1
|
Mishra AK, Chong B, Arunachalam SP, Oberg AL, Majumder S. Machine Learning Models for Pancreatic Cancer Risk Prediction Using Electronic Health Record Data-A Systematic Review and Assessment. Am J Gastroenterol 2024; 119:1466-1482. [PMID: 38752654 PMCID: PMC11296923 DOI: 10.14309/ajg.0000000000002870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/06/2024] [Indexed: 06/20/2024]
Abstract
INTRODUCTION Accurate risk prediction can facilitate screening and early detection of pancreatic cancer (PC). We conducted a systematic review to critically evaluate effectiveness of machine learning (ML) and artificial intelligence (AI) techniques applied to electronic health records (EHR) for PC risk prediction. METHODS Ovid MEDLINE(R), Ovid EMBASE, Ovid Cochrane Central Register of Controlled Trials, Ovid Cochrane Database of Systematic Reviews, Scopus, and Web of Science were searched for articles that utilized ML/AI techniques to predict PC, published between January 1, 2012, and February 1, 2024. Study selection and data extraction were conducted by 2 independent reviewers. Critical appraisal and data extraction were performed using the CHecklist for critical Appraisal and data extraction for systematic Reviews of prediction Modelling Studies checklist. Risk of bias and applicability were examined using prediction model risk of bias assessment tool. RESULTS Thirty studies including 169,149 PC cases were identified. Logistic regression was the most frequent modeling method. Twenty studies utilized a curated set of known PC risk predictors or those identified by clinical experts. ML model discrimination performance (C-index) ranged from 0.57 to 1.0. Missing data were underreported, and most studies did not implement explainable-AI techniques or report exclusion time intervals. DISCUSSION AI/ML models for PC risk prediction using known risk factors perform reasonably well and may have near-term applications in identifying cohorts for targeted PC screening if validated in real-world data sets. The combined use of structured and unstructured EHR data using emerging AI models while incorporating explainable-AI techniques has the potential to identify novel PC risk factors, and this approach merits further study.
Collapse
Affiliation(s)
- Anup Kumar Mishra
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Bradford Chong
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | - Ann L. Oberg
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Shounak Majumder
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
2
|
Xie F, Chang J, Luong T, Wu B, Lustigova E, Shrader E, Chen W. Identifying Symptoms Prior to Pancreatic Ductal Adenocarcinoma Diagnosis in Real-World Care Settings: Natural Language Processing Approach. JMIR AI 2024; 3:e51240. [PMID: 38875566 PMCID: PMC11041417 DOI: 10.2196/51240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/08/2023] [Accepted: 12/16/2023] [Indexed: 06/16/2024]
Abstract
BACKGROUND Pancreatic cancer is the third leading cause of cancer deaths in the United States. Pancreatic ductal adenocarcinoma (PDAC) is the most common form of pancreatic cancer, accounting for up to 90% of all cases. Patient-reported symptoms are often the triggers of cancer diagnosis and therefore, understanding the PDAC-associated symptoms and the timing of symptom onset could facilitate early detection of PDAC. OBJECTIVE This paper aims to develop a natural language processing (NLP) algorithm to capture symptoms associated with PDAC from clinical notes within a large integrated health care system. METHODS We used unstructured data within 2 years prior to PDAC diagnosis between 2010 and 2019 and among matched patients without PDAC to identify 17 PDAC-related symptoms. Related terms and phrases were first compiled from publicly available resources and then recursively reviewed and enriched with input from clinicians and chart review. A computerized NLP algorithm was iteratively developed and fine-trained via multiple rounds of chart review followed by adjudication. Finally, the developed algorithm was applied to the validation data set to assess performance and to the study implementation notes. RESULTS A total of 408,147 and 709,789 notes were retrieved from 2611 patients with PDAC and 10,085 matched patients without PDAC, respectively. In descending order, the symptom distribution of the study implementation notes ranged from 4.98% for abdominal or epigastric pain to 0.05% for upper extremity deep vein thrombosis in the PDAC group, and from 1.75% for back pain to 0.01% for pale stool in the non-PDAC group. Validation of the NLP algorithm against adjudicated chart review results of 1000 notes showed that precision ranged from 98.9% (jaundice) to 84% (upper extremity deep vein thrombosis), recall ranged from 98.1% (weight loss) to 82.8% (epigastric bloating), and F1-scores ranged from 0.97 (jaundice) to 0.86 (depression). CONCLUSIONS The developed and validated NLP algorithm could be used for the early detection of PDAC.
Collapse
Affiliation(s)
- Fagen Xie
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States
| | - Jenny Chang
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States
| | - Tiffany Luong
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States
| | - Bechien Wu
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States
| | - Eva Lustigova
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States
| | - Eva Shrader
- Pancreatic Cancer Action Network, Manhattan Beach, CA, United States
| | - Wansu Chen
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States
| |
Collapse
|
3
|
Ke TM, Lophatananon A, Muir KR. An Integrative Pancreatic Cancer Risk Prediction Model in the UK Biobank. Biomedicines 2023; 11:3206. [PMID: 38137427 PMCID: PMC10740416 DOI: 10.3390/biomedicines11123206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/20/2023] [Accepted: 11/26/2023] [Indexed: 12/24/2023] Open
Abstract
Pancreatic cancer (PaCa) is a lethal cancer with an increasing incidence, highlighting the need for early prevention strategies. There is a lack of a comprehensive PaCa predictive model derived from large prospective cohorts. Therefore, we have developed an integrated PaCa risk prediction model for PaCa using data from the UK Biobank, incorporating lifestyle-related, genetic-related, and medical history-related variables for application in healthcare settings. We used a machine learning-based random forest approach and a traditional multivariable logistic regression method to develop a PaCa predictive model for different purposes. Additionally, we employed dynamic nomograms to visualize the probability of PaCa risk in the prediction model. The top five influential features in the random forest model were age, PRS, pancreatitis, DM, and smoking. The significant risk variables in the logistic regression model included male gender (OR = 1.17), age (OR = 1.10), non-O blood type (OR = 1.29), higher polygenic score (PRS) (Q5 vs. Q1, OR = 2.03), smoking (OR = 1.82), alcohol consumption (OR = 1.27), pancreatitis (OR = 3.99), diabetes (DM) (OR = 2.57), and gallbladder-related disease (OR = 2.07). The area under the receiver operating curve (AUC) of the logistic regression model is 0.78. Internal validation and calibration performed well in both models. Our integrative PaCa risk prediction model with the PRS effectively stratifies individuals at future risk of PaCa, aiding targeted prevention efforts and supporting community-based cancer prevention initiatives.
Collapse
Affiliation(s)
| | | | - Kenneth R. Muir
- Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (T.-M.K.); (A.L.)
| |
Collapse
|
4
|
Hjaltelin JX, Novitski SI, Jørgensen IF, Siggaard T, Vulpius SA, Westergaard D, Johansen JS, Chen IM, Juhl Jensen L, Brunak S. Pancreatic cancer symptom trajectories from Danish registry data and free text in electronic health records. eLife 2023; 12:e84919. [PMID: 37988407 PMCID: PMC10662947 DOI: 10.7554/elife.84919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 10/19/2023] [Indexed: 11/23/2023] Open
Abstract
Pancreatic cancer is one of the deadliest cancer types with poor treatment options. Better detection of early symptoms and relevant disease correlations could improve pancreatic cancer prognosis. In this retrospective study, we used symptom and disease codes (ICD-10) from the Danish National Patient Registry (NPR) encompassing 6.9 million patients from 1994 to 2018,, of whom 23,592 were diagnosed with pancreatic cancer. The Danish cancer registry included 18,523 of these patients. To complement and compare the registry diagnosis codes with deeper clinical data, we used a text mining approach to extract symptoms from free text clinical notes in electronic health records (3078 pancreatic cancer patients and 30,780 controls). We used both data sources to generate and compare symptom disease trajectories to uncover temporal patterns of symptoms prior to pancreatic cancer diagnosis for the same patients. We show that the text mining of the clinical notes was able to complement the registry-based symptoms by capturing more symptoms prior to pancreatic cancer diagnosis. For example, 'Blood pressure reading without diagnosis', 'Abnormalities of heartbeat', and 'Intestinal obstruction' were not found for the registry-based analysis. Chaining symptoms together in trajectories identified two groups of patients with lower median survival (<90 days) following the trajectories 'Cough→Jaundice→Intestinal obstruction' and 'Pain→Jaundice→Abnormal results of function studies'. These results provide a comprehensive comparison of the two types of pancreatic cancer symptom trajectories, which in combination can leverage the full potential of the health data and ultimately provide a fuller picture for detection of early risk factors for pancreatic cancer.
Collapse
Affiliation(s)
- Jessica Xin Hjaltelin
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Sif Ingibergsdóttir Novitski
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Isabella Friis Jørgensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Troels Siggaard
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Siri Amalie Vulpius
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - David Westergaard
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | | | - Inna M Chen
- Department of Oncology, Copenhagen University Hospital - Herlev and GentofteHerlevDenmark
| | - Lars Juhl Jensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
- Copenhagen University Hospital, Rigshospitalet, BlegdamsvejCopenhagenDenmark
| |
Collapse
|
5
|
Dite GS, Spaeth E, Wong CK, Murphy NM, Allman R. Predicting 10-Year Risk of Pancreatic Cancer Using a Combined Genetic and Clinical Model. GASTRO HEP ADVANCES 2023; 2:979-989. [PMID: 39130772 PMCID: PMC11308393 DOI: 10.1016/j.gastha.2023.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/12/2023] [Indexed: 08/13/2024]
Abstract
Background and Aims Pancreatic cancer has the poorest 5-year survival rate of any major solid tumor, but when diagnosed at an early stage, survival rates improve. Population screening is impractical because pancreatic cancer is rare with a lifetime risk of 1.7%, but accurate risk stratification in the general population could enable health care providers to focus early detection strategies to at-risk individuals. Here, we validate a combined risk prediction model that integrates a polygenic risk score and a clinical risk model. Methods Using the UK Biobank, we conducted a prospective cohort study assessing 10-year pancreatic cancer risks based on a polygenic risk score, a clinical risk score, and a combined risk score. We assessed the association, discrimination, calibration, cumulative hazards, and standardized incidence ratios compared to population incidence rates for the risk scores. We also conducted net reclassification analyses. Results While all of the risk scores discriminated well between affected and unaffected participants, the combined risk score - with a Harrell's C-index of 0.714 (95% confidence interval [CI] = 0.698, 0.730) - discriminated better than both the polygenic risk score (P = .001) and the clinical risk score (P = .02). In terms of calibration, there was no problem with dispersion for the combined risk score (β = 0.952, 95% CI = 0.865-1.039, P = .3) and overall there was a small overestimation of risk (α = -0.089, 95% CI = -0.156 to -0.021, P = .009). Participants in the top decile of 10-year risk were at 1.413 (95% CI = 1.242-1.607) times population risk. Conclusion The combined risk score was able to identify individuals at substantially increased risk of pancreatic cancer and to whom targeted screening could be useful.
Collapse
Affiliation(s)
| | - Erika Spaeth
- Phenogen Sciences Inc, Charlotte, North Carolina
| | - Chi Kuen Wong
- Genetic Technologies Limited, Fitzroy, Victoria, Australia
| | | | - Richard Allman
- Genetic Technologies Limited, Fitzroy, Victoria, Australia
| |
Collapse
|
6
|
Zhang Y, Wang QL, Yuan C, Lee AA, Babic A, Ng K, Perez K, Nowak JA, Lagergren J, Stampfer MJ, Giovannucci EL, Sander C, Rosenthal MH, Kraft P, Wolpin BM. Pancreatic cancer is associated with medication changes prior to clinical diagnosis. Nat Commun 2023; 14:2437. [PMID: 37117188 PMCID: PMC10147931 DOI: 10.1038/s41467-023-38088-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 04/11/2023] [Indexed: 04/30/2023] Open
Abstract
Patients with pancreatic ductal adenocarcinoma (PDAC) commonly develop symptoms and signs in the 1-2 years before diagnosis that can result in changes to medications. We investigate recent medication changes and PDAC diagnosis in Nurses' Health Study (NHS; females) and Health Professionals Follow-up Study (HPFS; males), including up to 148,973 U.S. participants followed for 2,994,057 person-years and 991 incident PDAC cases. Here we show recent initiation of antidiabetic (NHS) or anticoagulant (NHS, HFS) medications and cessation of antihypertensive medications (NHS, HPFS) are associated with pancreatic cancer diagnosis in the next 2 years. Two-year PDAC risk increases as number of relevant medication changes increases (P-trend <1 × 10-5), with participants who recently start antidiabetic and stop antihypertensive medications having multivariable-adjusted hazard ratio of 4.86 (95%CI, 1.74-13.6). These changes are not associated with diagnosis of other digestive system cancers. Recent medication changes should be considered as candidate features in multi-factor risk models for PDAC, though they are not causally implicated in development of PDAC.
