1
|
Farinella R, Felici A, Peduzzi G, Testoni SGG, Costello E, Aretini P, Blazquez-Encinas R, Oz E, Pastore A, Tacelli M, Otlu B, Campa D, Gentiluomo M. From classical approaches to artificial intelligence, old and new tools for PDAC risk stratification and prediction. Semin Cancer Biol 2025; 112:71-92. [PMID: 40147701 DOI: 10.1016/j.semcancer.2025.03.004] [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: 11/28/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is recognized as one of the most lethal malignancies, characterized by late-stage diagnosis and limited therapeutic options. Risk stratification has traditionally been performed using epidemiological studies and genetic analyses, through which key risk factors, including smoking, diabetes, chronic pancreatitis, and inherited predispositions, have been identified. However, the multifactorial nature of PDAC has often been insufficiently addressed by these methods, leading to limited precision in individualized risk assessments. Advances in artificial intelligence (AI) have been proposed as a transformative approach, allowing the integration of diverse datasets-spanning genetic, clinical, lifestyle, and imaging data into dynamic models capable of uncovering novel interactions and risk profiles. In this review, the evolution of PDAC risk stratification is explored, with classical epidemiological frameworks compared to AI-driven methodologies. Genetic insights, including genome-wide association studies and polygenic risk scores, are discussed, alongside AI models such as machine learning, radiomics, and deep learning. Strengths and limitations of these approaches are evaluated, with challenges in clinical translation, such as data scarcity, model interpretability, and external validation, addressed. Finally, future directions are proposed for combining classical and AI-driven methodologies to develop scalable, personalized predictive tools for PDAC, with the goal of improving early detection and patient outcomes.
Collapse
Affiliation(s)
| | | | | | - Sabrina Gloria Giulia Testoni
- Division of Gastroenterology and Gastrointestinal Endoscopy, IRCCS Policlinico San Donato, Vita-Salute San Raffaele University, Milan, Italy
| | - Eithne Costello
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Paolo Aretini
- Fondazione Pisana per la Scienza, San Giuliano Terme, Italy
| | - Ricardo Blazquez-Encinas
- Department of Cell Biology, Physiology and Immunology, University of Cordoba / Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Elif Oz
- Department of Biostatistics and Bioinformatics, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Aldo Pastore
- Fondazione Pisana per la Scienza, San Giuliano Terme, Italy
| | - Matteo Tacelli
- Pancreas Translational & Clinical Research Center, Pancreato-Biliary Endoscopy and Endosonography Division, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Burçak Otlu
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, Ankara, Turkey
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | | |
Collapse
|
2
|
Ghosn B, Baniasadi MM, Jalalzadeh M, Esmaillzadeh A. Total, unprocessed, and processed red meat intake in relation to the risk of pancreatic cancer: A systematic review and dose-response meta-analysis of prospective cohort studies. Clin Nutr ESPEN 2025; 67:265-275. [PMID: 40118182 DOI: 10.1016/j.clnesp.2025.03.021] [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: 05/31/2024] [Revised: 02/19/2025] [Accepted: 03/12/2025] [Indexed: 03/23/2025]
Abstract
Data on the association between red meat from both processed and unprocessed sources and risk of pancreatic cancer is controversial. Therefore, this study summarized current evidence on the relationship between red and processed red meat intake and pancreatic cancer risk through a meta-analysis. A systematic search was conducted on PubMed, ISI Web of Science and Scopus for prospective cohorts up to October 2024. Hazard ratio (HR) and 95 % confidence intervals (CIs) for the highest vs. the lowest category of the exposures were combined using random-effects models. Dose-response relations were explored by one-stage weighted mixed effects meta-analysis. 19 studies involving 4,291,065 participants with 13,820 pancreatic cancer cases were included. The highest intake of total red meat was positively related to risk of pancreatic cancer (Pooled HR: 1.12, 95%CIs: 1.01, 1.24; I2: 36.5 %, PQ-test: 0.10). Such association was not significant for unprocessed (Pooled HR: 1.05, 95%CIs: 0.88, 1.24; I2: 66.6 %, PQ-test: 0.002) and processed (Pooled HR: 1.02, 95%CIs: 0.86, 1.21; I2: 66.0 %, PQ-test: 0.001) red meat intake. No significant association was observed between each 3 serving/wk of total, unprocessed, and processed red meat and risk of pancreatic cancer. Also, no evidence for a nonlinear association was found for all associations. This meta-analysis suggests a positive relation between the highest consumption of total red meat and pancreatic cancer. However, this relation was not substantial in terms of unprocessed and processed red meat.
Collapse
Affiliation(s)
- Batoul Ghosn
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Moradi Baniasadi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Moharam Jalalzadeh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Esmaillzadeh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran; Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular -Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Peduzzi G, Archibugi L, Farinella R, de Leon Pisani RP, Vodickova L, Vodicka P, Kraja B, Sainz J, Bars-Cortina D, Daniel N, Silvestri R, Uysal-Onganer P, Landi S, Dulińska-Litewka J, Comandatore A, Campa D, Hughes DJ, Rizzato C. The exposome and pancreatic cancer, lifestyle and environmental risk factors for PDAC. Semin Cancer Biol 2025; 113:100-129. [PMID: 40368260 DOI: 10.1016/j.semcancer.2025.05.004] [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: 01/17/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/16/2025]
Abstract
Pancreatic cancer (PC), particularly pancreatic ductal adenocarcinoma (PDAC), is a significant global health issue with high mortality rates. PDAC, though only 3 % of cancer diagnoses, causes 7 % of cancer deaths due to its severity and asymptomatic early stages. Risk factors include lifestyle choices, environmental exposures, and genetic predispositions. Conditions like new-onset type 2 diabetes and chronic pancreatitis also contribute significantly. Modifiable risk factors include smoking, alcohol consumption, non-alcoholic fatty pancreatic disease (NAFPD), and obesity. Smoking and heavy alcohol consumption increase PC risk, while NAFPD and obesity, particularly central adiposity, contribute through chronic inflammation and insulin resistance. Refined sugar and sugar-sweetened beverages (SSBs) are also linked to increased PC risk, especially among younger individuals. Hormonal treatments and medications like statins, aspirin, and metformin have mixed results on PC risk, with some showing protective effects. The gut microbiome influences PC through the gut-pancreas axis, with disruptions leading to inflammation and carcinogenesis. Exposure to toxic substances, including heavy metals and chemicals, is associated with increased PC risk. Glycome changes, such as abnormal glycosylation patterns, are significant in PDAC development and offer potential for early diagnosis. Interactions between environmental and genetic factors are crucial in PDAC susceptibility. Genome-wide association studies (GWAS) have identified several single nucleotide polymorphisms (SNPs) linked to PDAC, but gene-environment interactions remain largely unexplored. Future research should focus on polygenic risk scores (PRS) and large-scale studies to better understand these interactions and their impact on PDAC risk.
Collapse
Affiliation(s)
| | - Livia Archibugi
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Ruggero Ponz de Leon Pisani
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Ludmila Vodickova
- Biomedical Center Martin, Bioinformatic Center, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Slovakia; Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Vodicka
- Biomedical Center Martin, Bioinformatic Center, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Slovakia; Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czech Republic
| | - Bledar Kraja
- University Clinic of Gastrohepatology, University Hospital Center Mother Teresa, Tirana, Albania
| | - Juan Sainz
- Department of Biochemistry and Molecular Biology, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid 28029, Spain; GENYO. Centre for Genomics and Oncological Research. Genomic Oncology department, Granada, Spain; Instituto de Investigación Biosanitaria Ibs.Granada, Granada, Spain
| | - David Bars-Cortina
- Institut Català d'Oncologia (ICO) IDIBELL, Unit of Biomarkers and Susceptibility (UBS), Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain; Institut Català d'Oncologia (ICO) IDIBELL, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | | | - Pinar Uysal-Onganer
- Cancer Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London, UK
| | - Stefano Landi
- Department of Biology, University of Pisa, Pisa, Italy
| | | | - Annalisa Comandatore
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | | |
Collapse
|
4
|
Qin C, Xu C, Zhu Z, Song X, Wang X, Xu W, Zhu M. A study of the association between Helicobacter pylori infection type and pancreatic cancer risk: A systematic review and meta‑analysis. Oncol Lett 2025; 29:174. [PMID: 39975953 PMCID: PMC11837465 DOI: 10.3892/ol.2025.14920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025] Open
Abstract
Pancreatic cancer is a highly invasive malignant tumor with a complex pathogenesis that makes early diagnosis challenging. The potential association between Helicobacter pylori infection and pancreatic cancer risk has been noted; however, the available results are still highly divergent. The aim of the present study was to systematically evaluate the association between different types of H. pylori infection and pancreatic cancer risk as well as to explore the possible causes. A systematic search was conducted using the PubMed, Embase and Cochrane Library databases up to August 2023. The literature quality was evaluated using the Newcastle-Ottawa Scale. All studies that met the criteria were included in the overall meta-analysis to calculate the odds ratios (ORs) and corresponding 95% confidence intervals (CIs). In addition, subgroup analyses were performed based on factors such as diagnostic criteria for H. pylori infection, study region, type of study design and CagA status. The effect of publication bias on the quantitative synthesis results was assessed using the trim-and-fill analysis, and sensitivity analyses were used to verify the robustness of the quantitative synthesis results. A total of 17 studies involving 67,910 participants, including 64,372 controls and 3,538 patients with pancreatic cancer, were included in the present study. The overall analysis showed that no significant association was observed between H. pylori infection and pancreatic cancer risk (OR, 1.15; 95% CI, 0.93-1.41). Further subgroup analyses, which did not consider the effects of study quality, diagnostic criteria, geographical distribution and the type of study design, did not produce new findings that contradicted the results of the overall analysis. CagA+ H. pylori infection did not significantly affect the risk of pancreatic cancer (OR, 0.95; 95% CI, 0.78-1.16), whereas CagA- H. pylori infection may be a possible risk factor for pancreatic cancer (OR, 1.24; 95% CI, 1.004-1.541). The H. pylori infection did not significantly increase the risk of pancreatic cancer. However, it is noteworthy that CagA- H. pylori infection could be a potential factor that elevated the risk of pancreatic cancer.
