiple surface membrane proteins on plasma-derived sEVs. We discovered that the PDAC sEVs' phenotype signatures had high accuracy for PDAC diagnosis (100%) and showed strong correlation with cancer stages, which were further validated by the imaging techniques (e.g. computerized tomography and magnetic resonance imaging) and also the correlation of cancer stages with CA19-9 (gold standard biomarker) and the sEVs' phenotype signatures. The present proof-of-concept study thus provides an initial investigation of using the proposed SERS assay for PDAC diagnosis and early cancer stage prediction in the clinic.
Collapse
Affiliation(s)
- Wei Zhang
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW 2109, Australia.
| | - Ling Wang
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, Jilin, P. R. China
| | - Dan Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China.
| | | | - Bradley J Walsh
- Minomic International Ltd, Macquarie Park, NSW 2113, Australia
| | - Nicolle H Packer
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW 2109, Australia.
| | - Qing Dong
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China.
| | - Erkang Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China.
| | - Yuling Wang
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW 2109, Australia.
| |
Collapse
|
5
|
Gonda TA, Farrell J, Wallace M, Khanna L, Janec E, Kwon R, Saunders M, Siddiqui UD, Brand R, Simeone DM. Standardization of EUS imaging and reporting in high-risk individuals of pancreatic adenocarcinoma: consensus statement of the Pancreatic Cancer Early Detection Consortium. Gastrointest Endosc 2022; 95:723-732.e7. [PMID: 34736932 DOI: 10.1016/j.gie.2021.10.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 10/25/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Pancreatic ductal adenocarcinoma is an aggressive disease most often diagnosed after local progression or metastatic dissemination, precluding resection and resulting in a high mortality rate. For individuals with elevated personal risk of the development of pancreatic cancer, EUS is a frequently used advanced imaging and diagnostic modality. However, variability in the expertise and definition of EUS findings exists among gastroenterologists, as well as a lack of standardized reporting of relevant findings at the time of examination. Adoption of standardized EUS reporting, using a universally accepted and agreed on terminology, is needed. METHODS A consensus statement designed to create a standardized reporting template was authored by a multidisciplinary group of experts in pancreatic diseases that includes gastroenterologists, radiologists, surgeons, oncologists, and geneticists. This statement was developed using a modified Delphi process as part of the Pancreatic Cancer Early Detection Consortium, and >75% agreement was required to reach consensus. RESULTS We identified reporting elements and present standardized reporting templates for EUS indications, procedural data, EUS image capture, and descriptors of findings, tissue sampling, and postprocedural assessment of adequacy. CONCLUSIONS Adoption of this standardized EUS reporting template should improve consistency in clinical decision-making for individuals with elevated risk of pancreatic cancer by providing complete and accurate reporting of pancreatic abnormalities. Standardization will also help to facilitate research and clinical trial design by using clearly defined and consistent imaging descriptions, thus allowing for comparison of results across different centers.
Collapse
Affiliation(s)
- Tamas A Gonda
- Division of Gastroenterology and Hepatology, New York University Langone Health, New York, New York, USA
| | - James Farrell
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Michael Wallace
- Department of Gastroenterology & Hepatology, Mayo Clinic, Jacksonville, Florida, USA
| | - Lauren Khanna
- Division of Gastroenterology and Hepatology, New York University Langone Health, New York, New York, USA
| | - Eileen Janec
- Division of Gastroenterology and Hepatology, New York University Langone Health, New York, New York, USA
| | - Richard Kwon
- Gastroenterology, Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael Saunders
- Gastroenterology, Internal Medicine, University of Washington, Seattle, Washington, USA
| | - Uzma D Siddiqui
- Gastroenterology, Internal Medicine, University of Chicago, Chicago, Illinois, USA
| | - Randall Brand
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Diane M Simeone
- Gastroenterology, Surgical Oncology, New York University Langone Health, New York, New York, USA
| |
Collapse
|
6
|
Santos WD, Fernandes FCGDM, Souza DLBD, Aiquoc KM, Souza AMGD, Barbosa IR. Pancreatic cancer incidence and mortality trends: a population-based study. Rev Salud Publica (Bogota) 2022. [DOI: 10.15446/rsap.v24n1.89397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Objectives To analyze trends in pancreatic cancer incidence and mortality in Latin American countries.
Methods An ecological study with incidence data from the International Agency for Research on Cancer and mortality data from the World Health Organization. The trend of incidence by Joinpoint regression, the variation of the annual average and the 95% confidence interval were analyzed.
Results There were increasing trends in incidence in Brazil, in males, aged 40-59 years, and reduction in Costa Rica. In females, there was stability in all age groups. The mortality rates increased in the elderly in Brazil (AAPC: 1.09%; 95% CI: 0.76; 1.42), Peru (AAPC: 1.76%; 95% CI: 0.36; 3.17) and El Salvador (AAPC: 2.88%; 95% CI: 0.38; 5.43), while in Mexico, there was a reduction. In females, this rate increased in Brazil (AAPC: 1.38%; 95% CI: 1.07; 1.69), Peru (AAPC: 2.25%; 95% CI: 0.68; 3.85), Chile (AAPC: 3.62%; 95% CI:1.96; 5.31), Nicaragua (AAPC: 2.51%; 95% CI: 0.36; 4.71) and Paraguay (AAPC: 1.17%; 95% CI: 0.37; 1.98) and a downward trend was observed in Colombia and Ecuador.
Conclusions Pancreatic cancer had a higher incidence in the elderly population of both sexes and an increase of the mortality trend in females was noted.