Collapse
Affiliation(s)
- Yin Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Qiao-Li Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Department of Clinical Science, Intervention and Technology, Karolinka Institutet, Stockholm, Sweden
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Alice A Lee
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Kimberly Perez
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jesper Lagergren
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Meir J Stampfer
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Edward L Giovannucci
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Chris Sander
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Michael H Rosenthal
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Søreide K, Ismail W, Roalsø M, Ghotbi J, Zaharia C. Early Diagnosis of Pancreatic Cancer: Clinical Premonitions, Timely Precursor Detection and Increased Curative-Intent Surgery. Cancer Control 2023; 30:10732748231154711. [PMID: 36916724 PMCID: PMC9893084 DOI: 10.1177/10732748231154711] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The overall poor prognosis in pancreatic cancer is related to late clinical detection. Early diagnosis remains a considerable challenge in pancreatic cancer. Unfortunately, the onset of clinical symptoms in patients usually indicate advanced disease or presence of metastasis. ANALYSIS AND RESULTS Currently, there are no designated diagnostic or screening tests for pancreatic cancer in clinical use. Thus, identifying risk groups, preclinical risk factors or surveillance strategies to facilitate early detection is a target for ongoing research. Hereditary genetic syndromes are a obvious, but small group at risk, and warrants close surveillance as suggested by society guidelines. Screening for pancreatic cancer in asymptomatic individuals is currently associated with the risk of false positive tests and, thus, risk of harms that outweigh benefits. The promise of cancer biomarkers and use of 'omics' technology (genomic, transcriptomics, metabolomics etc.) has yet to see a clinical breakthrough. Several proposed biomarker studies for early cancer detection lack external validation or, when externally validated, have shown considerably lower accuracy than in the original data. Biopsies or tissues are often taken at the time of diagnosis in research studies, hence invalidating the value of a time-dependent lag of the biomarker to detect a pre-clinical, asymptomatic yet operable cancer. New technologies will be essential for early diagnosis, with emerging data from image-based radiomics approaches, artificial intelligence and machine learning suggesting avenues for improved detection. CONCLUSIONS Early detection may come from analytics of various body fluids (eg 'liquid biopsies' from blood or urine). In this review we present some the technological platforms that are explored for their ability to detect pancreatic cancer, some of which may eventually change the prospects and outcomes of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Kjetil Søreide
- Department of Gastrointestinal Surgery, HPB unit, 60496Stavanger University Hospital, Stavanger, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Gastrointestinal Translational Research Group, Laboratory for Molecular Medicine, 60496Stavanger University Hospital, Stavanger, Norway
| | - Warsan Ismail
- Department of Gastrointestinal Surgery, HPB unit, 60496Stavanger University Hospital, Stavanger, Norway
| | - Marcus Roalsø
- Department of Gastrointestinal Surgery, HPB unit, 60496Stavanger University Hospital, Stavanger, Norway.,Gastrointestinal Translational Research Group, Laboratory for Molecular Medicine, 60496Stavanger University Hospital, Stavanger, Norway.,Department of Quality and Health Technology, 60496University of Stavanger, Stavanger, Norway
| | - Jacob Ghotbi
- Department of Gastrointestinal Surgery, HPB unit, 60496Stavanger University Hospital, Stavanger, Norway
| | - Claudia Zaharia
- Gastrointestinal Translational Research Group, Laboratory for Molecular Medicine, 60496Stavanger University Hospital, Stavanger, Norway.,Department of Pathology, 60496Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
8
|
Clinical Prediction Models for Pancreatic Cancer in General and At-Risk Populations: A Systematic Review. Am J Gastroenterol 2023; 118:26-40. [PMID: 36148840 DOI: 10.14309/ajg.0000000000002022] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 09/16/2022] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Identifying high-risk individuals using a risk prediction model could be a crucial first stage of screening pathways to improve the early detection of pancreatic cancer. A systematic review was conducted to critically evaluate the published primary literature on the development or validation of clinical risk prediction models for pancreatic cancer risk. METHODS MEDLINE, Embase, and Web of Science were searched for relevant articles from the inception of each database up to November 2021. Study selection and data extraction were conducted by 2 independent reviewers. The Prediction model Risk Of Bias Assessment Tool (PROBAST) was applied to assess risk of bias. RESULTS In total, 33 studies were included, describing 38 risk prediction models. Excluding studies with an overlapping population, this study consist of 15,848,100 participants, of which 58,313 were diagnosed with pancreatic cancer. Eight studies externally validated their model, and 13 performed internal validation. The studies described risk prediction models for pancreatic cancer in the general population (n = 14), patients with diabetes (n = 8), and individuals with gastrointestinal (and other) symptoms (symptoms included abdominal pain, unexplained weight loss, jaundice, and change in bowel habits and indigestion; n = 11). The commonly used clinical risk factors in the model were cigarette smoking (n = 27), age (n = 25), diabetes history (n = 22), chronic pancreatitis (n = 18), and body mass index (n = 14). In the 25 studies that assessed model performance, C-statistics ranged from 0.61 to 0.98. Of the 33 studies included, 6 were rated as being at a low risk of bias based on PROBAST. DISCUSSION Many clinical risk prediction models for pancreatic cancer had been developed for different target populations. Although low risk-of-bias studies were identified, these require external validation and implementation studies to ensure that these will benefit clinical decision making.
Collapse
|
9
|
Lee HS, Chae W, Sung MJ, Keum J, Jo JH, Chung MJ, Park JY, Park SW, Song SY, Park EC, Nam CM, Jang SI, Bang S. Difference of risk of pancreatic cancer in new-onset diabetes and long-standing diabetes: population-based cohort study. J Clin Endocrinol Metab 2022; 108:1338-1347. [PMID: 36548964 DOI: 10.1210/clinem/dgac728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
CONTEXT Considering the absence of methods to find pancreatic cancer early, surveillance of high-risk groups is needed for early diagnosis. OBJECTIVE The study aimed to investigate the effect in the incidence of pancreatic cancer and the differences between new-onset DM (NODM) and long-standing DM (LSDM) since NODM group is a representative high-risk group. METHODS The Korean National Health Insurance Service-National Sample Cohort between 2002 and 2013 data was used. Regarding 88,396 people with DM (case group), we conducted a 1:1 propensity score matching to select a matched non-DM population (control group). To investigate the interaction between DM and the time variable distinguishing NODM and LSDM, we performed a multi-variable time-dependent Cox regression analysis. RESULTS The incidence of pancreatic cancer was higher in the DM group compared to the non-DM group (0.52% vs. 0.16%, P < 0.001). The DM group had shown different risk of pancreatic cancer development according to the duration since the DM diagnosis (NODM hazard ratio (HR): 3.81, 95% confidence interval (CI): 2.97-4.88, P < 0.001; LSDM HR: 1.53, 95% CI: 1.11-2.11, P < 0.001). When the NODM and the LSDM groups were compared, the risk of pancreatic cancer was higher in the NODM group than LSDM group (HR: 1.55, P = 0.020). In subgroup analysis, NODM group showed that men (HR = 4.42 95% CI: 3.15-6.19, P < 0.001) and patients who were in their 50 s (HR = 7.54, 95% CI: 3.24-17.56, P < 0.001) were at a higher risk of developing pancreatic cancer than matched same sex or age control group (non-DM population), respectively. CONCLUSION The risk of pancreatic cancer was greater in people with DM than non-DM population. Among people with DM, NODM showed a higher risk of pancreatic cancer than long standing DM.
Collapse
Affiliation(s)
- Hee Seung Lee
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Wonjeong Chae
- Department of Health Policy and Management, Yonsei University Graduate School of Public Health, Seoul, Republic of Korea
- Institute of Health Services Research, Yonsei University, Seoul, Republic of Korea
| | - Min Je Sung
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jiyoung Keum
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung Hyun Jo
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Moon Jae Chung
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Woo Park
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Si Young Song
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun-Cheol Park
- Institute of Health Services Research, Yonsei University, Seoul, Republic of Korea
- Department of Preventive Medicine, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Chung Mo Nam
- Department of Preventive Medicine, College of Medicine, Yonsei University, Seoul, Republic of Korea
- Department of Biostatics, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Sung-In Jang
- Institute of Health Services Research, Yonsei University, Seoul, Republic of Korea
- Department of Preventive Medicine, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Seungmin Bang
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
10
|
Huber MA, Nadella S, Cao H, Kallakury B, Tucker RD, Gay MD, Shivapurkar N, Edmondson EF, Yue Y, Dou W, Fang HB, Smith JP. Does Chronic Use of High Dose Proton Pump Inhibitors Increase Risk for Pancreatic Cancer? Pancreas 2022; 51:1118-1127. [PMID: 37078934 PMCID: PMC10119745 DOI: 10.1097/mpa.0000000000002145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
OBJECTIVES To analyze whether use of proton pump inhibitors increase the risk for pancreatic cancer in a mouse model and human clinical cohorts. METHODS p48-Cre/LSL-KrasG12D mice that develop precancerous pancreatic intraepithelial neoplasia (PanINs) were treated with low- or high-dose proton pump inhibitors (PPIs) orally for 1 and 4 months. The mechanism for the cholecystokinin receptor 2 (CCK-2R) activation was investigated in vitro. Two resources were employed to analyze the risk of pancreatic cancer in human subjects with PPI use. RESULTS Serum gastrin levels were increased 8-fold (P < 0.0001) in mice treated with chronic high-dose PPIs, and this change correlated with an increase (P = 0.02) in PanIN grade and the development of microinvasive cancer. The CCK-2R expression was regulated by microRNA-148a in the p48-Cre/LSL-KrasG12D mice pancreas and in human pancreatic cancer cells in vitro. Proton pump inhibitor consumption in human subjects was correlated with pancreatic cancer risk (odds ratio, 1.54). A validation analysis conducted using the large-scale United Kingdom Biobank database confirmed the correlation (odds ratio, 1.9; P = 0.00761) of pancreatic cancer risk with PPI exposure. CONCLUSIONS This investigation revealed in both murine models and human subjects, PPI use is correlated with a risk for development of pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | | | - Robin D Tucker
- Department of Pathology, Georgetown University, Washington, DC
| | | | | | | | - Yuanzhen Yue
- Biostatistics, Bioinformatics and Biomathematics, Georgetown University, Washington, DC
| | - Wenyu Dou
- Biostatistics, Bioinformatics and Biomathematics, Georgetown University, Washington, DC
| | - Hong-Bin Fang
- Biostatistics, Bioinformatics and Biomathematics, Georgetown University, Washington, DC
| | | |
Collapse
|
11
|
Zhou W, Chen X, Fan Q, Yu H, Jiang W. Using proton pump inhibitors increases the risk of hepato-biliary-pancreatic cancer. A systematic review and meta-analysis. Front Pharmacol 2022; 13:979215. [PMID: 36188583 PMCID: PMC9515471 DOI: 10.3389/fphar.2022.979215] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Background: More and more studies are focusing on the adverse effects and damage caused by PPI abuse, we carried out a systematic review and meta-analysis for assessing whether the proton pump inhibitor (PPI) leads to hepato-biliary-pancreatic cancer. Methods: PubMed, EMBASE and Web of Science were searched until 1 July 2022, 25 studies (17 case-control and 8 cohort studies; 2741853 individuals) included in this study. Pooled Odd Ratios (ORs) were used for random effect models. Sensitivity analysis and dose-response analysis, subgroup analysis were all conducted. Results: The aggregate OR of the meta-analysis was 1.69 (95% confidence interval (CI): 1.42–2.01, p = 0.01) and heterogeneity (I2 = 98.9%, p < 0.001) was substantial. According to stratified subgroup analyses, the incidence of hepato-biliary-pancreatic cancer was associated, expect for study design, study quality and region. Risk of hepato-biliary-pancreatic cancer is highest when people is treated with normal doses of PPI. The risks decrease and become insignificant when the cumulative defined daily dose (cDDD) increases. Conclusion: The use of PPI may be associated with an increased risk of hepato-biliary-pancreatic cancer. Hence, caution is needed when using PPIs among patients with a high risk of hepato-biliary-pancreatic cancer.
Collapse
Affiliation(s)
- Wence Zhou
- First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
- *Correspondence: Wence Zhou,
| | - Xinlong Chen
- First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Qigang Fan
- First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Haichuan Yu
- First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Wenkai Jiang
- First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
12
|
Yablecovitch D, Ben-Horin S, Picard O, Yavzori M, Fudim E, Nadler M, Levy I, Sakhnini E, Lang A, Engel T, Lahav M, Saker T, Neuman S, Selinger L, Dvir R, Raitses-Gurevich M, Golan T, Laish I. Serum Syndecan-1: A Novel Biomarker for Pancreatic Ductal Adenocarcinoma. Clin Transl Gastroenterol 2022; 13:e00473. [PMID: 35297817 PMCID: PMC9132524 DOI: 10.14309/ctg.0000000000000473] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 02/01/2022] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Syndecan-1 (SDC1) has multiple functions in tumorigenesis in general and specifically in pancreatic cancer. We aimed to evaluate SDC1 as a diagnostic and prognostic biomarker in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS In this case-control study, patients newly diagnosed with a biopsy-proven PDAC were enrolled alongside healthy individuals in a derivation-validation cohort design. Serum SDC1 was measured by enzyme-linked immunoassay. The diagnostic accuracy of SDC1 levels for diagnosing PDAC was computed. A unified cohort enriched with additional early-stage patients with PDAC was used to evaluate the association of SDC1 with survival outcomes and patient characteristics. RESULTS In the derivation cohort, serum SDC1 levels were significantly higher in patients with PDAC (n = 39) compared with healthy controls (n = 20) (40.1 ng/mL, interquartile range 29.8-95.3 vs 25.6 ng/mL, interquartile range 17.1-29.8, respectively; P < 0.001). The receiver operating characteristic analysis area under the curve was 0.847 (95% confidence interval 0.747-0.947, P < 0.001). These results were replicated in a separate age-matched validation cohort (n = 38 PDAC, n = 38 controls; area under the curve 0.844, 95% confidence interval 0.757-0.932, P < 0.001). In the combined-enriched PDAC cohort (n = 110), using a cutoff of 35 ng/mL, the median overall 5-year survival between patients below and above this cutoff was not significantly different, although a trend for better survival after 1 year was found in the lower level group (P = 0.06). There were 12 of the 110 patients with PDAC (11%) who had normal CA 19-9 in the presence of elevated SDC1. DISCUSSION These findings suggest serum SDC1 as a promising novel biomarker for early blood-based diagnosis of pancreatic cancer.
