1
|
Huang C, Shen Y, Galgano SJ, Goenka AH, Hecht EM, Kambadakone A, Wang ZJ, Chu LC. Advancements in early detection of pancreatic cancer: the role of artificial intelligence and novel imaging techniques. Abdom Radiol (NY) 2025; 50:1731-1743. [PMID: 39467913 DOI: 10.1007/s00261-024-04644-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024]
Abstract
Early detection is crucial for improving survival rates of pancreatic ductal adenocarcinoma (PDA), yet current diagnostic methods can often fail at this stage. Recently, there has been significant interest in improving risk stratification and developing imaging biomarkers, through novel imaging techniques, and most notably, artificial intelligence (AI) technology. This review provides an overview of these advancements, with a focus on deep learning methods for early detection of PDA.
Collapse
Affiliation(s)
| | - Yiqiu Shen
- New York University Langone Health, New York, USA
| | | | | | | | | | - Zhen Jane Wang
- University of California, San Francisco, San Francisco, USA
| | - Linda C Chu
- Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
2
|
Liu Y, Gao Y, Wu Y, Wu W, Yu J, Ma S, Shi J, Wang K, Ye H. Autoantibodies as Potential Liquid Biopsy Biomarker in Detection of Pancreatic Cancer: A Diagnostic Test Accuracy Review and Meta-Analysis. Scand J Immunol 2025; 101:e70012. [PMID: 40185645 DOI: 10.1111/sji.70012] [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: 02/25/2025] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 04/07/2025]
Abstract
Autoantibodies against tumour-associated antigens (TAA) are promising biomarkers for cancer diagnosis. This systematic review aims to evaluate the diagnostic values of tumour-associated autoantibodies (TAAbs) in patients with pancreatic cancer. A search was conducted in the PubMed, Web of Science, and Embase databases to collect eligible studies. The primary outcomes included sensitivity, specificity, and accuracy of the test. We used QUADAS-2 to evaluate the risk of bias in the included studies. Meta-analysis was performed using MetaDisc 1.4 and STATA 14.0 software to calculate the combined sensitivity and specificity. A total of 49 articles were included in the final analysis that reported over 100 different TAAbs that were studied for the detection of pancreatic cancer. p53, Ezrin, CLDN17, KCNN3, SLAMF7, SLC22A11 and OR51F2 were the most frequently investigated autoantibodies in these studies. Ezrin exhibited better diagnostic performance with the pooled sensitivity, specificity and summary area under the receiver operating characteristic (SROC) curves being 56%, 88% and 0.90, respectively. Moreover, certain autoantibody combinations achieved substantially higher sensitivity at reasonably high levels of specificity. For example, the combination of Ezrin and ENOA1.2 autoantibodies with CA19.9 yielded sensitivity, specificity and area under the SROC curve of 100%, 92% and 0.96, respectively. TAAb is a promising diagnostic biomarker for early detection of PC, especially when combining TAAb with other markers. The promising candidate markers identified in this review deserve further validation in a broad screening population.
Collapse
Affiliation(s)
- Yuqi Liu
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yuyi Gao
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yangxue Wu
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Wanyang Wu
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Jinyao Yu
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Siyao Ma
- Minhang Crosspoint High School, Shanghai, China
| | - Jianxiang Shi
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Keyan Wang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Hua Ye
- College of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Shi J, Zhao L, Wang K, Lin J, Shen J. Disulfidptosis classification of pancreatic carcinoma reveals correlation with clinical prognosis and immune profile. Hereditas 2025; 162:26. [PMID: 39987145 PMCID: PMC11846472 DOI: 10.1186/s41065-025-00381-z] [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: 11/20/2024] [Accepted: 01/27/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Disulfidptosis, a novel form of metabolism-related regulated cell death, is a promising intervention for cancer therapeutic intervention. Although aberrant expression of long-chain noncoding RNAs (lncRNAs) expression has been associated with pancreatic carcinoma (PC) development, the biological properties and prognostic potential of disulfidptosis-related lncRNAs (DRLs) remain unclear. METHODS We obtained RNA-seq data, clinical data, and genomic mutations of PC from the TCGA database, and then determined DRLs. We developed a risk score model and analyzed the role of risk score in the predictive ability, immune cell infiltration, immunotherapy response, and drug sensitivity. RESULTS We finally established a prognostic model including three DRLs (AP005233.2, FAM83A-AS1, and TRAF3IP2-AS1). According to Kaplan-Meier curve analysis, the survival time of patients in the low-risk group was significantly longer than that in the high-risk group. Based on enrichment analysis, significant associations between metabolic processes and differentially expressed genes were assessed in two risk groups. In addition, we observed significant differences in the tumor immune microenvironment landscape. Tumor Immune Dysfunction and Rejection (TIDE) analysis showed no statistically significant likelihood of immune evasion in both risk groups. Patients exhibiting both high risk and high tumor mutation burden (TMB) had the poorest survival times, while those falling into the low risk and low TMB categories showed the best prognosis. Moreover, the risk group identified by the 3-DRLs profile showed significant drug sensitivity. CONCLUSIONS Our proposed 3-DRLs-based feature could serve as a promising tool for predicting the prognosis, immune landscape, and treatment response of PC patients, thus facilitating optimal clinical decision-making.
Collapse
Affiliation(s)
- Jiangmin Shi
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China
| | - Liang Zhao
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China
| | - Kai Wang
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China
| | - Jieqiong Lin
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China
| | - Jianwei Shen
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China.
| |
Collapse
|
4
|
Delrue C, De Bruyne S, Speeckaert MM. The Promise of Infrared Spectroscopy in Liquid Biopsies for Solid Cancer Detection. Diagnostics (Basel) 2025; 15:368. [PMID: 39941298 PMCID: PMC11818004 DOI: 10.3390/diagnostics15030368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/15/2025] [Accepted: 02/01/2025] [Indexed: 02/16/2025] Open
Abstract
Attenuated total reflection-Fourier transform infrared (ATR-FTIR) spectroscopy has shown significant promise in the context of liquid biopsy, offering a potential tool for cancer diagnostics. Unlike traditional tissue biopsies, which may not fully capture the clonal heterogeneity of tumors, liquid biopsy reflects the dynamic state of the disease and its progression more comprehensively. Biofluids such as serum and plasma are low-cost, minimally invasive diagnostic media with well-established clinical uses. This review assesses the use of ATR-FTIR spectroscopy to detect biochemical changes in biofluids linked to various malignancies, including breast, ovarian, endometrial, prostate, bladder, kidney, pancreatic, colorectal, hepatic, esophageal, gastric, lung, and brain cancers. While ATR-FTIR offers the advantages of rapid, minimally invasive detection and real-time disease monitoring, its integration into clinical practice faces challenges, particularly in terms of reproducibility due to variability in sample preparation, spectral acquisition, and data processing. The translation of ATR-FTIR into routine diagnostics will require validation through large-scale cohort studies and multicenter trials to ensure its clinical reliability and effectiveness.
Collapse
Affiliation(s)
- Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Sander De Bruyne
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium;
- Department of Laboratory Medicine, AZ Sint-Blasius, 9200 Dendermonde, Belgium
| | - Marijn M. Speeckaert
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium;
- Research Foundation-Flanders (FWO), 1000 Brussels, Belgium
| |
Collapse
|
5
|
Blanco Abad C, Gomila Pons P, Campos Ramírez S, Álvarez Alejandro M, Torres Ramón MI, Miramar Gallart MD, Izquierdo Álvarez S, Polo Marques E, Pazo Cid R. Hereditary Pancreatic Cancer: Advances in Genetic Testing, Early Detection Strategies, and Personalized Management. J Clin Med 2025; 14:367. [PMID: 39860372 PMCID: PMC11766428 DOI: 10.3390/jcm14020367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/29/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with a five-year survival rate of approximately 13% for advanced stages. While the majority of PDAC cases are sporadic, a significant subset is attributable to hereditary and familial predispositions, accounting for approximately 25% of cases. This article synthesizes recent advancements in the understanding, detection, and management of hereditary pancreatic cancer (PC). Results: Our review highlights the critical role of genetic testing (GT) in identifying high-risk individuals (HRIs), with germline pathogenic variants (PVs) found in up to 20% of hereditary PDAC cases. Since the implementation of next-generation sequencing (NGS) panels in 2014, detection capabilities have been significantly enhanced. HRIs can be included in screening programs that facilitate the early detection of PDAC. Early detection strategies, including the use of microribonucleic acid (miRNAs) signatures and novel imaging techniques like hyperpolarized 13C-magnetic resonance spectroscopy (MRS) have shown promising results. The identification of germline pathogenic variants (PVs) or mutations in homologous recombination (HR) genes plays a predictive role in the response to various treatments, prolonging patient survival. Discussion: Universal germline testing for PDAC, as recommended by the National Comprehensive Cancer Network (NCCN), is now a standard practice, facilitating the identification of at-risk individuals and enabling targeted surveillance and intervention. Multidisciplinary management, integrating genetic counseling, imaging, and gastrointestinal services, is essential for optimizing outcomes. Conclusions: Advances in genetic testing and biomarker research are transforming the landscape of hereditary PC management. Early detection and personalized treatment strategies are pivotal in improving survival rates. Ongoing multi-institutional research efforts are crucial for validating biomarkers and developing preventive measures, ultimately aiming to reduce the burden of this aggressive cancer.
Collapse
Affiliation(s)
- Carmen Blanco Abad
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
| | - Paula Gomila Pons
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
| | - Sara Campos Ramírez
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
| | - María Álvarez Alejandro
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
- Medical Oncology Department, Hospital Clinico Universitario Lozano Blesa, 50009 Zaragoza, Spain
| | - María Irene Torres Ramón
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
- Medical Oncology Department, Hospital Clinico Universitario Lozano Blesa, 50009 Zaragoza, Spain
| | | | - Silvia Izquierdo Álvarez
- Genetics Unit, Biochemistry Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
| | - Eduardo Polo Marques
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
| | - Roberto Pazo Cid
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
- Medical Oncology Department, Hospital Clinico Universitario Lozano Blesa, 50009 Zaragoza, Spain
- Department of Medicine, Psychiatry and Dermatology, Faculty of Medicine, Zaragoza University, 50009 Zaragoza, Spain
| |
Collapse
|
6
|
Half E, Ovcharenko A, Shmuel R, Furman-Assaf S, Avdalimov M, Rabinowicz A, Arber N. Non-invasive multiple cancer screening using trained detection canines and artificial intelligence: a prospective double-blind study. Sci Rep 2024; 14:28204. [PMID: 39548246 PMCID: PMC11568277 DOI: 10.1038/s41598-024-79383-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024] Open
Abstract
The specificity and sensitivity of a simple non-invasive multi-cancer screening method in detecting breast, lung, prostate, and colorectal cancer in breath samples were evaluated in a double-blind study. Breath samples of 1386 participants (59.7% males, median age 56.0 years) who underwent screening for cancer using gold-standard screening methods, or a biopsy for a suspected malignancy were collected. The samples were analyzed using a bio-hybrid platform comprising trained detection canines and artificial intelligence tools. According to cancer screening/biopsy results, 1048 (75.6%) were negative for cancer and 338 (24.4%) were positive. Among the 338 positive samples, 261 (77.2%) were positive for one of the four cancer types that the bio-hybrid platform was trained to detect, with an overall sensitivity and specificity of 93.9% (95% confidence interval [CI] 90.3-96.2%) and 94.3% (95% CI 92.7%-95.5%), respectively. The sensitivity of each cancer type was similar; breast: 95.0% (95% CI 87.8-98.0%), lung: 95.0% (95% CI 87.8-98.0%), colorectal: 90.0% (95% CI 74.4-96.5%), prostate: 93.0% (95% CI 84.6-97.0%). The sensitivity of 14 other malignant tumors that the bio-hybrid platform was not trained to detect, but identified, was 81.8% (95% CI 71.8%-88.8%). Early cancer (0-2) detection sensitivity was 94.8% (95% CI 91.0%-97.1%). This bio-hybrid multi-cancer screening platform demonstrated high sensitivity and specificity and enables early-stage cancer detection.
Collapse
Affiliation(s)
- Elizabeth Half
- Gastroenterology Unit, Rambam Health Care Campus, Haifa, Israel
| | | | - Ronit Shmuel
- Medical consultant (independent), Tel Aviv, Israel
| | | | | | | | - Nadir Arber
- Integrated Cancer Prevention Center, Tel Aviv Souraski Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
7
|
Maurer E, Lehman B, Matthäi E, Denzer U, Figiel J, Jesinghaus M, Slater EP, Stefenelli U, Gress TM, Bartsch DK. Pancreatic cancer screening is effective in individuals at risk with predisposing germline gene variants, but not in gene variant-negative familial pancreatic cancer families. United European Gastroenterol J 2024; 12:1211-1221. [PMID: 39031472 PMCID: PMC11578844 DOI: 10.1002/ueg2.12631] [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: 03/17/2024] [Accepted: 06/20/2024] [Indexed: 07/22/2024] Open
Abstract
OBJECTIVE To evaluate the diagnostic yield of pancreatic cancer screening in individuals at risk (IAR) from familial pancreatic cancer (FPC) families with respect to the presence or absence of pathogenic germline variants predisposing to pancreatic adenocarcinoma (PDAC). DESIGN In a 20 years period, IAR from FPC families were enrolled in a prospective screening program of the national case collection for FPC of Germany, including magnet resonance imaging (MRI) and endoscopic ultrasound (EUS). The diagnostic yield was analyzed regarding significant pancreatic lesions such as PDAC, high-grade pancreatic-intraepithelial-neoplasia (PanIN3) and intraductal-papillary-mucinous-neoplasia (IPMN) with high-grade dysplasia. Screening results were compared between carriers of pathogenic variants and variant-negative IAR. RESULTS 337 IAR, including 74 (22%) variant-carriers and 263 IAR of variant-negative FPC families (mean age 49; standard deviation [SD] + 8.9) were followed 64 (SD + 55) months. IAR underwent 5.1 (SD + 3.9) screening visits with 1733 MRI (5.1,SD + 3.9 per IAR) and 728 EUS (2.2,SD + 1.7 per IAR). In 12 (4%) cases, significant pancreatic lesions were detected, including 4 PDAC, 3 PanIN3 and 5 high-grade IPMN. Three of 4 IAR with PDAC died after a mean of 27 months postoperatively, and one IAR is alive without evidence of disease after 31 months. The diagnostic yield for significant lesions was 13.5% (10/74) for variant carriers compared to 0.8% (2/263) for IAR of variant-negative FPC families (p < 0.001). Logistic regression analysis revealed that a negative variant status was almost always accompanied by the absence of a significant lesion over time with a negative predictive value of 99.2% (95% CI 97.3%-99.9%). CONCLUSION The diagnostic yield seems to justify PDAC screening in IAR of FPC-families with pathogenic germline variants in PDAC predisposing genes, not in IAR of variant-negative families.