Collapse
Affiliation(s)
- Chao Qin
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Chonghe Xu
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, P.R. China
| | - Zhongqi Zhu
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Xixi Song
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Xin Wang
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Wei Xu
- Department of Blood Transfusion, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Mei Zhu
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| |
Collapse
|
5
|
Devasahayam Arokia Balaya R, Sen P, Grant CW, Zenka R, Sappani M, Lakshmanan J, Athreya AP, Kandasamy RK, Pandey A, Byeon SK. An integrative multi-omics analysis reveals a multi-analyte signature of pancreatic ductal adenocarcinoma in serum. J Gastroenterol 2025; 60:496-511. [PMID: 39666045 DOI: 10.1007/s00535-024-02197-6] [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: 02/16/2024] [Accepted: 12/01/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains a formidable health challenge due to its detection at a late stage and a lack of reliable biomarkers for early detection. Although levels of carbohydrate antigen 19-9 are often used in conjunction with imaging-based tests to aid in the diagnosis of PDAC, there is still a need for more sensitive and specific biomarkers for early detection of PDAC. METHODS We obtained serum samples from 88 subjects (patients with PDAC (n = 58) and controls (n = 30)). We carried out a multi-omics analysis to measure cytokines and related proteins using proximity extension technology and lipidomics and metabolomics using tandem mass spectrometry. Statistical analysis was carried out to find molecular alterations in patients with PDAC and a machine learning model was used to derive a molecular signature of PDAC. RESULTS We quantified 1,462 circulatory proteins along with 873 lipids and 1,001 metabolites. A total of 505 proteins, 186 metabolites and 33 lipids including bone marrow stromal antigen 2 (BST2), keratin 18 (KRT18), and cholesteryl ester(20:5) were found to be significantly altered in patients. We identified different levels of sphingosine, sphinganine, urobilinogen and lactose indicating that glycosphingolipid and galactose metabolisms were significantly altered in patients compared to controls. In addition, elevated levels of diacylglycerols and decreased cholesteryl esters were observed in patients. Using a machine learning model, we identified a signature of 38 biomarkers for PDAC, composed of 21 proteins, 4 lipids, and 13 metabolites. CONCLUSIONS Overall, this study identified several proteins, metabolites and lipids involved in various pathways including cholesterol and lipid metabolism to be changing in patients. In addition, we discovered a multi-analyte signature that could be further tested for detection of PDAC.
Collapse
Affiliation(s)
| | - Partho Sen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Caroline W Grant
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Roman Zenka
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Marimuthu Sappani
- Department of Biostatistics, Christian Medical College, Vellore, Tamil Nadu, 632002, India
| | - Jeyaseelan Lakshmanan
- College of Medicine, Mohammad Bin Rashid University of Medicine and Health Sciences, Dubai, 505055, UAE
| | - Arjun P Athreya
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Richard K Kandasamy
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
- Manipal Academy of Higher Education, Manipal, Karnataka, 5761904, India
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
- Manipal Academy of Higher Education, Manipal, Karnataka, 5761904, India.
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
6
|
Chandra S, Halfdanarson TR, Carlson EE, Rabe KG, Mahipal A, Majumder S, Bamlet WR, Horibe M, Tella SH, Shariq O, Carr RM, Cleary SP, Oberg AL, Antwi SO. Discordant risk factors between pancreatic neuroendocrine neoplasms and pancreatic ductal adenocarcinoma. Endocr Relat Cancer 2025; 32:e240142. [PMID: 39932006 PMCID: PMC11896652 DOI: 10.1530/erc-24-0142] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/13/2025]
Abstract
Pancreatic neuroendocrine neoplasm (panNEN) is a rare malignancy and the second most common type of pancreatic cancer after pancreatic ductal adenocarcinoma (PDAC), but its etiology is poorly understood. We investigated whether the risk factors of panNEN are concordant with those known for PDAC. We performed the largest case-control study to date on panNENs, comprising 927 sporadic nonfunctional panNEN cases and 1807 frequency-matched controls, using data from the Mayo Clinic Biospecimen Resource for Pancreas Research. We assessed associations for obesity, first-degree family history of pancreatic cancer, cigarette smoking, overall type II diabetes mellitus (T2DM), new-onset T2DM (<1 year before panNEN diagnosis), longstanding T2DM (≥5 years), alcohol intake and aspirin use. Multivariable logistic regression was used to calculate odds ratios and 95% confidence intervals (CIs). Our results show that overall T2DM (OR = 1.71, 95% CI: 1.37-2.14) and new-onset T2DM (OR = 2.65, 95% CI: 1.92-3.69) are associated with higher odds of panNEN, but not longstanding T2DM (OR = 1.29, 95% CI: 0.94-1.75). A non-significant elevated odds of panNEN was observed among participants with a positive family history of pancreatic cancer (OR = 1.44, 95% CI: 0.96-2.14). Alcohol use was inversely related to panNEN (OR = 0.52, 95% CI: 0.42-0.66, ever-vs-never). No association was observed for smoking, obesity or aspirin use. These findings indicate that overall T2DM and new-onset T2DM are associated with higher odds of panNEN. Unlike PDAC, alcohol use was inversely related to panNEN, and we found no associations for cigarette smoking, obesity or aspirin use. These results indicate differences in the risk factor profiles of panNEN and PDAC.
Collapse
Affiliation(s)
- Shruti Chandra
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Thorvardur R Halfdanarson
- Division of Medical Oncology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Erin E Carlson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Kari G Rabe
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Amit Mahipal
- Department of Oncology, Seidman Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - William R Bamlet
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Masayasu Horibe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Sri Harsha Tella
- Department of Hematology and Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Omair Shariq
- Department of General Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ryan M Carr
- Division of Medical Oncology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Sean P Cleary
- Department of General Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Division of Hepatobiliary and Pancreatic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ann L Oberg
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Samuel O Antwi
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida, USA
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
7
|
Song L, Wang G, Chen Z, Wang G. Research status of risk factors and prevention of pancreatic cancer: A bibliometric and visual analysis. Medicine (Baltimore) 2025; 104:e41831. [PMID: 40101086 PMCID: PMC11922469 DOI: 10.1097/md.0000000000041831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/21/2025] [Indexed: 03/20/2025] Open
Abstract
One of the biggest public health issues facing the globe today is pancreatic cancer (PC). To serve as a guide for clinically identifying existing research hotspots and conducting related studies in the future, bibliometric and visualization analyses of the literature on risk factors and PC prevention were carried out in this work. Results of published research from 2004 to 2024 were retrieved using the Web of Science database as a search platform. CiteSpace and VOSviewer were used for bibliometric and visual analysis. Based on the exclusion criteria, 868 articles in all were screened. Between 2004 and 2024, the quantity of articles published varied. Between 2017 and 2023, there was a consistent upward trend in the quantity of published literature. Cancer epidemiology biomarkers and prevention, cancers, and the Asian Pacific Journal of Cancer Prevention were the 3 journals with the most publications. The 2 nations with the most publications are China and the United States. The 2 nations with the most publications are China and the United States. The top 3 most published universities are Harvard University, the National Institutes of Health (NIH), and the National Cancer Institute (NCI). Buzzwords include body mass index, obesity, diabetes, smoking, and exercise.