Collapse
|
7
|
Santos Apolonio J, Lima de Souza Gonçalves V, Cordeiro Santos ML, Silva Luz M, Silva Souza JV, Rocha Pinheiro SL, de Souza WR, Sande Loureiro M, de Melo FF. Oncolytic virus therapy in cancer: A current review. World J Virol 2021; 10:229-255. [PMID: 34631474 PMCID: PMC8474975 DOI: 10.5501/wjv.v10.i5.229] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/19/2021] [Accepted: 08/09/2021] [Indexed: 02/06/2023] Open
Abstract
In view of the advancement in the understanding about the most diverse types of cancer and consequently a relentless search for a cure and increased survival rates of cancer patients, finding a therapy that is able to combat the mechanism of aggression of this disease is extremely important. Thus, oncolytic viruses (OVs) have demonstrated great benefits in the treatment of cancer because it mediates antitumor effects in several ways. Viruses can be used to infect cancer cells, especially over normal cells, to present tumor-associated antigens, to activate "danger signals" that generate a less immune-tolerant tumor microenvironment, and to serve transduction vehicles for expression of inflammatory and immunomodulatory cytokines. The success of therapies using OVs was initially demonstrated by the use of the genetically modified herpes virus, talimogene laherparepvec, for the treatment of melanoma. At this time, several OVs are being studied as a potential treatment for cancer in clinical trials. However, it is necessary to be aware of the safety and possible adverse effects of this therapy; after all, an effective treatment for cancer should promote regression, attack the tumor, and in the meantime induce minimal systemic repercussions. In this manuscript, we will present a current review of the mechanism of action of OVs, main clinical uses, updates, and future perspectives on this treatment.
Collapse
Affiliation(s)
- Jonathan Santos Apolonio
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | - Maria Luísa Cordeiro Santos
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Marcel Silva Luz
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | - João Victor Silva Souza
- Universidade Estadual do Sudoeste da Bahia, Campus Vitória da Conquista, Vitória da Conquista 45083-900, Bahia, Brazil
| | - Samuel Luca Rocha Pinheiro
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Wedja Rafaela de Souza
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Matheus Sande Loureiro
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| |
Collapse
|
8
|
Wang X, Wang C, Zhang H. Improvement of Diagnostic Accuracy for Pancreatic Cancer with Serum Lactate Dehydrogenase. Cancer Manag Res 2021; 13:4879-4886. [PMID: 34188541 PMCID: PMC8232858 DOI: 10.2147/cmar.s312312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/29/2021] [Indexed: 12/15/2022] Open
Abstract
Purpose Due to the lack of early-stage detection, pancreatic cancer (PC) remains a devastating disease worldwide. Lactate dehydrogenase (LDH) is associated with tumorigenesis and cancer progression. This study aims to analyze the diagnostic improvements in serum LDH levels combined with other common tumor biomarkers, including carbohydrate antigen 19–9 (CA19–9) and carcinoembryonic antigen (CEA), for monitoring PC. Patients and Methods A retrospective analysis was performed on 73 patients with newly diagnosed PC, 90 patients with pancreatic benign diseases (PBD), and 92 people with healthy physical examination (HPE) at Zhongda Hospital, Southeast University from July 2013 to July 2020. The diagnostic efficiencies of serum levels of LDH, CA19–9, and CEA were analyzed through receiver operating characteristic (ROC) curves for PC. The sensitivity and specificity were evaluated at an optimal cutoff. The prognostic impacts of LDH on PC patients were also assessed. Results The LDH level was elevated in 21 (28.77%) patients with PC, 3 (3.33%) PBD patients, and no HPE individuals (P<0.05). The sensitivities of LDH, CA19–9, and CEA for the diagnosis of PC were 63.0%, 78.1%, and 72.6%, respectively, but the combination of these three markers increased predictive sensitivity significantly to 87.6%. The specificities of LDH, CA19–9, and CEA for the diagnosis of PC were 93.4%, 84.1%, and 73.1%, respectively. The combined specificity reached up to 96.7%. The medium survival time of PC patients with low-level LDH was 21 ± 5.1 months, whereas that of patients with high-level LDH was only 7 ± 0.92 months (P<0.05). Conclusion The serum LDH level was higher in PC patients than in PBD patients and HPE individuals and was associated with a poor prognosis. The combined assessment of LDH, CEA, and CA19–9 showed higher sensitivity and specificity for the diagnosis of PC.
Collapse
Affiliation(s)
- Xi Wang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| | - Chunbin Wang
- Department of Oncology, Yancheng Third People's Hospital, The Affiliated Yancheng Hospital of Southeast University Medical College, Yancheng, Jiangsu Province, People's Republic of China
| | - Haijun Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| |
Collapse
|
9
|
Abstract
Screening for pancreatic cancer (PC) in high-risk groups aimed to detect early cancers is currently done only in the research setting, and data on psychological outcomes of screening in these populations is scarce. To determine the psychological impact of a national Australian pancreatic screening program, a prospective study was conducted using validated psychological measures: impact of events scale (IES), psychological consequences questionnaire (PCQ) and the cancer worry scale. Measures were administered at baseline, 1-month and at 1-year post-enrolment and correlations with abnormal endoscopic ultrasound (EUS) results were calculated. Over a 6-year period, 102 participants were recruited to the screening program. Thirty-nine patients (38.2%) had an abnormal endoscopic ultrasound, and two patients (2.0%) were diagnosed with PC and two with other malignancies. Those with a personal history of cancer or a positive BRCA2 mutation demonstrated significantly increased worry about developing other types of cancer at baseline (p < 0.01). Irrespective of EUS result, there was a significant decrease of total IES score at 1 year (Z = - 2.0, p = 0.041). In patients with abnormal EUS results, there was a decrease in the total IES score at 1 year (Z = - 2.5, p = 0.011). In participants deemed to be most distressed at baseline based on their negative PCQ score, there was a significant decrease of the total PCQ (Z = - 3.2, p = 0.001), emotional (Z = - 3.0, p = 0.001), social (Z = 3.0, p = 0.001) and physical (Z = - 2.8, p = 0.002) subscale at 1-year post-intervention. This study provides evidence of the long-term psychological benefits of PC screening in high-risk patients. There was no negative impact of screening in the short-term and the positive benefits appeared at 1-year post-intervention irrespective of screening result.