Collapse
Affiliation(s)
- Doron Yablecovitch
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Shomron Ben-Horin
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Orit Picard
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Miri Yavzori
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Ella Fudim
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Moshe Nadler
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Idan Levy
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Emad Sakhnini
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Alon Lang
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Tal Engel
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Maor Lahav
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Talia Saker
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
- Shalvata Mental Health Center, Hod Hasharon, Israel;
| | - Sandra Neuman
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Limor Selinger
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Revital Dvir
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Maria Raitses-Gurevich
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
- Department of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel.
| | - Talia Golan
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
- Department of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel.
| | - Ido Laish
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| |
Collapse
|
13
|
Prediction Model for Pancreatic Cancer-A Population-Based Study from NHIRD. Cancers (Basel) 2022; 14:cancers14040882. [PMID: 35205630 PMCID: PMC8870511 DOI: 10.3390/cancers14040882] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 01/06/2023] Open
Abstract
Simple Summary Pancreatic cancer has been ranked seventh in the top ten cancer mortality rates for the past three year in Taiwan. It is one of the more difficult cancers to detect early due to the lack of early diagnostic tools. This is a population-based study from NHIRD. A higher performance pancreatic cancer prediction model has been established. This predictive model can improve the awareness of the risk of pancreatic cancer and give patients with pancreatic cancer a simpler tool for early screening in the golden period when the disease can still be eradicated. Abstract (1) Background: Cancer has been the leading cause of death in Taiwan for 39 years, and among them, pancreatic cancer has been ranked seventh in the top ten cancer mortality rates for the past three years. While the incidence rate of pancreatic cancer is ranked at the bottom of the top 10 cancers, the survival rate is very low. Pancreatic cancer is one of the more difficult cancers to detect early due to the lack of early diagnostic tools. Early screening is important for the treatment of pancreatic cancer. Only a few studies have designed predictive models for pancreatic cancer. (2) Methods: The Taiwan Health Insurance Database was used in this study, covering over 99% of the population in Taiwan. The subset sample was not significantly different from the original NHIRD sample. A machine learning approach was used to develop a predictive model for pancreatic cancer disease. Four models, including logistic regression, deep neural networks, ensemble learning, and voting ensemble were used in this study. The ROC curve and a confusion matrix were used to evaluate the accuracy of the pancreatic cancer prediction models. (3) Results: The AUC of the LR model was higher than the other three models in the external testing set for all three of the factor combinations. Sensitivity was best measured by the stacking model for the first factor combinations, and specificity was best measured by the DNN model for the second factor combination. The result of the model that used only nine factors (third factor combinations) was equal to the other two factor combinations. The AUC of the previous models for the early assessment of pancreatic cancer ranged from approximately 0.57 to 0.71. The AUC of this study was higher than that of previous studies and ranged from 0.71 to 0.76, which provides higher accuracy. (4) Conclusions: This study compared the performances of LR, DNN, stacking, and voting models for pancreatic cancer prediction and constructed a pancreatic cancer prediction model with accuracy higher than that of previous studies. This predictive model will improve awareness of the risk of pancreatic cancer and give patients with pancreatic cancer a simpler tool for early screening in the golden period when the disease can still be eradicated.
Collapse
|
14
|
Jeon CY, Kim S, Lin YC, Risch HA, Goodarzi MO, Nuckols TK, Freedland SJ, Pandol SJ, Pisegna JR. Prediction of Pancreatic Cancer in Diabetes Patients with Worsening Glycemic Control. Cancer Epidemiol Biomarkers Prev 2022; 31:242-253. [PMID: 34728468 PMCID: PMC8759109 DOI: 10.1158/1055-9965.epi-21-0712] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/25/2021] [Accepted: 10/22/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Worsening glycemic control indicates elevated risk of pancreatic ductal adenocarcinoma (PDAC). We developed prediction models for PDAC among those with worsening glycemic control after diabetes diagnosis. METHODS In 2000-2016 records within the Veterans Affairs Health System (VA), we identified three cohorts with progression of diabetes: (i) insulin initiation (n = 449,685), (ii) initiation of combination oral hypoglycemic medication (n = 414,460), and (iii) hemoglobin A1c (HbA1c) ≥8% with ≥Δ1% within 15 months (n = 593,401). We computed 12-, 36-, and 60-month incidence of PDAC and developed prediction models separately for males and females, with consideration of >30 demographic, behavioral, clinical, and laboratory variables. Models were selected to optimize Akaike's Information Criterion, and performance for predicting 12-, 36-, and 60-month incident PDAC was evaluated by bootstrap. RESULTS Incidence of PDAC was highest for insulin initiators and greater in males than in females. Optimism-corrected c-indices of the models for predicting 36-month incidence of PDAC in the male population were: (i) 0.72, (ii) 0.70, and (iii) 0.71, respectively. Models performed better for predicting 12-month incident PDAC [c-index (i) 0.78, (ii) 0.73, (iii) 0.76 for males], and worse for predicting 60-month incident PDAC [c-index (i) 0.69, (ii) 0.67, (iii) 0.68 for males]. Model performance was lower among females. For subjects whose model-predicted 36-month PDAC risks were ≥1%, the observed incidences were (i) 1.9%, (ii) 2.2%, and (iii) 1.8%. CONCLUSIONS Sex-specific models for PDAC can estimate risk of PDAC at the time of progression of diabetes. IMPACT Our models can identify diabetes patients who would benefit from PDAC screening.
Collapse
Affiliation(s)
- Christie Y. Jeon
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California.,Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California.,Corresponding Author: Christie Y. Jeon, Department of Medicine, Cedars-Sinai Medical Center, 700 N San Vicente Boulevard, Pacific Design Center G596, West Hollywood, CA 90069. Phone: 310-423-6345; E-mail:
| | - Sungjin Kim
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California
| | - Yu-Chen Lin
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California
| | - Harvey A. Risch
- Department of Epidemiology, Yale School of Public Health, Los Angeles, California
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, California
| | - Teryl K. Nuckols
- Division of General Internal Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Stephen J. Freedland
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California.,Section of Urology, Durham VA Medical Center, Durham, North Carolina
| | - Stephen J. Pandol
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California.,Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Joseph R. Pisegna
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| |
Collapse
|
15
|
Wu Y, Zeng H, Yu Q, Huang H, Fervers B, Chen ZS, Lu L. A Circulating Exosome RNA Signature Is a Potential Diagnostic Marker for Pancreatic Cancer, a Systematic Study. Cancers (Basel) 2021; 13:cancers13112565. [PMID: 34073722 PMCID: PMC8197236 DOI: 10.3390/cancers13112565] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/19/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Most patients with pancreatic cancer are diagnosed at an advanced stage due to the lack of tools with high sensitivity and specificity for early detection. Aberrant gene expression occurs in pancreatic cancer, which can be packaged into nanoparticles (also known as exosomes or nano-sized extracellular vesicles) and then released into blood. In this study, we aimed to evaluate the diagnostic value of a circulating exosome RNA signature in pancreatic cancer. Our findings indicate that the circulating exosome RNA signature is a potential marker for the early detection or diagnosis of pancreatic cancer. Abstract Several exosome proteins, miRNAs and KRAS mutations have been investigated in the hope of carrying out the early detection of pancreatic cancer with high sensitivity and specificity, but they have proven to be insufficient. Exosome RNAs, however, have not been extensively evaluated in the diagnosis of pancreatic cancer. The purpose of this study was to investigate the potential of circulating exosome RNAs in pancreatic cancer detection. By retrieving RNA-seq data from publicly accessed databases, differential expression and random-effects meta-analyses were performed. The results showed that pancreatic cancer had a distinct circulating exosome RNA signature in healthy individuals, and that the top 10 candidate exosome RNAs could distinguish patients from healthy individuals with an area under the curve (AUC) of 1.0. Three (HIST2H2AA3, LUZP6 and HLA-DRA) of the 10 genes in exosomes had similar differential patterns to those in tumor tissues based on RNA-seq data. In the validation dataset, the levels of these three genes in exosomes displayed good performance in distinguishing cancer from both chronic pancreatitis (AUC = 0.815) and healthy controls (AUC = 0.8558), whereas a slight difference existed between chronic pancreatitis and healthy controls (AUC = 0.586). Of the three genes, the level of HIST2H2AA3 was positively associated with KRAS status. However, there was no significant difference in the levels of the three genes across the disease stages (stages I–IV). These findings indicate that circulating exosome RNAs have a potential early detection value in pancreatic cancer, and that a distinct exosome RNA signature exists in distinguishing pancreatic cancer from healthy individuals.
Collapse
Affiliation(s)
- Yixing Wu
- Department of Endocrinology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China;
| | - Hongmei Zeng
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Qing Yu
- Center for Cancer and Blood Disorders, Children’s National Medical Center, Washington, DC 20010, USA;
| | - Huatian Huang
- Department of Imaging, Guizhou Qianxinan People’s Hospital, Xingyi 652400, China;
| | - Beatrice Fervers
- Département Prévention Cancer Environnement, Centre Léon Bérard—Université Lyon 1, 69008 Lyon, France;
- UMR Inserm 1296 “Radiations: Défense, Santé, Environnement”, Centre Léon Bérard, 69008 Lyon, France
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, St. John’s University, New York, NY 11439, USA;
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, School of Medicine, New Haven, CT 06520, USA
- Center for Biomedical Data Science, Yale University, 60 College Street, New Haven, CT 06520, USA
- Yale Cancer Center, Yale University, 60 College Street, New Haven, CT 06520, USA
- Correspondence:
| |
Collapse
|
16
|
Abstract
OBJECTIVES The purpose of this study was to investigate the association of syndecan-1 (SDC1) and KRAS molecular characteristics with patient survival in pancreatic cancer. METHODS Both SDC1 mRNA and methylation and KRAS mRNA and somatic mutations, as well as clinical data were retrieved from The Cancer Genome Alta pancreatic cancer data set for survival analyses. Kyoto Encyclopedia of Gene and Genomes pathway analysis for coexpressed genes for either SDC1 or KRAS was performed, respectively. RESULTS A significantly negative correlation existed between SDC1 mRNA and DNA methylation. Patients with KRAS somatic mutations had a significantly higher SDC1 mRNA but lower methylation than those without the mutations. Compared with patients with KRASSDC1 signature, those with a high level of KRAS and SDC1 alone or both had a significantly elevated mortality. The adjusted hazard ratios (95% confidence interval) were 2.30 (1.16-4.54, P = 0.017) for KRASSDC1, 2.85 (1.48-5.49, P = 0.002) for KRASSDC1, and 2.48 (1.31-4.70, P = 0.005) for KRASSDC1, respectively. Several Kyoto Encyclopedia of Gene and Genomes pathways were shared, whereas there were distinct pathways between KRAS and SDC1 coexpressed genes. CONCLUSIONS SDC1 interplays with KRAS, and targeting both KRAS and SDC1 in combination may be more beneficial to pancreatic cancer patients.
Collapse
|
17
|
Streicher SA, Klein AP, Olson SH, Kurtz RC, Amundadottir LT, DeWan AT, Zhao H, Risch HA. A pooled genome-wide association study identifies pancreatic cancer susceptibility loci on chromosome 19p12 and 19p13.3 in the full-Jewish population. Hum Genet 2020; 140:309-319. [PMID: 32671597 DOI: 10.1007/s00439-020-02205-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/07/2020] [Indexed: 12/20/2022]
Abstract
Jews are estimated to be at increased risk of pancreatic cancer compared to non-Jews, but their observed 50-80% excess risk is not explained by known non-genetic or genetic risk factors. We conducted a GWAS in a case-control sample of American Jews, largely Ashkenazi, including 406 pancreatic cancer patients and 2332 controls, identified in the dbGaP, PanScan I/II, PanC4 and GERA data sets. We then examined resulting SNPs with P < 10-7 in an expanded sample set, of 539 full- or part-Jewish pancreatic cancer patients and 4117 full- or part-Jewish controls from the same data sets. Jewish ancestries were genetically determined using seeded FastPCA. Among the full Jews, a novel genome-wide significant association was detected on chromosome 19p12 (rs66562280, per-allele OR = 1.55, 95% CI = 1.33-1.81, P = 10-7.6). A suggestive relatively independent association was detected on chromosome 19p13.3 (rs2656937, OR = 1.53, 95% CI = 1.31-1.78, P = 10-7.0). Similar associations were seen for these SNPs among the full and part Jews combined. This is the first GWAS conducted for pancreatic cancer in the increased-risk Jewish population. The SNPs rs66562280 and rs2656937 are located in introns of ZNF100-like and ARRDC5, respectively, and are known to alter regulatory motifs of genes that play integral roles in pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Samantha A Streicher
- Department of Chronic Disease Epidemiology, Yale School of Public Health, 60 College Street, New Haven, CT, 06520-8034, USA
| | - Alison P Klein
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 401 North Broadway, Baltimore, MD, 21287, USA.,Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, 401 North Broadway, Baltimore, MD, 21287, USA
| | - Sara H Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 485 Lexington Avenue, New York, NY, 10017, USA
| | - Robert C Kurtz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health,, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Andrew T DeWan
- Department of Chronic Disease Epidemiology, Yale School of Public Health, 60 College Street, New Haven, CT, 06520-8034, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, 60 College Street, New Haven, Connecticut, 06520, USA.,Program of Computational Biology and Bioinformatics, Yale University, 260 Whitney Avenue, New Haven, CT, 06520, USA
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, 60 College Street, New Haven, CT, 06520-8034, USA.
| |
Collapse
|
18
|
Pereira SP, Oldfield L, Ney A, Hart PA, Keane MG, Pandol SJ, Li D, Greenhalf W, Jeon CY, Koay EJ, Almario CV, Halloran C, Lennon AM, Costello E. Early detection of pancreatic cancer. Lancet Gastroenterol Hepatol 2020; 5:698-710. [PMID: 32135127 PMCID: PMC7380506 DOI: 10.1016/s2468-1253(19)30416-9] [Citation(s) in RCA: 254] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma is most frequently detected at an advanced stage. Such late detection restricts treatment options and contributes to a dismal 5-year survival rate of 3-15%. Pancreatic ductal adenocarcinoma is relatively uncommon and screening of the asymptomatic adult population is not feasible or recommended with current modalities. However, screening of individuals in high-risk groups is recommended. Here, we review groups at high risk for pancreatic ductal adenocarcinoma, including individuals with inherited predisposition and patients with pancreatic cystic lesions. We discuss studies aimed at finding ways of identifying pancreatic ductal adenocarcinoma in high-risk groups, such as among individuals with new-onset diabetes mellitus and people attending primary and secondary care practices with symptoms that suggest this cancer. We review early detection biomarkers, explore the potential of using social media for detection, appraise prediction models developed using electronic health records and research data, and examine the application of artificial intelligence to medical imaging for the purposes of early detection.