Collapse
Affiliation(s)
- Lichen Song
- School of Clinical Medicine, Dali University, Dali, China
| | - Guihua Wang
- Respiratory and Critical Care Medicine Department, The First Affiliated Hospital of Dali University, Dali, China
| | - Ziyi Chen
- School of Clinical Medicine, Dali University, Dali, China
| | - Guangming Wang
- Center of Genetic Testing, The First Affiliated Hospital of Dali University, Dali, China
| |
Collapse
|
8
|
Zhu Z, Wang X. Causal relationship and potential common pathogenic mechanisms between hidradenitis suppurativa and related cancer. Discov Oncol 2025; 16:304. [PMID: 40072688 PMCID: PMC11904070 DOI: 10.1007/s12672-025-02075-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Hidradenitis suppurativa (HS) is a chronic, inflammatory and common skin disease. Observation studies have reported the association between HS and cancers, however no studies reported whether a causal relationship exists between HS and cancers. This study aimed to explore the causal relationship between HS and differential subtypes of cancers by conducting a bidirectional Mendelian randomization (MR) analysis. METHOD Genome-wide association study (GWAS) data related to HS and 16 subtypes of cancers were collected. The inverse variance weighted (IVW) method was primarily applied for our MR analysis, MR-Egger, weighted median, simple mode, and weighted mode methods were used additionally. Heterogeneity, horizontal pleiotropy, and potential outliers were assessed for the MR analysis results. Subsequently, disease-related genes were retrieved from the GeneCards database. To investigate the potential functions of these associated genes, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted. RESULT The results of our MR analysis indicated a causal association between HS and pancreatic cancer (PAC). Specifically, HS was found to elevate the risk of developing PAC (odds ratio (OR), 1.074; 95% confidence interval (CI) 1.015-1.135; p = 0.013). Conversely, reverse MR analysis demonstrated that PAC does not exert a causal effect on HS. Furthermore, our findings did not reveal any significant causal relationships between HS and other types of cancer. No evidence of heterogeneity or pleiotropy was identified in the analysis. Additionally, we identified disease-related genes, and subsequent GO and KEGG enrichment analyses indicated that the genes common to both HS and PAC are implicated in pathways associated with immune and inflammatory processes. CONCLUSION The results of this study offer novel evidence regarding the causal relationship between HA and PAC. Our Mendelian randomization analysis indicates that HS may have a causal influence on PAC, which could inform the development of improved treatment strategies for patients suffering from HS. However, the underlying mechanisms warrant further exploration.
Collapse
Affiliation(s)
- Zexin Zhu
- Department of Surgical Oncology, the Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoxue Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
9
|
Srinivasan S, Mehra S, Jinka S, Bianchi A, Singh S, Dosch AR, Amirian H, Krishnamoorthy V, Silva IDC, Patel M, Box EW, Garrido V, Totiger TM, Zhou Z, Ban Y, Datta J, VanSaun M, Merchant N, Nagathihalli NS. Activation of CREB drives acinar cells to ductal reprogramming and promotes pancreatic cancer progression in animal models of alcoholic pancreatitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.01.05.574376. [PMID: 38903082 PMCID: PMC11188065 DOI: 10.1101/2024.01.05.574376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
BACKGROUND & AIMS Alcoholic chronic pancreatitis (ACP) exacerbates pancreatic damage through acinar cell injury, fibroinflammation, and cyclic adenosine monophosphate response element binding protein 1 (CREB) activation, surpassing the damage by alcohol (A) alone or cerulein-induced CP. The molecular cooperativity between CREB and oncogenic Kras G12D/+ (Kras*) in promoting pancreatic cancer progression within the context of ACP remains unclear. METHODS Experimental ACP induction was established in multiple mouse models, with euthanasia during the recovery stage to assess tumor latency. We established CREB deletion (Creb fl/fl ) in Ptf1a CreERTM/+ ;LSL-Kras G12D/+ (KC) genetic mouse models (KCC -/- ). Pancreata from Ptf1a CreERTM/+ , KC, and KCC -/- mice were analyzed using western blotting, phosphokinase array, and quantitative PCR. Single-cell RNA sequencing was performed in ACP-induced KC mice. Lineage tracing of acinar cell explant cultures and analysis of tissue samples from human pancreatic diseases (CP and pancreatic ductal adenocarcinoma [PDAC]) were conducted. RESULTS ACP induction in KC mice impaired the pancreas' repair mechanism. Acinar cell-derived ductal lesions demonstrated prolonged hyperactivated CREB in acinar-to-ductal metaplasia (ADM)/pancreatic intraepithelial neoplasia (PanIN) lesions associated with pancreatitis and in PDAC. Persistent CREB activation reprogrammed acinar cells, increasing profibrotic inflammation. In ACP-induced models, acinar-specific Creb ablation reduced advanced PanIN lesions, hindered tumor progression, and improved acinar cell function. Pharmacological targeting of CREB significantly reduced the primary tumor burden in a PDAC mouse model with ACP. CONCLUSIONS Our findings demonstrate that CREB and Kras* promote irreversible ADM, accelerating pancreatic cancer progression with ACP. Targeting CREB offers a promising strategy to address the clinical need for effective treatments for inflammation-driven pancreatic cancer.
Collapse
Affiliation(s)
- Supriya Srinivasan
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Siddharth Mehra
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Sudhakar Jinka
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Anna Bianchi
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Samara Singh
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Austin R Dosch
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Haleh Amirian
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | | | - Iago De Castro Silva
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Manan Patel
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Edmond Worley Box
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Vanessa Garrido
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Tulasigeri M Totiger
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Zhiqun Zhou
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Yuguang Ban
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, Florida
| | - Jashodeep Datta
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Michael VanSaun
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Nipun Merchant
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Nagaraj S Nagathihalli
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| |
Collapse
|
10
|
Yang H, Chou W, Nguyen P, Nguyen NTH, Phuong NT, Wang C, Hsu JC, Lin M, Huang C. The protective role of anti-parkinsonian drugs in pancreatic cancer risk: A comprehensive case-control study in Taiwan. Cancer Sci 2025; 116:783-791. [PMID: 39629516 PMCID: PMC11875767 DOI: 10.1111/cas.16422] [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: 09/09/2024] [Revised: 11/01/2024] [Accepted: 11/20/2024] [Indexed: 03/05/2025] Open
Abstract
Pancreatic cancer is among the deadliest cancers, with a grim prognosis despite advances in treatment. We conducted a population-based case-control study from Taiwan, linking Health and Welfare Data Science Center data to the Taiwan Cancer Registry, which offers a promising strategy for its treatment through drug repurposing. The study aims to identify the association of anti-parkinsonian drugs with pancreatic cancer risk across different age groups. The analysis encompassed 18,921 pancreatic cancer cases and 75,684 matched controls, employing conditional logistic regression to assess the impact of anti-parkinsonian drugs on the risk of pancreatic cancer. Key findings revealed a statistically significant association of the administration with specific anti-parkinsonian medications, including anticholinergic agents, tertiary amines, dopa derivatives, and dopamine receptor agonists, with a reduction in pancreatic cancer risk. These associations were represented as adjusted odds ratios (aORs), ranging from 0.620 (95% CI 0.470-0.810) to 0.764 (95% CI 0.655-0.891). Further, age-stratified analysis revealed variations in efficacy across different age groups. Anticholinergic agents and tertiary amines exhibited greater effectiveness in the 40-64-year age group (aOR, 0.653; 95% CI, 0.489-0.872), whereas dopa derivatives and dopamine receptor agonists were particularly efficacious in the cohort aged ≥65 years (aOR, 0.728; 95% CI, 0.624-0.850 and aOR, 0.665; 95% CI, 0.494-0.894, respectively). Notably, specific drugs such as trihexyphenidyl, levodopa/dopa decarboxylase inhibitor (DDCI), and pramipexole demonstrated a significant decrease in cancer risk, especially in the elderly population. These preliminary findings can contribute to the possible therapeutic role of anti-parkinsonian drugs in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Hsuan‐Chia Yang
- Graduate Institute of Biomedical Informatics, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- International Center for Health Information Technology (ICHIT), College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- Clinical Big Data Research Center, Taipei Medical University HospitalTaipei Medical UniversityTaipeiTaiwan
- Research Center of Big Data and Meta‐Analysis, Wan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Wen‐Chi Chou
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou and College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Phung‐Anh Nguyen
- Clinical Big Data Research Center, Taipei Medical University HospitalTaipei Medical UniversityTaipeiTaiwan
- Graduate Institute of Data Science, College of ManagementTaipei Medical UniversityTaipeiTaiwan
- Clinical Data Center, Office of Data ScienceTaipei Medical UniversityTaipeiTaiwan
- Research Center of Health Care Industry Data Science, College of ManagementTaipei Medical UniversityTaipeiTaiwan
| | - Nhi Thi Hong Nguyen
- School of Nutrition and Health SciencesTaipei Medical UniversityTaipeiTaiwan
- Health Personnel Training InstituteUniversity of Medicine and Pharmacy, Hue UniversityHueVietnam
| | - Nguyen Thi Phuong
- Graduate Institute of Biomedical Informatics, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
| | - Ching‐Huan Wang
- Biomedical Informatics and Data Science (BIDS) Section, School of MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jason C. Hsu
- Clinical Big Data Research Center, Taipei Medical University HospitalTaipei Medical UniversityTaipeiTaiwan
- Clinical Data Center, Office of Data ScienceTaipei Medical UniversityTaipeiTaiwan
- Research Center of Health Care Industry Data Science, College of ManagementTaipei Medical UniversityTaipeiTaiwan
- International Ph.D. Program in Biotech and Healthcare Management, College of ManagementTaipei Medical UniversityTaipeiTaiwan
| | - Ming‐Chin Lin
- Graduate Institute of Biomedical Informatics, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- Department of Neurosurgery, Wang‐Fang HospitalTaipei Medical UniversityTaipeiTaiwan
- Taipei Neuroscience InstituteTaipei Medical UniversityTaipeiTaiwan
| | - Chih‐Wei Huang
- Graduate Institute of Biomedical Informatics, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- International Center for Health Information Technology (ICHIT), College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- Clinical Big Data Research Center, Taipei Medical University HospitalTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
11
|
Wang L, Wang Q, Li L, Kaelber DC, Xu R. Glucagon-like peptide-1 receptor agonists and pancreatic cancer risk: target trial emulation using real-world data. J Natl Cancer Inst 2025; 117:476-485. [PMID: 39418202 PMCID: PMC11884861 DOI: 10.1093/jnci/djae260] [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: 08/09/2024] [Revised: 09/16/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Data on the effects of glucagon-like peptide-1 receptor agonists (GLP-1RAs) on pancreatic cancer incidence are limited and inconsistent. Here we evaluate the association of GLP-1RAs, alone and in combinations, with incident pancreatic cancer risk in a real-world population, stratified by obesity and smoking status. METHODS This retrospective cohort included patients with type 2 diabetes mellitus who were prescribed GLP-1RAs or other nonglucagon-like peptide-1 receptor agonist antidiabetes medications between January 2013 and March 2019 and had no prior diagnosis of pancreatic cancer. The incident (first-time) diagnosis of pancreatic cancer during a 5-year follow-up was compared between propensity-score matched cohorts of patients prescribed GLP-1RAs vs other nonglucagon-like peptide-1 receptor agonist antidiabetes medications. Subgroup analyses were performed in patients stratified by the status of obesity and tobacco use disorder. We also compared GLP-1RA combination therapies with monotherapies. Time-to-first-event analysis was performed using Cox proportional hazards and Kaplan-Meier survival analysis, with the hazard ratio and 95% confidence interval calculated. RESULTS The study population comprised 1 636 056 eligible patients including 167 091 prescribed GLP-1RAs and 1 468 965 prescribed other antidiabetes medications. GLP-1RAs were associated with a statistically significant decreased risk for pancreatic cancer incidence compared with each of 6 nonglucagon-like peptide-1 receptor agonist antidiabetes medications with hazard ratios ranging from 0.42 to 0.82. The reduction was greater in patients with obesity and tobacco use disorder than in those without. GLP-1RA combination therapies were associated with lower pancreatic cancer risk compared with monotherapies. CONCLUSIONS GLP-1RAs were associated with reduced pancreatic cancer incidence in patients with type 2 diabetes mellitus. Further studies and trials are needed to explore mechanisms and confirm causal effects.