Collapse
|
10
|
Prospective Study Using Plasma Apolipoprotein A2-Isoforms to Screen for High-Risk Status of Pancreatic Cancer. Cancers (Basel) 2020; 12:cancers12092625. [PMID: 32937962 PMCID: PMC7564617 DOI: 10.3390/cancers12092625] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Apolipoprotein A2 isoforms (apoA2-i) have been identified as minimally invasive biomarkers for detecting pancreatic cancer (PC) and high-risk individuals for PC. We investigated the efficiency of an enrichment strategy for high-risk individuals using a combination of blood testing for apoA2-i with imaging examinations in the general population. We enrolled 5120 subjects in experimental pancreatic cancer screening, with 84 subjects (1.3%) showing abnormal results for apoA2-i. Pancreatic diseases were recognized in about 30% of subjects with an apoA2-ATQ/AT level of ≤35 μg/mL. Among them, 1 pancreatic cancer and 15 high-risk individuals with intraductal papillary mucinous neoplasm were detected. ApoA2-i has the potential to enrich PC and high-risk status by increasing the diagnostic probability before imaging examinations. Abstract Apolipoprotein A2-ATQ/AT (apoA2-ATQ/AT) has been identified as a minimally invasive biomarker for detecting pancreatic cancer (PC) and high-risk (HR) individuals for PC. To establish an efficient enrichment strategy for HR, we carried out a plasma apoA2-ATQ/AT level-based prospective screening study among the general population. The subjects for the screening study were recruited at six medical check-up facilities in Japan between October 2015 and January 2017. We evaluated the positive predictive value (PPV) of the plasma apoA2-ATQ/AT level of ≤35 μg/mL for detecting PC and HR. Furthermore, we prospectively confirmed its diagnostic accuracy with another post-diagnosis population in a cross-sectional study. We enrolled 5120 subjects in experimental screening, with 84 subjects (1.3%) showing positive results for apoA2-ATQ/AT. Pancreatic abnormalities were recognized in 26 of the 84 subjects from imaging examinations. Pancreatic abnormalities detected included 1 PC and 15 HR abnormalities, such as cystic lesions including intraductal papillary mucinous neoplasm. The PPV of apoA2-ATQ/AT for detecting PC and HR was 33.3%. Moreover, a combination study with another cross-sectional study revealed that the area under the curve for apoA2-ATQ/AT to distinguish PC from healthy controls was 0.903. ApoA2-ATQ/AT has the potential to enrich PC and HR by increasing the diagnostic probability before imaging examinations.
Collapse
|
11
|
Rhee H, Park MS. The Role of Imaging in Current Treatment Strategies for Pancreatic Adenocarcinoma. Korean J Radiol 2020; 22:23-40. [PMID: 32901458 PMCID: PMC7772381 DOI: 10.3348/kjr.2019.0862] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 04/30/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023] Open
Abstract
In pancreatic cancer, imaging plays an essential role in surveillance, diagnosis, resectability evaluation, and treatment response evaluation. Pancreatic cancer surveillance in high-risk individuals has been attempted using endoscopic ultrasound (EUS) or magnetic resonance imaging (MRI). Imaging diagnosis and resectability evaluation are the most important factors influencing treatment decisions, where computed tomography (CT) is the preferred modality. EUS, MRI, and positron emission tomography play a complementary role to CT. Treatment response evaluation is of increasing clinical importance, especially in patients undergoing neoadjuvant therapy. This review aimed to comprehensively review the role of imaging in relation to the current treatment strategy for pancreatic cancer, including surveillance, diagnosis, evaluation of resectability and treatment response, and prediction of prognosis.
Collapse
Affiliation(s)
- Hyungjin Rhee
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Mi Suk Park
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
12
|
Zhu Y, Zhang H, Chen N, Hao J, Jin H, Ma X. Diagnostic value of various liquid biopsy methods for pancreatic cancer: A systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e18581. [PMID: 32011436 PMCID: PMC7220382 DOI: 10.1097/md.0000000000018581] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Liquid biopsy is a novel method for cancer diagnosis, which has been applied in lung and breast cancers, demonstrating high diagnostic value. However, clinical value of it in pancreatic cancer (PC) remains to be verified. The aim of this meta-analysis was to evaluate overall diagnostic value of various liquid biopsy methods (circulating tumor DNA, circulating tumor cells and exosomes) in detecting PC. METHODS We comprehensively searched relevant studies in PubMed, Medline, Embase, and Web of Science without time limitation according to PRISMA. Data necessary for reconstructing a 2 × 2 table was calculated from the original articles. The methodological quality of included studies was evaluated by QUADAS-2. Statistical analysis including was performed by the software Meta-Disc version 1.4, and STATA 14.2. RESULTS A total of 19 studies including 1872 individuals were included in this meta-analysis. In which, 7 were studies about ctDNA, 7 were on CTCs and 6 were about exosomes (Sefrioui D, studied diagnostic accuracy of both ctDNA and CTCs, with no common patients in these 2 groups). The pooled sensitivity estimates for ctDNA, CTCs and exosomes in detecting PC with their 95% confidential intervals (95% CI) were 0.64 (95%CI 0.58-0.70), 0.74 (95%CI 0.68-0.79) and 0.93 (95%CI 0.90-0.95), respectively. The pooled specificity estimates were 0.92(95%CI 0.88-0.95), 0.83 (95%CI 0.78-0.88) and 0.92 (95%CI 0.88-0.95), respectively. The area under curve (AUC) of the sROC for ctDNA, CTCs and exosomes in detecting PC were 0.9478, 0.8166, and 0.9819, respectively. The overall sensitivity, specificity and AUC of the sROC curve for overall liquid biopsy in detecting PC were 0.80 (95%CI 0.77-0.82), 0.89 (95%CI 0.87-0.91) and 0.9478, respectively. CONCLUSION This meta-analysis confirmed that liquid biopsy had high diagnostic value in detecting PC. In ctDNA, CTCs and exosomes these 3 subgroups, exosomes showed highest sensitivity and specificity.