Collapse
Affiliation(s)
- Elisabeth Maurer
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| | - Bettina Lehman
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| | - Elvira Matthäi
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| | - Ulrike Denzer
- Department of GastroenterologyEndocrinologyMetabolism and InfectiologyPhilipps University MarburgMarburgGermany
| | - Jens Figiel
- Department of Diagnostic and Interventional RadiologyPhilipps University MarburgMarburgGermany
| | | | - Emily P. Slater
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| | | | - Thomas M. Gress
- Department of GastroenterologyEndocrinologyMetabolism and InfectiologyPhilipps University MarburgMarburgGermany
| | - Detlef K. Bartsch
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| |
Collapse
|
8
|
Ahmed TM, Kawamoto S, Lopez-Ramirez F, Yasrab M, Hruban RH, Fishman EK, Chu LC. Early detection of pancreatic cancer in the era of precision medicine. Abdom Radiol (NY) 2024; 49:3559-3573. [PMID: 38761272 DOI: 10.1007/s00261-024-04358-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/20/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related mortality and it is often diagnosed at advanced stages due to non-specific clinical presentation. Disease detection at localized disease stage followed by surgical resection remains the only potentially curative treatment. In this era of precision medicine, a multifaceted approach to early detection of PDAC includes targeted screening in high-risk populations, serum biomarkers and "liquid biopsies", and artificial intelligence augmented tumor detection from radiologic examinations. In this review, we will review these emerging techniques in the early detection of PDAC.
Collapse
Affiliation(s)
- Taha M Ahmed
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Felipe Lopez-Ramirez
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Mohammad Yasrab
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Ralph H Hruban
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Linda C Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA.
| |
Collapse
|
9
|
Zaman M, Li JH, Dhir M. Malpractice Claims Following Major Liver and Pancreatic Surgeries: What Can we Learn? J Surg Res 2024; 298:291-299. [PMID: 38640614 DOI: 10.1016/j.jss.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/08/2024] [Accepted: 03/21/2024] [Indexed: 04/21/2024]
Abstract
INTRODUCTION General surgery is a highly litigious specialty. Lawsuits can be a source of emotional distress and burnout for surgeons. Major hepatic and pancreatic surgeries are technically challenging general surgical oncology procedures associated with an increased risk of complications and mortality. It is unclear whether these operations are associated with an increased risk of lawsuits. The objective of the present study was to summarize the medical malpractice claims surrounding pancreatic and hepatic surgeries from publicly available court records. METHODS The Westlaw legal database was searched and analyzed for relevant malpractice claims from the last two decades. RESULTS Of 165 search results, 30 (18.2%) cases were eligible for inclusion. Appellant cases comprised 53.3% of them. Half involved a patient death. Including co-defendants, a majority (n = 21, 70%) named surgeons as defendants, whereas several claims (n = 13, 43%) also named non-surgeons. The most common cause of alleged malpractice was a delay in diagnosis (n = 12, 40%). In eight of these, surgery could not be performed. The second most common were claims alleging the follow-up surgery was due to negligence (n = 6). Collectively, 20 claims were found in favor of the defendant. Seven verdicts (23.3%) returned in favor of the plaintiff, two of which resulted in monetary awards (totaling $1,608,325 and $424,933.85). Three cases went to trial or delayed motion for summary judgment. There were no settlements. CONCLUSIONS A defendant verdict was reached in two-thirds of malpractice cases involving major hepatic or pancreatic surgery. A delay in diagnosis was the most cited claim in hepatopancreaticobiliary lawsuits, and defendants may often practice in nonsurgical specialties. While rulings favoring plaintiffs are less frequent, the payouts may be substantial.
Collapse
Affiliation(s)
- Muizz Zaman
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, New York; Department of Surgery, SUNY Upstate Medical University, Syracuse, New York.
| | - Jian Harvard Li
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, New York
| | - Mashaal Dhir
- Department of Surgery, SUNY Upstate Medical University, Syracuse, New York.
| |
Collapse
|
10
|
Wang L, Wang L, Sun X, Fu L, Wang X, Wang X, Chen L, Huang Y. Detection of uridine diphosphate glucuronosyltransferase 1A1 for pancreatic cancer imaging and treatment via a "turn-on" fluorescent probe. Analyst 2024; 149:2877-2886. [PMID: 38567989 DOI: 10.1039/d4an00035h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) is expressed ubiquitously in cancer cells and can metabolize exogenous substances. Studies show higher UGT1A1 levels in pancreatic cancer cells than normal cells. Therefore, we need a method to monitor the activity level of UGT1A1 in pancreatic cancer cells and in vivo. Here, we report a fluorescent probe, BCy-panc, for UGT1A1 imaging in cells and in vivo. Compared with other molecular probes, this probe is readily prepared, with high selectivity and sensitivity for the detection of UGT1A1. Our results show that BCy-panc rapidly detects UGT1A1 in pancreatic cancer. In addition, there is an urgent need for evidence to clarify the relationship between UGT1A1 and pancreatic cancer development. The present investigation found that the increase of UGT1A1 by chrysin was effective in inducing apoptosis in pancreatic cancer cells. These results indicate that the synergistic effect of chrysin and cisplatin at the cellular level is superior to that of cisplatin alone. The UGT1A1 level may be a biomarker for early diagnosis of cancer. Meanwhile, UGT1A1 plays a crucial role in pancreatic cancer, and the combination of chrysin and cisplatin may provide effective ideas for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Lingxiao Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| | - Lingyun Wang
- Jinan Zhangqiu District People's Hospital, Jinan 250000, China
| | - Xiao Sun
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| | - Lili Fu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Shandong Key Laboratory of Coastal Environmental Processes, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| | - Xinlei Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| | - Xiaoyan Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Shandong Key Laboratory of Coastal Environmental Processes, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| | - Lingxin Chen
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Shandong Key Laboratory of Coastal Environmental Processes, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Yan Huang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Shandong Key Laboratory of Coastal Environmental Processes, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| |
Collapse
|
11
|
Reshkin SJ, Cardone RA, Koltai T. Genetic Signature of Human Pancreatic Cancer and Personalized Targeting. Cells 2024; 13:602. [PMID: 38607041 PMCID: PMC11011857 DOI: 10.3390/cells13070602] [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: 02/12/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
Pancreatic cancer is a highly lethal disease with a 5-year survival rate of around 11-12%. Surgery, being the treatment of choice, is only possible in 20% of symptomatic patients. The main reason is that when it becomes symptomatic, IT IS the tumor is usually locally advanced and/or has metastasized to distant organs; thus, early diagnosis is infrequent. The lack of specific early symptoms is an important cause of late diagnosis. Unfortunately, diagnostic tumor markers become positive at a late stage, and there is a lack of early-stage markers. Surgical and non-surgical cases are treated with neoadjuvant and/or adjuvant chemotherapy, and the results are usually poor. However, personalized targeted therapy directed against tumor drivers may improve this situation. Until recently, many pancreatic tumor driver genes/proteins were considered untargetable. Chemical and physical characteristics of mutated KRAS are a formidable challenge to overcome. This situation is slowly changing. For the first time, there are candidate drugs that can target the main driver gene of pancreatic cancer: KRAS. Indeed, KRAS inhibition has been clinically achieved in lung cancer and, at the pre-clinical level, in pancreatic cancer as well. This will probably change the very poor outlook for this disease. This paper reviews the genetic characteristics of sporadic and hereditary predisposition to pancreatic cancer and the possibilities of a personalized treatment according to the genetic signature.
Collapse
Affiliation(s)
- Stephan J. Reshkin
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Tomas Koltai
- Oncomed, Via Pier Capponi 6, 50132 Florence, Italy
| |
Collapse
|
12
|
Sagami R, Yamao K, Minami R, Nakahodo J, Akiyama H, Nishikiori H, Mizukami K, Yamao K, Bhatia V, Amano Y, Murakami K. Endoscopic Ultrasound Can Differentiate High-Grade Pancreatic Intraepithelial Neoplasia, Small Pancreatic Ductal Adenocarcinoma, and Benign Stenosis. Gut Liver 2024; 18:338-347. [PMID: 37165770 PMCID: PMC10938144 DOI: 10.5009/gnl220521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/06/2023] [Accepted: 02/15/2023] [Indexed: 05/12/2023] Open
Abstract
Background/Aims High-grade pancreatic intraepithelial neoplasia and invasive pancreatic ductal adenocarcinoma ≤10 mm are targets for early detection of pancreatic cancer. However, their imaging characteristics are unknown. We aimed to identify endoscopic ultrasound findings for the detection of these lesions. Methods Patients diagnosed with high-grade pancreatic intraepithelial neoplasia (n=29), pancreatic ductal adenocarcinoma ≤10 mm (n=11) (who underwent surgical resection), or benign main pancreatic duct stenosis (n=20) between January 2014 and January 2021 were retrospectively included. Six features differentiating these lesions were examined by endoscopic ultrasonography: main pancreatic duct stenosis, upstream main pancreatic duct dilation, hypoechoic areas surrounding the main pancreatic duct irregularities (mottled areas without demarcation or round areas with demarcation), branch duct dilation, prominent lobular segmentation, and atrophy. Interobserver agreement was assessed by two independent observers. Results Hypoechoic areas surrounding the main pancreatic duct irregularities were observed more frequently in high-grade pancreatic intraepithelial neoplasia (82.8%) and pancreatic ductal adenocarcinoma ≤10 mm (90.9%) than in benign stenosis (15.0%) (p<0.001). High-grade pancreatic intraepithelial neoplasia exhibited mottled hypoechoic areas more frequently (79.3% vs 18.9%, p<0.001), and round hypoechoic areas less frequently (3.4% vs 72.7%, p<0.001), than pancreatic ductal adenocarcinoma ≤10 mm. The sensitivity and specificity of hypoechoic areas for differentiating high-grade pancreatic intraepithelial neoplasia, pancreatic ductal adenocarcinoma ≤10 mm, and benign stenosis were both 85.0%, with moderate interobserver agreement. Conclusions The hypoechoic areas surrounding main pancreatic duct irregularities on endoscopic ultrasound may differentiate between high-grade pancreatic intraepithelial neoplasia, pancreatic ductal adenocarcinoma ≤10 mm, and benign stenosis (Trial Registration: UMIN Clinical Trials Registry (UMIN000044789).
Collapse
Affiliation(s)
- Ryota Sagami
- Department of Gastroenterology, Oita San-ai Medical Center, Oita, Japan
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Japan
| | - Kentaro Yamao
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Japan
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Ryuki Minami
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Japan
- Department of Gastroenterology, Tenri Hospital, Nara, Japan
| | - Jun Nakahodo
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Japan
- Department of Gastroenterology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Hidetoshi Akiyama
- Department of Gastroenterology, Oita Red Cross Hospital, Oita, Japan
| | | | - Kazuhiro Mizukami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Kenji Yamao
- Department of Gastroenterology, Narita Memorial Hospital, Aichi, Japan
| | - Vikram Bhatia
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Yuji Amano
- Department of Endoscopy, Urawa Kyosai Hospital, Saitama, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| |
Collapse
|
13
|
Wang L, Larki NR, Dobkin J, Salgado S, Ahmad N, Kaplan DE, Yang W, Yang YX. A Clinical Prediction Model to Assess Risk for Pancreatic Cancer Among Patients With Acute Pancreatitis. Pancreas 2024; 53:e254-e259. [PMID: 38266222 PMCID: PMC11214820 DOI: 10.1097/mpa.0000000000002295] [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: 01/26/2024]
Abstract
OBJECTIVES We aimed to develop and validate a prediction model as the first step in a sequential screening strategy to identify acute pancreatitis (AP) individuals at risk for pancreatic cancer (PC). MATERIALS AND METHODS We performed a population-based retrospective cohort study among individuals 40 years or older with a hospitalization for AP in the US Veterans Health Administration. For variable selection, we used least absolute shrinkage and selection operator regression with 10-fold cross-validation to identify a parsimonious logistic regression model for predicting the outcome, PC diagnosed within 2 years after AP. We evaluated model discrimination and calibration. RESULTS Among 51,613 eligible study patients with AP, 801 individuals were diagnosed with PC within 2 years. The final model (area under the receiver operating curve, 0.70; 95% confidence interval, 0.67-0.73) included histories of gallstones, pancreatic cyst, alcohol use, smoking, and levels of bilirubin, triglycerides, alkaline phosphatase, aspartate aminotransferase, alanine aminotransferase, and albumin. If the predicted risk threshold was set at 2% over 2 years, 20.3% of the AP population would undergo definitive screening, identifying nearly 50% of PC associated with AP. CONCLUSIONS We developed a prediction model using widely available clinical factors to identify high-risk patients with PC-associated AP, the first step in a sequential screening strategy.