Collapse
Affiliation(s)
- Lindsey Wang
- Center for Science, Health, and Society, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States
| | - QuanQiu Wang
- Center for Artificial Intelligence in Drug Discovery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States
| | - Li Li
- Department of Family Medicine, University of Virginia, Charlottesville, VA 22903, United States
- University of Virginia Comprehensive Cancer Center, University of Virginia, Charlottesville, VA 22908, United States
| | - David C Kaelber
- Center for Clinical Informatics Research and Education, The MetroHealth System, Cleveland, OH 44109, United States
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States
| |
Collapse
|
12
|
Sridharan A, Dotan E, Dorta M, Vemula N, Handorf E, Deng M, Renning A, Sorice K, Laderman L, Whittington K, Cukierman E, Astsaturov I, Vijayvergia N, Meyer JE, Reddy SS, Lynch SM. Racial/Ethnic Differences and Effects of Clinical/Socioeconomic Factors on Time from Diagnosis to Treatment in Pancreatic Cancer. J Gastrointest Cancer 2025; 56:67. [PMID: 39954184 PMCID: PMC11829832 DOI: 10.1007/s12029-025-01188-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
PURPOSE Five-year survival for pancreatic adenocarcinoma (PDAC) is < 10% but can vary by a patient's race, socioeconomic status (SES), and the factors related to the neighborhood where a patient lives (nSES) . Prolonged time from diagnosis to first treatment (T2T) is another important disparity indicator. Here, we examined the effect of race, nSES, and patient-level clinical factors on T2T and survival in metastatic PDAC (mPDAC) patients. METHODS Patients with mPDAC treated at an academic cancer center between 2010 and 2018 (n = 334) were evaluated for nSES measures related to racial concentration, neighborhood deprivation, stability, immigration status, and transportation access from the US Census. We assessed and reported the effects of nSES and patient-level variables (age, race, gender, Charlson Comorbidity Index (CCI), etc.) on T2T and survival using univariate and multivariate Cox proportional hazards regression, hazard ratios (HR), confidence intervals (CI). RESULTS 82.9% of the patients were White; 17.1% were Black. Median T2T was 26 days with no significant difference in T2T and survival by race. In multivariable models, no nSES variables were significantly associated with T2T. T2T did not significantly impact survival; however, receipt of chemotherapy (HR = 0.14 [95% CI = 0.06, 0.30]) was associated with better survival outcomes. CONCLUSION Among patients with mPDAC, T2T was not associated with race/ethnic disparities or survival in a mostly White, high SES population treated at a comprehensive cancer center. Future investigations into pancreatic cancer disparities may be warranted in other hospital settings and in larger, more diverse study samples.
Collapse
Affiliation(s)
- Anush Sridharan
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Efrat Dotan
- Fox Chase Cancer Center, Cancer Prevention and Control, Young Pavilion P4159, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Marianna Dorta
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Navya Vemula
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Elizabeth Handorf
- Population Studies Facility, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Mengying Deng
- Population Studies Facility, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Ashley Renning
- Fox Chase Cancer Center, Cancer Prevention and Control, Young Pavilion P4159, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Kristen Sorice
- Fox Chase Cancer Center, Cancer Prevention and Control, Young Pavilion P4159, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Lauren Laderman
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Kate Whittington
- Fox Chase Cancer Center, Cancer Prevention and Control, Young Pavilion P4159, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Edna Cukierman
- Program of Cancer Signaling and Epigenetic, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Igor Astsaturov
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Namrata Vijayvergia
- Fox Chase Cancer Center, Cancer Prevention and Control, Young Pavilion P4159, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Joshua E Meyer
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Sanjay S Reddy
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Shannon M Lynch
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
- Fox Chase Cancer Center, Cancer Prevention and Control, Young Pavilion P4159, 333 Cottman Avenue, Philadelphia, PA, 19111, USA.
| |
Collapse
|
13
|
Kalra A, Meltzer SJ. The Role of DNA Methylation in Gastrointestinal Disease: An Expanded Review of Malignant and Nonmalignant Gastrointestinal Diseases. Gastroenterology 2025; 168:245-266. [PMID: 38971197 PMCID: PMC11698954 DOI: 10.1053/j.gastro.2024.07.001] [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: 10/10/2023] [Revised: 06/20/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Esophageal, colorectal, pancreatic, hepatocellular, and gastric cancer together impact millions of patients worldwide each year, with high overall mortality rates, and are increasing in incidence. Additionally, premalignant gastrointestinal diseases, such as Barrett's esophagus and inflammatory bowel disease, are also increasing in incidence. However, involvement of aberrant DNA methylation in these diseases is incompletely understood, especially given recent research advancements in this field. Here, we review knowledge of this epigenetic mechanism in gastrointestinal preneoplasia and neoplasia, considering mechanisms of action, genetic and environmental factors, and 5'-C-phosphate-G-3' island methylator phenotype. We also highlight developments in translational research, focusing on genomic-wide data, methylation-based biomarkers and diagnostic tests, machine learning, and therapeutic epigenetic strategies.
Collapse
Affiliation(s)
- Andrew Kalra
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Stephen J Meltzer
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
14
|
Wu Z, Zeng L, Fang Z, Yuan Y, Zhou Y, Chen R. Life's Essential 8, genetic susceptibility, and risk of incident pancreatic cancer: A prospective cohort study. Int J Cancer 2025; 156:566-574. [PMID: 39279141 DOI: 10.1002/ijc.35184] [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: 05/21/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/18/2024]
Abstract
The association between the American Heart Association (AHA) Life's Essential 8 (LE8) and the risk of pancreatic cancer (PC) remains unclear. Our goal was to assess the relationships between LE8, genetic susceptibility, and PC risk. This cohort consisted of 234,102 participants from the UK Biobank. The components of LE8 include diet, nicotine exposure, sleep, physical activity, blood glucose, body mass index, blood lipids, and blood pressure. LE8 is classified into three categories: low cardiovascular health (CVH), moderate CVH, and high CVH. Measurements were made using Cox proportional risk models to estimate impact of associations between LE8, genetic susceptibility, and incidence of PC in participants. Compared to participants with low LE8 scores, those with moderate and high LE8 scores had a 53% (HR, 0.47; 95% CI, 0.39-0.57) and 70% (HR, 0.30; 95% CI, 0.22-0.41) lower risk of developing PC, respectively. Interestingly, among individuals with high genetic risk, high LE8 scores were associated with greater benefits (HR, 0.24; 95% CI, 0.15-0.40), whereas the protective effect was weaker among those with low genetic risk (HR, 0.40; 95% CI, 0.21-0.75). Participants with a high LE8 score and a low polygenic risk score (PRS) had the lowest risk of PC (HR, 0.19; 95% CI: 0.11-0.33). Furthermore, we observed a significant additive interaction between LE8 and PRS. A higher LE8 score is associated with a lower risk of PC, especially for participants with a high PRS. These findings have important implications for participants most genetically predisposed to PC and for targeted strategies for PC prevention.