Collapse
Affiliation(s)
- Yuzhou Zhu
- Department of Biotherapy, Cancer Center
- Department of Gastrointestinal Surgery
| | - Hao Zhang
- Department of Pancreatic Surgery, West China Hospital, Sichuan University
| | | | | | | | - Xuelei Ma
- Department of Biotherapy, Cancer Center
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
13
|
Lorenzo D, Rebours V, Maire F, Palazzo M, Gonzalez JM, Vullierme MP, Aubert A, Hammel P, Lévy P, Mestier LD. Role of endoscopic ultrasound in the screening and follow-up of high-risk individuals for familial pancreatic cancer. World J Gastroenterol 2019; 25:5082-5096. [PMID: 31558858 PMCID: PMC6747297 DOI: 10.3748/wjg.v25.i34.5082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/04/2019] [Accepted: 08/24/2019] [Indexed: 02/06/2023] Open
Abstract
Managing familial pancreatic cancer (FPC) is challenging for gastroenterologists, surgeons and oncologists. High-risk individuals (HRI) for pancreatic cancer (PC) (FPC or with germline mutations) are a heterogeneous group of subjects with a theoretical lifetime cumulative risk of PC over 5%. Screening is mainly based on annual magnetic resonance imaging (MRI) and endoscopic ultrasound (EUS). The goal of screening is to identify early-stage operable cancers or high-risk precancerous lesions (pancreatic intraepithelial neoplasia or intraductal papillary mucinous neoplasms with high-grade dysplasia). In the literature, target lesions are identified in 2%-5% of HRI who undergo screening. EUS appears to provide better identification of small solid lesions (0%-46% of HRI) and chronic-pancreatitis-like parenchymal changes (14%-77% of HRI), while MRI is probably the best modality to identify small cystic lesions (13%-49% of HRI). There are no specific studies in HRI on the use of contrast-enhanced harmonic EUS. EUS can also be used to obtain tissue samples. Nevertheless, there is still limited evidence on the accuracy of imaging procedures used for screening or agreement on which patients to treat. The cost-effectiveness of screening is also unclear. Certain new EUS-related techniques, such as searching for DNA abnormalities or protein markers in pancreatic fluid, appear to be promising.
Collapse
Affiliation(s)
- Diane Lorenzo
- Pancreatology Department, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, and Paris Diderot University, Paris 75013, France
| | - Vinciane Rebours
- Pancreatology Department, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, and Paris Diderot University, Paris 75013, France
- INSERM, UMR1149, Paris 92110, France
| | - Frédérique Maire
- Pancreatology Department, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, and Paris Diderot University, Paris 75013, France
| | - Maxime Palazzo
- Pancreatology Department, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, and Paris Diderot University, Paris 75013, France
| | - Jean-Michel Gonzalez
- Departement of Gastroenterology, Aix Marseille university - APHM - Hôpital Nord, Marseille 13000, France
| | - Marie-Pierre Vullierme
- Radiology Department, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, and Paris Diderot University, Paris 92110, France
| | - Alain Aubert
- Pancreatology Department, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, and Paris Diderot University, Paris 75013, France
| | - Pascal Hammel
- Oncology Department, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, and Paris Diderot University, Paris 92110, France
| | - Philippe Lévy
- Pancreatology Department, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, and Paris Diderot University, Paris 75013, France
| | - Louis de Mestier
- Pancreatology Department, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, and Paris Diderot University, Paris 75013, France
- INSERM, UMR1149, Paris 92110, France
| |
Collapse
|
14
|
Kobayashi T, Honda K. Trends in biomarker discoveries for the early detection and risk stratification of pancreatic cancer using omics studies. Expert Rev Mol Diagn 2019; 19:651-654. [PMID: 31298060 DOI: 10.1080/14737159.2019.1643718] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Takashi Kobayashi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine , Kobe , Hyogo , Japan
| | - Kazufumi Honda
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute , Tokyo , Japan
| |
Collapse
|
15
|
de Mestier L, Muller M, Cros J, Vullierme MP, Vernerey D, Maire F, Dokmak S, Rebours V, Sauvanet A, Lévy P, Hammel P. Appropriateness of pancreatic resection in high-risk individuals for familial pancreatic ductal adenocarcinoma: a patient-level meta-analysis and proposition of the Beaujon score. United European Gastroenterol J 2019; 7:358-368. [PMID: 31019704 DOI: 10.1177/2050640618824910] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 12/19/2018] [Indexed: 02/06/2023] Open
Abstract
Background About 5% of pancreatic ductal adenocarcinomas are inherited due to a deleterious germline mutation detected in 20% or fewer families. Pancreatic screening in high-risk individuals is proposed to allow early surgical treatment of (pre)malignant lesions. The outcomes of pancreatic surgery in high-risk individuals have never been correctly explored. Objectives To evaluate surgical appropriateness and search for associated factors in high-risk individuals. Methods A patient-level meta-analysis was performed including studies published since 1999. Individual classification distinguished the highest risk imaging abnormality into low-risk or high-risk abnormality, and the highest pathological degree of malignancy of lesions into no/low malignant potential or potentially/frankly malignant. Surgical appropriateness was considered when potentially/frankly malignant lesions were resected. Results Thirteen out of 24 studies were selected, which reported 90 high-risk individuals operated on. Low-risk/high-risk abnormalities were preoperatively detected in 46.7%/53.3% of operated high-risk individuals, respectively. Surgical appropriateness was consistent in 38 (42.2%) high-risk individuals, including 20 pancreatic ductal adenocarcinomas (22.2%). Identification of high-risk abnormalities was strongly associated with surgical appropriateness at multivariate analysis (P = 0.001). We proposed a score and nomogram predictive of surgical appropriateness, including high-risk abnormalities, age and existence of deleterious germline mutation. Conclusion Overall, 42.2% of high-risk individuals underwent appropriate surgery. The proposed score might help selecting the best candidates among high-risk individuals for pancreatic resection.