Collapse
Affiliation(s)
- Louise Wang
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Navid Rahimi Larki
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Jane Dobkin
- Columbia Irving Medical Center, New York City, NY
| | - Sanjay Salgado
- Division of Gastroenterology and Hepatology, New York Presbyterian Hospital/Weill Cornell Medical College, New York, New York, USA
| | - Nuzhat Ahmad
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, Philadelphia, PA
| | - David E. Kaplan
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, Philadelphia, PA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Wei Yang
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, Philadelphia, PA
| | - Yu-Xiao Yang
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, Philadelphia, PA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| |
Collapse
|
14
|
Peters MLB, Eckel A, Seguin CL, Davidi B, Howard DH, Knudsen AB, Pandharipande PV. Cost-Effectiveness Analysis of Screening for Pancreatic Cancer Among High-Risk Populations. JCO Oncol Pract 2024; 20:278-290. [PMID: 38086003 PMCID: PMC10911581 DOI: 10.1200/op.23.00495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/05/2023] [Accepted: 10/30/2023] [Indexed: 12/19/2023] Open
Abstract
PURPOSE We evaluated the potential cost-effectiveness of combined magnetic resonance imaging (MRI) and endoscopic ultrasound (EUS) screening for pancreatic ductal adenocarcinoma (PDAC) among populations at high risk for the disease. METHODS We used a microsimulation model of the natural history of PDAC to estimate the lifetime health benefits, costs, and cost-effectiveness of PDAC screening among populations with specific genetic risk factors for PDAC, including BRCA1 and BRCA2, PALB2, ATM, Lynch syndrome, TP53, CDKN2A, and STK11. For each high-risk population, we simulated 29 screening strategies, defined by starting age and frequency. Screening included MRI with follow-up EUS in a subset of patients. Costs of tests were based on Medicare reimbursement for MRI, EUS, fine-needle aspiration biopsy, and pancreatectomy. Cancer-related cost by stage of disease and phase of treatment was based on the literature. For each high-risk population, we performed an incremental cost-effectiveness analysis, assuming a willingness-to-pay (WTP) threshold of $100,000 US dollars (USD) per quality-adjusted life year (QALY) gained. RESULTS For men with relative risk (RR) 12.33 (CDKN2A) and RR 28 (STK11), annual screening was cost-effective, starting at age 55 and 40 years, respectively. For women, screening was only cost-effective for those with RR 28 (STK11), with annual screening starting at age 45 years. CONCLUSION Combined MRI/EUS screening may be a cost-effective approach for the highest-risk populations (among mutations considered, those with RR >12). However, for those with moderate risk (RR, 5-12), screening would only be cost-effective at higher WTP thresholds (eg, $200K USD/QALY) or with once-only screening.
Collapse
Affiliation(s)
- Mary Linton B. Peters
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA
| | - Andrew Eckel
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA
| | - Claudia L. Seguin
- Department of Radiology, The Ohio State University College of Medicine, Columbus, OH
| | - Barak Davidi
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA
| | - David H. Howard
- Department of Health Policy and Management, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Amy B. Knudsen
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA
- Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Pari V. Pandharipande
- Department of Radiology, The Ohio State University College of Medicine, Columbus, OH
| |
Collapse
|
15
|
Farooq M, Leevan E, Ahmed J, Ko B, Shin S, De Souza A, Takebe N. Blood-based multi-cancer detection: A state-of-the-art update. Curr Probl Cancer 2024; 48:101059. [PMID: 38181630 DOI: 10.1016/j.currproblcancer.2023.101059] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/12/2023] [Accepted: 12/26/2023] [Indexed: 01/07/2024]
Abstract
The early detection of cancer is a key goal of the National Cancer Plan formally released by the National Institutes of Health's (NIH) National Cancer Institute (NCI) in April 2023. To support this effort, many laboratories and vendors are developing multi-cancer detection (MCD) assays that interrogate blood and other bodily fluids for cancer-related biomarkers, most commonly circulating tumor DNA (ctDNA). While this approach holds promise for non-invasively detecting early signals of multiple different cancers and potentially reducing cancer-related mortality, there is a dearth of prospective clinical data to inform the deployment of MCD assays for cancer screening in the general adult population. In this review we highlight differing technologies that underpin various MCD assays in clinical development, the importance of achieving adequate performance specifications for MCD assays, ongoing clinical studies investigating the utility of MCD assays in cancer screening and detection, and efforts by the NCI's Division of Cancer Prevention (DCP) to establish a network infrastructure that has the capacity to comprehensively address the scientific and logistical challenges of evaluating blood-based MCD approaches and other cancer screening tools.
Collapse
Affiliation(s)
- Maria Farooq
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elyse Leevan
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jibran Ahmed
- Developmental Therapeutics Clinic, Early Phase Clinical Trials Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Brian Ko
- Developmental Therapeutics Clinic, Early Phase Clinical Trials Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sarah Shin
- Developmental Therapeutics Clinic, Early Phase Clinical Trials Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andre De Souza
- Developmental Therapeutics Clinic, Early Phase Clinical Trials Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Naoko Takebe
- Developmental Therapeutics Clinic, Early Phase Clinical Trials Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
16
|
Grobbelaar C, Kgomo M, Mabeta P. Angiogenesis and Pancreatic Cancer: Novel Approaches to Overcome Treatment Resistance. Curr Cancer Drug Targets 2024; 24:1116-1127. [PMID: 38299403 DOI: 10.2174/0115680096284588240105051402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 02/02/2024]
Abstract
Pancreatic cancer (PCa) is acknowledged as a significant contributor to global cancer- related mortality and is widely recognized as one of the most challenging malignant diseases to treat. Pancreatic ductal adenocarcinoma (PDAC), which is the most common type of PCa, is highly aggressive and is mostly incurable. The poor prognosis of this neoplasm is exacerbated by the prevalence of angiogenic molecules, which contribute to stromal stiffness and immune escape. PDAC overexpresses various proangiogenic proteins, including vascular endothelial growth factor (VEGF)-A, and the levels of these molecules correlate with poor prognosis and treatment resistance. Moreover, VEGF-targeting anti-angiogenesis treatments are associated with the onset of resistance due to the development of hypoxia, which in turn induces the production of angiogenic molecules. Furthermore, excessive angiogenesis is one of the hallmarks of the second most common form of PCa, namely, pancreatic neuroendocrine tumor (PNET). In this review, the role of angiogenesis regulators in promoting disease progression in PCa, and the impact of these molecules on resistance to gemcitabine and various therapies against PCa are discussed. Finally, the use of anti-angiogenic agents in combination with chemotherapy and other targeted therapeutic molecules is discussed as a novel solution to overcome current treatment limitations in PCa.
Collapse
Affiliation(s)
- Craig Grobbelaar
- Department of Physiology, University of Pretoria, CNR Lynnwood Road and Roper Street, Hatfield, 0028, South Africa
| | - Mpho Kgomo
- Department of Internal Medicine, Faculty of Health Sciences, University of Pretoria, 9 Bophelo Road, Arcadia, CNR Lynnwood Road and Roper Street, Hatfield, 0028, South Africa
| | - Peace Mabeta
- Department of Physiology, University of Pretoria, CNR Lynnwood Road and Roper Street, Hatfield, 0028South Africa
| |
Collapse
|
17
|
Paiella S, Secchettin E, Lionetto G, Archibugi L, Azzolina D, Casciani F, Simeone DM, Overbeek KA, Goggins M, Farrell J, Ponz de Leon Pisani R, Tridenti M, Corciulo MA, Malleo G, Arcidiacono PG, Falconi M, Gregori D, Bassi C, Salvia R, Capurso G. Surveillance of Individuals at High Risk of Developing Pancreatic Cancer: A Prevalence Meta-analysis to Estimate the Rate of Low-yield Surgery. Ann Surg 2024; 279:37-44. [PMID: 37681303 DOI: 10.1097/sla.0000000000006094] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
OBJECTIVE To quantify the rate of low-yield surgery, defined as no high-grade dysplastic precursor lesions or T1N0M0 pancreatic cancer at pathology, during pancreatic cancer surveillance. BACKGROUND Global efforts have been made in pancreatic cancer surveillance to anticipate the diagnosis of pancreatic cancer at an early stage and improve survival in high-risk individuals (HRIs) with a hereditary predisposition. The negative impact of pancreatic cancer surveillance when surgery is performed for low-grade dysplasia or a non-neoplastic condition is not well quantified. MATERIALS AND METHODS A systematic search and prevalence meta-analysis was performed for studies reporting surgery with final diagnoses other than those defined by the Cancer of the Pancreas Screening (CAPS) goals from January 2000 to July 2023. The secondary outcome was the pooled proportion of final diagnoses matching the CAPS goals (PROSPERO: #CRD42022300408). RESULTS Twenty-three articles with 5027 patients (median 109 patients/study, interquartile range 251) were included. The pooled prevalence of low-yield surgery was 2.1% (95% CI: 0.9-3.7, I2 : 83%). In the subgroup analysis, this prevalence was nonsignificantly higher in studies that only included familial pancreatic cancer subjects without known pathogenic variants, compared with those enrolling pathogenic variant carriers. No effect modifiers were found. Overall, the pooled prevalence of subjects under surveillance who had a pancreatic resection that contained target lesions was 0.8% (95% CI, 0.3-1.5, I2 : 24%]. The temporal analysis showed that the rate of low-yield surgeries decreased in the last decades and stabilized at around 1% (test for subgroup differences P <0.01). CONCLUSIONS The risk of "low-yield" surgery during pancreatic cancer surveillance is relatively low but should be thoroughly discussed with individuals under surveillance.
Collapse
Affiliation(s)
- Salvatore Paiella
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Erica Secchettin
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Gabriella Lionetto
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Livia Archibugi
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| | - Danila Azzolina
- Department of Environmental and Preventive Science, University of Ferrara, Ferrara, Italy
| | - Fabio Casciani
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Diane M Simeone
- Department of Surgery, New York University, New York, NY
- Perlmutter Cancer Center, New York University, New York, NY
| | - Kasper A Overbeek
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Michael Goggins
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - James Farrell
- Yale Center for Pancreatic Disease, Yale University School of Medicine, New Haven, CT
| | - Ruggero Ponz de Leon Pisani
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| | - Maddalena Tridenti
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| | - Maria Assunta Corciulo
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, Padova, Italy
| | - Giuseppe Malleo
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Paolo Giorgio Arcidiacono
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| | - Massimo Falconi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Pancreatic Surgery and Transplantation Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Dario Gregori
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, Padova, Italy
| | - Claudio Bassi
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Roberto Salvia
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| |
Collapse
|
18
|
Sirtl S, Vornhülz M, Hofmann FO, Mayerle J, Beyer G. [Pancreatic cancer-screening or surveillance?]. RADIOLOGIE (HEIDELBERG, GERMANY) 2023; 63:908-915. [PMID: 37878016 DOI: 10.1007/s00117-023-01227-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
BACKGROUND Despite continuous improvement of diagnostic and therapeutic procedures, the number of new pancreatic ductal adenocarcinoma (PDAC) cases diagnosed annually almost equals the number of PDAC-related deaths. Prerequisite for curative treatment is a resectable tumor at the time of diagnosis. Individuals with genetic and/or familial risk profiles should therefore be screened and included in structured surveillance programs. OBJECTIVES Description of the status quo and usefulness of current PDAC screening and surveillance concepts. METHODS A selective literature search of current national and international guidelines including underlying literature was performed. RESULTS Nearly half of pancreatic cancer cases are missed by currently available surveillance programs, even in high-risk cohorts. Magnetic resonance imaging and endoscopic ultrasound supplemented by CA19‑9 (± HbA1c) are not accurate enough to ensure robust earlier pancreatic cancer detection. Complementary biomarker panels will take on a crucial diagnostic role in the future.
Collapse
Affiliation(s)
- Simon Sirtl
- Medizinische Klinik und Poliklinik II, LMU Klinikum, 81377, München, Deutschland.
| | - Marlies Vornhülz
- Medizinische Klinik und Poliklinik II, LMU Klinikum, 81377, München, Deutschland
| | - Felix O Hofmann
- Klinik für Allgemein‑, Viszeral- und Transplantationschirurgie, LMU Klinikum, München, Deutschland
| | - Julia Mayerle
- Medizinische Klinik und Poliklinik II, LMU Klinikum, 81377, München, Deutschland.
| | - Georg Beyer
- Medizinische Klinik und Poliklinik II, LMU Klinikum, 81377, München, Deutschland
| |
Collapse
|
19
|
Lee White L, Smith-Gagen J, Elliott L, Lu M. Patient characteristics associated with definitive diagnosis of metastatic pancreatic cancer in those initially diagnosed with cancer of unknown primary. Mol Clin Oncol 2023; 19:101. [PMID: 38022848 PMCID: PMC10666077 DOI: 10.3892/mco.2023.2697] [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: 07/06/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Cancer of unknown primary (CUP) and pancreatic cancer (PC) are malignancies associated with poor prognosis. CUP is the fourth most common cause of cancer mortality in the US, and median survival time is 3-4 months. PC is the third most common cause of cancer mortality in the US, and median survival time for patients with stage 3 or 4 PC is 2-3 months. The present study aimed to understand the patient characteristics of those initially misdiagnosed with CUP who ultimately received a diagnosis of PC. The present study used 2010-2015 Surveillance, Epidemiology, and End Results-Medicare data, a US population-based cancer registry linked to Medicare health insurance claims. Odds ratios (ORs) and 95% confidence intervals were calculated using two binary logistic regression models to compare the characteristics of patients who received definitive diagnosis between the CUP-PC group (those with an initial diagnosis of CUP who eventually received a stage 3 or 4 PC diagnosis) and the PC group (those diagnosed with stage 3 or 4 PC only). Approximately 26% of patients who received a definitive diagnosis of metastatic PC started with an initial diagnosis of CUP (n=17,565). The odds of definitive PC diagnosis in patients with CUP were lower for those with a comorbidity score of 0 [OR, 0.85 (95% CI: 0.79, 0.91)] and epithelial/unspecified histology [OR, 0.76 (95% CI: 0.71, 0.82)]. The odds of definitive PC diagnosis in patients with CUP were higher for patients of other race [OR, 1.27 (95% CI: 1.13, 1.43)] compared with white patients. Definitive diagnosis of PC in patients with CUP was lower in patients who were older with fewer or no comorbidities and unspecified histology. The complexity of CUP diagnosis and patient performance status may influence delays in diagnosis to a known primary site.