Collapse
Affiliation(s)
- Zhuo Wu
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Liangtang Zeng
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhou Fang
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yuan Yuan
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yu Zhou
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Rufu Chen
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
15
|
Wang H, Sun W, Zuo J, Wang G, Deng Z, Jiang Z. Gender Difference in the Trend and Forecast Analysis of Changes in the Burden of Disease of Pancreatic Cancer Attributable to Smoking in China, 1990-2021. Cancer Control 2025; 32:10732748251341521. [PMID: 40347157 PMCID: PMC12065983 DOI: 10.1177/10732748251341521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 04/09/2025] [Accepted: 04/23/2025] [Indexed: 05/12/2025] Open
Abstract
BackgroundPancreatic cancer is one of the most aggressive malignant tumors worldwide, often resulting in poor prognosis. Smoking is widely recognized as a significant risk factor for this disease.MethodsThis study extracted data on the disease burden of pancreatic cancer attributable to smoking in China from the Global Burden of Disease 2021 (GBD 2021) database. A total of 447 334 individuals were included in the analysis. A joinpoint regression model was used to calculate the annual percentage change (APC) and average annual percentage change (AAPC) to assess trends in the disease burden from 1990 to 2021. The age-period-cohort model was applied to explain the epidemiological trends of the disease by controlling for variables associated with age, period, and birth cohort. Decomposition analysis was conducted to quantify the relative contributions of population growth, population aging, and epidemiological changes. The Autoregressive Integrated Moving Average (ARIMA) model was utilized to forecast the burden of pancreatic cancer due to smoking in China over the next 15 years.ResultsFrom 1990 to 2021, the age-standardized mortality rate (ASMR) and the age-standardized disability-adjusted life years rate (ASDR) due to smoking-related pancreatic cancer in China exhibited an upward trend. By 2021, the total number of deaths and disability-adjusted life years (DALYs) had almost doubled compared to 1990. The mortality and DALY rates increased with age, peaking at approximately 85 years and 70 years, respectively, indicating a substantial burden on the elderly population. Furthermore, we predict that female ASMR and ASDR may start to decline in the next 15 years; however, the absolute burden remains unchanged. Conversely, the ASMR and ASDR for males continue to rise, indicating an increasing burden, with the male burden consistently surpassing that of females.ConclusionsThis study demonstrates that from 1990 to 2021, the burden of pancreatic cancer attributable to smoking in China has intensified, with significant disparities related to sex and age. The future outlook appears quite severe, given the large population base and accelerating population aging. Strong smoking cessation and control measures specifically targeting elderly males are imperative to limit tobacco exposure among high-risk groups, as these measures are crucial for alleviating the burden of pancreatic cancer in China.
Collapse
Affiliation(s)
- Haodi Wang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wenhao Sun
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jiaqian Zuo
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Gang Wang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Zhengming Deng
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Zhiwei Jiang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
16
|
Assarian BA, Byrne CD, McDonnell D, Hamady Z. Physical Activity and Incident Pancreatic Cancer: Results From the UK Biobank Prospective Cohort. Cureus 2025; 17:e78040. [PMID: 39872920 PMCID: PMC11770282 DOI: 10.7759/cureus.78040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2025] [Indexed: 01/30/2025] Open
Abstract
Background The relationship between physical activity and incident pancreatic cancer is poorly defined, and the evidence to date is inconsistent, largely due to small sample sizes and insufficient incident outcomes. Using the UK Biobank cohort dataset, the association between physical activity levels at recruitment and incident pancreatic ductal adenocarcinoma (PDAC) at follow-up was analysed. Method Physical activity, the key exposure, was quantified using Metabolic Equivalent Task (MET) values and categorised into walking, moderate, and vigorous activity. These categories were each analysed in quartiles. Summed activity was analysed both in quartiles and using International Physical Activity Questionnaire (IPAQ) activity levels (low, moderate, high). Univariate hazard ratios (HRs) and multivariable-adjusted HRs (aHRs) with 95% confidence intervals (CIs) were calculated using Cox regression analyses. Results A total of 542 incident PDAC cases and 2,139 controls (1:4 matching for age and sex) with a median (IQR) follow-up of 6.8 (1.7) years were analysed. No significant association was found in walking, moderate, and vigorous activities. In summed activity, only the 3rd quartile showed a statistically significant inverse association with PDAC risk (aHR 0.67, 95% CI 0.52-0.86, p<0.01). For IPAQ activity, the moderate and high activity groups showed borderline statistically significant associations with incident PDAC (aHR 0.80, 95% CI 0.63-1.00, p=0.05, and aHR 0.80, 95% CI 0.64-1.01, p=0.06, respectively). Conclusion The large UK Biobank cohort study did not show a strong association between physical activity levels and the development of incident PDAC.
Collapse
Affiliation(s)
- Borna A Assarian
- Human Development and Health, Faculty of Medicine University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, GBR
| | - Christopher D Byrne
- Human Development and Health, Faculty of Medicine University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, GBR
| | - Declan McDonnell
- Hepato-Pancreato-Biliary (HPB) Unit, University Hospital Southampton NHS Foundation Trust, Southampton, GBR
| | - Zaed Hamady
- Hepato-Pancreato-Biliary (HPB) Unit, University Hospital Southampton NHS Foundation Trust, Southampton, GBR
| |
Collapse
|
17
|
Harne PS, Harne V, Wray C, Thosani N. Endoscopic innovations in diagnosis and management of pancreatic cancer: a narrative review and future directions. Therap Adv Gastroenterol 2024; 17:17562848241297434. [PMID: 39664230 PMCID: PMC11632891 DOI: 10.1177/17562848241297434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/15/2024] [Indexed: 12/13/2024] Open
Abstract
Pancreatic cancer serves as the third leading cause of cancer-associated morbidity and mortality in the United States, with a 5-year survival rate of only 12% with an expected increase in incidence and mortality in the coming years. Pancreatic ductal adenocarcinomas constitute most pancreatic malignancies. Certain genetic syndromes, including Lynch syndrome, hereditary breast and ovarian cancer syndrome, hereditary pancreatitis, familial adenomatous polyposis, Peutz-Jeghers syndrome, familial pancreatic cancer mutation, and ataxia telangiectasia, confer a significantly higher risk. Screening for pancreatic malignancies currently targets patients with germline mutations or those with significant family history. Screening the general population is not currently viable owing to overall low incidence and lack of specific tests. Endoscopic ultrasound (EUS) and its applied advances are increasingly being used for surveillance, diagnosis, and management of pancreatic malignancies and have now become an indispensable tool in their management. For patients with risk factors, EUS in combination with magnetic resonance imaging/magnetic resonance cholangiopancreatography is used for screening. The role of endoscopic modalities has been expanding with the increased utilization of endoscopic retrograde cholangiopancreatography, EUS-directed therapies include EUS-guided fine-needle aspiration and EUS-fine-needle biopsy (FNB). EUS combined with FNB has the highest specificity and sensitivity for detecting pancreatic cancer amongst available modalities. Studies also recognize that artificial intelligence assisted EUS in the early detection of pancreatic cancer. At the same time, surgical resection has been historically considered the only curative treatment for pancreatic cancer, over 80% of patients present with unresectable disease. We also discuss EUS-guided therapies of physicochemicals (radiofrequency ablation, brachytherapy, and intratumor chemotherapy), biological agents (gene therapies and oncolytic viruses), and immunotherapy. We aim to perform a detailed review of the current burden, risk factors, role of screening, diagnosis, and endoscopic advances in the treatment modalities available for pancreatic cancer.
Collapse
Affiliation(s)
- Prateek Suresh Harne
- Division of Gastroenterology, Allegheny Health Network, Pittsburgh, PA 15212, USA
| | - Vaishali Harne
- Division of Pediatric Gastroenterology, The University of Texas
- Health Science Center and McGovern School of Medicine, Houston, TX, USA
| | - Curtis Wray
- Department of Surgery, The University of Texas Health Science Center and McGovern School of Medicine, Houston, TX, USA
| | - Nirav Thosani
- Department of Surgery and Interventional Gastroenterology, The University of Texas
- Health Science Center and McGovern School of Medicine, Houston, TX, USA
| |
Collapse
|
18
|
Grahovac J, Đurić A, Tanić M, Krivokuća A. Sex-Related Differences in Pancreatic Ductal Adenocarcinoma Progression and Response to Therapy. Int J Mol Sci 2024; 25:12669. [PMID: 39684385 DOI: 10.3390/ijms252312669] [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: 10/15/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most deadly malignancies with an increasing incidence rate and limited therapeutic options. Biological sex has an impact on many aspects of PDAC development and response to therapy, yet it is highly unappreciated in both basic and translational research, and worryingly in PDAC clinical trials. In this review, we summarize how biological sex influences PDAC incidence and mortality, genetic and epigenetic landscapes, anti-tumor immunity, responses to hormones, cachexia, and the efficacy of therapy. We highlight the importance of sex as a variable and discuss how to implement it into preclinical and clinical research. These considerations should be of use to researchers aiming at improving understanding of PDAC biology and developing precision medicine therapeutic strategies.