Collapse
Affiliation(s)
- Louis de Mestier
- Department of Gastroenterology and Pancreatology, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Marie Muller
- Department of Digestive Oncology and Genetic Counselling, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Jérôme Cros
- Department of Pathology, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Marie-Pierre Vullierme
- Department of Radiology, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Dewi Vernerey
- Department of Methodology and Quality of Life in Oncology Unit, EA 3181, Minjoz University Hospital, Besançon, France
| | - Frédérique Maire
- Department of Gastroenterology and Pancreatology, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Safi Dokmak
- Department of Hepatobiliary and Pancreatic Surgery, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Vinciane Rebours
- Department of Gastroenterology and Pancreatology, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Alain Sauvanet
- Department of Hepatobiliary and Pancreatic Surgery, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Philippe Lévy
- Department of Gastroenterology and Pancreatology, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Pascal Hammel
- Department of Digestive Oncology and Genetic Counselling, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| |
Collapse
|
16
|
Patel K, Siraj S, Smith C, Nair M, Vishwanatha JK, Basha R. Pancreatic Cancer: An Emphasis on Current Perspectives in Immunotherapy. Crit Rev Oncog 2019; 24:105-118. [PMID: 31679206 PMCID: PMC8038975 DOI: 10.1615/critrevoncog.2019031417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pancreatic cancer affects both male and female individuals with higher incidences and death rates among the male population. Detection of this malignancy is delayed due to the lack of symptoms in the early-stage cancer, which makes it extremely difficult to treat. Identifying effective strategies has been a challenge for improving the survival rates in pancreatic cancer patients. Resistance to chemotherapy is often developed in pancreatic cancer treatment. Although many strategies are under clinical trials to target certain markers associated with cancer, immunotherapeutic approaches are currently gaining importance. Immunotherapy for pancreatic cancer is in the limelight after preclinical research showed some promise. Immunotherapy approaches were tested along with other treatment options to enhance the treatment effect. Adoptive cell transfer and immune checkpoint inhibitors are currently in clinical trials. The Food and Drug Administration approved pembrolizumab in a fast-tracked review for advanced pancreatic cancer patients. Pembrolizumab blocks the checkpoint protein, programmed cell death protein 1 (PD-1), on T cells to boost the response of the immune system against cancer cells, thereby shrinking tumors. The recent developments in immunotherapy and the early success in other cancers are encouraging to further test immunotherapy in pancreatic cancer. The combination of pembrolizumab and pelareorep, an isolate of human reovirus, is in phase II clinical study in metastatic disease. Depending on the results of current clinical trials and testing, the strategies in the pipeline are expected to increase the use of immunotherapy in the clinical testing setting. Success in immunotherapy is urgently needed to address the side-effects, treating patients with advanced disease and reducing metastasis for increasing the survival rate in pancreatic cancer patients.
Collapse
Affiliation(s)
| | | | - Chloe Smith
- Old Dominion University, Norfolk, Virginia 23529
| | - Maya Nair
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, Texas 76107
| | - Jamboor K. Vishwanatha
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, Texas 76107
| | | |
Collapse
|
17
|
Sheel ARG, Harrison S, Sarantitis I, Nicholson JA, Hanna T, Grocock C, Raraty M, Ramesh J, Farooq A, Costello E, Jackson R, Chapman M, Smith A, Carter R, Mckay C, Hamady Z, Aithal GP, Mountford R, Ghaneh P, Hammel P, Lerch MM, Halloran C, Pereira SP, Greenhalf W. Identification of Cystic Lesions by Secondary Screening of Familial Pancreatic Cancer (FPC) Kindreds Is Not Associated with the Stratified Risk of Cancer. Am J Gastroenterol 2019; 114:155-164. [PMID: 30353057 DOI: 10.1038/s41395-018-0395-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Intraductal papillary mucinous neoplasms (IPMNs) are associated with risk of pancreatic ductal adenocarcinoma (PDAC). It is unclear if an IPMN in individuals at high risk of PDAC should be considered as a positive screening result or as an incidental finding. Stratified familial pancreatic cancer (FPC) populations were used to determine if IPMN risk is linked to familial risk of PDAC. METHODS This is a cohort study of 321 individuals from 258 kindreds suspected of being FPC and undergoing secondary screening for PDAC through the European Registry of Hereditary Pancreatitis and Familial Pancreatic Cancer (EUROPAC). Computerised tomography, endoscopic ultrasound of the pancreas and magnetic resonance imaging were used. The risk of being a carrier of a dominant mutation predisposing to pancreatic cancer was stratified into three even categories (low, medium and high) based on: Mendelian probability, the number of PDAC cases and the number of people at risk in a kindred. RESULTS There was a median (interquartile range (IQR)) follow-up of 2 (0-5) years and a median (IQR) number of investigations per participant of 4 (2-6). One PDAC, two low-grade neuroendocrine tumours and 41 cystic lesions were identified, including 23 IPMN (22 branch-duct (BD)). The PDAC case occurred in the top 10% of risk, and the BD-IPMN cases were evenly distributed amongst risk categories: low (6/107), medium (10/107) and high (6/107) (P = 0.63). CONCLUSIONS The risk of finding BD-IPMN was independent of genetic predisposition and so they should be managed according to guidelines for incidental finding of IPMN.
Collapse
Affiliation(s)
- A R G Sheel
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| | - S Harrison
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| | - I Sarantitis
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| | - J A Nicholson
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| | - T Hanna
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| | - C Grocock
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| | - M Raraty
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| | - J Ramesh
- Department of Gastroenterology, The Royal Liverpool University Hospital, London, UK
| | - A Farooq
- Department of Radiology, The Royal Liverpool University Hospital, London, UK
| | - E Costello
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| | - R Jackson
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| | - M Chapman
- Institute for Liver & Digestive Health, University College London, London, UK
| | - A Smith
- Department of Pancreatico-Biliary Surgery, Leeds Teaching Hospital Trust, Leeds, UK
| | - R Carter
- West of Scotland Pancreatic unit, Glasgow Royal Infirmary, Glasgow, UK
| | - C Mckay
- West of Scotland Pancreatic unit, Glasgow Royal Infirmary, Glasgow, UK
| | - Z Hamady
- Department of Hepatobiliary and Pancreatic Diseases, University Hospital Southampton, Southampton, UK
| | - G P Aithal
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, NG7 2UH, UK
| | - R Mountford
- Mersey Regional Molecular Genetics Laboratory, Liverpool Women's Hospital, Liverpool, UK
| | - P Ghaneh
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| | - P Hammel
- Service de Gastroentérologie-Pancréatologie, Pôle des Maladies de l'Appareil Digestif, Hôpital Beaujon, 92118, Clichy Cedex, France
| | - M M Lerch
- Department of Medicine A, University Medicine Greifswald, Sauerbruch-Strasse, 17475, Greifswald, Germany
| | - C Halloran
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| | - S P Pereira
- Institute for Liver & Digestive Health, University College London, London, UK
| | - W Greenhalf
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| |
Collapse
|
18
|
Benzel J, Fendrich V. Familial Pancreatic Cancer. Oncol Res Treat 2018; 41:611-618. [PMID: 30269130 DOI: 10.1159/000493473] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/04/2018] [Indexed: 12/13/2022]
Abstract
Familial pancreatic cancer accounts for 10% of all patients with pancreatic cancer. Because the 5-year survival rate of pancreatic cancer is only 7%, screening programs for high-risk individuals are essential and might be advantageous. Pancreatic ductal adenocarcinoma mostly shows symptoms at an advanced state and treatment is not efficient enough to cure most patients. People with hereditary tumor syndromes or their affected relatives can also be included in such screening programs. Besides the collection of data to investigate the background of the disease, these screening programs aim to diagnose and treat precursor lesions so that more dangerous, invasive lesions are prevented. These precursor lesions can be pancreatic intraepithelial neoplasia, intraductal papillary mucinous neoplasm, and mucinous cystic neoplasm. This review summarizes the latest knowledge of pancreatic screening programs, shows the procedure of pancreatic cancer screening, and gives an overview of current guidelines.