Collapse
Affiliation(s)
- Larissa Lee White
- Kaiser Permanente Colorado, Institute for Health Research, Aurora, CO 80014, USA
| | | | - Leslie Elliott
- School of Public Health, University of Nevada, Reno, NV 89557, USA
| | - Minggen Lu
- School of Public Health, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
20
|
Kawakami Y, Koshita S, Kanno Y, Ogawa T, Kusunose H, Sakai T, Yonamine K, Miyamoto K, Kozakai F, Okada T, Oikawa M, Tsuchiya T, Noda Y, Sawai T, Nakase H, Ito K. Pancreatic ductal adenocarcinomas concomitant with intraductal papillary mucinous neoplasms of the pancreas: A investigation of those clinicopathological features by analyzing 48 patients who underwent surgery for those lesions. Pancreatology 2023; 23:674-681. [PMID: 37604732 DOI: 10.1016/j.pan.2023.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND Differences between pancreatic ductal adenocarcinomas (PDACs) concomitant with intraductal papillary mucinous neoplasm (IPMN) (C-PDACs), those without IPMN (NC-PDACs) and invasive cancers derived from IPMN (IC-Ds) have not been fully clarified. METHODS Forty-eight patients with C-PDAC were included to investigate the differences in 1) clinicopathological features and 2) post-operative courses among the three invasive cancer groups. RESULTS 1) Characteristics of C-PDACs were mostly similar to those of NC-PDACs; whereas, between C-PDACs and IC-Ds, the rate of mucinous carcinoma (2%/25%, p = 0.003) and pathological stage (IA, 15%/36%, p = 0.033; III, 31%/4%, p = 0.015) significantly differed. Most C-PDACs coexisted with small, multifocal IPMNs without mural nodules. 2) Cumulative 5-year recurrence-free survival (RFS) rate related to extra-pancreatic recurrence was significantly worse in C-PDACs than in IC-Ds (35%/69%, p = 0.008) and was not significantly different between C-PDACs and NC-PDACs (35%/18%). This related to intra-pancreatic recurrence tended to be poor in the order of IC-Ds, C-PDACs, and NC-PDACs (69%/82%/93%). CONCLUSIONS Because characteristics of IPMNs remarkably differed between C-PDACs and IC-Ds, another algorithm specific to the early detection of C-PDACs is necessary. Appropriate post-operative managements according to the two types of recurrences may contribute to the improvement in the prognoses of C-PDACs/IC-Ds.
Collapse
Affiliation(s)
- Yujiro Kawakami
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan; Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shinsuke Koshita
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan.
| | - Yoshihide Kanno
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan
| | - Takahisa Ogawa
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan
| | - Hiroaki Kusunose
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan
| | - Toshitaka Sakai
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan
| | - Keisuke Yonamine
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan
| | - Kazuaki Miyamoto
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan
| | - Fumisato Kozakai
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan
| | - Takaho Okada
- Department of Surgery, Sendai City Medical Center, Sendai, Japan
| | - Masaya Oikawa
- Department of Surgery, Sendai City Medical Center, Sendai, Japan
| | - Takashi Tsuchiya
- Department of Surgery, Sendai City Medical Center, Sendai, Japan
| | - Yutaka Noda
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan; Department of Pathology, Sendai City Medical Center, Sendai, Japan
| | - Takashi Sawai
- Department of Pathology, Sendai City Medical Center, Sendai, Japan
| | - Hiroshi Nakase
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kei Ito
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan
| |
Collapse
|
21
|
Xu J, Quan G, Huang W, Jiang J. VSIG2 promotes malignant progression of pancreatic ductal adenocarcinoma by enhancing LAMTOR2-mediated mTOR activation. Cell Commun Signal 2023; 21:223. [PMID: 37626304 PMCID: PMC10463957 DOI: 10.1186/s12964-023-01209-x] [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: 04/05/2023] [Accepted: 06/30/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most intractable malignancies to overcome clinically due to its insidious onset as well as rapid progression. It is urgent to seek new diagnostic markers and therapeutic targets in order to furthest ameliorate the prognosis of patients with PDAC. V-set and immunoglobulin domain containing 2 (VSIG2) belongs to immunoglobulin superfamily (IgSF), which function as coinhibitory molecule to mediate immune evasion of tumors. Nevertheless, the role of VSIG2 in PDAC and related mechanism still keep unclear. METHODS Different expression of VSIG2 in PDAC tissues and cells were detected by bioinformatic analysis, immunohistochemistry, real-time quantitative PCR as well as western blotting. CCK-8, colony formation, Transwell assay, and scratch experiment were utilized to assess proliferation, invasion and migration properties of PDAC cells. The relationship of VSIG2 with late endosomal/lysosomal adaptor, MAPK and MTOR activator 2 (LAMTOR2) and mechanistic target of rapamycin (mTOR) was identified using mass spectrometry, co-immunoprecipitation and immunofluorescence. GO and KEGG enrichment analysis were performed for further pathway verification using western blotting. Additionally, subcutaneous xenograft tumor model and clinical samples analysis were implemented to further elucidate the oncogenic effect of VSIG2 on PDAC in vivo and clinically. RESULTS VSIG2 was highly expressed in PDAC tissues and cells. Overexpression of VSIG2 facilitated the proliferation, invasion and migration abilities of PDAC cells, while VSIG2-inhibition exerted opposite effects. Mechanistically, VSIG2 could simultaneously bind to LAMTOR2 and mTOR, thereby enhancing interaction between two molecules, which resulted in elevated phosphorylation-modificatory activation of mTOR and downstream key molecules. Clinically, up-regulation of VSIG2 was positively associated with advanced stage, overall survival and disease-free survival of PDAC patients. CONCLUSIONS Our study disclosed that VSIG2 was overexpressed in PDAC, which promoted the proliferation, invasion and metastasis. Mechanically, VSIG2 acted as a scaffold to recruit LAMTOR2 and mTOR simultaneously, stabilize the interaction between them, thus enhancing LAMTOR2-mediated mTOR phosphorylated activation. Collectively, VSIG2 could be exploited as a biomarker for diagnosis and prognosis monitor of PDAC in the future, meanwhile, targeting VSIG2 in PDAC management is expected to be a novel strategy. Video Abstract. Video Abstract.
Collapse
Affiliation(s)
- Jichuan Xu
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 78 Wandao Road, Wanjiang Street, Dongguan City, Guangdong Province, 523058, People's Republic of China
| | - Gang Quan
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 78 Wandao Road, Wanjiang Street, Dongguan City, Guangdong Province, 523058, People's Republic of China
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Huang
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 78 Wandao Road, Wanjiang Street, Dongguan City, Guangdong Province, 523058, People's Republic of China
| | - Jianxin Jiang
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), 78 Wandao Road, Wanjiang Street, Dongguan City, Guangdong Province, 523058, People's Republic of China.
| |
Collapse
|
22
|
Bahrani HMH, Ghobeh M, Homayouni Tabrizi M. The anticancer, anti-oxidant, and antibacterial activities of chitosan-lecithin-coated parthenolide/tyrosol hybrid nanoparticles. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:1603-1617. [PMID: 36755525 DOI: 10.1080/09205063.2023.2177473] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/29/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023]
Abstract
Tyrosol (TYR) and parthenolide (PLT) have been used as synthetic antioxidant and natural anticancer compounds. In the current study, we aimed to synthesize an encapsulated complex of both PLT and TYR in a hybrid coating layer consisting of lecithin and chitosan molecules, a proper biocompatible drug delivery system to evaluate its antibacterial and anticancer potentials on human liver HepG2 and pancreatic Panc cancer cell lines. The chitosan-lecithin-coated PLT/TYR nanoparticles (clPT-NPs) were synthesized applying an auto-self-assembling method. The clPT-NPs were characterized utilizing DLS, FTIR, zeta potential, and TEM analysis. The clPT-NPs' antioxidant activity was measured by running ABTS and DPPH antioxidant assays. Moreover, the antibacterial potential of clPT-NPs was evaluated by applying disk diffusion, MIC, and MBC assays. Finally, the nanoparticles' cytotoxicity and apoptotic activity were studied by conducting MTT, Flow cytometry, AO/PI cell staining, and real-time PCR techniques. The clPT-NPs (38 nm) exhibited significant antioxidant activity by inhibiting ABTS and DPPH radicals at 187 and 290 μg/mL IC50 concentrations, respectively. Also, the nanoparticles induced a notable antibacterial activity against Staphylococcus aureus at 0.0625 mg/mL MIC and 0.125 mg/mL MBC concentrations. The clPT-NPs selectively decreased the cancer cells' survival and increased the apoptotic dead cells by up-regulating apoptotic gene expression (BAX and Cas-8) and down-regulating BCL-2 anti-apoptotic gene expression. The PLT toxicity has been merged with improved TYR antioxidant activity, which has been functionalized in a safe, biocompatible hybrid nano-delivery system.
Collapse
Affiliation(s)
| | - Maryam Ghobeh
- Department of Biology, Science and Research branch, Islamic Azad University, Tehran, Iran
| | | |
Collapse
|
23
|
Bures J, Kohoutova D, Skrha J, Bunganic B, Ngo O, Suchanek S, Skrha P, Zavoral M. Diabetes Mellitus in Pancreatic Cancer: A Distinct Approach to Older Subjects with New-Onset Diabetes Mellitus. Cancers (Basel) 2023; 15:3669. [PMID: 37509329 PMCID: PMC10377806 DOI: 10.3390/cancers15143669] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/02/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is associated with a very poor prognosis, with near-identical incidence and mortality. According to the World Health Organization Globocan Database, the estimated number of new cases worldwide will rise by 70% between 2020 and 2040. There are no effective screening methods available so far, even for high-risk individuals. The prognosis of PDAC, even at its early stages, is still mostly unsatisfactory. Impaired glucose metabolism is present in about 3/4 of PDAC cases. METHODS Available literature on pancreatic cancer and diabetes mellitus was reviewed using a PubMed database. Data from a national oncology registry (on PDAC) and information from a registry of healthcare providers (on diabetes mellitus and a number of abdominal ultrasound investigations) were obtained. RESULTS New-onset diabetes mellitus in subjects older than 60 years should be an incentive for a prompt and detailed investigation to exclude PDAC. Type 2 diabetes mellitus, diabetes mellitus associated with chronic non-malignant diseases of the exocrine pancreas, and PDAC-associated type 3c diabetes mellitus are the most frequent types. Proper differentiation of particular types of new-onset diabetes mellitus is a starting point for a population-based program. An algorithm for subsequent steps of the workup was proposed. CONCLUSIONS The structured, well-differentiated, and elaborately designed approach to the elderly with a new onset of diabetes mellitus could improve the current situation in diagnostics and subsequent poor outcomes of therapy of PDAC.
Collapse
Affiliation(s)
- Jan Bures
- Institute of Gastrointestinal Oncology, Military University Hospital Prague, 169 02 Prague, Czech Republic
- Department of Medicine, First Faculty of Medicine, Charles University, Prague and Military University Hospital Prague, 169 02 Prague, Czech Republic
- Biomedical Research Centre, University Hospital Hradec Kralove, 500 03 Hradec Kralove, Czech Republic
| | - Darina Kohoutova
- Biomedical Research Centre, University Hospital Hradec Kralove, 500 03 Hradec Kralove, Czech Republic
- The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK
| | - Jan Skrha
- Third Department of Internal Medicine-Endocrinology and Metabolism, First Faculty of Medicine, Charles University, Prague and General University Hospital in Prague, 128 08 Prague, Czech Republic
| | - Bohus Bunganic
- Department of Medicine, First Faculty of Medicine, Charles University, Prague and Military University Hospital Prague, 169 02 Prague, Czech Republic
| | - Ondrej Ngo
- Institute of Health Information and Statistics of the Czech Republic, 128 01 Prague, Czech Republic
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, 602 00 Brno, Czech Republic
| | - Stepan Suchanek
- Institute of Gastrointestinal Oncology, Military University Hospital Prague, 169 02 Prague, Czech Republic
- Department of Medicine, First Faculty of Medicine, Charles University, Prague and Military University Hospital Prague, 169 02 Prague, Czech Republic
| | - Pavel Skrha
- Department of Medicine, Third Faculty of Medicine, Charles University, Prague and University Hospital Kralovske Vinohrady, 100 00 Prague, Czech Republic
| | - Miroslav Zavoral
- Institute of Gastrointestinal Oncology, Military University Hospital Prague, 169 02 Prague, Czech Republic
- Department of Medicine, First Faculty of Medicine, Charles University, Prague and Military University Hospital Prague, 169 02 Prague, Czech Republic
| |
Collapse
|
24
|
White L, Smith Gagen J, Elliott L, Lu M. Patient Characteristics Associated with Definitive Diagnosis of Metastatic Pancreatic Cancer in Those Initially Diagnosed with Cancer of Unknown Primary. RESEARCH SQUARE 2023:rs.3.rs-2833560. [PMID: 37131591 PMCID: PMC10153360 DOI: 10.21203/rs.3.rs-2833560/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Purpose Cancer of unknown primary (CUP) is the fourth most common cause of cancer mortality in the U.S. Median survival after CUP diagnosis is 3-4 months. As CUP and metastatic pancreatic cancer (PC) are comparable in prevalence and survival, PC diagnosis is a useful endpoint to assess patient characteristics associated with definitive diagnosis in older patients who initially present with CUP. Methods This study used 2010-2015 SEER-Medicare data. Logistic regression models compared patient characteristics who received definitive diagnosis in two subsets: CUP-PC and PC only. Results Approximately 26% of patients who received a definitive diagnosis of metastatic pancreatic cancer started with an initial diagnosis of CUP (n=17,565). The odds of definitive diagnosis in CUP-PC were lower for those with a comorbidity score of 0 (OR 0.85 [0.79, 0.91]) and epithelial/unspecified histology (OR 0.76 [0.71, 0.82]). The odds of definitive diagnosis in CUP-PC were higher for patients of Other race (OR 1.27 [1.13, 1.43]) compared to White patients. Conclusion Definitive diagnosis of CUP-PC was favorable in patients in the Other race category with fewer or no comorbidities. Unfavorable characteristics included older patients and those with epithelial/unspecified histology. Future studies will focus on patterns of care and survival in patients with CUP-PC.