Collapse
Affiliation(s)
- Jelena Grahovac
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Ana Đurić
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Miljana Tanić
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Ana Krivokuća
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| |
Collapse
|
19
|
Paranal RM, Wood LD, Klein AP, Roberts NJ. Understanding familial risk of pancreatic ductal adenocarcinoma. Fam Cancer 2024; 23:419-428. [PMID: 38609521 PMCID: PMC11660179 DOI: 10.1007/s10689-024-00383-2] [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: 01/27/2024] [Accepted: 03/24/2024] [Indexed: 04/14/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease that is the result of an accumulation of sequential genetic alterations. These genetic alterations can either be inherited, such as pathogenic germline variants that are associated with an increased risk of cancer, or acquired, such as somatic mutations that occur during the lifetime of an individual. Understanding the genetic basis of inherited risk of PDAC is essential to advancing patient care and outcomes through improved clinical surveillance, early detection initiatives, and targeted therapies. In this review we discuss factors associated with an increased risk of PDAC, the prevalence of genetic variants associated with an increased risk in patients with PDAC, estimates of PDAC risk in carriers of pathogenic germline variants in genes associated with an increased risk of PDAC. The role of common variants in pancreatic cancer risk will also be discussed.
Collapse
Affiliation(s)
- Raymond M Paranal
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Human Genetics Predoctoral Training Program, the McKusick-Nathans Department of Genetic Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laura D Wood
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alison P Klein
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, MD, USA.
| | - Nicholas J Roberts
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
20
|
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
|
21
|
Song L, Chen Z, Li Y, Ran L, Liao D, Zhang Y, Wang G. Trend and forecast analysis of the changing disease burden of pancreatic cancer attributable to high fasting glucose in China, 1990-2021. Front Oncol 2024; 14:1471699. [PMID: 39493456 PMCID: PMC11527594 DOI: 10.3389/fonc.2024.1471699] [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: 07/29/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024] Open
Abstract
Background Pancreatic cancer (PC) is a malignant tumour with poor prognosis and high mortality, and high fasting plasma glucose (HFPG) is considered to be one of its important risk factors. Methods PC disease burden data were obtained from the Global Burden of Disease Study 2021 (GBD 2021) database. Annual percent change (APC), average APC (AAPC), and 95% confidence interval (95% CI) were analysed using joinpoint linkpoint regression models to assess the trend of PC burden of disease between 1990 and 2021. An age-period-cohort model was used to estimate the independent effects of age, period, and cohort on PC burden, and data on PC mortality attributable to HFPG in China from 2022 to 2032 were analysed on the basis of a Bayesian age-period-cohort model projection. Results The number of Pc deaths due to HFPG continue to rise in China from 1990 to 2021, with age-standardised mortality (ASMR) and age-standardised disability-adjusted life-year rates with increasing AAPC values of 1.12% (95% CI, 0.73-1.52) and 1.00% (95% CI, 0.63-1.37), respectively. Throughout the study, we found that the overall level of PC disease burden was significantly higher in men than that in women. In age-period-cohort analyses, the age effect of PC showed an increasing and then decreasing trend, the period effect showed an overall increasing trend during the study period, and the cohort effect showed an overall slow decreasing trend. In addition, the BAPC model predicted that ASMR is expected to decline significantly in both men and women from 2022 to 2032. Conclusions It was found that PC attributable to HFPG was generally on the rise in China from 1990 to 2021 and has been on the decline in recent years, and projections suggest that the country's future PC disease burden will continue to show a downward trend. Age and period of birth are the main factors affecting the disease burden, especially in men and older age groups. Early prevention, regular screening, and research into the pathogenesis of PC have, therefore, become particularly important.
Collapse
Affiliation(s)
- Lichen Song
- School of Clinical Medicine, Dali University, Dali, Yunnan, China
| | - Ziyi Chen
- School of Clinical Medicine, Dali University, Dali, Yunnan, China
| | - Yongjie Li
- School of Clinical Medicine, Dali University, Dali, Yunnan, China
| | - Lirong Ran
- School of Clinical Medicine, Dali University, Dali, Yunnan, China
| | - Dongwei Liao
- School of Clinical Medicine, Dali University, Dali, Yunnan, China
| | - Yuanyuan Zhang
- Medicine Department, School of Clinical Medicine, Dali University, Dali, Yunnan, China
| | - Guangming Wang
- Center of Genetic Testing, The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| |
Collapse
|
22
|
Wang X, Wang Z, Wang X. Passive smoking and risk of pancreatic cancer: an updated systematic review and meta-analysis. PeerJ 2024; 12:e18017. [PMID: 39399427 PMCID: PMC11468807 DOI: 10.7717/peerj.18017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/09/2024] [Indexed: 10/15/2024] Open
Abstract
Background Previous meta-analysis has demonstrated that no association was validated between passive smoking and pancreatic cancer. However, there is growing evidence on this issue recently. This study aimed to confirm this association. Methods PubMed, Embase, Web of Science, and Cochrane Library databases were searched up to April 2024 for retrieval of full articles. Studies with the exposure of passive smoking and outcome of pancreatic cancer were eligible for the analysis. We generated pooled relative risks (RRs) and 95% confidence intervals (CIs) using DerSimonian-Laird random-effects models. Quality of evidence was assessed using the GRADE system. Results Fourteen studies were included, with 5,560 pancreatic cancer patients. Passive smoking was associated with a moderate increased risk of pancreatic cancer (RR = 1.20, 95% CI: 1.11-1.30, p < 0.001). The results were consistent in both case-control (p=0.013) and cohort studies (p < 0.001) and in studies with high (p = 0.007) and moderate quality (p < 0.001). In subgroup analysis, the risk was significant for both current (RR=1.91, 95% CI: 1.45-2.51, p < 0.001) and non-current smokers (RR = 1.17, 95% CI: 1.01-1.36, p = 0.037), for exposure both in adulthood (RR = 1.18, 95% CI: 1.06-1.31, p = 0.002) and childhood (RR = 1.20, 95% CI: 1.08-1.34, p = 0.001). However, only regular or daily exposure (RR=1.28, 95% CI: 1.08-1.50, p = 0.003), rather than exposing occasionally, seldom or few times per week (p = 0.421), to passive smoking could increase the risk of pancreatic cancer. Conclusion Passive smoking exposure confers a significant increased risk for pancreatic cancer. The risk was valid in both case-control and cohort, high and moderate quality studies, in current and non-current smokers, and for both childhood and adulthood exposure. Regular or daily exposure rather than exposing occasionally, seldom or few times per week could exert a detrimental effect on pancreatic cancer.
Collapse
Affiliation(s)
- Xudong Wang
- Minimally Invasive Interventional Therapy Center, Qingdao Hospital University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Zihan Wang
- Department of Ultrasound, Qingdao Hospital University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Xujie Wang
- Minimally Invasive Interventional Therapy Center, Qingdao Hospital University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| |
Collapse
|
23
|
Turner KM, Patel SH. Pancreatic Cancer Screening among High-risk Individuals. Surg Clin North Am 2024; 104:951-964. [PMID: 39237170 DOI: 10.1016/j.suc.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) continues to remain one of the leading causes of cancer-related death. Unlike other malignancies where universal screening is recommended, the same cannot be said for PDAC. The purpose of this study is to review which patients are at high risk of developing PDAC and therefore candidates for screening, methods/frequency of screening, and risk for these groups of patients.
Collapse
Affiliation(s)
- Kevin M Turner
- Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0558, USA
| | - Sameer H Patel
- Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0558, USA; Division of Surgical Oncology, Medical Science Building 231 Albert Sabin Way, Cincinnati, OH 45267-0558, USA.
| |
Collapse
|
24
|
Netto D, Frizziero M, Foy V, McNamara MG, Backen A, Hubner RA. Systemic Therapy for Metastatic Pancreatic Cancer-Current Landscape and Future Directions. Curr Oncol 2024; 31:5206-5223. [PMID: 39330013 PMCID: PMC11430697 DOI: 10.3390/curroncol31090385] [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: 07/03/2024] [Revised: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 09/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a significant cause of cancer-associated mortality, with a rising global incidence. A paucity of strong predictive risk factors mean screening programmes are difficult to implement. Historically, a lack of identifiable and actionable driver mutations, coupled with a relatively immunosuppressed tumour microenvironment, has led to a reliance on cytotoxic chemotherapy. The NAPOLI-3 trial has reported data supporting consideration of NALIRIFOX as a new first-line standard of care. Kirsten Rat Sarcoma Virus (KRAS) G12D mutations are present in >90% of all PDAC's; exciting breakthroughs in small molecule inhibitors targeting KRAS G12D may open new modalities of treatment, and therapies targeting multiple KRAS mutations are also in early clinical trials. Although immunotherapy strategies to date have been disappointing, combination with chemotherapy and/or small molecule inhibitors hold promise and warrant further exploration.