Collapse
|
19
|
Rong D, Mao Y, Hu W, Xu S, Wang J, He H, Li S, Zhang R. Intravoxel incoherent motion magnetic resonance imaging for differentiating metastatic and non-metastatic lymph nodes in pancreatic ductal adenocarcinoma. Eur Radiol 2018; 28:2781-2789. [PMID: 29404768 DOI: 10.1007/s00330-017-5259-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/14/2017] [Accepted: 12/20/2017] [Indexed: 01/27/2023]
Abstract
OBJECTIVES To evaluate the diagnostic potential of intravoxel incoherent motion (IVIM) DWI for differentiating metastatic and non-metastatic lymph node stations (LNS) in pancreatic ductal adenocarcinoma (PDAC). METHODS 59 LNS histologically diagnosed following surgical resection from 15 patients were included. IVIM DWI with 12 b values was added to the standard MRI protocol. Evaluation of parameters was performed pre-operatively and included the apparent diffusion coefficient (ADC), pure diffusion coefficient (D), pseudo-diffusion coefficient (D*) and perfusion fraction (f). Diagnostic performance of ADC, D, D* and f for differentiating between metastatic and non-metastatic LNS was evaluated using ROC analysis. RESULTS Metastatic LNS had significantly lower D, D*, f and ADC values than the non-metastatic LNS (p< 0.01). The best diagnostic performance was found in D, with an area under the ROC curve of 0.979, while the area under the ROC curve values of D*, f and ADC were 0.867, 0.855 and 0.940, respectively. The optimal cut-off values for distinguishing metastatic and non-metastatic lymph nodes were D = 1.180 × 10-3 mm2/s; D* = 14.750 × 10-3 mm2/s, f = 20.65 %, and ADC = 1.390 × 10-3 mm2/s. CONCLUSION IVIM DWI is useful for differentiating between metastatic and non-metastatic LNS in PDAC. KEY POINTS • IVIM DWI is feasible for diagnosing LN metastasis in PDAC. • Metastatic LNS has lower D, D*, f, ADC values than non-metastatic LNS. • D-value from IVIM model has best diagnostic performance, followed by ADC value. • D* has the lowest AUC value.
Collapse
Affiliation(s)
- Dailin Rong
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China
- Department of Radiology, Sun Yat-sen University Cancer Center, No. 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Yize Mao
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China
- Department of Hepato-Biliary-Pancreatic Oncology, Sun Yat-Sen University Cancer Center, No. 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Wanming Hu
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Center, No. 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Shuhang Xu
- Department of Ultrasound, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong Province, People's Republic of China
| | - Jun Wang
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China
- Department of Hepato-Biliary-Pancreatic Oncology, Sun Yat-Sen University Cancer Center, No. 651 Dongfeng Road East, Guangzhou, 510060, China
- Department of Ultrasound, Sun Yat-sen University Cancer Center, No. 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Haoqiang He
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China
- Department of Radiology, Sun Yat-sen University Cancer Center, No. 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Shengping Li
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China.
- Department of Hepato-Biliary-Pancreatic Oncology, Sun Yat-Sen University Cancer Center, No. 651 Dongfeng Road East, Guangzhou, 510060, China.
| | - Rong Zhang
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China.
- Department of Radiology, Sun Yat-sen University Cancer Center, No. 651 Dongfeng Road East, Guangzhou, 510060, China.
| |
Collapse
|
20
|
Zhang L, Sanagapalli S, Stoita A. Challenges in diagnosis of pancreatic cancer. World J Gastroenterol 2018; 24:2047-2060. [PMID: 29785074 PMCID: PMC5960811 DOI: 10.3748/wjg.v24.i19.2047] [Citation(s) in RCA: 330] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/28/2018] [Accepted: 05/11/2018] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is a growing source of cancer related death, yet has poor survival rates which have not improved in the last few decades. Its high mortality rate is attributed to pancreatic cancer biology, difficulty in early diagnosis and the lack of standardised international guidelines in assessing suspicious pancreatic masses. This review aims to provide an update in the current state of play in pancreatic cancer diagnosis and to evaluate the benefits and limitations of available diagnostic technology. The main modalities discussed are imaging with computed tomography, magnetic resonance imaging, endoscopic ultrasound and positron emission tomography and tissue acquisition with fine needle aspiration. We also review the improvements in the techniques used for tissue acquisition and the opportunity for personalised cancer medicine. Screening of high risk individuals, promising biomarkers and common mimickers of pancreatic cancer are also explored, as well as suggestions for future research directions to allow for earlier detection of pancreatic cancer. Timely and accurate diagnosis of pancreatic cancer can lead to improvements in the current poor outcome of this disease.