Collapse
Affiliation(s)
- Larissa White
- Kaiser Permanente Colorado Institute for Health Research
| | | | | | | |
Collapse
|
25
|
Clinical Features and Outcomes of Patients with Pancreaticobiliary Malignancies in Los Angeles County and Their Association with CA 19-9 Levels. Cancers (Basel) 2023; 15:cancers15061723. [PMID: 36980609 PMCID: PMC10046349 DOI: 10.3390/cancers15061723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/23/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Although CA 19-9 is a commonly used tumor marker in the management of PBMs, the literature describing outcomes in patients with PBMs who have undetectable or low (hereinafter “low”) CA 19-9 levels remains scarce. In this study, we sought to compare clinical features and outcomes in patients with PBMs and low CA 19-9 levels to those with normal and elevated CA 19-9 levels. Methods: We retrospectively collected data on patients with biopsy-confirmed PBMs and stratified patients into categories based on their CA 19-9 level at diagnosis. Survival curves were estimated for patients in each of the three aforementioned CA 19-9 groups using the Kaplan–Meier method and compared using a Cox proportional hazards regression model. Results: Of the 283 patients identified, 23 (8.1%) had low, 70 (24.7%) had normal, and 190 (67.1%) had elevated CA 19-9 levels. After controlling for sex, age, BMI, the presence of metastases at the time of diagnosis, and treatment with curative intent, the hazard ratio for death in the elevated CA 19-9 group compared to the low CA 19-9 group was 1.993 (95% CI 1.089–3.648; p = 0.025). Conclusion: The elevated CA 19-9 level compared to the low CA 19-9 level and the presence of metastases were associated with an increased hazard of death, while treatment with curative intent was associated with a decreased hazard of death.
Collapse
|
26
|
Del Nero L, Dabizzi E, Ceglie AD, Ziola S, Zerbi A, Baron TH, Conio M. Familial pancreatic cancer. Clin Res Hepatol Gastroenterol 2023; 47:102079. [PMID: 36681116 DOI: 10.1016/j.clinre.2023.102079] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/19/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023]
Abstract
Pancreatic cancer (PC) carries a poor prognosis with an overall 5-year survival of less than 10%. Early diagnosis, though cumbersome, is essential to allow complete surgical resection. Therefore, primary and secondary prevention are critical to reduce the incidence and to potentially prevent mortality. Given a relatively low lifetime risk of developing PC, identification of high-risk individuals is crucial to allow identification of pre-malignant lesions and small, localized tumors. Although 85-90% of PC cases are sporadic, we could consider risk stratification for the 5-10% of patients with a family history and the 3-5% of cases due to inherited genetic syndromes. These high-risk populations should be considered for screening and surveillance of PC. MRI/MRCP and EUS are the preferred modalities, due to their high sensitivity in lesion detection. Surveillance should be personalized, considering genetics and family history, and assessment of risk factors that may increase cancer risk. Screening programs should be limited to tertiary referral center, with high-volumes and adequate facilities to manage these patients.
Collapse
Affiliation(s)
- Lorenzo Del Nero
- Gastroenterology Department, Santa Corona Hospital, Pietra Ligure, ASL 2 Savonese, Italy.
| | - Emanuele Dabizzi
- Gastroenterology and Interventional Endoscopy Unit, AUSL Bologna, Surgical Department, Bologna, Italy
| | - Antonella De Ceglie
- Gastroenterology Department, Sanremo General Hospital, Sanremo (IM), ASL1 Imperiese, Italy
| | - Sebastiano Ziola
- Gastroenterology Department, Santa Corona Hospital, Pietra Ligure, ASL 2 Savonese, Italy
| | - Alessandro Zerbi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy. IRCCS Istituto Clinico Humanitas, Rozzano, Milan, Italy
| | - Todd H Baron
- Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Massimo Conio
- Gastroenterology Department, Santa Corona Hospital, Pietra Ligure, ASL 2 Savonese, Italy; Endoscopie, Clinique Saint George, Nice, France
| |
Collapse
|
27
|
Shetty NS, Agarwal U, Choudhari A, Gupta A, PG N, Bhandare M, Gala K, Chandra D, Ramaswamy A, Ostwal V, Shrikhande SV, Kulkarni SS. Imaging Recommendations for Diagnosis, Staging, and Management of Pancreatic Cancer. Indian J Med Paediatr Oncol 2023. [DOI: 10.1055/s-0042-1759521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
AbstractPancreatic cancer is the fourth most prevalent cause of cancer-related death worldwide, with a fatality rate equal to its incidence rate. Pancreatic cancer is a rare malignancy with a global incidence and death ranking of 14th and 7th, respectively. Pancreatic cancer cases are divided into three categories without metastatic disease: resectable, borderline resectable, or locally advanced disease. The category is determined by the tumor's location in the pancreas and whether it is abutting or encasing the adjacent arteries and/or vein/s.The stage of disease and the location of the primary tumor determine the clinical presentation: the pancreatic head, neck, or uncinate process, the body or tail, or multifocal disease. Imaging plays a crucial role in the diagnosis and follow-up of pancreatic cancers. Various imaging modalities available for pancreatic imaging are ultrasonography (USG), contrast-enhanced computed tomography (CECT), magnetic resonance imaging (MRI), and 18-fluoro-deoxy glucose positron emission tomography (FDG PET).Even though surgical resection is possible in both resectable and borderline resectable non-metastatic cases, neoadjuvant chemotherapy with or without radiotherapy has become the standard practice for borderline resectable cases as it gives a high yield of R0 resection.
Collapse
Affiliation(s)
- Nitin Sudhakar Shetty
- Department of Radio-Diagnosis, Tata Memorial Hospital, Homi Bhabha National University (HBNI), Mumbai, Maharashtra, India
| | - Ujjwal Agarwal
- Department of Radio-Diagnosis, Tata Memorial Hospital, Homi Bhabha National University (HBNI), Mumbai, Maharashtra, India
| | - Amit Choudhari
- Department of Radio-Diagnosis, Tata Memorial Hospital, Homi Bhabha National University (HBNI), Mumbai, Maharashtra, India
| | - Anurag Gupta
- Department of Radio-Diagnosis, Tata Memorial Hospital, Homi Bhabha National University (HBNI), Mumbai, Maharashtra, India
| | - Nandakumar PG
- Department of Radio-Diagnosis, Tata Memorial Hospital, Homi Bhabha National University (HBNI), Mumbai, Maharashtra, India
| | - Manish Bhandare
- Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National University (HBNI), Mumbai, Maharashtra, India
| | - Kunal Gala
- Department of Radio-Diagnosis, Tata Memorial Hospital, Homi Bhabha National University (HBNI), Mumbai, Maharashtra, India
| | - Daksh Chandra
- Department of Radio-Diagnosis, Tata Memorial Hospital, Homi Bhabha National University (HBNI), Mumbai, Maharashtra, India
| | - Anant Ramaswamy
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National University (HBNI), Mumbai, Maharashtra, India
| | - Vikas Ostwal
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National University (HBNI), Mumbai, Maharashtra, India
| | - Shailesh V. Shrikhande
- Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National University (HBNI), Mumbai, Maharashtra, India
| | - Suyash S. Kulkarni
- Department of Radio-Diagnosis, Tata Memorial Hospital, Homi Bhabha National University (HBNI), Mumbai, Maharashtra, India
| |
Collapse
|
28
|
Granata V, Fusco R, Setola SV, Galdiero R, Maggialetti N, Silvestro L, De Bellis M, Di Girolamo E, Grazzini G, Chiti G, Brunese MC, Belli A, Patrone R, Palaia R, Avallone A, Petrillo A, Izzo F. Risk Assessment and Pancreatic Cancer: Diagnostic Management and Artificial Intelligence. Cancers (Basel) 2023; 15:351. [PMID: 36672301 PMCID: PMC9857317 DOI: 10.3390/cancers15020351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Pancreatic cancer (PC) is one of the deadliest cancers, and it is responsible for a number of deaths almost equal to its incidence. The high mortality rate is correlated with several explanations; the main one is the late disease stage at which the majority of patients are diagnosed. Since surgical resection has been recognised as the only curative treatment, a PC diagnosis at the initial stage is believed the main tool to improve survival. Therefore, patient stratification according to familial and genetic risk and the creation of screening protocol by using minimally invasive diagnostic tools would be appropriate. Pancreatic cystic neoplasms (PCNs) are subsets of lesions which deserve special management to avoid overtreatment. The current PC screening programs are based on the annual employment of magnetic resonance imaging with cholangiopancreatography sequences (MR/MRCP) and/or endoscopic ultrasonography (EUS). For patients unfit for MRI, computed tomography (CT) could be proposed, although CT results in lower detection rates, compared to MRI, for small lesions. The actual major limit is the incapacity to detect and characterize the pancreatic intraepithelial neoplasia (PanIN) by EUS and MR/MRCP. The possibility of utilizing artificial intelligence models to evaluate higher-risk patients could favour the diagnosis of these entities, although more data are needed to support the real utility of these applications in the field of screening. For these motives, it would be appropriate to realize screening programs in research settings.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, 41012 Napoli, Italy
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via della Signora 2, 20122 Milan, Italy
| | - Sergio Venanzio Setola
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Roberta Galdiero
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Nicola Maggialetti
- Department of Medical Science, Neuroscience and Sensory Organs (DSMBNOS), University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Lucrezia Silvestro
- Division of Clinical Experimental Oncology Abdomen, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Mario De Bellis
- Division of Gastroenterology and Digestive Endoscopy, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Elena Di Girolamo
- Division of Gastroenterology and Digestive Endoscopy, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Giulia Grazzini
- Department of Emergency Radiology, University Hospital Careggi, Largo Brambilla 3, 50134 Florence, Italy
| | - Giuditta Chiti
- Department of Emergency Radiology, University Hospital Careggi, Largo Brambilla 3, 50134 Florence, Italy
| | - Maria Chiara Brunese
- Diagnostic Imaging Section, Department of Medical and Surgical Sciences & Neurosciences, University of Molise, 86100 Campobasso, Italy
| | - Andrea Belli
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Renato Patrone
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Raffaele Palaia
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Antonio Avallone
- Division of Clinical Experimental Oncology Abdomen, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Antonella Petrillo
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Francesco Izzo
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| |
Collapse
|
29
|
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: 3.0] [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
|
30
|
Sagami R, Hayasaka K, Ujihara T, Iwaki T, Katsuyama Y, Harada H, Ome Y, Honda G, Horiguchi SI, Murakami K, Amano Y. Role of EUS combined with a newly modified scoring system to detect pancreatic high-grade precancerous lesions. Endosc Ultrasound 2023; 12:111-119. [PMID: 36861510 PMCID: PMC10134925 DOI: 10.4103/eus-d-21-00187] [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: 08/24/2021] [Accepted: 08/01/2022] [Indexed: 03/03/2023] Open
Abstract
Backgrounds and Objectives Although pancreatic cancer (PC) has an extremely poor prognosis, the 5-year survival rate of patients with pancreatic high-grade precancerous lesion without invasive carcinoma (PHP) is favorable. PHP diagnosis and identification of patients requiring intervention are needed. We aimed to validate a modified PC detection scoring system regarding its detection ability for PHP and PC in the general population. Subjects and Methods We modified an existing PC detection scoring system that incorporates low-grade risk (LGR) factors (family history, presence of diabetes mellitus [DM] or worsening DM, heavy drinking, smoking, stomach symptoms, weight loss, and pancreatic enzyme) and high-grade risk (HGR) factors (new-onset DM, familial PC, jaundice, tumor biomarkers, chronic pancreatitis, intraductal papillary mucinous neoplasm, cysts, hereditary PC syndrome, and hereditary pancreatitis). Each factor was scored as one point; LGR score ≥3 points and/or HGR score ≥1 point (positive scores) were indicative of PC. The newly modified scoring system incorporated main pancreatic duct dilation as an HGR factor. The PHP diagnosis rate using this scoring system combined with EUS was prospectively analyzed. Results Among 544 patients with positive scores, 10 had PHP. The diagnosis rates were 1.8% for PHP and 4.2% for invasive PC. Although the number of LGR and HGR factors tended to increase with PC progression, none of the individual factors were significantly different between patients with PHP and those without lesions. Conclusion The newly modified scoring system evaluating multiple factors associated with PC could potentially identify patients with higher risk of PHP or PC.
Collapse
Affiliation(s)
- Ryota Sagami
- Department of Gastroenterology, Oita San-ai Medical Center, Oita, Japan
| | - Kenji Hayasaka
- Department of Gastroenterology, New Tokyo Hospital, Matsudo, Chiba, Japan
| | - Tetsuro Ujihara
- Department of Gastroenterology, New Tokyo Hospital, Matsudo, Chiba, Japan
| | - Tomoyuki Iwaki
- Department of Gastroenterology, Urawa Kyosai Hospital, Saitama, Japan
| | - Yasushi Katsuyama
- Department of Gastroenterology, Urawa Kyosai Hospital, Saitama, Japan
| | - Hideaki Harada
- Department of Gastroenterology, Urawa Kyosai Hospital, Saitama, Japan
| | - Yusuke Ome
- Department of Surgery, Institute of Gastroenterology, Tokyo Women’s Medical University, Shinjuku-Ku, Tokyo, Japan
| | - Goro Honda
- Department of Surgery, Institute of Gastroenterology, Tokyo Women’s Medical University, Shinjuku-Ku, Tokyo, Japan
| | - Shin-ichiro Horiguchi
- Department of Pathology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Bunkyo, Tokyo, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Yuji Amano
- Department of Gastroenterology, Urawa Kyosai Hospital, Saitama, Japan
| |
Collapse
|
31
|
Park HY, Suh CH, Kim SO. Use of "Diagnostic Yield" in Imaging Research Reports: Results from Articles Published in Two General Radiology Journals. Korean J Radiol 2022; 23:1290-1300. [PMID: 36447417 PMCID: PMC9747267 DOI: 10.3348/kjr.2022.0741] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE "Diagnostic yield," also referred to as the detection rate, is a parameter positioned between diagnostic accuracy and diagnosis-related patient outcomes in research studies that assess diagnostic tests. Unfamiliarity with the term may lead to incorrect usage and delivery of information. Herein, we evaluate the level of proper use of the term "diagnostic yield" and its related parameters in articles published in Radiology and Korean Journal of Radiology (KJR). MATERIALS AND METHODS Potentially relevant articles published since 2012 in these journals were identified using MEDLINE and PubMed Central databases. The initial search yielded 239 articles. We evaluated whether the correct definition and study setting of "diagnostic yield" or "detection rate" were used and whether the articles also reported companion parameters for false-positive results. We calculated the proportion of articles that correctly used these parameters and evaluated whether the proportion increased with time (2012-2016 vs. 2017-2022). RESULTS Among 39 eligible articles (19 from Radiology and 20 from KJR), 17 (43.6%; 11 from Radiology and 6 from KJR) correctly defined "diagnostic yield" or "detection rate." The remaining 22 articles used "diagnostic yield" or "detection rate" with incorrect meanings such as "diagnostic performance" or "sensitivity." The proportion of correctly used diagnostic terms was higher in the studies published in Radiology than in those published in KJR (57.9% vs. 30.0%). The proportion improved with time in Radiology (33.3% vs. 80.0%), whereas no improvement was observed in KJR over time (33.3% vs. 27.3%). The proportion of studies reporting companion parameters was similar between journals (72.7% vs. 66.7%), and no considerable improvement was observed over time. CONCLUSION Overall, a minority of articles accurately used "diagnostic yield" or "detection rate." Incorrect usage of the terms was more frequent without improvement over time in KJR than in Radiology. Therefore, improvements are required in the use and reporting of these parameters.