Collapse
Affiliation(s)
- Daniel Netto
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Melissa Frizziero
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Victoria Foy
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Mairéad G. McNamara
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Alison Backen
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Richard A. Hubner
- The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| |
Collapse
|
25
|
Elebo N, Mpinganjira MG, Baichan P, Devar J, Omoshoro-Jones J, Francis JM, Smith M, Nweke EE. The need for research targeting the link between occupational carcinogens and hepatopancreatobiliary cancers in Africa: A systematic review. Transl Oncol 2024; 47:102036. [PMID: 38878612 PMCID: PMC11225925 DOI: 10.1016/j.tranon.2024.102036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/25/2024] [Accepted: 06/11/2024] [Indexed: 07/08/2024] Open
Abstract
INTRODUCTION Hepatopancreatobiliary (HPB) cancers encompassing malignancies of the liver, pancreas, gall bladder, and bile ducts pose a significant health burden in Africa. While the association of certain occupational carcinogens in cancer is well established globally, their potential role in HPB cancers remains understudied, especially in an African context. AIM This systematic review delves into the association between occupational carcinogens and HPB cancer in Africa. It examines the current state of research on occupational carcinogens and HPB cancers in Africa, identifying key challenges and knowledge gaps. METHODS This systematic review examined publications (published between 01 January 2012 and 31 May 2023) that highlight occupational carcinogens and HBP cancers in Africa. The search was conducted on electronic databases namely PubMed, Web of Science, and Africa Wide Information. RESULT Due to the lack of information on the association between occupational carcinogens and HPB cancers in Africa, as a result of the paucity of published studies, only four articles were included in this study. Hepatocellular carcinoma (HCC) was the predominant cancer associated with the occupational carcinogen, aflatoxin. Agricultural workers, especially those involved in the production and processing of maize and peanuts, appear to be the most exposed to aflatoxin. CONCLUSION Despite the sample size limitations due to the paucity of research studies on occupational carcinogens and HPB cancers in Africa, this study provides a reasonable tool for subsequent epidemiological studies. There is a need for more research on the association of occupational carcinogens and HPB cancers in Africa, especially with the growing industrialization.
Collapse
Affiliation(s)
- Nnenna Elebo
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand. Johannesburg 2193, Gauteng, South Africa; International Centre for Genetic Engineering and Biotechnology, Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Mafuno Grace Mpinganjira
- Department of Family Medicine, Faculty of Health Sciences, University of Witwatersrand. Johannesburg 2193, Gauteng, South Africa
| | - Pavan Baichan
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand. Johannesburg 2193, Gauteng, South Africa
| | - John Devar
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand. Johannesburg 2193, Gauteng, South Africa; Hepatopancreatobiliary unit, Department of Surgery, Chris Hani-Baragwanath Academic Hospital, Soweto Johannesburg, South Africa
| | - Jones Omoshoro-Jones
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand. Johannesburg 2193, Gauteng, South Africa; Hepatopancreatobiliary unit, Department of Surgery, Chris Hani-Baragwanath Academic Hospital, Soweto Johannesburg, South Africa
| | - Joel Msafiri Francis
- Department of Family Medicine, Faculty of Health Sciences, University of Witwatersrand. Johannesburg 2193, Gauteng, South Africa
| | - Martin Smith
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand. Johannesburg 2193, Gauteng, South Africa; Hepatopancreatobiliary unit, Department of Surgery, Chris Hani-Baragwanath Academic Hospital, Soweto Johannesburg, South Africa
| | - Ekene Emmanuel Nweke
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand. Johannesburg 2193, Gauteng, South Africa; Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Florida, Roodepoort, South Africa.
| |
Collapse
|
26
|
Tan JY, Yeo YH, Ng WL, Chiang CH, Stucky CC, Wasif N, Fong ZV. Pancreatic cancer mortality trends in the United States: how much have we moved the needle? J Gastrointest Oncol 2024; 15:1789-1795. [PMID: 39279936 PMCID: PMC11399823 DOI: 10.21037/jgo-24-213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/27/2024] [Indexed: 09/18/2024] Open
Abstract
Background Despite advances made in pancreatic cancer treatment, the extent of progress made in pancreatic cancer mortality at the population level remains unclear. Our cross-sectional study sought to measure trends in pancreatic cancer mortality in the United States in the last 2 decades. Methods Patients with pancreatic cancer mortality from 1999 to 2020 were analyzed from the Centers for Disease Control and Prevention's Wide-Ranging Online Data for Epidemiologic Research (CDC WONDER). Age-adjusted mortality rates (AAMRs) per 100,000 individuals were measured. We used joinpoint trend analysis to determine average annual percent change (AAPC) in AAMR trends. Results From 1999 to 2020, pancreatic cancer accounted for 809,197 deaths. Overall, the AAMRs of pancreatic cancer increased from 20.74 per 100,000 individuals in 1999 to 21.60 per 100,000 individuals in 2020. The highest AAMR was recorded in non-Hispanic Black males (30.11 per 100,000 individuals), and the lowest, in non-Hispanic White females (18.51 per 100,000 individuals). Patients aged 75-84 years had the highest AAMR (6.87 per 100,000 individuals) compared to the younger patients. The highest AAMR was observed in the Northeast region (22.07 per 100,000 individuals) and rural regions (21.29 per 100,000 individuals). Conclusions There was no improvement in pancreatic cancer mortality in the last two decades. These findings emphasize the importance of efforts to increase access to multidisciplinary cancer care with the realization that without it, improvements in treatment standards will not translate to lower cancer mortality at the population level.
Collapse
Affiliation(s)
- Jia Yi Tan
- Department of Internal Medicine, New York Medical College at Saint Michael’s Medical Center, Newark, NJ, USA
| | - Yong Hao Yeo
- Department of Internal Medicine/Pediatrics, Corewell Health, Royal Oak, MI, USA
| | - Wern Lynn Ng
- Department of Internal Medicine, University of Pittsburgh Medical Center (UPMC) Harrisburg, Harrisburg, PA, USA
| | - Cho Han Chiang
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Chee-Chee Stucky
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Nabil Wasif
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Zhi Ven Fong
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, Mayo Clinic Arizona, Phoenix, AZ, USA
| |
Collapse
|
27
|
Dinagde TA, Abubeker Z. Surgical management of pancreatic cancer in Tikur Anbessa Specialized Hospital, Addis Ababa, Ethiopia: a 5 years retrospective descriptive study. BMC Surg 2024; 24:223. [PMID: 39103810 DOI: 10.1186/s12893-024-02503-2] [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: 08/28/2023] [Accepted: 02/12/2024] [Indexed: 08/07/2024] Open
Abstract
INTRODUCTION The incidence of Pancreatic cancer is different in different parts of the world. It is a cancer with the worst prognosis of all malignancies. Pancreatic cancer is predominantly a disease of an older population. There are different environmental (modifiable) and non-modifiable risk factors associated with the development of pancreatic cancer. At present, surgical resection is the only potential cure for pancreatic cancer. However, as only 10-20% of the patients have resectable disease at the time of diagnosis. The morbidities associated with surgeries for pancreatic cancers remain high though the post-operative mortality has shown significant reduction in the past few decades. So far, no study has been conducted to investigate pancreatic cancer in Ethiopia. OBJECTIVES To assess the clinico-pathologic profile, associated factors, surgical management and short-term outcome of patients with pancreatic cancer in Tikur Anbessa Specialized hospital. METHODS A 5 years retrospective hospital-based cross-sectional study was conducted on 52 patients operated with the diagnosis of pancreatic cancer with either curative or palliative intents. The study period was from April 2016 to July 2021. The data collected includes demographic profile, associated risk factors and comorbidities, clinical presentations, biochemical parameters, pathologic features of the tumors as well as type of treatment offered and short term treatment outcome. The data was analyzed using SPSS version 25. RESULT The mean and median age of patients was 54.1 and 54.5% respectively. Males constitute about 52% the patients. 21% of the patients have potential risk factors; whereas only 10 (19.2%) of the patients had medical comorbidities. Median duration of symptoms at diagnosis was 12 weeks. Abdominal pain (88.5%) was the most common presenting symptom followed by anorexia (80.8%) and significant weight loss (78.8%), while 71.2% of the patients have jaundice. On clinical evaluation, 69.2% were jaundiced, while 34.6% had a palpable gallbladder. More than two third of patients presented with advanced disease. 76.9% of the tumors are located in the head of pancreas. More than three quarters (77%) of the surgeries performed were palliative. Postoperative morbidity and mortality were 19.2% and 3.8% respectively. CONCLUSION Age at first diagnosis of pancreatic cancer is relatively earlier in our setup. Most patients present with advanced condition, only amenable for palliative measures. The post-operative morbidity and mortality are more or less comparable with similar studies. The need for adjuvant therapy in pancreatic cancer should be emphasized.
Collapse
Affiliation(s)
- Tesfaye Aga Dinagde
- Department of Surgery, Tikur Anbessa Specialized Hospital, Addis Ababa University, Addis Ababa, Ethiopia.
| | - Zeki Abubeker
- Department of Surgery, Tikur Anbessa Specialized Hospital, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
28
|
Grigorescu RR, Husar-Sburlan IA, Gheorghe C. Pancreatic Cancer: A Review of Risk Factors. Life (Basel) 2024; 14:980. [PMID: 39202722 PMCID: PMC11355429 DOI: 10.3390/life14080980] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Pancreatic adenocarcinoma is one of the most lethal types of gastrointestinal cancer despite the latest medical advances. Its incidence has continuously increased in recent years in developed countries. The location of the pancreas can result in the initial symptoms of neoplasia being overlooked, which can lead to a delayed diagnosis and a subsequent reduction in the spectrum of available therapeutic options. The role of modifiable risk factors in pancreatic cancer has been extensively studied in recent years, with smoking and alcohol consumption identified as key contributors. However, the few screening programs that have been developed focus exclusively on genetic factors, without considering the potential impact of modifiable factors on disease occurrence. Thus, fully understanding and detecting the risk factors for pancreatic cancer represents an important step in the prevention and early diagnosis of this type of neoplasia. This review reports the available evidence on different risk factors and identifies the areas that could benefit the most from additional studies.