Collapse
Affiliation(s)
- Stephen P Pereira
- Institute for Liver and Digestive Health, University College London, London, UK
| | - Lucy Oldfield
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, UK
| | - Alexander Ney
- Institute for Liver and Digestive Health, University College London, London, UK
| | - Phil A Hart
- Division of Gastroenterology, Hepatology, and Nutrition, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Margaret G Keane
- Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, MD, USA
| | - Stephen J Pandol
- Department of Medicine, Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Debiao Li
- Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - William Greenhalf
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, UK
| | - Christie Y Jeon
- Department of Medicine, Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Eugene J Koay
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher V Almario
- Department of Medicine, Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Christopher Halloran
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, UK
| | - Anne Marie Lennon
- Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, MD, USA
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, UK.
| |
Collapse
|
19
|
Wu BU, Butler RK, Lustigova E, Lawrence JM, Chen W. Association of Glycated Hemoglobin Levels With Risk of Pancreatic Cancer. JAMA Netw Open 2020; 3:e204945. [PMID: 32530471 PMCID: PMC7292999 DOI: 10.1001/jamanetworkopen.2020.4945] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
IMPORTANCE New-onset diabetes after the age of 50 years is a potential indicator of pancreatic cancer. Understanding the associations between hyperglycemia, diabetes, and pancreatic cancer, including pancreatic ductal adenocarcinoma, is key to developing an approach to early detection. OBJECTIVE To assess the association of elevation in glycated hemoglobin (HbA1c) with the risk of pancreatic cancer. DESIGN, SETTING, AND PARTICIPANTS This cohort study was conducted using data collected from an integrated health care system in California. A total of 851 402 patients aged 50 to 84 years who had HbA1c measurements taken between 2010 and 2014 were identified as the base cohort, with 12 contemporaneous cohorts created based on varying HbA1c thresholds (ie, 6.1%, 6.3%, 6.5%, and 6.7%) and prior diabetes status. Data analysis was conducted from August 2018 to September 2019. MAIN OUTCOMES AND MEASURES New cases of pancreatic cancer identified through cancer registry and California death files during a 3-year period. Three-year risk, incidence rate, sensitivity, number of patients needed to screen to detect 1 case, timing, and stage at diagnosis were determined. RESULTS Among 851 402 patients in the base cohort, 447 502 (52.5%) were women, 255 441 (30.0%) were Hispanic participants, 383 685 (45.1%) were non-Hispanic white participants, 100 477 (11.8%) were Asian participants, and 88 969 (10.4%) were non-Hispanic black participants, with a median (interquartile range) age of 62 (56-69) years and a median (interquartile range) HbA1c level of 6.0% (5.7%-6.6%). The incidence rate of pancreatic cancer was 0.45 (95% CI, 0.43-0.49) per 1000 person-years. After excluding prior diabetes as well as confirmation of new-onset hyperglycemia based on an HbA1c level of 6.5%, a total of 20 012 patients remained, with 74 of 1041 pancreatic ductal adenocarcinoma cases (7.1%) from the base cohort included. The rate of pancreatic cancer was 0.72 (95% CI, 0.32-1.42) per 1000 person-years among Asian patients, 0.83 (95% CI, 0.35-1.71) per 1000 person-years among non-Hispanic black patients, 0.84 (95% CI, 0.48-1.37) per 1000 person-years among Hispanic patients, and 2.37 (95% CI, 1.75-3.14) per 1000 person-years among non-Hispanic white patients. Overall, 42 of 74 cancers (56.8%) were diagnosed within 1 year of the index laboratory test. Among 1041 total cases, 708 (68.0%) had staging information available, of whom 465 (65.7%) had stage III or IV disease at diagnosis. In the base cohort, the number needed to undergo evaluation to identify a single case of pancreatic ductal adenocarcinoma was 818 (95% CI, 770-869), with estimates ranging from 206 (95% CI, 160-264) to 600 (95% CI, 540-666) in the subcohorts. CONCLUSIONS AND RELEVANCE The findings of this study suggest that screening patients for pancreatic cancer based solely on elevation in HbA1c level is unlikely to represent an effective strategy. Future efforts to identify a high-risk population based on changes in glycemic parameters should account for racial/ethnic differences.
Collapse
Affiliation(s)
- Bechien U. Wu
- Center for Pancreatic Care, Division of Gastroenterology, Kaiser Permanente Los Angeles Medical Center, Los Angeles, California
| | - Rebecca K. Butler
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | - Eva Lustigova
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | - Jean M. Lawrence
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | - Wansu Chen
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| |
Collapse
|
20
|
Kim J, Yuan C, Babic A, Bao Y, Clish CB, Pollak MN, Amundadottir LT, Klein AP, Stolzenberg-Solomon RZ, Pandharipande PV, Brais LK, Welch MW, Ng K, Giovannucci EL, Sesso HD, Manson JE, Stampfer MJ, Fuchs CS, Wolpin BM, Kraft P. Genetic and Circulating Biomarker Data Improve Risk Prediction for Pancreatic Cancer in the General Population. Cancer Epidemiol Biomarkers Prev 2020; 29:999-1008. [PMID: 32321713 PMCID: PMC8020898 DOI: 10.1158/1055-9965.epi-19-1389] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/31/2020] [Accepted: 02/07/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Pancreatic cancer is the third leading cause of cancer death in the United States, and 80% of patients present with advanced, incurable disease. Risk markers for pancreatic cancer have been characterized, but combined models are not used clinically to identify individuals at high risk for the disease. METHODS Within a nested case-control study of 500 pancreatic cancer cases diagnosed after blood collection and 1,091 matched controls enrolled in four U.S. prospective cohorts, we characterized absolute risk models that included clinical factors (e.g., body mass index, history of diabetes), germline genetic polymorphisms, and circulating biomarkers. RESULTS Model discrimination showed an area under ROC curve of 0.62 via cross-validation. Our final integrated model identified 3.7% of men and 2.6% of women who had at least 3 times greater than average risk in the ensuing 10 years. Individuals within the top risk percentile had a 4% risk of developing pancreatic cancer by age 80 years and 2% 10-year risk at age 70 years. CONCLUSIONS Risk models that include established clinical, genetic, and circulating factors improved disease discrimination over models using clinical factors alone. IMPACT Absolute risk models for pancreatic cancer may help identify individuals in the general population appropriate for disease interception.
Collapse
Affiliation(s)
- Jihye Kim
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ying Bao
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Clary B Clish
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
| | - Michael N Pollak
- Cancer Prevention Research Unit, Department of Oncology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Alison P Klein
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Rachael Z Stolzenberg-Solomon
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Pari V Pandharipande
- Department of Radiology and Institute for Technology Assessment, Massachusetts General Hospital, Boston, Massachusetts
| | - Lauren K Brais
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marisa W Welch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Howard D Sesso
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Division of Prevention Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - JoAnn E Manson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Division of Prevention Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Meir J Stampfer
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Charles S Fuchs
- Department of Medical Oncology, Yale Cancer Center, New Haven, Connecticut
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut
- Department of Medical Oncology, Smilow Cancer Hospital, New Haven, Connecticut
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
21
|
Risch HA. Diabetes and Pancreatic Cancer: Both Cause and Effect. J Natl Cancer Inst 2020; 111:1-2. [PMID: 29917095 DOI: 10.1093/jnci/djy093] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 04/17/2018] [Indexed: 12/25/2022] Open
Affiliation(s)
- Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT
| |
Collapse
|
22
|
Alkhushaym N, Almutairi AR, Althagafi A, Fallatah SB, Oh M, Martin JR, Babiker HM, McBride A, Abraham I. Exposure to proton pump inhibitors and risk of pancreatic cancer: a meta-analysis. Expert Opin Drug Saf 2020; 19:327-334. [PMID: 31928106 DOI: 10.1080/14740338.2020.1715939] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/10/2020] [Indexed: 02/07/2023]
Abstract
Objectives: To estimate the pancreatic cancer risk among subjects exposed versus not exposed to proton pump inhibitors.Methods: The authors searched PubMed, EMBASE, Scopus, Cochrane Library, and clinicaltrials.gov to identify relevant studies. The authors quantified pancreatic cancer risk among subjects exposed versus not exposed to PPIs, expressed as the pooled (adjusted) odds ratio (OR/aOR) and 95% confidence interval (95%CI) in overall and sensitivity analyses.Results: One randomized trial, two cohort, four case-control, and five nested case-control studies with 700,178 subjects (73,985 cases; 626,193 controls) were retained. PPI exposure was associated with pancreatic cancer risk (OR = 1.75, 95%CI = 1.12-2.72, I2 = 99%); confirmed in sensitivity analyses for high-quality studies, observational studies, case-control studies, studies with pancreatic cancer as the primary outcome, and in sensitivity analyses for diabetes and obesity but not for pancreatitis and smoking. This association was independent of the duration and Defined Daily Dose of PPI exposure. Rabeprazole had a singular significant association with pancreatic cancer (OR = 5.40, 95%CI = 1.98-14.703, I2 = 87.9%).Conclusion: The class of PPIs is associated with a 1.75-fold increase in pancreatic cancer risk, confirmed in sensitivity analyses.
Collapse
Affiliation(s)
- Nasser Alkhushaym
- Center for Health Outcomes and PharmacoEconomic Research, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
- Department of Clinical Pharmacy, Royal Commission Health Services Program, Jubail, Saudi Arabia
| | - Abdulaali R Almutairi
- Center for Health Outcomes and PharmacoEconomic Research, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
- SFD-Drug sector, Saudi Food and Drug Authority, Riyadh, Saudi Arabia
| | - Abdulhamid Althagafi
- Clinical Pharmacy Department, College of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Saad B Fallatah
- Center for Health Outcomes and PharmacoEconomic Research, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
- Clinical and Hospital Pharmacy Department, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Mok Oh
- Center for Health Outcomes and PharmacoEconomic Research, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Jennifer R Martin
- Arizona Health Sciences Library, The University of Arizona, Tucson, AZ, USA
- Department of Pharmacy Practice and Science, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Hani M Babiker
- Department of Hematology & Oncology, College of Medicine, The University of Arizona, Tucson, AZ, USA
- Department of Pharmacy Practice and Science, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Ali McBride
- Department of Pharmacy Practice and Science, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
- Department of Pharmacy Practice and Science, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
- University of Arizona Cancer Center, Tucson, AZ, USA
| | - Ivo Abraham
- Center for Health Outcomes and PharmacoEconomic Research, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
- Department of Pharmacy Practice and Science, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
- Department of Pharmacy Practice and Science, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
- Department of Family and Community Medicine, College of Medicine, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
23
|
Abstract
OBJECTIVES Abnormalities of the main pancreatic duct may be an early indicator of pancreatic ductal adenocarcinoma (PDAC). We develop and validate algorithms that predict the risk of PDAC using features identified on cross-sectional imaging and other clinical characteristics collected through electronic medical records. METHODS Adult patients with abdominal computed tomography or magnetic resonance imaging in January 2006 to June 2016 demonstrating dilatation of main pancreatic duct were identified. Pancreas-related morphologic features were extracted from radiology reports using natural language processing. The cumulative incidence of PDAC with death as a competing risk was estimated using multistate models. Model discrimination was assessed using c-index. The models were internally validated using bootstrapping. RESULTS The cohort consisted of 7819 patients (mean age, 71 years; 65% female). A total of 781 patients (10%) developed PDAC within 3 years after the first eligible imaging study. The final models achieved reasonable discrimination (c-index, 0.825-0.833). The 3-year average risk of PDAC in the top 5% of the total eligible patients was 56.0%, more than 20 times of the average risk among the bottom 50% of patients. CONCLUSIONS Prediction models combining imaging features and clinical measures can be used to further stratify the risk of pancreatic cancer among patients with pancreas ductal dilatation.
Collapse
|
24
|
Blyuss O, Zaikin A, Cherepanova V, Munblit D, Kiseleva EM, Prytomanova OM, Duffy SW, Crnogorac-Jurcevic T. Development of PancRISK, a urine biomarker-based risk score for stratified screening of pancreatic cancer patients. Br J Cancer 2019; 122:692-696. [PMID: 31857725 PMCID: PMC7054390 DOI: 10.1038/s41416-019-0694-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 12/04/2019] [Indexed: 12/15/2022] Open
Abstract
Background An accurate and simple risk prediction model that would facilitate earlier detection of pancreatic adenocarcinoma (PDAC) is not available at present. In this study, we compare different algorithms of risk prediction in order to select the best one for constructing a biomarker-based risk score, PancRISK. Methods Three hundred and seventy-nine patients with available measurements of three urine biomarkers, (LYVE1, REG1B and TFF1) using retrospectively collected samples, as well as creatinine and age, were randomly split into training and validation sets, following stratification into cases (PDAC) and controls (healthy patients). Several machine learning algorithms were used, and their performance characteristics were compared. The latter included AUC (area under ROC curve) and sensitivity at clinically relevant specificity. Results None of the algorithms significantly outperformed all others. A logistic regression model, the easiest to interpret, was incorporated into a PancRISK score and subsequently evaluated on the whole data set. The PancRISK performance could be even further improved when CA19-9, commonly used PDAC biomarker, is added to the model. Conclusion PancRISK score enables easy interpretation of the biomarker panel data and is currently being tested to confirm that it can be used for stratification of patients at risk of developing pancreatic cancer completely non-invasively, using urine samples.