Collapse
Affiliation(s)
- Ho Young Park
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chong Hyun Suh
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seon-Ok Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
32
|
Jatoi I. Mitigating Cancer Overdiagnosis. Indian J Surg Oncol 2022; 13:671-673. [PMID: 36687223 PMCID: PMC9845438 DOI: 10.1007/s13193-022-01546-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/03/2022] [Indexed: 01/25/2023] Open
Abstract
Overdiagnosis refers to the detection of cancers that pose no threat to life and would never have been detected in the absence of screening. It is now a major public health concern throughout the world because advancements in screening technology have substantially increased the detection rates of non-lethal cancers. Cancer overdiagnosis leads to unnecessary treatments, putting patients unnecessarily at risk for the potential morbidity and mortality that may arise from those treatments, and it wastes healthcare resources. Patients may also suffer unnecessary anxiety, job discrimination, financial hardships, and other detrimental effects on quality-of-life following cancer overdiagnosis. In this article, I provide examples of cancer overdiagnosis as a consequence of screening for thyroid, breast, and pancreatic cancers. To mitigate the adverse effects of cancer overdiagnosis, we should adopt only those screening strategies that have been shown in randomized trials to reduce cancer mortality, and we should inform patients about the potential benefits and risks of cancer screening, particularly the risk for overdiagnosis.
Collapse
Affiliation(s)
- Ismail Jatoi
- Division of Surgical Oncology and Endocrine Surgery, University of Texas Health Science Center, San Antonio, TX 78229 USA
| |
Collapse
|
33
|
Rosenthal M, Schawkat K, Wolpin B. A Growing Hope for Earlier Detection of Pancreatic Cancer. Gastroenterology 2022; 163:1170-1172. [PMID: 35961377 DOI: 10.1053/j.gastro.2022.07.080] [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: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/02/2022]
Affiliation(s)
- Michael Rosenthal
- Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Khoschy Schawkat
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Brian Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
34
|
Hong MZ, Li JM, Chen ZJ, Lin XY, Pan JS, Gong LL. Global burden of major gastrointestinal cancers and its association with socioeconomics, 1990-2019. Front Oncol 2022; 12:942035. [PMID: 36387124 PMCID: PMC9664003 DOI: 10.3389/fonc.2022.942035] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/19/2022] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND To understand the impact of common cancers of the gastrointestinal tract and help to formulate evidence-based policy, we evaluate the relationship between the burden of GI tract cancers and socioeconomics. METHODS Data on GI tract cancer burden were obtained from the Global Burden of Disease (GBD) 2019 including mortality and incidence rates. According to the Socio-demographic Index (SDI) level, country and territory, and sex, etc., the data were further stratified. The association between the burden of GI tract cancer and socioeconomics, indicated by SDI, was described. Uncertainty analysis was estimated using bootstrap draw. RESULTS In 2019, five major cancers of the gastrointestinal tract led to an age-standardized incidence rate (ASIR) of 61.9 (95% CI 56.1-67.6) per 100 000 person-years. From 1990 to 2019, five common tumors of the gastrointestinal tract related age-standardized death rates (ASDRs) decreased by -22.7% (-31.1 to -13.5). For the five common tumors, ASIRs and ASDRs were both higher in males than those in females. Globally, Mongolia, and several East Asia countries exhibited the highest ASIRs in 2019. The high SDI, and high-middle SDI locations recorded the highest incidence rate and death rate of colon and rectum cancer and pancreatic cancer. On the contrary, the low-middle SDI, and low SDI locations possessed the highest incidence rate and death rate of stomach cancer and esophageal cancer. CONCLUSION There is a profound association between socioeconomics and burden of common cancers of the gastrointestinal tract. It would be helpful for the high SDI, and high-middle SDI locations to pay special attention to the screening of colon and rectum cancer and pancreatic cancer while the low-middle SDI, and low SDI locations should pay more attention to the screening of stomach cancer and esophageal cancer.
Collapse
Affiliation(s)
- Mei-Zhu Hong
- Department of Traditional Chinese Medicine, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jing-Mao Li
- Department of Statistics, School of Economics, Xiamen University, Xiamen, Fujian, China
| | - Zhi-Jian Chen
- Department of Hepatology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xiao-Yun Lin
- Department of Hepatology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jin-Shui Pan
- Department of Hepatology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Li-Li Gong
- Department of Health Care, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
35
|
Dbouk M, Katona BW, Brand RE, Chak A, Syngal S, Farrell JJ, Kastrinos F, Stoffel EM, Blackford AL, Rustgi AK, Dudley B, Lee LS, Chhoda A, Kwon R, Ginsberg GG, Klein AP, Kamel I, Hruban RH, He J, Shin EJ, Lennon AM, Canto MI, Goggins M. The Multicenter Cancer of Pancreas Screening Study: Impact on Stage and Survival. J Clin Oncol 2022; 40:3257-3266. [PMID: 35704792 PMCID: PMC9553376 DOI: 10.1200/jco.22.00298] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/29/2022] [Accepted: 05/11/2022] [Indexed: 01/21/2023] Open
Abstract
PURPOSE To report pancreas surveillance outcomes of high-risk individuals within the multicenter Cancer of Pancreas Screening-5 (CAPS5) study and to update outcomes of patients enrolled in prior CAPS studies. METHODS Individuals recommended for pancreas surveillance were prospectively enrolled into one of eight CAPS5 study centers between 2014 and 2021. The primary end point was the stage distribution of pancreatic ductal adenocarcinoma (PDAC) detected (stage I v higher-stage). Overall survival was determined using the Kaplan-Meier method. RESULTS Of 1,461 high-risk individuals enrolled into CAPS5, 48.5% had a pathogenic variant in a PDAC-susceptibility gene. Ten patients were diagnosed with PDAC, one of whom was diagnosed with metastatic PDAC 4 years after dropping out of surveillance. Of the remaining nine, seven (77.8%) had a stage I PDAC (by surgical pathology) detected during surveillance; one had stage II, and one had stage III disease. Seven of these nine patients with PDAC were alive after a median follow-up of 2.6 years. Eight additional patients underwent surgical resection for worrisome lesions; three had high-grade and five had low-grade dysplasia in their resected specimens. In the entire CAPS cohort (CAPS1-5 studies, 1,731 patients), 26 PDAC cases have been diagnosed, 19 within surveillance, 57.9% of whom had stage I and 5.2% had stage IV disease. By contrast, six of the seven PDACs (85.7%) detected outside surveillance were stage IV. Five-year survival to date of the patients with a screen-detected PDAC is 73.3%, and median overall survival is 9.8 years, compared with 1.5 years for patients diagnosed with PDAC outside surveillance (hazard ratio [95% CI]; 0.13 [0.03 to 0.50], P = .003). CONCLUSION Most pancreatic cancers diagnosed within the CAPS high-risk cohort in the recent years have had stage I disease with long-term survival.
Collapse
Affiliation(s)
- Mohamad Dbouk
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Bryson W. Katona
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Randall E. Brand
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Amitabh Chak
- Division of Gastroenterology and Liver Disease, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH
| | - Sapna Syngal
- Cancer Genetics and Prevention Division, Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Gastroenterology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| | - James J. Farrell
- Yale Center for Pancreatic Disease, Section of Digestive Disease, Yale University, New Haven, CT
| | - Fay Kastrinos
- Division of Digestive and Liver Diseases, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Elena M. Stoffel
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Amanda L. Blackford
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Anil K. Rustgi
- Yale Center for Pancreatic Disease, Section of Digestive Disease, Yale University, New Haven, CT
| | - Beth Dudley
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Linda S. Lee
- Cancer Genetics and Prevention Division, Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Gastroenterology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| | - Ankit Chhoda
- Yale Center for Pancreatic Disease, Section of Digestive Disease, Yale University, New Haven, CT
| | - Richard Kwon
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Gregory G. Ginsberg
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Alison P. Klein
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Ihab Kamel
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Bloomberg School of Public Health, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Ralph H. Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Jin He
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Radiology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Eun Ji Shin
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Anne Marie Lennon
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Bloomberg School of Public Health, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Radiology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Marcia Irene Canto
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Michael Goggins
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|
36
|
Akushevich I, Yashkin A, Kovtun M, Yashin AI, Kravchenko J. Underlying mechanisms of change in cancer prevalence in older U.S. adults: contributions of incidence, survival, and ascertainment at early stages. Cancer Causes Control 2022; 33:1161-1172. [PMID: 35799033 PMCID: PMC9360135 DOI: 10.1007/s10552-022-01595-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022]
Abstract
PURPOSE To quantitatively evaluate contributions of trends in incidence, relative survival, and stage at diagnosis to the dynamics in the prevalence of major cancers (lung, prostate, colon, breast, urinary bladder, ovaries, stomach, pancreas, esophagus, kidney, liver, and skin melanoma) among older U.S. adults age 65 +. METHODS Trend partitioning was applied to the Surveillance, Epidemiology, and End Results Program data for 1973-2016. RESULTS Growth of cancer prevalence in older adults decelerated or even decreased over time for all studied cancers due to decreasing incidence and improving survival for most of cancers, with a smaller contribution of the stage at cancer diagnosis. Changes in the prevalence of cancers of the lung, colon, stomach, and breast were predominantly due to decreasing incidence, increasing survival and more frequent diagnoses at earlier stages. Changes in prevalence of some other cancers demonstrated adverse trends such as decreasing survival in localized and regional stages (urinary bladder and ovarian) and growing impact of late-stage diagnoses (esophageal cancer). CONCLUSION While decelerating or decreasing prevalence of many cancers were due to a beneficial combination of decreasing incidence and increasing survival, there are cancers for which decelerating prevalence is due to lack of improvement in their stage-specific survival and/or increasing frequency of diagnosis at advanced stages. Overall, if the observed trends persist, it is likely that the burden associated with cancer prevalence in older U.S. adults will be lower comparing to projections based on constant increasing prevalence have previously estimated.
Collapse
Affiliation(s)
- I Akushevich
- Center for Population Health and Aging, Duke University, Durham, NC, USA.
| | - A Yashkin
- Center for Population Health and Aging, Duke University, Durham, NC, USA
| | - M Kovtun
- Center for Population Health and Aging, Duke University, Durham, NC, USA
| | - A I Yashin
- Center for Population Health and Aging, Duke University, Durham, NC, USA
| | - J Kravchenko
- Department of Surgery, Duke University School of Medicine, Duke University, Durham, NC, USA
| |
Collapse
|
37
|
Peters MLB, Eckel A, Lietz A, Seguin C, Mueller P, Hur C, Pandharipande PV. Genetic testing to guide screening for pancreatic ductal adenocarcinoma: Results of a microsimulation model. Pancreatology 2022; 22:760-769. [PMID: 35752568 PMCID: PMC9474673 DOI: 10.1016/j.pan.2022.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND First-degree relatives (FDRs) of patients with pancreatic ductal adenocarcinoma (PDAC) have elevated PDAC risk, partially due to germline genetic variants. We evaluated the potential effectiveness of genetic testing to target MRI-based screening among FDRs. METHODS We used a microsimulation model of PDAC, calibrated to Surveillance, Epidemiology, and End Results (SEER) data, to estimate the potential life expectancy (LE) gain of screening for each of the following groups of FDRs: individuals who test positive for each of eight variants associated with elevated PDAC risk (e.g., BRCA2, CDKN2A); individuals who test negative; and individuals who do not test. Screening was assumed to take place if LE gains were achievable. We simulated multiple screening approaches, defined by starting age and frequency. Sensitivity analysis evaluated changes in results given varying model assumptions. RESULTS For women, 92% of mutation carriers had projected LE gains from screening for PDAC, if screening strategies (start age, frequency) were optimized. Among carriers, LE gains ranged from 0.1 days (ATM+ women screened once at age 70) to 510 days (STK11+ women screened annually from age 40). For men, LE gains were projected for all mutation carriers, ranging from 0.2 days (BRCA1+ men screened once at age 70) to 620 days (STK11+ men screened annually from age 40). For men and women who did not undergo genetic testing, or for whom testing showed no variant, screening yielded small LE benefit (0-2.1 days). CONCLUSIONS Genetic testing of FDRs can inform targeted PDAC screening by identifying which FDRs may benefit.