Collapse
Affiliation(s)
- Lulu Zhang
- Department of Gastroenterology, St. Vincent’s Hospital Sydney, Darlinghurst 2010, NSW, Australia
| | - Santosh Sanagapalli
- Department of Gastroenterology, St. Vincent’s Hospital Sydney, Darlinghurst 2010, NSW, Australia
| | - Alina Stoita
- Department of Gastroenterology, St. Vincent’s Hospital Sydney, Darlinghurst 2010, NSW, Australia
| |
Collapse
|
21
|
Signoretti M, Bruno MJ, Zerboni G, Poley JW, Delle Fave G, Capurso G. Results of surveillance in individuals at high-risk of pancreatic cancer: A systematic review and meta-analysis. United European Gastroenterol J 2018; 6:489-499. [PMID: 29881603 DOI: 10.1177/2050640617752182] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 12/03/2017] [Indexed: 12/20/2022] Open
Abstract
Background Data on surveillance for pancreatic ductal adenocarcinoma (PDAC) in high-risk individuals (HRIs) with "familial pancreatic cancer" (FPC) and specific syndromes are limited and heterogeneous. Objective We conducted a systematic review and meta-analysis of PDAC surveillance studies in HRIs. Methods Prevalence of solid/cystic pancreatic lesions and of lesions considered a successful target of surveillance (proven resectable PDAC and high-grade precursors) was pooled across studies. The rate of lesions diagnosed by endoscopic ultrasonography (EUS)/magnetic resonance imaging (MRI) and across different HRI groups was calculated. Results Sixteen studies incorporating 1588 HRIs were included. The pooled prevalence of pancreatic solid and cystic lesions was 5.8% and 20.2%, respectively. The pooled prevalence of patients with lesions considered a successful target of surveillance was 3.3%, being similar to EUS or MRI and varying across subgroups, being 3% in FPC, 4% in hereditary pancreatitis, 5% in familial melanoma, 6.3% in hereditary breast/ovarian cancer, and 12.2% in Peutz-Jeghers syndrome. The pooled estimated rate of lesions considered a successful target of surveillance during follow-up was 5/1000 person-years. Conclusion Surveillance programs identify successful target lesions in 3.3% of HRIs with a similar yield of EUS and MRI and an annual risk of 0.5%. A higher rate of target lesions was reported in HRIs with specific DNA mutations.
Collapse
Affiliation(s)
| | - Marco J Bruno
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, University Medical Center, Rotterdam, The Netherlands
| | - Giulia Zerboni
- Digestive and Liver Disease Unit, S. Andrea Hospital, Rome, Italy
| | - Jan-Werner Poley
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, University Medical Center, Rotterdam, The Netherlands
| | | | - Gabriele Capurso
- Digestive and Liver Disease Unit, S. Andrea Hospital, Rome, Italy
| |
Collapse
|
22
|
Clinical value of ctDNA in upper-GI cancers: A systematic review and meta-analysis. Biochim Biophys Acta Rev Cancer 2017; 1868:394-403. [PMID: 28801248 DOI: 10.1016/j.bbcan.2017.08.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/05/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND The recent expanding technical possibilities to detect tumor derived mutations in blood, so-called circulating tumor DNA (ctDNA), has rapidly increased the interest in liquid biopsies. This review and meta-analysis explores the clinical value of ctDNA in malignancies of the upper gastro-intestinal tract. METHODS PubMed, Cochrane and Embase databases were searched to identify studies reporting the diagnostic, prognostic or predictive value of ctDNA in patients with esophageal, gastric and pancreatic cancer, until January 2017. The diagnostic accuracy and, using random-effect pair-wise meta-analyses, the prognostic value of ctDNA was assessed. RESULTS A total of 34 studies met the inclusion criteria. For esophageal and gastric cancer, amplification of oncogenes in blood, such as HER2 and MYC, can be relevant for diagnostic purposes, and to predict treatment response in certain patient subpopulations. Given the limited number of studies assessing the role of ctDNA in esophageal and gastric cancer, the meta-analysis estimated the diagnostic accuracy and predictive value of ctDNA in pancreatic cancer only (n=10). The pooled sensitivity and specificity of ctDNA as a diagnostic tool in pancreatic cancer were 28% and 95%, respectively. Patients with pancreatic cancer and detectable ctDNA demonstrated a worse overall survival compared to patients with undetectable ctDNA (HR 1.92, 95% confidence interval (CI) 1.15-3.22, p=0.01). CONCLUSION The presence of ctDNA is significantly associated with a poor prognosis in patients with pancreatic cancer. The use of ctDNA in clinical practice is promising, although standardization of sequencing techniques and further development of high-sensitive detection methods is needed.
Collapse
|
23
|
Moravec R, Divi R, Verma M. Detecting circulating tumor material and digital pathology imaging during pancreatic cancer progression. World J Gastrointest Oncol 2017; 9:235-250. [PMID: 28656074 PMCID: PMC5472554 DOI: 10.4251/wjgo.v9.i6.235] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/04/2017] [Accepted: 03/24/2017] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer (PC) is a leading cause of cancer-related death worldwide. Clinical symptoms typically present late when treatment options are limited and survival expectancy is very short. Metastatic mutations are heterogeneous and can accumulate up to twenty years before PC diagnosis. Given such genetic diversity, detecting and managing the complex states of disease progression may be limited to imaging modalities and markers present in circulation. Recent developments in digital pathology imaging show potential for early PC detection, making a differential diagnosis, and predicting treatment sensitivity leading to long-term survival in advanced stage patients. Despite large research efforts, the only serum marker currently approved for clinical use is CA 19-9. Utility of CA 19-9 has been shown to improve when it is used in combination with PC-specific markers. Efforts are being made to develop early-screening assays that can detect tumor-derived material, present in circulation, before metastasis takes a significant course. Detection of markers that identify circulating tumor cells and tumor-derived extracellular vesicles (EVs) in biofluid samples offers a promising non-invasive method for this purpose. Circulating tumor cells exhibit varying expression of epithelial and mesenchymal markers depending on the state of tumor differentiation. This offers a possibility for monitoring disease progression using minimally invasive procedures. EVs also offer the benefit of detecting molecular cargo of tumor origin and add the potential to detect circulating vesicle markers from tumors that lack invasive properties. This review integrates recent genetic insights of PC progression with developments in digital pathology and early detection of tumor-derived circulating material.