Collapse
Affiliation(s)
- Oleg Blyuss
- Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK. .,School of Physics, Astronomy and Mathematics, University of Hertfordshire, Hatfield, UK. .,Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child Health, Sechenov First Moscow State Medical University, Moscow, Russia.
| | - Alexey Zaikin
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child Health, Sechenov First Moscow State Medical University, Moscow, Russia.,Department of Mathematics and Institute for Women's Health, University College London, London, UK.,Department of Applied Mathematics, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Valeriia Cherepanova
- Department of Mathematics and Institute for Women's Health, University College London, London, UK
| | - Daniel Munblit
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child Health, Sechenov First Moscow State Medical University, Moscow, Russia.,Inflammation, Repair and Development Section, National Heart & Lung Institute, Imperial College London, London, UK
| | | | | | - Stephen W Duffy
- Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | | |
Collapse
|
25
|
Overbeek KA, Alblas M, Gausman V, Kandel P, Schweber AB, Brooks C, Van Riet PA, Wallace MB, Gonda TA, Cahen DL, Bruno MJ. Development of a stratification tool to identify pancreatic intraductal papillary mucinous neoplasms at lowest risk of progression. Aliment Pharmacol Ther 2019; 50:789-799. [PMID: 31429105 PMCID: PMC6772152 DOI: 10.1111/apt.15440] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/11/2019] [Accepted: 07/06/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Because most pancreatic intraductal papillary mucinous neoplasms (IPMNs) will never become malignant, currently advocated long-term surveillance is low-yield for most individuals. AIM To develop a score chart identifying IPMNs at lowest risk of developing worrisome features or high-risk stigmata. METHODS We combined prospectively maintained pancreatic cyst surveillance databases of three academic institutions. Patients were included if they had a presumed side-branch IPMN, without worrisome features or high-risk stigmata at baseline (as defined by the 2012 international Fukuoka guidelines), and were followed ≥ 12 months. The endpoint was development of one or more worrisome features or high-risk stigmata during follow-up. We created a multivariable prediction model using Cox-proportional logistic regression analysis and performed an internal-external validation. RESULTS 875 patients were included. After a mean follow-up of 50 months (range 12-157), 116 (13%) patients developed worrisome features or high-risk stigmata. The final model included cyst size (HR 1.12, 95% CI 1.09-1.15), cyst multifocality (HR 1.49, 95% CI 1.01-2.18), ever having smoked (HR 1.40, 95% CI 0.95-2.04), history of acute pancreatitis (HR 2.07, 95% CI 1.21-3.55), and history of extrapancreatic malignancy (HR 1.34, 95% CI 0.91-1.97). After validation, the model had good discriminative ability (C-statistic 0.72 in the Mayo cohort, 0.71 in the Columbia cohort, 0.64 in the Erasmus cohort). CONCLUSION In presumed side branch IPMNs without worrisome features or high-risk stigmata at baseline, the Dutch-American Risk stratification Tool (DART-1) successfully identifies pancreatic lesions at low risk of developing worrisome features or high-risk stigmata.
Collapse
Affiliation(s)
- Kasper A. Overbeek
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
| | - Maaike Alblas
- Department of Public HealthErasmus University Medical CenterRotterdamThe Netherlands
| | - Valerie Gausman
- Department of MedicineNYU – Langone Medical CenterNew YorkUSA
| | - Pujan Kandel
- Department of Gastroenterology and HepatologyMayo ClinicJacksonvilleUSA
| | - Adam B. Schweber
- Division of Digestive and Liver Diseases, Department of MedicineColumbia University Medical CenterNew YorkUSA
| | - Christian Brooks
- Division of Digestive and Liver Diseases, Department of MedicineColumbia University Medical CenterNew YorkUSA
| | - Priscilla A. Van Riet
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
| | | | - Tamas A. Gonda
- Division of Digestive and Liver Diseases, Department of MedicineColumbia University Medical CenterNew YorkUSA
| | - Djuna L. Cahen
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
| | - Marco J. Bruno
- Department of Gastroenterology & HepatologyErasmus University Medical CenterRotterdamThe Netherlands
| |
Collapse
|
26
|
Environmental Risk Factors of Pancreatic Cancer. J Clin Med 2019; 8:jcm8091427. [PMID: 31510046 PMCID: PMC6780233 DOI: 10.3390/jcm8091427] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/06/2019] [Accepted: 09/07/2019] [Indexed: 02/06/2023] Open
Abstract
Despite the advancement in medical knowledge that has improved the survival rate of many cancers, the survival rate of pancreatic cancer has remained dismal with a five-year survival rate of only 9%. The poor survival of pancreatic cancer emphasizes the urgent need to identify the causes or the risk factors of pancreatic cancer in order to establish effective preventive strategies. This review summarizes the current evidence regarding the environmental (non-genetic, including lifestyle, and clinical factors) risk factors of pancreatic cancer. Based on the current evidence, the established risk factors of pancreatic cancer are cigarette smoking, chronic diabetes, and obesity. Other strong risk factors include low consumption of fruits and vegetables, excess consumption of alcohol, poor oral hygiene, and the lack of allergy history. In the future, more studies are needed to identify additional risk factors of pancreatic cancer, especially the modifiable risk factors that could be included in a public health campaign to educate the public in order to reduce the incidence of pancreatic cancer.
Collapse
|
27
|
Baecker A, Kim S, Risch HA, Nuckols TK, Wu BU, Hendifar AE, Pandol SJ, Pisegna JR, Jeon CY. Do changes in health reveal the possibility of undiagnosed pancreatic cancer? Development of a risk-prediction model based on healthcare claims data. PLoS One 2019; 14:e0218580. [PMID: 31237889 PMCID: PMC6592596 DOI: 10.1371/journal.pone.0218580] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/04/2019] [Indexed: 12/22/2022] Open
Abstract
Background and objective Early detection methods for pancreatic cancer are lacking. We aimed to develop a prediction model for pancreatic cancer based on changes in health captured by healthcare claims data. Methods We conducted a case-control study on 29,646 Medicare-enrolled patients aged 68 years and above with pancreatic ductal adenocarcinoma (PDAC) reported to the Surveillance Epidemiology an End Results (SEER) tumor registries program in 2004–2011 and 88,938 age and sex-matched controls. We developed a prediction model using multivariable logistic regression on Medicare claims for 16 risk factors and pre-diagnostic symptoms of PDAC present within 15 months prior to PDAC diagnosis. Claims within 3 months of PDAC diagnosis were excluded in sensitivity analyses. We evaluated the discriminatory power of the model with the area under the receiver operating curve (AUC) and performed cross-validation by bootstrapping. Results The prediction model on all cases and controls reached AUC of 0.68. Excluding the final 3 months of claims lowered the AUC to 0.58. Among new-onset diabetes patients, the prediction model reached AUC of 0.73, which decreased to 0.63 when claims from the final 3 months were excluded. Performance measures of the prediction models was confirmed by internal validation using the bootstrap method. Conclusion Models based on healthcare claims for clinical risk factors, symptoms and signs of pancreatic cancer are limited in classifying those who go on to diagnosis of pancreatic cancer and those who do not, especially when excluding claims that immediately precede the diagnosis of PDAC.
Collapse
Affiliation(s)
- Aileen Baecker
- UCLA Fielding School of Public Health, Los Angeles, CA, United States of America
| | - Sungjin Kim
- Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Harvey A. Risch
- Yale School of Public Health, New Haven, CT, United States of America
| | - Teryl K. Nuckols
- Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Bechien U. Wu
- Kaiser Permanente Southern California, Research and Evaluation, Pasadena, CA, United States of America
| | | | - Stephen J. Pandol
- Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Joseph R. Pisegna
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States of America
| | - Christie Y. Jeon
- UCLA Fielding School of Public Health, Los Angeles, CA, United States of America
- Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States of America
- * E-mail:
| |
Collapse
|
28
|
Pang Y, Holmes MV, Chen Z, Kartsonaki C. A review of lifestyle, metabolic risk factors, and blood-based biomarkers for early diagnosis of pancreatic ductal adenocarcinoma. J Gastroenterol Hepatol 2019; 34:330-345. [PMID: 30550622 PMCID: PMC6378598 DOI: 10.1111/jgh.14576] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/09/2018] [Accepted: 12/12/2018] [Indexed: 12/28/2022]
Abstract
We aimed to review the epidemiologic literature examining lifestyle and metabolic risk factors, and blood-based biomarkers including multi-omics (genomics, proteomics, and metabolomics) and to discuss how these predictive markers can inform early diagnosis of pancreatic ductal adenocarcinoma (PDAC). A search of the PubMed database was conducted in June 2018 to review epidemiologic studies of (i) lifestyle and metabolic risk factors for PDAC, genome-wide association studies, and risk prediction models incorporating these factors and (ii) blood-based biomarkers for PDAC (conventional diagnostic markers, metabolomics, and proteomics). Prospective cohort studies have reported at least 20 possible risk factors for PDAC, including smoking, heavy alcohol drinking, adiposity, diabetes, and pancreatitis, but the relative risks and population attributable fractions of individual risk factors are small (mostly < 10%). High-throughput technologies have continued to yield promising genetic, metabolic, and protein biomarkers in addition to conventional biomarkers such as carbohydrate antigen 19-9. Nonetheless, most studies have utilized a hospital-based case-control design, and the diagnostic accuracy is low in studies that collected pre-diagnostic samples. Risk prediction models incorporating lifestyle and metabolic factors as well as other clinical parameters have shown good discrimination and calibration. Combination of traditional risk factors, genomics, and blood-based biomarkers can help identify high-risk populations and inform clinical decisions. Multi-omics investigations can provide valuable insights into disease etiology, but prospective cohort studies that collect pre-diagnostic samples and validation in independent studies are warranted.
Collapse
Affiliation(s)
- Yuanjie Pang
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Michael V Holmes
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- National Institute for Health Research Oxford Biomedical Research CentreOxford University HospitalOxfordUK
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Christiana Kartsonaki
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| |
Collapse
|
29
|
Setiawan VW, Stram DO, Porcel J, Chari ST, Maskarinec G, Le Marchand L, Wilkens LR, Haiman CA, Pandol SJ, Monroe KR. Pancreatic Cancer Following Incident Diabetes in African Americans and Latinos: The Multiethnic Cohort. J Natl Cancer Inst 2019; 111:27-33. [PMID: 29917105 PMCID: PMC6335114 DOI: 10.1093/jnci/djy090] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/08/2018] [Accepted: 04/17/2018] [Indexed: 12/18/2022] Open
Abstract
Background Diabetes has been proposed to be a risk factor for and a consequence of pancreatic cancer (PC). The relationship between recent-onset diabetes and PC is not well understood, and data in minorities are sparse. We examined the relationships between recent-onset diabetes and PC incidence in African Americans and Latinos in the Multiethnic Cohort. Methods A total of 48 995 African Americans and Latinos without prior diabetes and cancer at baseline (1993-1996) were included in the study. Questionnaires, Medicare data, and California hospital discharge files were used to identify new diabetes diagnoses. Cox regressions were used to calculate hazard ratios (HRs) and 95% confidence intervals (CIs) for cancer associated with diabetes and with diabetes duration. Results A total of 15 833 (32.3%) participants developed diabetes between baseline and 2013. A total of 408 incident PC cases were identified during follow-up. Diabetes was associated with PC (HRage75 = 2.39, 95% CI = 1.91 to 2.98). Individuals with recent-onset diabetes (within three or fewer years of PC diagnosis) had a greater risk compared with those with long-term diabetes across all ages. The HRage75 for recent-onset diabetes was 4.08 (95% CI = 2.76 to 6.03) in Latinos and 3.38 (95% CI = 2.30 to 4.98) in African Americans. Conclusions Diabetes was associated with a more than twofold higher risk of PC in African Americans and Latinos, but recent-onset diabetes was associated with a 2.3-fold greater increase in risk of PC than long-standing diabetes. Our findings support the hypothesis that recent-onset diabetes is a manifestation of PC and that long-standing diabetes is a risk factor for this malignancy.
Collapse
Affiliation(s)
- Veronica Wendy Setiawan
- Department of Preventive Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA
- Norris Comprehensive Cancer Center, Keck School of Medicine of University of Southern California, Los Angeles, CA
| | - Daniel O Stram
- Department of Preventive Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA
| | - Jacqueline Porcel
- Norris Comprehensive Cancer Center, Keck School of Medicine of University of Southern California, Los Angeles, CA
| | - Suresh T Chari
- Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, MN
| | | | - Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI
| | - Lynne R Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA
- Norris Comprehensive Cancer Center, Keck School of Medicine of University of Southern California, Los Angeles, CA
| | - Stephen J Pandol
- Division of Gastroenterology, Departments of Medicine, Cedars-Sinai Medical Center and Department of Veterans Affairs, Los Angeles, CA
| | - Kristine R Monroe
- Department of Preventive Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA
| |
Collapse
|
30
|
Hwang IC, Chang J, Park SM. Association between proton pump inhibitor use and the risk of pancreatic cancer: A Korean nationwide cohort study. PLoS One 2018; 13:e0203918. [PMID: 30208110 PMCID: PMC6135510 DOI: 10.1371/journal.pone.0203918] [Citation(s) in RCA: 21] [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: 07/18/2017] [Accepted: 08/30/2018] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Proton pump inhibitor (PPI) therapy causes hypergastrinemia, which could promote the development and progression of pancreatic cancer. Accordingly, this study aimed to investigate the association between PPI exposure and the risk of pancreatic cancer. METHODS We conducted a twelve-year longitudinal population-based study (2002-2013) using the Korean National Health Insurance Corporation claims database merged with national health examination data. The study cohort included 453,655 cancer-free individuals in January 2007 (index date). Incident pancreatic cancer was assessed throughout follow up until December 2013. The exposure to PPIs before the index date was assessed using a standardized Defined Daily Dose (DDD) system. We calculated the hazard ratios (HRs) and their 95% confidence intervals (CIs) for pancreatic cancer risk associated with cumulative PPI use using Cox proportional hazard regression models. RESULTS There were 3,086 cases of pancreatic cancer during the period of 2,920,000 person-years. PPI users exceeding 60 DDDs were at a higher risk of pancreatic cancer compared with non-users (HR, 1.34; 95% CI, 1.04-1.72). Subgroup analyses revealed that a significant association existed between PPI use and pancreatic cancer in low risk groups including individuals who were female, engaged in healthy lifestyle habits, and had no history of diabetes or chronic pancreatitis. CONCLUSION Exposure to PPI appears to increase the risk of pancreatic cancer, independent of conventional risk factors.