Collapse
Affiliation(s)
- Mary Linton B Peters
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, USA.
| | - Andrew Eckel
- Institute for Technology Assessment, Massachusetts General Hospital, USA
| | - Anna Lietz
- Institute for Technology Assessment, Massachusetts General Hospital, USA
| | - Claudia Seguin
- Institute for Technology Assessment, Massachusetts General Hospital, USA
| | - Peter Mueller
- Institute for Technology Assessment, Massachusetts General Hospital, USA
| | - Chin Hur
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Current Affiliation: Division of Gastroenterology, Columbia University College of Physicians and Surgeons, USA
| | - Pari V Pandharipande
- Institute for Technology Assessment and Department of Radiology, Massachusetts General Hospital, USA
| |
Collapse
|
38
|
Vanek P, Urban O, Zoundjiekpon V, Falt P. Current Screening Strategies for Pancreatic Cancer. Biomedicines 2022; 10:biomedicines10092056. [PMID: 36140157 PMCID: PMC9495594 DOI: 10.3390/biomedicines10092056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a dreaded malignancy with a dismal 5-year survival rate despite maximal efforts on optimizing treatment strategies. Radical surgery is the only potential curative procedure. Unfortunately, the majority of patients are diagnosed with locally advanced or metastatic disease, which renders them ineligible for curative resection. Early detection of PDAC is thus considered to be the most effective way to improve survival. In this regard, pancreatic screening has been proposed to improve results by detecting asymptomatic stages of PDAC and its precursors. There is now evidence of benefits of systematic surveillance in high-risk individuals, and the current guidelines emphasize the potential of screening to affect overall survival in individuals with genetic susceptibility syndromes or familial occurrence of PDAC. Here we aim to summarize the current knowledge about screening strategies for PDAC, including the latest epidemiological data, risk factors, associated hereditary syndromes, available screening modalities, benefits, limitations, as well as management implications.
Collapse
|
39
|
Kandiah J, Lo T, Jin D, Melchior L, Krebs TL, Anand N, Ingram S, Krumholtz P, Pandya D, Trinidad A, Dong X(E, Seshadri R, Bauman J, Lee R, Frank RC. A Community-Based Pancreatic Cancer Screening Study in High-Risk Individuals: Preliminary Efficacy and Safety Results. Clin Transl Gastroenterol 2022; 13:e00516. [PMID: 35854467 PMCID: PMC9400932 DOI: 10.14309/ctg.0000000000000516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 06/09/2022] [Accepted: 06/30/2022] [Indexed: 11/28/2022] Open
Abstract
INTRODUCTION Pancreatic cancer (PC) screening recommendations have been based on studies performed solely at high-volume academic centers. To make PC screening more widely available, community-based efforts are essential. We implemented a prospective PC screening study in the community of Fairfield County, CT, and report our early safety and efficacy results. METHODS Eligible individuals were enrolled into an investigator-initiated study and underwent a baseline and 3 annual magnetic resonance imagings/magnetic resonance cholangiopancreatographies (MRIs/MRCPs) with gadolinium, biannual blood donations for biobanking, and assessments for anxiety and depression. All MRIs were presented at a multidisciplinary board to determine whether further investigation was warranted. RESULTS Seventy-five individuals have been enrolled and 201 MRIs performed over a 2.6-year average length of follow-up. Abnormal pancreatic findings (predominantly small cysts) were detected in 58.7% of the participants. Among these, 6.7% underwent endoscopic ultrasound, with 1 case complicated by postprocedural pancreatitis. One surgical resection was performed on a 4.7-cm intraductal papillary mucinous neoplasm with a focus on low-grade pancreatic intraepithelial neoplasia. One incidental finding of fibrosing mediastinitis was detected. Anxiety and depression scores decreased over the course of this study from 21.4% to 5.4% and 10.7% to 3.6%, respectively. DISCUSSION This preliminary report supports the feasibility of performing MRI/magnetic resonance cholangiopancreatographies-based PC screening as part of a clinical trial in a community setting. A longer follow-up is needed to better assess safety and efficacy. To the best of our knowledge, this is the first report from a community-based PC screening effort ( clinicaltrials.gov ID: NCT03250078).
Collapse
Affiliation(s)
- Jonathan Kandiah
- Department of Gastroenterology and Hepatology, Nuvance Health, Danbury, Connecticut, USA
| | - Tammy Lo
- Department of Medicine, Nuvance Health, Danbury, Connecticut, USA
| | - Dugho Jin
- Department of Radiology, Nuvance Health, Danbury, Connecticut, USA
| | - Landon Melchior
- Department of Radiology, Nuvance Health, Danbury, Connecticut, USA
| | | | - Naveen Anand
- Department of Gastroenterology and Hepatology, Nuvance Health, Danbury, Connecticut, USA
| | - Susan Ingram
- Department of Genetics, Nuvance Health, Danbury, Connecticut, USA
| | | | - Deep Pandya
- Rudy L. Ruggles Biomedical Research Institute, Nuvance Health, Danbury, Connecticut, USA
| | - Antolin Trinidad
- Department of Psychiatry, Nuvance Health, Danbury, Connecticut, USA
| | | | | | - James Bauman
- Department of Radiology, Nuvance Health, Danbury, Connecticut, USA
| | - Ronald Lee
- Department of Radiology, Nuvance Health, Danbury, Connecticut, USA
| | - Richard C. Frank
- Department of Gastroenterology and Hepatology, Nuvance Health, Danbury, Connecticut, USA
- Rudy L. Ruggles Biomedical Research Institute, Nuvance Health, Danbury, Connecticut, USA
| |
Collapse
|
40
|
Gili Ortiz E. Factors influencing mortality trends of pancreatic cancer in Spain, 1955-2020. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2022; 114:474-480. [PMID: 35548865 DOI: 10.17235/reed.2022.8559/2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
OBJECTIVES The aim of this study was to analyze the trends of pancreatic cancer mortality in Spain from 1955-2020 in both genders and every age group, in order to describe the changes in the prevalence of some risk factors and their possible influence on mortality. METHODS Direct standardized mortality rates were calculated using the World Standard Population 2000-2025 and Joinpoint analysis was performed for age-specific and age-standardized mortality trends for the period from 1955-2020. RESULTS Mortality rates increased with age in both genders, with a marked increase in older groups. During 2020, 71.5% of male deaths and 81.5% of female deaths from pancreatic cancer occurred among those aged 65 years or more. Spanish National Health Surveys since 1987 show decreasing trends in daily smoking, but striking increases in obesity and diabetes mellitus rates in both genders, and rates of daily smoking and obesity are remarkably higher among disadvantaged social classes. CONCLUSIONS Pancreatic cancer mortality rates have increased uninterruptedly in Spain during the last decades. Increasing trends in obesity and diabetes mellitus, particularly among males, and the different prevalence of obesity and smoking according to social class are public health problems of great concern. Smoking and obesity are potentially avoidable risk factors. Thus, educational programs and legislative measures should be implemented more widely, such as programs for smoking prevention, healthy nutrition and physical exercise, and would be applied more intensively in the most disadvantaged social classes.
Collapse
|
41
|
Wood LD, Canto MI, Jaffee EM, Simeone DM. Pancreatic Cancer: Pathogenesis, Screening, Diagnosis, and Treatment. Gastroenterology 2022; 163:386-402.e1. [PMID: 35398344 PMCID: PMC9516440 DOI: 10.1053/j.gastro.2022.03.056] [Citation(s) in RCA: 392] [Impact Index Per Article: 130.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/13/2022] [Accepted: 03/25/2022] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a clinically challenging cancer, due to both its late stage at diagnosis and its resistance to chemotherapy. However, recent advances in our understanding of the biology of PDAC have revealed new opportunities for early detection and targeted therapy of PDAC. In this review, we discuss the pathogenesis of PDAC, including molecular alterations in tumor cells, cellular alterations in the tumor microenvironment, and population-level risk factors. We review the current status of surveillance and early detection of PDAC, including populations at high risk and screening approaches. We outline the diagnostic approach to PDAC and highlight key treatment considerations, including how therapeutic approaches change with disease stage and targetable subtypes of PDAC. Recent years have seen significant improvements in our approaches to detect and treat PDAC, but large-scale, coordinated efforts will be needed to maximize the clinical impact for patients and improve overall survival.
Collapse
Affiliation(s)
- Laura D Wood
- Departments of Pathology and Oncology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Marcia Irene Canto
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth M Jaffee
- Sidney Kimmel Cancer Center, Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Diane M Simeone
- Departments of Surgery and Pathology, Perlmutter Cancer Center, NYU Langone Health, New York, New York
| |
Collapse
|
42
|
Cai J, Chen H, Lu M, Zhang Y, Lu B, Luo C, Feng X, You L, Dai M, Zhao Y. Association between Temporal Glycemic Change and Risk of Pancreatic Cancer in Men: A Prospective Cohort Study. Cancers (Basel) 2022; 14:3403. [PMID: 35884465 PMCID: PMC9323305 DOI: 10.3390/cancers14143403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 12/04/2022] Open
Abstract
Hyperglycemia has been reported to increase the risk of pancreatic cancer (PC), while the association between glycemic change and PC risk has rarely been explored. Using data from a prospective cohort study conducted in China since 2006, 138,870 males with available fasting blood glucose (FBG) levels, including 106,632 males with at least two FBG measurements, were analyzed. The associations between FBG (level, change, and stability) and PC incidence were evaluated using Cox proportional hazard regression and restricted cubic splines. Baseline (p = 0.109) and recent (p = 0.070) FBG levels and incident PC were not significantly associated. U-shaped associations were observed between the annual FBG change and PC risk. Compared with stable FBG, participants with annual FBG change rates <−0.05 mmol/L or >0.15 mmol/L had about four-fold (HR, 4.010; 95% CI: 1.920−8.375) and six-fold (HR, 5.897; 95% CI: 2.935−11.848) higher PC risks, respectively. The PC risk increased by 2.5% (HRlinear = 1.025, 95% CI:1.009−1.042) for every 1% increase in the coefficient of variation for FBG. A subgroup analysis of males without diabetes at baseline showed stronger associations. Temporal FBG changes may be an important factor for identifying populations with high PC risks.
Collapse
Affiliation(s)
- Jie Cai
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (J.C.); (L.Y.)
| | - Hongda Chen
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (H.C.); (M.L.); (Y.Z.); (B.L.); (C.L.)
| | - Ming Lu
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (H.C.); (M.L.); (Y.Z.); (B.L.); (C.L.)
| | - Yuhan Zhang
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (H.C.); (M.L.); (Y.Z.); (B.L.); (C.L.)
| | - Bin Lu
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (H.C.); (M.L.); (Y.Z.); (B.L.); (C.L.)
| | - Chenyu Luo
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (H.C.); (M.L.); (Y.Z.); (B.L.); (C.L.)
| | - Xiaoshuang Feng
- Genomic Epidemiology Branch, International Agency for Research on Cancer, 69372 Lyon, France;
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (J.C.); (L.Y.)
| | - Min Dai
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (H.C.); (M.L.); (Y.Z.); (B.L.); (C.L.)
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (J.C.); (L.Y.)
| |
Collapse
|
43
|
Park JH, Hong JY, Han K, Kang W, Park JK. Increased risk of pancreatic cancer in individuals with non-alcoholic fatty liver disease. Sci Rep 2022; 12:10681. [PMID: 35739172 PMCID: PMC9226051 DOI: 10.1038/s41598-022-14856-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 06/14/2022] [Indexed: 11/25/2022] Open
Abstract
The association between non-alcoholic fatty liver disease (NAFLD) and the risk of pancreatic cancer in the general population remains unclear. This nationwide cohort study included 8,120,674 adults who underwent a national health screening in 2009 from the Korean National Health Insurance Service database. Participants were followed-up until December 2017 for the development of pancreatic cancer. NAFLD was assessed using the fatty liver index: ≥ 60, NAFLD and < 30, no NAFLD. Multivariable Cox proportional hazards regression was performed. During the follow-up of 59.1 million person-years, 10,470 participants were newly diagnosed with pancreatic cancer. NAFLD was significantly associated with an increased risk of pancreatic cancer compared to no NAFLD (adjusted hazard ratio [aHR], 1.17; 95% CI 1.09–1.26). This association was significant in both the obese (aHR, 1.14; 95% CI 1.05–1.23) and non-obese groups (aHR, 1.14; 95% CI 1.003–1.29). Individuals with fatty liver index 30–59 also had an increased risk (aHR, 1.10; 95% CI 1.05–1.16). The risk of pancreatic cancer increased with increasing fatty liver index scores (P for trend < 0.001). This study demonstrated that NAFLD was independently associated with an increased risk of pancreatic cancer, regardless of obesity. Our finding suggests that NAFLD may be a modifiable risk factor for pancreatic cancer.
Collapse
Affiliation(s)
- Joo-Hyun Park
- Department of Family Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea.,Department of Healthcare Administration and Policy, School of Public Health, University of Nevada Las Vegas, Las Vegas, USA
| | - Jung Yong Hong
- Department of Healthcare Administration and Policy, School of Public Health, University of Nevada Las Vegas, Las Vegas, USA.,Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea
| | - Wonseok Kang
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.
| | - Joo Kyung Park
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.
| |
Collapse
|
44
|
Overbeek KA, Levink IJM, Koopmann BDM, Harinck F, Konings ICAW, Ausems MGEM, Wagner A, Fockens P, van Eijck CH, Groot Koerkamp B, Busch ORC, Besselink MG, Bastiaansen BAJ, van Driel LMJW, Erler NS, Vleggaar FP, Poley JW, Cahen DL, van Hooft JE, Bruno MJ. Long-term yield of pancreatic cancer surveillance in high-risk individuals. Gut 2022; 71:1152-1160. [PMID: 33820756 PMCID: PMC9120399 DOI: 10.1136/gutjnl-2020-323611] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE We aimed to determine the long-term yield of pancreatic cancer surveillance in hereditary predisposed high-risk individuals. DESIGN From 2006 to 2019, we prospectively enrolled asymptomatic individuals with an estimated 10% or greater lifetime risk of pancreatic ductal adenocarcinoma (PDAC) after obligatory evaluation by a clinical geneticist and genetic testing, and subjected them to annual surveillance with both endoscopic ultrasonography (EUS) and MRI/cholangiopancreatography (MRI/MRCP) at each visit. RESULTS 366 individuals (201 mutation-negative familial pancreatic cancer (FPC) kindreds and 165 PDAC susceptibility gene mutation carriers; mean age 54 years, SD 9.9) were followed for 63 months on average (SD 43.2). Ten individuals developed PDAC, of which four presented with a symptomatic interval carcinoma and six underwent resection. The cumulative PDAC incidence was 9.3% in the mutation carriers and 0% in the FPC kindreds (p<0.001). Median PDAC survival was 18 months (range 1-32). Surgery was performed in 17 individuals (4.6%), whose pathology revealed 6 PDACs (3 T1N0M0), 7 low-grade precursor lesions, 2 neuroendocrine tumours <2 cm, 1 autoimmune pancreatitis and in 1 individual no abnormality. There was no surgery-related mortality. EUS detected more solid lesions than MRI/MRCP (100% vs 22%, p<0.001), but less cystic lesions (42% vs 83%, p<0.001). CONCLUSION The diagnostic yield of PDAC was substantial in established high-risk mutation carriers, but non-existent in the mutation-negative proven FPC kindreds. Nevertheless, timely identification of resectable lesions proved challenging despite the concurrent use of two imaging modalities, with EUS outperforming MRI/MRCP. Overall, surveillance by imaging yields suboptimal results with a clear need for more sensitive diagnostic markers, including biomarkers.