Collapse
|
24
|
Affiliation(s)
- MÔNICA SOLDAN
- Federal University of Rio de Janeiro, Brazil; São Vicente de Paulo Hospital, Brazil
| |
Collapse
|
25
|
Abstract
Pancreatic cancer (PC) remains a deadly disease and early detection through screening is likely to be our best hope to improve survival. Considering the low incidence of PC, population-based screening is not feasible, but is advisable for high-risk patients. Screening individuals at high risk for developing PC leads to the detection of premalignant lesions. High-grade pancreatic intraepithelial neoplasia and intraductal papillary mucinous neoplasm are the targets for early detection of PC. Endoscopic ultrasound (EUS) and magnetic resonance imaging are considered the most accurate techniques for pancreatic imaging; in particular EUS has emerged as a promising imaging test given its potential for tissue sampling to obtain diagnosis and to provide material for molecular profiling of PC. At the moment, screening should be performed within research protocols at experienced centers with a specific clinical and research interest, where a multidisciplinary team of specialists is available.
Collapse
Affiliation(s)
- Maria Chiara Petrone
- a Pancreato-Biliary Endoscopy and Endosonography Division, San Raffaele Scientific Institute , Vita Salute San Raffaele University , Milan , Italy
| | - Paolo Giorgio Arcidiacono
- a Pancreato-Biliary Endoscopy and Endosonography Division, San Raffaele Scientific Institute , Vita Salute San Raffaele University , Milan , Italy
| |
Collapse
|
26
|
Herreros-Villanueva M, Bujanda L. Non-invasive biomarkers in pancreatic cancer diagnosis: what we need versus what we have. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:134. [PMID: 27162784 DOI: 10.21037/atm.2016.03.44] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pancreatic cancer (PC) is probably the most lethal tumor being forecast as the second most fatal cancer by 2020 in developed countries. Only the earliest forms of the disease are a curable disease but it has to be diagnosed before symptoms starts. Detection at curable phase demands screening intervention for early detection and differential diagnosis. Unfortunately, no successful strategy or image technique has been concluded as effective approach and currently non-invasive biomarkers are the hope. Multiple translational research studies have explored minimally or non-invasive biomarkers in biofluids-blood, urine, stool, saliva or pancreatic juice, but diagnostic performance has not been validated yet. Nowadays no biomarker, alone or in combination, has been superior to carbohydrate antigen 19-9 (CA19-9) in sensitivity and specificity. Although the number of novel biomarkers for early diagnosis of PC has been increasing during the last couple of years, no molecular signature is ready to be implemented in clinical routine. Under the uncertain future, miRNAs profiling and methylation status seem to be the most promising biomarkers. However, good results in larger validations are urgently needed before application. Industry efforts through biotech and pharmaceutical companies are urgently required to demonstrate accuracy and validate promising results from basic and translational results.
Collapse
Affiliation(s)
- Marta Herreros-Villanueva
- Department of Gastroenterology, Hospital Donostia/Biodonostia Institute, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco UPV/EHU, San Sebastian, Spain
| | - Luis Bujanda
- Department of Gastroenterology, Hospital Donostia/Biodonostia Institute, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco UPV/EHU, San Sebastian, Spain
| |
Collapse
|
27
|
Riva F, Dronov OI, Khomenko DI, Huguet F, Louvet C, Mariani P, Stern MH, Lantz O, Proudhon C, Pierga JY, Bidard FC. Clinical applications of circulating tumor DNA and circulating tumor cells in pancreatic cancer. Mol Oncol 2016; 10:481-93. [PMID: 26856794 DOI: 10.1016/j.molonc.2016.01.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/12/2016] [Accepted: 01/18/2016] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most frequent pancreatic cancer type and is characterized by a dismal prognosis due to late diagnosis, local tumor invasion, frequent distant metastases and poor sensitivity to current therapy. In this context, circulating tumor cells and circulating tumor DNA constitute easily accessible blood-borne tumor biomarkers that may prove their clinical interest for screening, early diagnosis and metastatic risk assessment of PDAC. Moreover these markers represent a tool to assess PDAC mutational landscape. In this review, together with key biological findings, we summarize the clinical results obtained using "liquid biopsies" at the different stages of the disease, for early and metastatic diagnosis as well as monitoring during therapy.
Collapse
Affiliation(s)
- Francesca Riva
- Institut Curie, PSL Research University, SiRIC, Laboratory of Circulating Tumor Biomarkers, Paris, France; San Gerardo Hospital, Department of Medical Oncology, Monza, Italy
| | - Oleksii I Dronov
- Bogomolets National Medical University, Department of General Surgery No. 1, Kiev, Ukraine
| | - Dmytro I Khomenko
- Bogomolets National Medical University, Department of General Surgery No. 1, Kiev, Ukraine
| | - Florence Huguet
- Hopital Tenon, Pierre and Marie Curie Paris VI University, Department of Radiation Oncology, Paris, France
| | - Christophe Louvet
- Institut Mutualiste Montsouris, Department of Medical Oncology, Paris, France
| | - Pascale Mariani
- Institut Curie, PSL Research University, Department of Surgery, Paris, France
| | - Marc-Henri Stern
- Institut Curie, PSL Research University, INSERM U830, Paris, France
| | - Olivier Lantz
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Charlotte Proudhon
- Institut Curie, PSL Research University, SiRIC, Laboratory of Circulating Tumor Biomarkers, Paris, France
| | - Jean-Yves Pierga
- Institut Curie, PSL Research University, SiRIC, Laboratory of Circulating Tumor Biomarkers, Paris, France; Institut Curie, PSL Research University, Department of Medical Oncology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Francois-Clement Bidard
- Institut Curie, PSL Research University, SiRIC, Laboratory of Circulating Tumor Biomarkers, Paris, France; Institut Curie, PSL Research University, Department of Medical Oncology, Paris, France.
| |
Collapse
|