Collapse
Affiliation(s)
- In Cheol Hwang
- Department of Family Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Jooyoung Chang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Sang Min Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
- Department of Family Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
31
|
Nakatochi M, Lin Y, Ito H, Hara K, Kinoshita F, Kobayashi Y, Ishii H, Ozaka M, Sasaki T, Sasahira N, Morimoto M, Kobayashi S, Ueno M, Ohkawa S, Egawa N, Kuruma S, Mori M, Nakao H, Wang C, Nishiyama T, Kawaguchi T, Takahashi M, Matsuda F, Kikuchi S, Matsuo K. Prediction model for pancreatic cancer risk in the general Japanese population. PLoS One 2018; 13:e0203386. [PMID: 30192808 PMCID: PMC6128543 DOI: 10.1371/journal.pone.0203386] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/20/2018] [Indexed: 12/17/2022] Open
Abstract
Genome-wide association studies (GWASs) have identified many single nucleotide polymorphisms (SNPs) that are significantly associated with pancreatic cancer susceptibility. We sought to replicate the associations of 61 GWAS-identified SNPs at 42 loci with pancreatic cancer in Japanese and to develop a risk model for the identification of individuals at high risk for pancreatic cancer development in the general Japanese population. The model was based on data including directly determined or imputed SNP genotypes for 664 pancreatic cancer case and 664 age- and sex-matched control subjects. Stepwise logistic regression uncovered five GWAS-identified SNPs at five loci that also showed significant associations in our case-control cohort. These five SNPs were included in the risk model and also applied to calculation of the polygenic risk score (PRS). The area under the curve determined with the leave-one-out cross-validation method was 0.63 (95% confidence interval, 0.60–0.66) or 0.61 (0.58–0.64) for versions of the model that did or did not include cigarette smoking and family history of pancreatic cancer in addition to the five SNPs, respectively. Individuals in the lowest and highest quintiles for the PRS had odds ratios of 0.62 (0.42–0.91) and 1.98 (1.42–2.76), respectively, for pancreatic cancer development compared with those in the middle quintile. We have thus developed a risk model for pancreatic cancer that showed moderately good discriminatory ability with regard to differentiation of pancreatic cancer patients from control individuals. Our findings suggest the potential utility of a risk model that incorporates replicated GWAS-identified SNPs and established demographic or environmental factors for the identification of individuals at increased risk for pancreatic cancer development.
Collapse
Affiliation(s)
- Masahiro Nakatochi
- Division of Data Science, Data Coordinating Center, Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Yingsong Lin
- Department of Public Health, Aichi Medical University School of Medicine, Nagakute, Japan
- * E-mail:
| | - Hidemi Ito
- Division of Cancer Information and Control, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Kazuo Hara
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Fumie Kinoshita
- Division of Data Science, Data Coordinating Center, Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Yumiko Kobayashi
- Division of Data Science, Data Coordinating Center, Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Hiroshi Ishii
- Clinical Research Center, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Masato Ozaka
- Department of Hepato-biliary-pancreatic Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takashi Sasaki
- Department of Hepato-biliary-pancreatic Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Naoki Sasahira
- Department of Hepato-biliary-pancreatic Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Manabu Morimoto
- Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center Hospital, Kanagawa, Japan
| | - Satoshi Kobayashi
- Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center Hospital, Kanagawa, Japan
| | - Makoto Ueno
- Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center Hospital, Kanagawa, Japan
| | - Shinichi Ohkawa
- Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center Hospital, Kanagawa, Japan
| | - Naoto Egawa
- Tokyo Metropolitan Hiroo Hospital, Tokyo, Japan
| | - Sawako Kuruma
- Department of Internal Medicine, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | - Mitsuru Mori
- Hokkaido Chitose College of Rehabilitation, Hokkaido, Japan
| | - Haruhisa Nakao
- Division of Hepatology and Pancreatology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Chaochen Wang
- Department of Public Health, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Takeshi Nishiyama
- Department of Public Health, Nagoya City University Graduate School of Medicine, Nagoya, Japan
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Meiko Takahashi
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shogo Kikuchi
- Department of Public Health, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| |
Collapse
|
32
|
Sánchez-García J, Candanedo-González F, Félix-Félix AK, Sánchez-Ramírez D, Medrano-Guzmán R, Quintana-Quintana M, Baas-Cabrera YB, Flores-Figueroa E. Retrospective cohort of pancreatic and Vater ampullary adenocarcinoma from a reference center in Mexico. Ann Med Surg (Lond) 2018; 30:7-12. [PMID: 29707208 PMCID: PMC5918165 DOI: 10.1016/j.amsu.2018.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 03/25/2018] [Accepted: 04/03/2018] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) and ampulla of Vater adenocarcinomas (AVAC) are periampullary tumors. These tumors have overlapping symptoms and a common treatment, but present differences in their survival and biology. No recent studies in Mexico have been published that describe the clinicopathological characteristics of these tumors. Therefore, the aim of this study was to describe the clinicopathological characteristics of PDAC and AVAC in patients at a reference center in Mexico. METHODS A retrospective cohort of patients with PDAC or AVAC was analyzed at our institution (July 2007 to June 2016). Inferential analysis of the clinical data was performed with Student's t-test or a χ2 test with odds ratios (OR) and confidence intervals (CI), depending on the variables. Overall survival was compared using Kaplan-Meier curves with log-rank p values. RESULTS Forty patients with PDAC and 76 with AVAC were analyzed, including 77 females and 39 males with a mean age of 60.6 years and a mean evolution time of 5.7 months. PDAC patients had more abdominal pain, a larger tumor size and more advanced stages than AVAC patients. In contrast, AVAC patients had more jaundice, a higher percentage of complete resections and higher overall survival. Up to 70% of patients were overweight. PDAC cohort included a higher proportion of smokers. CONCLUSIONS Our cohort was slightly younger, had a larger percentage of females, and a greater percentage of obese patients than those in many international reports. A high proportion of PDAC patients are diagnosed in advanced stages and have a low likelihood of resectability.
Collapse
Affiliation(s)
| | | | | | | | | | - Miguel Quintana-Quintana
- Medical Oncology Service, Oncology Hospital, National Medical Center Century XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Yair Benjamín Baas-Cabrera
- Medical Oncology Service, Oncology Hospital, National Medical Center Century XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | | |
Collapse
|
33
|
Peres LC, Risch H, Terry KL, Webb PM, Goodman MT, Wu AH, Alberg AJ, Bandera EV, Barnholtz-Sloan J, Bondy ML, Cote ML, Funkhouser E, Moorman PG, Peters ES, Schwartz AG, Terry PD, Manichaikul A, Abbott SE, Camacho F, Jordan SJ, Nagle CM, Rossing MA, Doherty JA, Modugno F, Moysich K, Ness R, Berchuck A, Cook L, Le N, Brooks-Wilson A, Sieh W, Whittemore A, McGuire V, Rothstein J, Anton-Culver H, Ziogas A, Pearce CL, Tseng C, Pike M, Schildkraut JM. Racial/ethnic differences in the epidemiology of ovarian cancer: a pooled analysis of 12 case-control studies. Int J Epidemiol 2018; 47:460-472. [PMID: 29211900 PMCID: PMC5913601 DOI: 10.1093/ije/dyx252] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 11/02/2017] [Accepted: 11/09/2017] [Indexed: 12/25/2022] Open
Abstract
Background Ovarian cancer incidence differs substantially by race/ethnicity, but the reasons for this are not well understood. Data were pooled from the African American Cancer Epidemiology Study (AACES) and 11 case-control studies in the Ovarian Cancer Association Consortium (OCAC) to examine racial/ethnic differences in epidemiological characteristics with suspected involvement in epithelial ovarian cancer (EOC) aetiology. Methods We used multivariable logistic regression to estimate associations for 17 reproductive, hormonal and lifestyle characteristics and EOC risk by race/ethnicity among 10 924 women with invasive EOC (8918 Non-Hispanic Whites, 433 Hispanics, 911 Blacks, 662 Asian/Pacific Islanders) and 16 150 controls (13 619 Non-Hispanic Whites, 533 Hispanics, 1233 Blacks, 765 Asian/Pacific Islanders). Likelihood ratio tests were used to evaluate heterogeneity in the risk factor associations by race/ethnicity. Results We observed statistically significant racial/ethnic heterogeneity for hysterectomy and EOC risk (P = 0.008), where the largest odds ratio (OR) was observed in Black women [OR = 1.64, 95% confidence interval (CI) = 1.34-2.02] compared with other racial/ethnic groups. Although not statistically significant, the associations for parity, first-degree family history of ovarian or breast cancer, and endometriosis varied by race/ethnicity. Asian/Pacific Islanders had the greatest magnitude of association for parity (≥3 births: OR = 0.38, 95% CI = 0.28-0.54), and Black women had the largest ORs for family history (OR = 1.77, 95% CI = 1.42-2.21) and endometriosis (OR = 2.42, 95% CI = 1.65-3.55). Conclusions Although racial/ethnic heterogeneity was observed for hysterectomy, our findings support the validity of EOC risk factors across all racial/ethnic groups, and further suggest that any racial/ethnic population with a higher prevalence of a modifiable risk factor should be targeted to disseminate information about prevention.
Collapse
Affiliation(s)
- Lauren C Peres
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Harvey Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Kathryn L Terry
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Penelope M Webb
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Marc T Goodman
- Samuel Oschin Comprehensive Cancer Institute
- Community and Population Health Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anna H Wu
- Department of Preventive Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Anthony J Alberg
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Elisa V Bandera
- Cancer Prevention and Control Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Jill Barnholtz-Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Melissa L Bondy
- Cancer Prevention and Population Sciences Program, Baylor College of Medicine, Houston, TX, USA
| | - Michele L Cote
- Karmanos Cancer Institute Population Studies and Disparities Research Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ellen Funkhouser
- Division of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Patricia G Moorman
- Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA
| | - Edward S Peters
- Department of Epidemiology, Louisiana State University Health Sciences Center School of Public Health, New Orleans, LA, USA
| | - Ann G Schwartz
- Karmanos Cancer Institute Population Studies and Disparities Research Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Paul D Terry
- Graduate School of Medicine, University of Tennessee Medical Center, Knoxville, TN, USA
| | - Ani Manichaikul
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Sarah E Abbott
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Fabian Camacho
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Susan J Jordan
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Christina M Nagle
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | - Mary Anne Rossing
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | | | - Francesmary Modugno
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
- Ovarian Cancer Center of Excellence, Magee-Womens Research Institute and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Kirsten Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Roberta Ness
- University of Texas School of Public Health, Houston, TX, USA
| | - Andrew Berchuck
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Linda Cook
- Division of Epidemiology and Biostatistics, University of New Mexico, Albuquerque, NM, USA
| | | | - Angela Brooks-Wilson
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Weiva Sieh
- Department of Population Health Science and Policy and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alice Whittemore
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, CA, USA
| | - Valerie McGuire
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph Rothstein
- Department of Population Health Science and Policy and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hoda Anton-Culver
- Department of Epidemiology
- Genetic Epidemiology Research Institute, University of California Irvine, Irvine, CA, USA
| | | | - Celeste L Pearce
- Department of Preventive Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Chiuchen Tseng
- Department of Preventive Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Malcom Pike
- Department of Preventive Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Joellen M Schildkraut
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | | |
Collapse
|
34
|
Dasic G, Jones T, Frajzyngier V, Rojo R, Madsen A, Valdez H. Safety signal detection and evaluation in clinical development programs: A case study of tofacitinib. Pharmacol Res Perspect 2018; 6. [PMID: 29417755 PMCID: PMC5817838 DOI: 10.1002/prp2.371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 09/11/2017] [Indexed: 12/30/2022] Open
Abstract
Adverse events are anticipated during a clinical development program. Tofacitinib is an oral Janus kinase inhibitor for the treatment of rheumatoid arthritis (RA). We describe here the process undertaken by Pfizer to investigate a safety signal for pancreatic cancer with tofacitinib. Potential cases of pancreatic cancer across indications from Pfizer's clinical trials and safety databases were identified and underwent in‐depth case review and external expert consultation. The magnitude of the signal was quantified. The feasibility of formal signal evaluation via a hypothesis‐testing study was explored. As of July 2016, 14 cases of potential pancreatic cancer were identified: eight cases in clinical development trials (psoriasis n = 6; RA n = 1; psoriatic arthritis n = 1), four cases in a postmarketing study in RA patients in Japan, and two spontaneous reports. Incidence rates (95% confidence intervals) per 100 patient‐years ranged from 0 (0, 0.02) to 0.14 in RA, 0.05 (0.01, 0.15) to 0.07 (0.02, 0.16) in psoriasis, and 0.25 (0.01, 1.37) in psoriatic arthritis. The majority of patients had established risk factors for pancreatic cancer. The pharmaceutical industry's rapid and transparent response to safety signals is essential for ensuring patient safety and enabling physicians and patients to adequately assess a drug's risk:benefit. Safety signals emerging through pharmacovigilance may be true or false indicators of a causative association with drug exposure. In this example, it was determined that tofacitinib exposure was unlikely to be related to induction and promotion of pancreatic cancer; however, a relationship with pancreatic cancer promotion could not be excluded.
Collapse
|
35
|
Liu G, Mukherjee B, Lee S, Lee AW, Wu AH, Bandera EV, Jensen A, Rossing MA, Moysich KB, Chang-Claude J, Doherty JA, Gentry-Maharaj A, Kiemeney L, Gayther SA, Modugno F, Massuger L, Goode EL, Fridley BL, Terry KL, Cramer DW, Ramus SJ, Anton-Culver H, Ziogas A, Tyrer JP, Schildkraut JM, Kjaer SK, Webb PM, Ness RB, Menon U, Berchuck A, Pharoah PD, Risch H, Pearce CL. Robust Tests for Additive Gene-Environment Interaction in Case-Control Studies Using Gene-Environment Independence. Am J Epidemiol 2018; 187:366-377. [PMID: 28633381 PMCID: PMC5860584 DOI: 10.1093/aje/kwx243] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 05/24/2017] [Accepted: 06/02/2017] [Indexed: 12/20/2022] Open
Abstract
There have been recent proposals advocating the use of additive gene-environment interaction instead of the widely used multiplicative scale, as a more relevant public health measure. Using gene-environment independence enhances statistical power for testing multiplicative interaction in case-control studies. However, under departure from this assumption, substantial bias in the estimates and inflated type I error in the corresponding tests can occur. In this paper, we extend the empirical Bayes (EB) approach previously developed for multiplicative interaction, which trades off between bias and efficiency in a data-adaptive way, to the additive scale. An EB estimator of the relative excess risk due to interaction is derived, and the corresponding Wald test is proposed with a general regression setting under a retrospective likelihood framework. We study the impact of gene-environment association on the resultant test with case-control data. Our simulation studies suggest that the EB approach uses the gene-environment independence assumption in a data-adaptive way and provides a gain in power compared with the standard logistic regression analysis and better control of type I error when compared with the analysis assuming gene-environment independence. We illustrate the methods with data from the Ovarian Cancer Association Consortium.