Collapse
Affiliation(s)
- Kasper A Overbeek
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Iris J M Levink
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Brechtje D M Koopmann
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Femme Harinck
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ingrid C A W Konings
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Margreet G E M Ausems
- Division Laboratories, Pharmacy and Biomedical Genetics, Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anja Wagner
- Department of Clinical Genetics, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Paul Fockens
- Department of Gastroenterology & Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Casper H van Eijck
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Olivier R C Busch
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marc G Besselink
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Barbara A J Bastiaansen
- Department of Gastroenterology & Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Lydi M J W van Driel
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nicole S Erler
- Department of Biostatistics, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Frank P Vleggaar
- Department of Gastroenterology & Hepatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan-Werner Poley
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Djuna L Cahen
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jeanin E van Hooft
- Department of Gastroenterology & Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marco J Bruno
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
45
|
Raoof S, Lee RJ, Jajoo K, Mancias JD, Rebbeck TR, Skates SJ. Multicancer Early Detection Technologies: A Review Informed by Past Cancer Screening Studies. Cancer Epidemiol Biomarkers Prev 2022; 31:1139-1145. [PMID: 35320352 DOI: 10.1158/1055-9965.epi-21-1443] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 12/14/2022] Open
Abstract
More than 75% of cancer-related deaths occur from cancers for which we do not screen. New screening liquid biopsies may help fill these clinical gaps, although evidence of benefit still needs to be assessed. Which lessons can we learn from previous efforts to guide those of the future? Screening trials for ovarian, prostate, pancreatic, and esophageal cancers are revisited to assess the evidence, which has been limited by small effect sizes, short duration of early-stage disease relative to screening frequency, study design, and confounding factors. Randomized controlled trials (RCT) to show mortality reduction have required millions of screening-years, two-decade durations, and been susceptible to external confounding. Future RCTs with late-stage incidence as a surrogate endpoint could substantially reduce these challenges, and clinical studies demonstrating safety and effectiveness of screening in high-risk populations may enable extrapolation to broader average-risk populations. Multicancer early detection tests provide an opportunity to advance these practical study designs. Conditional approvals based on RCTs with surrogate endpoints, contingent upon real world evidence generation and continuation of trials to definitive endpoints, may lower practical barriers to innovation in cancer screening and enable greater progress.
Collapse
Affiliation(s)
- Sana Raoof
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard J Lee
- Massachusetts General Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Kunal Jajoo
- Harvard Medical School, Boston, Massachusetts.,Brigham and Women's Hospital, Boston, Massachusetts
| | - Joseph D Mancias
- Harvard Medical School, Boston, Massachusetts.,Brigham and Women's Hospital, Boston, Massachusetts.,Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Timothy R Rebbeck
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard TH Chan School of Public Health, Boston, Massachusetts
| | - Steven J Skates
- Massachusetts General Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
46
|
Chen CH, Tsai MK, Lee JH, Lin RT, Hsu CY, Wen C, Wu X, Chu TW, Wen CP. "Sugar-Sweetened Beverages" Is an Independent Risk From Pancreatic Cancer: Based on Half a Million Asian Cohort Followed for 25 Years. Front Oncol 2022; 12:835901. [PMID: 35463371 PMCID: PMC9022008 DOI: 10.3389/fonc.2022.835901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/07/2022] [Indexed: 11/23/2022] Open
Abstract
Although the link between sugar-sweetened beverages (SSB) and pancreatic cancer has been suggested for its insulin-stimulating connection, most epidemiological studies showed inconclusive relationship. Whether the result was limited by sample size is explored. This prospective study followed 491,929 adults, consisting of 235,427 men and 256,502 women (mean age: 39.9, standard deviation: 13.2), from a health surveillance program and there were 523 pancreatic cancer deaths between 1994 and 2017. The individual identification numbers of the cohort were matched with the National Death file for mortality, and Cox models were used to assess the risk. The amount of SSB intake was recorded based on the average consumption in the month before interview by a structured questionnaire. We classified the amount of SSB intake into 4 categories: 0–<0.5 serving/day, ≥0.5–<1 serving per day, ≥1–<2 servings per day, and ≥2 servings per day. One serving was defined as equivalent to 12 oz and contained 35 g added sugar. We used the age and the variables at cohort enrolment as the reported risks of pancreatic cancers. The cohort was divided into 3 age groups, 20–39, 40–59, and ≥60. We found young people (age <40) had higher prevalence and frequency of sugar-sweetened beverages than the elderly. Those consuming 2 servings/day had a 50% increase in pancreatic cancer mortality (HR = 1.55, 95% CI: 1.08–2.24) for the total cohort, but a 3-fold increase (HR: 3.09, 95% CI: 1.44–6.62) for the young. The risk started at 1 serving every other day, with a dose–response relationship. The association of SSB intake of ≥2 servings/day with pancreatic cancer mortality among the total cohort remained significant after excluding those who smoke or have diabetes (HR: 2.12, 97% CI: 1.26–3.57), are obese (HR: 1.57, 95% CI: 1.08–2.30), have hypertension (HR: 1.90, 95% CI: 1.20–3.00), or excluding who died within 3 years after enrollment (HR: 1.67, 95% CI: 1.15–2.45). Risks remained in the sensitivity analyses, implying its independent nature. We concluded that frequent drinking of SSB increased pancreatic cancer in adults, with highest risk among young people.
Collapse
Affiliation(s)
- Chien Hua Chen
- Digestive Disease Center, Changbing Show-Chwan Memorial Hospital, Lukang, Taiwan.,Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan.,Department of Food Science and Technology, Hungkuang University, Taichung, Taiwan
| | - Min Kuang Tsai
- Institue of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - June Han Lee
- Institue of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Ro-Ting Lin
- College of Public Health, China Medical University, Taichung, Taiwan
| | - Chung Y Hsu
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Christopher Wen
- Long Beach VAMC Hospital, University of Irvine Medical Center, Irvine, CA, United States
| | - Xifeng Wu
- Center for Biostatistics, Bioinformatics and Big Data, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China.,National Institute for Data Science in Health and Medicine, Zhejiang University, Hangzhou, China
| | - Ta-Wei Chu
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Taipei MJ Health Screening Center, Taipei, Taiwan
| | - Chi Pang Wen
- Institue of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan.,Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| |
Collapse
|
47
|
Gonda TA, Farrell J, Wallace M, Khanna L, Janec E, Kwon R, Saunders M, Siddiqui UD, Brand R, Simeone DM. Standardization of EUS imaging and reporting in high-risk individuals of pancreatic adenocarcinoma: consensus statement of the Pancreatic Cancer Early Detection Consortium. Gastrointest Endosc 2022; 95:723-732.e7. [PMID: 34736932 DOI: 10.1016/j.gie.2021.10.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 10/25/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Pancreatic ductal adenocarcinoma is an aggressive disease most often diagnosed after local progression or metastatic dissemination, precluding resection and resulting in a high mortality rate. For individuals with elevated personal risk of the development of pancreatic cancer, EUS is a frequently used advanced imaging and diagnostic modality. However, variability in the expertise and definition of EUS findings exists among gastroenterologists, as well as a lack of standardized reporting of relevant findings at the time of examination. Adoption of standardized EUS reporting, using a universally accepted and agreed on terminology, is needed. METHODS A consensus statement designed to create a standardized reporting template was authored by a multidisciplinary group of experts in pancreatic diseases that includes gastroenterologists, radiologists, surgeons, oncologists, and geneticists. This statement was developed using a modified Delphi process as part of the Pancreatic Cancer Early Detection Consortium, and >75% agreement was required to reach consensus. RESULTS We identified reporting elements and present standardized reporting templates for EUS indications, procedural data, EUS image capture, and descriptors of findings, tissue sampling, and postprocedural assessment of adequacy. CONCLUSIONS Adoption of this standardized EUS reporting template should improve consistency in clinical decision-making for individuals with elevated risk of pancreatic cancer by providing complete and accurate reporting of pancreatic abnormalities. Standardization will also help to facilitate research and clinical trial design by using clearly defined and consistent imaging descriptions, thus allowing for comparison of results across different centers.
Collapse
Affiliation(s)
- Tamas A Gonda
- Division of Gastroenterology and Hepatology, New York University Langone Health, New York, New York, USA
| | - James Farrell
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Michael Wallace
- Department of Gastroenterology & Hepatology, Mayo Clinic, Jacksonville, Florida, USA
| | - Lauren Khanna
- Division of Gastroenterology and Hepatology, New York University Langone Health, New York, New York, USA
| | - Eileen Janec
- Division of Gastroenterology and Hepatology, New York University Langone Health, New York, New York, USA
| | - Richard Kwon
- Gastroenterology, Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael Saunders
- Gastroenterology, Internal Medicine, University of Washington, Seattle, Washington, USA
| | - Uzma D Siddiqui
- Gastroenterology, Internal Medicine, University of Chicago, Chicago, Illinois, USA
| | - Randall Brand
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Diane M Simeone
- Gastroenterology, Surgical Oncology, New York University Langone Health, New York, New York, USA
| |
Collapse
|
48
|
Fatty Pancreas-Centered Metabolic Basis of Pancreatic Adenocarcinoma: From Obesity, Diabetes and Pancreatitis to Oncogenesis. Biomedicines 2022; 10:biomedicines10030692. [PMID: 35327494 PMCID: PMC8945032 DOI: 10.3390/biomedicines10030692] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest types of cancer, and it is currently the third most common cause of cancer death in the U.S.A. Progress in the fight against PDAC has been hampered by an inability to detect it early in the overwhelming majority of patients, and also by the reduced oxygen levels and nutrient perfusion caused by new matrix formation through the activation of stromal cells in the context of desmoplasia. One harbinger of PDAC is excess intrapancreatic fat deposition, namely, fatty pancreas, which specifically affects the tumor macro- and microenvironment in the organ. Over half of PDAC patients have diabetes mellitus (DM) at the time of diagnosis, and fatty pancreas is associated with subsequent DM development. Moreover, there is a strong association between fatty pancreas and fatty liver through obesity, and a higher intrapancreatic fat percentage has been noted in acute pancreatitis patients with DM than in those without DM. All these findings suggest that the link between fatty pancreas and PDAC might occur through metabolic alterations, either DM-related or non-DM-related. Based on clinical, in vivo and in vitro evidence, the current review highlights the etiologies of fatty pancreas (including fatty infiltration and replacement) and the fatty pancreas-associated metabolic alterations involved in oncogenesis to provide crucial targets to prevent, detect, and/or effectively treat PDAC.
Collapse
|
49
|
Nissinen SI, Venäläinen M, Kumpulainen P, Roine A, Häkkinen MR, Vepsäläinen J, Oksala N, Rantanen T. Discrimination between Pancreatic Cancer, Pancreatitis and Healthy Controls Using Urinary Polyamine Panel. Cancer Control 2022; 28:10732748211039762. [PMID: 35135363 PMCID: PMC8832577 DOI: 10.1177/10732748211039762] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKROUND Polyamines play an important role in cellular proliferation, and the change in polyamine metabolism is reported in various cancers. We searched for urinary polyamine signature for distinguishing between pancreatic cancer, premalignant lesions of the pancreas (PLP), acute and chronic pancreatitis, and controls. METHODS Patients and controls were prospectively recruited in three Finnish hospitals between October 2013 and June 2016. The patients provided a urine sample at the time of the diagnosis. The panel of 14 polyamines was obtained in a single run with mass spectrometry. The polyamine concentrations were analysed with quadratic discriminant analysis and cross-validated with leave-one-out cross-validation. RESULTS Sixty-eight patients with pancreatic cancer, 36 with acute pancreatitis, 18 with chronic pancreatitis and 7 with PLP were recruited, as were 53 controls. The combination of 4 polyamines - acetylputrescine, diacetylspermidine, N8-acetylspermidine and diacetylputrescine - distinguished pancreatic cancer and PLP from controls (sensitivity = 94%, specificity = 68% and AUC = 0.88). The combination of diacetylspermidine, N8-acetylspermidine and diacetylspermine distinguished acute pancreatitis from controls (sensitivity = 94%, specificity = 92%, AUC = 0.98). The combination of acetylputrescine, diacetylspermidine and diacetylputrescine distinguished chronic pancreatitis from controls (sensitivity = 98%, specificity = 71%, AUC = 0.93). CONCLUSIONS Optimally selected urinary polyamine panels discriminate between pancreatic cancer and controls, as well as between acute and chronic pancreatitis and controls.
Collapse
Affiliation(s)
- Samuli I Nissinen
- Department of Internal Medicine, School of Medicine, 205537University of Eastern Finland, Kuopio, Finland.,Department of Internal Medicine, 3701Kanta-Häme Central Hospital, Hämeenlinna, Finland
| | - Markus Venäläinen
- Department of Internal Medicine, School of Medicine, 205537University of Eastern Finland, Kuopio, Finland
| | | | - Antti Roine
- Faculty of Medicine and Health Technology, 7840Tampere University, Tampere, Finland
| | - Merja R Häkkinen
- School of Pharmacy, Biocenter Kuopio, 205537University of Eastern Finland, Kuopio, Finland
| | - Jouko Vepsäläinen
- School of Pharmacy, Biocenter Kuopio, 205537University of Eastern Finland, Kuopio, Finland
| | - Niku Oksala
- Faculty of Medicine and Health Technology, 7840Tampere University, Tampere, Finland.,Centre for Vascular Surgery and Interventional Radiology, 60670Tampere University Hospital, Tampere, Finland
| | - Tuomo Rantanen
- Department of Surgery, School of Medicine, 205537University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
50
|
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: 3.7] [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
|