Collapse
Affiliation(s)
- Gang Liu
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Bhramar Mukherjee
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Seunggeun Lee
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Alice W Lee
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Anna H Wu
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Elisa V Bandera
- Cancer Prevention and Control Research Program, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Allan Jensen
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Mary Anne Rossing
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington
| | - Kirsten B Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
- University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jennifer A Doherty
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, New Hampshire
| | - Aleksandra Gentry-Maharaj
- Gynaecological Cancer Research Centre, Women’s Cancer, Institute for Women’s Health, University College London, London, United Kingdom
| | - Lambertus Kiemeney
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, the Netherlands
| | - Simon A Gayther
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Francesmary Modugno
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Gynecologic Oncology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
- Womens Cancer Research Program, Magee-Womens Research Institute and University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Leon Massuger
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ellen L Goode
- Department of Health Sciences Research, Division of Epidemiology, Mayo Clinic, Rochester, Minnesota
| | | | - Kathryn L Terry
- Obstetrics and Gynecology Center, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Daniel W Cramer
- Obstetrics and Gynecology Center, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Susan J Ramus
- School of Women’s and Children’s Health, University of New South Wales, Sydney, New South Wales, Australia
- Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Hoda Anton-Culver
- Genetic Epidemiology Research Institute, Center for Cancer Genetics Research and Prevention, School of Medicine, University of California, Irvine, Irvine, California
| | - Argyrios Ziogas
- Genetic Epidemiology Research Institute, Center for Cancer Genetics Research and Prevention, School of Medicine, University of California, Irvine, Irvine, California
| | - Jonathan P Tyrer
- Strangeways Research Laboratory, Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Joellen M Schildkraut
- Department of Public Health Sciences, School of Medicine, University of Virginia, Charlottesville, Virginia
| | - Susanne K Kjaer
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Gynecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Penelope M Webb
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Roberta B Ness
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas, Houston, Texas
| | - Usha Menon
- Gynaecological Cancer Research Centre, Women’s Cancer, Institute for Women’s Health, University College London, London, United Kingdom
| | - Andrew Berchuck
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina
| | - Paul D Pharoah
- Strangeways Research Laboratory, Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Oncology, Center for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, United Kingdom
| | - Harvey Risch
- Department of Chronic Disease Epidemiology, School of Public Health, Yale University, New Haven, Connecticut
| | - Celeste Leigh Pearce
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|
36
|
Coté GA, Xu H, Easler JJ, Imler TD, Teal E, Sherman S, Korc M. Informative Patterns of Health-Care Utilization Prior to the Diagnosis of Pancreatic Ductal Adenocarcinoma. Am J Epidemiol 2017; 186:944-951. [PMID: 28541521 PMCID: PMC5860250 DOI: 10.1093/aje/kwx168] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/08/2016] [Accepted: 12/09/2016] [Indexed: 12/15/2022] Open
Abstract
Early-detection tests for pancreatic ductal adenocarcinoma (PDAC) are needed. Since a hypothetical screening test would be applied during antecedent clinical encounters, we sought to define the variability in health-care utilization leading up to PDAC diagnosis. This was a retrospective cohort study that included patients diagnosed with PDAC in the Indianapolis, Indiana, area between 1999 and 2013 with at least 1 health-care encounter during the antecedent 36-month period (n = 1,023). Patients were classified by unique patterns of health-care utilization using a group-based trajectory model. The prevalences of PDAC signals, such as diabetes mellitus (DM) and chronic pancreatitis, were compared. Four distinct trajectories were identified, the most common (42.0%) being having few clinical encounters more than 6 months prior to PDAC diagnosis (late acceleration). In all cases, a minority of persons had DM (30.6%, with 9.5% <1.5 years before PDAC) or any pancreatic disorder (39.9%); these were least common in the late-acceleration group (DM, 14.7%; any pancreatic disorder, 32.1% (P < 0.001)). The most common pattern of antecedent care was having few clinical encounters until shortly before PDAC diagnosis. Since the majority of patients diagnosed with PDAC do not have an antecedent PDAC signal, early-detection strategies limited to these groups may not apply to the majority of cases.
Collapse
Affiliation(s)
- Gregory A Coté
- Correspondence to Dr. Gregory A. Coté, Department of Medicine, Medical University of South Carolina, 114 Doughty Street, MSC 702, Suite 249, Charleston, SC 29425 (e-mail: )
| | | | | | | | | | | | | |
Collapse
|
37
|
Streicher SA, Klein AP, Olson SH, Amundadottir LT, DeWan AT, Zhao H, Risch HA. Impact of Sixteen Established Pancreatic Cancer Susceptibility Loci in American Jews. Cancer Epidemiol Biomarkers Prev 2017; 26:1540-1548. [PMID: 28754795 PMCID: PMC5626623 DOI: 10.1158/1055-9965.epi-17-0262] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/19/2017] [Accepted: 07/20/2017] [Indexed: 01/04/2023] Open
Abstract
Background: The higher risk of pancreatic cancer in Ashkenazi Jews compared with non-Jews is only partially explained by the increased frequency of BRCA1 and BRCA2 mutations in Ashkenazi Jews.Methods: We evaluated the impact of 16 established pancreatic cancer susceptibility loci in a case-control sample of American Jews, largely Ashkenazi, including 406 full-Jewish pancreatic cancer patients and 2,332 full-Jewish controls, genotyped as part of the Pancreatic Cancer Cohort and Case-Control Consortium I/II (PanScan I/II), Pancreatic Cancer Case-Control Consortium (PanC4), and Resource for Genetic Epidemiology Research on Adult Health and Aging (GERA) datasets. We compared risk in full-Jewish subjects with risk in part-Jewish; non-Jewish Southern European; and in the combined non-Jewish Eastern, Northern, Southern, and Western European (non-Jewish white European) subjects from the same datasets. Jewish ancestries were genetically identified using seeded Fast principal component analysis. Data were analyzed by unconditional logistic regression, and adjusted for age, sex, and principal components.Results: One SNP on chromosome 13q22.1 (rs9543325; OR, 1.36; 95% confidence interval, 1.16-1.58; P = 10-4.1) was significant in full-Jews. Individual ORs and minor allele frequencies were similar between Jewish and non-Jewish white European subjects. The average ORs across the 16 pancreatic cancer susceptibility loci for full-Jewish, full- plus part-Jewish, non-Jewish Southern European, and non-Jewish white European subjects were 1.25, 1.30, 1.31, and 1.26, respectively.Conclusions: The 16 pancreatic cancer susceptibility loci similarly impact Jewish and non-Jewish white European subjects, both individually and as summary odds.Impact: These 16 pancreatic cancer susceptibility loci likely do not explain the higher risk seen in Ashkenazi Jews. Cancer Epidemiol Biomarkers Prev; 26(10); 1540-8. ©2017 AACR.
Collapse
Affiliation(s)
- Samantha A Streicher
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - Alison P Klein
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Sara H Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Andrew T DeWan
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut.
| |
Collapse
|
38
|
Duell EJ, Lujan-Barroso L, Sala N, McElyea SD, Overvad K, Tjonneland A, Olsen A, Weiderpass E, Busund LT, Moi L, Muller D, Vineis P, Aune D, Matullo G, Naccarati A, Panico S, Tagliabue G, Tumino R, Palli D, Kaaks R, Katzke VA, Boeing H, Bueno-de-Mesquita H, Peeters PH, Trichopoulou A, Lagiou P, Kotanidou A, Travis RC, Wareham N, Khaw KT, Quiros JR, Rodríguez-Barranco M, Dorronsoro M, Chirlaque MD, Ardanaz E, Severi G, Boutron-Ruault MC, Rebours V, Brennan P, Gunter M, Scelo G, Cote G, Sherman S, Korc M. Plasma microRNAs as biomarkers of pancreatic cancer risk in a prospective cohort study. Int J Cancer 2017; 141:905-915. [PMID: 28542740 PMCID: PMC5536971 DOI: 10.1002/ijc.30790] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/27/2017] [Accepted: 04/19/2017] [Indexed: 02/06/2023]
Abstract
Noninvasive biomarkers for early pancreatic ductal adenocarcinoma (PDAC) diagnosis and disease risk stratification are greatly needed. We conducted a nested case-control study within the Prospective Investigation into Cancer and Nutrition (EPIC) cohort to evaluate prediagnostic microRNAs (miRs) as biomarkers of subsequent PDAC risk. A panel of eight miRs (miR-10a, -10b, -21-3p, -21-5p, -30c, -106b, -155 and -212) based on previous evidence from our group was evaluated in 225 microscopically confirmed PDAC cases and 225 controls matched on center, sex, fasting status and age/date/time of blood collection. MiR levels in prediagnostic plasma samples were determined by quantitative RT-PCR. Logistic regression was used to model levels and PDAC risk, adjusting for covariates and to estimate area under the receiver operating characteristic curves (AUC). Plasma miR-10b, -21-5p, -30c and -106b levels were significantly higher in cases diagnosed within 2 years of blood collection compared to matched controls (all p-values <0.04). Based on adjusted logistic regression models, levels for six miRs (miR-10a, -10b, -21-5p, -30c, -155 and -212) overall, and for four miRs (-10a, -10b, -21-5p and -30c) at shorter follow-up time between blood collection and diagnosis (≤5 yr, ≤2 yr), were statistically significantly associated with risk. A score based on the panel showed a linear dose-response trend with risk (p-value = 0.0006). For shorter follow-up (≤5 yr), AUC for the score was 0.73, and for individual miRs ranged from 0.73 (miR-212) to 0.79 (miR-21-5p).
Collapse
Affiliation(s)
- Eric J. Duell
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | - Leila Lujan-Barroso
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | - Núria Sala
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | - Samantha Deitz McElyea
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Kim Overvad
- Aarhus University, Department of Public Health, Section for Epidemiology, Aarhus C, Denmark
| | | | - Anja Olsen
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Elisabete Weiderpass
- Department of Research, Cancer Registry of Norway, Institute of Population-Based Cancer Research, Oslo, Norway
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Community Medicine, University of Tromsø, The Arctic University of Norway, Tromsø, Norway
- Genetic Epidemiology Group, Folkhälsan Research Center, Helsinki, Finland
| | - Lill-Tove Busund
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Line Moi
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - David Muller
- School of Public Health, Epidemiology & Biostatistics, Imperial College London, London, United Kingdom
| | - Paolo Vineis
- School of Public Health, Epidemiology & Biostatistics, Imperial College London, London, United Kingdom
| | - Dagfinn Aune
- School of Public Health, Epidemiology & Biostatistics, Imperial College London, London, United Kingdom
| | - Giuseppe Matullo
- Human Genetics Foundation (HuGeF), Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Salvatore Panico
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Giovanna Tagliabue
- Lombardy Cancer Registry Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Unit, "Civic - M.P. Arezzo" Hospital, ASP Ragusa, Italy
| | - Domenico Palli
- Cancer Risk Factors and Life-Style Epidemiology Unit, Cancer Research and Prevention Institute – ISPO, Florence- Italy
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Verena A. Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany
| | - H.B(as) Bueno-de-Mesquita
- Dt. for Determinants of Chronic Diseases (DCD), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Dt. of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, United Kingdom
- Dt. of Social & Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Petra H. Peeters
- Dept of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
- MRC-PHE Centre for Environment and Health, Dept of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, UK
| | - Antonia Trichopoulou
- Hellenic Health Foundation, Athens, Greece
- WHO Collaborating Center for Nutrition and Health, Unit of Nutritional Epidemiology and Nutrition in Public Health, Dept. of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Pagona Lagiou
- Hellenic Health Foundation, Athens, Greece
- WHO Collaborating Center for Nutrition and Health, Unit of Nutritional Epidemiology and Nutrition in Public Health, Dept. of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Greece
- Department of Epidemiology, Harvard School of Public Health, Boston, USA
| | - Anastasia Kotanidou
- Hellenic Health Foundation, Athens, Greece
- Department of Critical Care Medicine & Pulmonary Services, University of Athens Medical School, Evangelismos Hospital, Athens, Greece
| | - Ruth C. Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Nick Wareham
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Kay-Tee Khaw
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | | | - Miguel Rodríguez-Barranco
- Andalusian School of Public Health, Research Insititute Biosanitary Granada, University Hospital Granada/University of Granada, Granada
- CIBER Epidemiology and Public Health (CIBERESP), Madrid
| | - Miren Dorronsoro
- CIBER Epidemiology and Public Health (CIBERESP), Madrid
- Basque Regional Health Department, San Sebatian, Spain
| | - María-Dolores Chirlaque
- CIBER Epidemiology and Public Health (CIBERESP), Madrid
- Department of Epidemiology, Murcia Regional Health Authority, Murcia, Spain
| | - Eva Ardanaz
- CIBER Epidemiology and Public Health (CIBERESP), Madrid
- Navarra Public Health Institute, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Gianluca Severi
- Université Paris-Saclay, Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
- Gustave Roussy, Villejuif, France
| | | | - Vinciane Rebours
- Pancreatology Unit, Beaujon Hospital, Clichy, France
- INSERM, University Paris, France
| | - Paul Brennan
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Marc Gunter
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Ghislaine Scelo
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Greg Cote
- Medical University of South Carolina, Charleston, USA
| | - Stuart Sherman
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Murray Korc
- Departments of Medicine and Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, USA
- Pancreatic Cancer Signature Center, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, USA
| |
Collapse
|
39
|
Ouyang H, Ma W, Liu F, Yue Z, Fang M, Quan M, Pan Z. Factors influencing survival of patients with pancreatic adenocarcinoma and synchronous liver metastases receiving palliative care. Pancreatology 2017; 17:773-781. [PMID: 28734721 DOI: 10.1016/j.pan.2017.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 07/07/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Patients with pancreatic ductal adenocarcinoma and synchronous liver metastases (PACLM) have an extremely limited life expectancy. We performed a single-center analysis to explore the clinical results and prognostic factors of patients with PACLM receiving palliative care. METHODS We retrospectively reviewed 189 patients undergoing palliative care at Tianjin Medical University Cancer Hospital over a 15-year period. Clinical characteristics, survival condition, and factors associated with survival were analyzed. Treatment methods included palliative bypass surgery, percutaneous transhepatic cholangiodrainage, drug analgesia, symptomatic treatment, and other nutritional or supportive measures. RESULTS The overall survival (OS) was 3.6 months for all patients. Multivariate analysis for clinical features showed that Karnofsky performance score (KPS), ascites, cigarette smoking, primary tumor size, and lactate dehydrogenase (LDH) were prognostic variables with statistical significance (P < 0.05). The patients were classified into three groups of patients according to how many of these 5 risk factors were present: 0-1, 2, or 3-5 risk factors. The median OS of the 3 groups of patients were 5.0, 3.3, and 2.5 months, respectively, with a notable statistical significance (P < 0.0001). CONCLUSIONS KPS<80, ascites, cigarette smoking, primary tumor size≥5 cm, and LDH≥250U/L are effective predictive factors of poor prognosis for patients with PACLM. The stratification of treatment outcome groups based on these factors facilitates evaluation of individual prognosis and can guide clinical decisions.
Collapse
Affiliation(s)
- Huaqiang Ouyang
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Weidong Ma
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China; Department of Pancreatic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Fang Liu
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China; Department of Interventional Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhensong Yue
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Minghui Fang
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Manman Quan
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhanyu Pan
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
40
|
Coté GA. The Countdown to a Paradigm Shift in Diagnosing Pancreatic Ductal Adenocarcinoma. Clin Gastroenterol Hepatol 2017; 15:1000-1002. [PMID: 28300695 PMCID: PMC5474177 DOI: 10.1016/j.cgh.2017.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/07/2017] [Accepted: 03/08/2017] [Indexed: 02/07/2023]
Affiliation(s)
- Gregory A. Coté
- Division of Gastroenterology & Hepatology, Department of Medicine, Medical University of South Carolina, Charleston, USA
| |
Collapse
|
41
|
Chase HS, Mitrani LR, Lu GG, Fulgieri DJ. Early recognition of multiple sclerosis using natural language processing of the electronic health record. BMC Med Inform Decis Mak 2017; 17:24. [PMID: 28241760 PMCID: PMC5329909 DOI: 10.1186/s12911-017-0418-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 02/10/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Diagnostic accuracy might be improved by algorithms that searched patients' clinical notes in the electronic health record (EHR) for signs and symptoms of diseases such as multiple sclerosis (MS). The focus this study was to determine if patients with MS could be identified from their clinical notes prior to the initial recognition by their healthcare providers. METHODS An MS-enriched cohort of patients with well-established MS (n = 165) and controls (n = 545), was generated from the adult outpatient clinic. A random sample cohort was generated from randomly selected patients (n = 2289) from the same adult outpatient clinic, some of whom had MS (n = 16). Patients' notes were extracted from the data warehouse and signs and symptoms mapped to UMLS terms using MedLEE. Approximately 1000 MS-related terms occurred significantly more frequently in MS patients' notes than controls'. Synonymous terms were manually clustered into 50 buckets and used as classification features. Patients were classified as MS or not using Naïve Bayes classification. RESULTS Classification of patients known to have MS using notes of the MS-enriched cohort entered after the initial ICD9[MS] code yielded an ROC AUC, sensitivity, and specificity of 0.90 [0.87-0.93], 0.75[0.66-0.82], and 0.91 [0.87-0.93], respectively. Similar classification accuracy was achieved using the notes from the random sample cohort. Classification of patients not yet known to have MS using notes of the MS-enriched cohort entered before the initial ICD9[MS] documentation identified 40% [23-59%] as having MS. Manual review of the EHR of 45 patients of the random sample cohort classified as having MS but lacking an ICD9[MS] code identified four who might have unrecognized MS. CONCLUSIONS Diagnostic accuracy might be improved by mining patients' clinical notes for signs and symptoms of specific diseases using NLP. Using this approach, we identified patients with MS early in the course of their disease which could potentially shorten the time to diagnosis. This approach could also be applied to other diseases often missed by primary care providers such as cancer. Whether implementing computerized diagnostic support ultimately shortens the time from earliest symptoms to formal recognition of the disease remains to be seen.
Collapse
Affiliation(s)
- Herbert S Chase
- Department of Biomedical Informatics, Columbia University Medical Center, PH-20, 622 West 168th street, New York, NY, 10032, USA.
| | - Lindsey R Mitrani
- Department of Biomedical Informatics, Columbia University Medical Center, PH-20, 622 West 168th street, New York, NY, 10032, USA
| | - Gabriel G Lu
- Department of Biomedical Informatics, Columbia University Medical Center, PH-20, 622 West 168th street, New York, NY, 10032, USA
| | - Dominick J Fulgieri
- Department of Biomedical Informatics, Columbia University Medical Center, PH-20, 622 West 168th street, New York, NY, 10032, USA
| |
Collapse
|
42
|
Overbeek KA, Cahen DL, Canto MI, Bruno MJ. Surveillance for neoplasia in the pancreas. Best Pract Res Clin Gastroenterol 2016; 30:971-986. [PMID: 27938791 PMCID: PMC5552042 DOI: 10.1016/j.bpg.2016.10.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 10/25/2016] [Accepted: 10/31/2016] [Indexed: 01/31/2023]
Abstract
Despite its low incidence in the general population, pancreatic cancer is one of the leading causes of cancer-related mortality. Survival greatly depends on operability, but most patients present with unresectable disease. Therefore, there is great interest in the early detection of pancreatic cancer and its precursor lesions by surveillance. Worldwide, several programs have been initiated for individuals at high risk for pancreatic cancer. Their first results suggest that surveillance in high-risk individuals is feasible, but their effectiveness in decreasing mortality remains to be proven. This review will discuss which individuals are eligible for surveillance, which lesions are aimed to be detected, and which surveillance modalities are being used in current clinical practice. Furthermore, it addresses the management of abnormalities found during surveillance and topics for future research.
Collapse
Affiliation(s)
- Kasper A. Overbeek
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, ‘s Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands,Corresponding author. Fax: +31 10 703 03 31
| | - Djuna L. Cahen
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, ‘s Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Marcia Irene Canto
- Division of Gastroenterology and Hepatology, The Johns Hopkins Medical Institutions, 1800 Orleans St., Blalock 407, Baltimore, MD, 21287, USA
| | - Marco J. Bruno
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, ‘s Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| |
Collapse
|
43
|
Doi SAR, Furuya-Kanamori L, Engel JM, Jamal MH, Stankowski RV, Barkun J, Onitilo AA. The McGill Brisbane Symptom Score in relation to survival in pancreatic adenocarcinoma: a validation study. Cancer Causes Control 2016; 27:941-6. [DOI: 10.1007/s10552-016-0761-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 05/12/2016] [Indexed: 01/14/2023]
|
44
|
Škrha P, Hořínek A, Pazourková E, Hajer J, Frič P, Škrha J, Anděl M. Serum microRNA-196 and microRNA-200 in pancreatic ductal adenocarcinoma of patients with diabetes mellitus. Pancreatology 2016; 16:839-43. [PMID: 27267055 DOI: 10.1016/j.pan.2016.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/28/2016] [Accepted: 05/20/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES Our aim was to compare expressions of 6 microRNAs (miRNAs) in patients with pancreatic ductal adenocarcinoma (PAC) and non-cancer patients, moreover according to the presence or absence of diabetes mellitus. METHODS Expressions of miRNA-192, -196, -200, -21, -30 and -423 were measured in 77 patients with PAC and 64 non-cancer patients (34 patients with type 2 DM and 30 control persons). 60 patients with PAC (78%) had DM or prediabetes and it was of new-onset (less than 2 years before the cancer diagnosis) in 44 out of them. RESULTS The expressions of all microRNAs were 1.4-3.7 times higher (significantly) in the PAC group compared to non-cancer patients. No difference was found between PAC diabetic and PAC non-diabetic patients. MicroRNA-200 was significantly higher in PAC patients with significant body weight loss against those without weight loss. Adding miRNA-196 and -200 to the current marker CA 19-9 improved the discriminative ability of the test (compared to CA 19-9 alone). CONCLUSION MicroRNA-196 and -200 could be used as additional markers in PAC diagnosis.
Collapse
Affiliation(s)
- Pavel Škrha
- 2nd Department of Internal Medicine, 3rd Faculty of Medicine, Charles University, Faculty Hospital Královské Vinohrady, Prague, Czech Republic.
| | - Aleš Hořínek
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, General Faculty Hospital, Prague, Czech Republic
| | - Eva Pazourková
- Institute of Biology and Medical Genetics, 1st Faculty of Medicine, Charles University, General Faculty Hospital, Prague, Czech Republic
| | - Jan Hajer
- 2nd Department of Internal Medicine, 3rd Faculty of Medicine, Charles University, Faculty Hospital Královské Vinohrady, Prague, Czech Republic
| | - Přemysl Frič
- Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Military University Hospital, Prague, Czech Republic
| | - Jan Škrha
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, General Faculty Hospital, Prague, Czech Republic
| | - Michal Anděl
- 2nd Department of Internal Medicine, 3rd Faculty of Medicine, Charles University, Faculty Hospital Královské Vinohrady, Prague, Czech Republic
| |
Collapse
|
45
|
Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, Neale RE, Tempero M, Tuveson DA, Hruban RH, Neoptolemos JP. Pancreatic cancer. Nat Rev Dis Primers 2016; 2:16022. [PMID: 27158978 DOI: 10.1038/nrdp.2016.22] [Citation(s) in RCA: 1202] [Impact Index Per Article: 150.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pancreatic cancer is a major cause of cancer-associated mortality, with a dismal overall prognosis that has remained virtually unchanged for many decades. Currently, prevention or early diagnosis at a curable stage is exceedingly difficult; patients rarely exhibit symptoms and tumours do not display sensitive and specific markers to aid detection. Pancreatic cancers also have few prevalent genetic mutations; the most commonly mutated genes are KRAS, CDKN2A (encoding p16), TP53 and SMAD4 - none of which are currently druggable. Indeed, therapeutic options are limited and progress in drug development is impeded because most pancreatic cancers are complex at the genomic, epigenetic and metabolic levels, with multiple activated pathways and crosstalk evident. Furthermore, the multilayered interplay between neoplastic and stromal cells in the tumour microenvironment challenges medical treatment. Fewer than 20% of patients have surgically resectable disease; however, neoadjuvant therapies might shift tumours towards resectability. Although newer drug combinations and multimodal regimens in this setting, as well as the adjuvant setting, appreciably extend survival, ∼80% of patients will relapse after surgery and ultimately die of their disease. Thus, consideration of quality of life and overall survival is important. In this Primer, we summarize the current understanding of the salient pathophysiological, molecular, translational and clinical aspects of this disease. In addition, we present an outline of potential future directions for pancreatic cancer research and patient management.
Collapse
Affiliation(s)
- Jorg Kleeff
- NIHR Pancreas Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Royal Liverpool and Broadgreen University Hospitals NHS Trust, Duncan Building, Daulby Street, Liverpool L69 3GA, UK
- Department of General, Visceral and Pediatric Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Murray Korc
- Departments of Medicine, and Biochemistry and Molecular Biology, Indiana University School of Medicine, the Melvin and Bren Simon Cancer Center, and the Pancreatic Cancer Signature Center, Indianapolis, Indiana, USA
| | - Minoti Apte
- SWS Clinical School, University of New South Wales, and Ingham Institute for Applied Medical Research, Sydney, New South Wales, Australia
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Colin D Johnson
- University Surgical Unit, University Hospital Southampton, Southampton, UK
| | - Andrew V Biankin
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Garscube Estate, Bearsden, Glasgow, Scotland, UK
| | - Rachel E Neale
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Margaret Tempero
- UCSF Pancreas Center, University of California San Francisco - Mission Bay Campus/Mission Hall, San Francisco, California, USA
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, New York, USA
| | - Ralph H Hruban
- The Sol Goldman Pancreatic Cancer Research Center, Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John P Neoptolemos
- NIHR Pancreas Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Royal Liverpool and Broadgreen University Hospitals NHS Trust, Duncan Building, Daulby Street, Liverpool L69 3GA, UK
| |
Collapse
|
46
|
Lu L, Risch HA. Exosomes: potential for early detection in pancreatic cancer. Future Oncol 2016; 12:1081-90. [PMID: 26860951 DOI: 10.2217/fon-2015-0005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Progress in the treatment of patients with pancreatic cancer at earlier stages has motivated research in identifying novel noninvasive or minimally invasive biomarkers for early detection. Exosomes, which contain bioactive molecules (such as proteins, RNAs and lipids), are membrane-structured nanovesicles that are secreted from living cells and are found in human body fluids. As functional mediators, exosomes play key roles in cell-cell communications, regulating diverse biological processes. Here we aim to examine recent findings in the potential diagnostic value of serum exosomes in pancreatic cancer.
Collapse
Affiliation(s)
- Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale School of Medicine, Yale Cancer Center, New Haven, CT 06520-8034, USA
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale School of Medicine, Yale Cancer Center, New Haven, CT 06520-8034, USA
| |
Collapse
|
47
|
Wentzensen N, Eldridge RC. Invited Commentary: Clinical Utility of Prediction Models for Rare Outcomes--The Example of Pancreatic Cancer. Am J Epidemiol 2015; 182:35-8. [PMID: 26049862 DOI: 10.1093/aje/kwv028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 12/17/2014] [Indexed: 12/12/2022] Open
Abstract
Translating relative risk estimates into absolute risks is important in evaluating the potential clinical and public health relevance of etiologic discoveries. Predicting high absolute risk is challenging, particularly for rare endpoints such as pancreatic cancer. Recent efforts to develop risk prediction models for pancreatic cancer have found moderate risk levels for very small parts of the population. A new approach in which clinical symptoms and medication use are evaluated in addition to information on risk factors is presented by Risch et al. in this issue of the Journal (Am J Epidemiol. 2015;182(1):26-34). The authors estimated absolute risks based on the relative risks obtained from their case-control study. Their absolute risk estimates were higher than those from previous approaches but remained restricted to a very small proportion of the general population. In the present commentary, we address issues of absolute risk stratification (particularly for rare diseases), specific analytic methods, and how actionable information will differ based on the disease and possible intervention. We suggest that moving from cancer-specific models to broader models used to predict risk for multiple outcomes can make risk prediction for rare diseases more effective. When considering translational goals, it is important to estimate absolute risk at the early stages of etiologic research. The results can be sobering but allow focusing on the most promising goals.
Collapse
|
48
|
Risch HA, Yu H, Lu L, Kidd MS. Risch et al. Respond to "Clinical Utility of Prediction Models for Rare Outcomes: The Example of Pancreatic Cancer". Am J Epidemiol 2015; 182:39-40. [PMID: 26049861 DOI: 10.1093/aje/kwv025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 01/22/2015] [Indexed: 11/13/2022] Open
|