Collapse
Affiliation(s)
- Raluca Roxana Grigorescu
- Gastroenterology Department, “Sfanta Maria” Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | | | - Cristian Gheorghe
- Center for Digestive Disease and Liver Transplantation, Fundeni Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
29
|
Zhao L, Kan Y, Wang L, Pan J, Li Y, Zhu H, Yang Z, Xiao L, Fu X, Peng F, Ren H. Roles of long non‑coding RNA SNHG16 in human digestive system cancer (Review). Oncol Rep 2024; 52:106. [PMID: 38940337 PMCID: PMC11234248 DOI: 10.3892/or.2024.8765] [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: 05/23/2023] [Accepted: 04/26/2024] [Indexed: 06/29/2024] Open
Abstract
The incidence of tumors in the human digestive system is relatively high, including esophageal cancer, liver cancer, pancreatic cancer, gastric cancer and colorectal cancer. These malignancies arise from a complex interplay of environmental and genetic factors. Among them, long non‑coding RNAs (lncRNAs), which cannot be translated into proteins, serve an important role in the development, progression, migration and prognosis of tumors. Small nucleolar RNA host gene 16 (SNHG16) is a typical lncRNA, and its relationship with digestive system tumors has been widely explored. The prevailing hypothesis suggests that the principal molecular mechanism of SNHG16 in digestive system tumors involves it functioning as a competitive endogenous RNA that interacts with other proteins, regulates various genes and influences a downstream target molecule. The present review summarizes recent research on the relationship between SNHG16 and numerous types of digestive system cancer, encompassing its biological functions, underlying mechanisms and potential clinical implications. Furthermore, it outlines the association between SNHG16 expression and pertinent risk factors, such as smoking, infection and diet. The present review indicated the promise of SNHG16 as a potential biomarker and therapeutic target in human digestive system cancer.
Collapse
Affiliation(s)
- Lujie Zhao
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Yuling Kan
- Central Laboratory of Binzhou People's Hospital, Binzhou, Shandong 256600, P.R. China
| | - Lu Wang
- School of Clinical Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Jiquan Pan
- School of Clinical Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Yun Li
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Haiyan Zhu
- Department of Medical Oncology, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
- Department of Medical Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Zhongfa Yang
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Lin Xiao
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Xinhua Fu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Fujun Peng
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
- Weifang Key Laboratory of Collaborative Innovation of Intelligent Diagnosis and Treatment and Molecular Diseases, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Haipeng Ren
- Department of Medical Oncology, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
- Department of Medical Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
30
|
Abushamma S, Chen LS, Chen J, Smock N, Pham G, Chen CH. Enabling tobacco treatment for gastroenterology patients via a novel low-burden point-of-care model. BMC Health Serv Res 2024; 24:752. [PMID: 38902682 PMCID: PMC11188289 DOI: 10.1186/s12913-024-11092-y] [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: 04/24/2023] [Accepted: 05/08/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND & AIM Smoking is a major risk factor for multiple gastrointestinal cancers, and adversely affects peptic ulcer disease, gastroesophageal reflux, pancreatitis and Crohn's disease. Despite key recommendations for diagnosing and treating tobacco use disorder in healthcare settings, the degree to which this is implemented in Gastroenterology (GI) clinics is unknown. We aimed to assess our providers' practices, identify barriers for implementing evidence-based smoking cessation treatments, and address these barriers by implementing a novel low-burden point of care Electronic health record-enabled evidence-based tobacco treatment (ELEVATE), in GI clinics. METHODS An online survey was distributed to clinic gastroenterologists. ELEVATE module training was implemented in 1/2021. Data were evaluated during pre (7/2020-12/2020) and post (1/2021-12/2021) implementation periods to evaluate the reach and effectiveness of ELEVATE. Generalized estimating equations (GEE) were used to generate rate ratios (RR) to evaluate the intervention. RESULTS 91% (20/22) of GI physicians responded to our survey, and only 20% often assisted patients who smoke with counseling. Lack of a systematic program to offer help to patients was reported by 80% of providers as an extremely/very important barrier limiting their smoking cessation practices. The proportion of current patients who smoke receiving cessation treatment increased from pre-ELEVATE to post-ELEVATE (14.36-27.47%, RR = 1.90, 95% CI 1.60-2.26, p < .001). Post-ELEVATE, 14.4% (38/264) of patients with treatment quit smoking, compared to 7.9% (55/697) of patients without treatment (RR = 1.89, 95% CI 1.26-2.82, p = .0021). CONCLUSION Smoking practices are frequently assessed in GI clinics but barriers limiting cessation treatment exist. The use of a low burden point of care EHR enabled smoking cessation treatment module has led to a significant improvement in the treatment of smoking and subsequent cessation in our clinics. This study sheds light on an often under-recognized source of morbidity in GI patients and identifies an efficient, effective, and scalable strategy to combat tobacco use and improve clinical outcomes in our patients.
Collapse
Affiliation(s)
- Suha Abushamma
- Division of Gastroenterology, John T. Milliken Department of Medicine, Washington University School of Medicine, 600 S. Euclid Avenue, MSC-8124-21-427, Saint Louis, MO, 63110, USA.
| | - Li-Shiun Chen
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jingling Chen
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nina Smock
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital, Washington University School of Medicine, Saint Louis, MO, USA
| | - Giang Pham
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Chien-Huan Chen
- Division of Gastroenterology, John T. Milliken Department of Medicine, Washington University School of Medicine, 600 S. Euclid Avenue, MSC-8124-21-427, Saint Louis, MO, 63110, USA
| |
Collapse
|
31
|
Brehany S, Colton M, Duarte C, Baliton M, McCranie AS, Okuyama S. Hereditary Cancer Screening and Outcomes at an Urban Safety-Net Hospital. JCO Precis Oncol 2024; 8:e2300699. [PMID: 38935898 PMCID: PMC11371097 DOI: 10.1200/po.23.00699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/07/2024] [Accepted: 04/26/2024] [Indexed: 06/29/2024] Open
Abstract
PURPOSE Patients with hereditary cancer syndromes (HCS) have a high lifetime risk of developing cancer. Historically underserved populations have lower rates of genetic evaluation. We sought to characterize demographic factors that are associated with undergoing HCS evaluation in an urban safety-net patient population. METHODS All patients who met inclusion criteria for this study from 2016 to 2021 at an urban safety-net hospital were included in this analysis. Inclusion criteria were pathologically confirmed breast, ovarian/fallopian tube, colon, pancreatic, and prostate cancers. Patients also qualified for hereditary breast and ovarian cancers or Lynch syndrome on the basis of National Comprehensive Cancer Network guidelines. Institutional review board approval was obtained. Demographic and oncologic data were collected through retrospective chart review. Univariable and multivariable logistic regression models were constructed. RESULTS Of the 637 patients included, 40% underwent genetic testing. Variables associated with receiving genetic testing on univariable analysis included patients living at the time of data collection, female sex, Latinx ethnicity, Spanish language, family history of cancer, and referral for genetic testing. Patients identifying as Black, having Medicare, having pancreatic or prostate cancer, having stage IV disease, having Eastern Cooperative Oncology Group (ECOG) prognostic score ≥1, having medium or high Charlson comorbidity index, with current or previous cigarette use, and with previous alcohol use were negatively associated with testing. On multivariable modeling, family history of cancer was positively associated with testing. Patients identifying as Black, having colon or prostate cancer, and having ECOG score of 2 had significantly lower association with genetic testing. CONCLUSION Uptake of HCS was lower in patients identifying as Black, those with colon or prostate cancer, and those with an ECOG score of 2. Efforts to increase HCS testing in these patients will be important to advance equitable cancer care.
Collapse
Affiliation(s)
| | - Meryl Colton
- University of Colorado Cancer Center, Aurora, CO
| | | | | | | | | |
Collapse
|
32
|
Akumuo RC, Reddy SP, Westwood C, Devarajan K, Barrak D, Reddy SS, Villano AM. Smoking history is associated with reduced efficacy of neoadjuvant therapy in pancreatic adenocarcinoma. J Gastrointest Surg 2024; 28:605-610. [PMID: 38704197 DOI: 10.1016/j.gassur.2024.01.007] [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: 08/25/2023] [Revised: 12/06/2023] [Accepted: 01/12/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Differential responses to neoadjuvant therapy (NAT) exist in pancreatic ductal adenocarcinoma (PDAC); however, contributing factors are poorly understood. Tobacco smoke is a common risk factor for PDAC, with nicotine-induced chemoresistance observed in other cancers. This study aimed to explore the potential association between tobacco use and NAT efficacy in PDAC. METHODS A single-center, retrospective analysis was conducted that included all consecutive patients with PDAC who underwent surgical resection after NAT with a documented smoking history (N = 208). NAT response was measured as percentage fibrosis in the surgical specimen. Multivariable models controlled for covariates and survival were modeled using the Kaplan-Meier method. RESULTS Postoperatively, major responses to NAT (>95% fibrosis) were less frequently observed in smokers than in nonsmokers (13.7% vs 30.4%, respectively; P = .021). Pathologic complete responses were similarly less frequent in smokers than in nonsmokers (2.1% vs 9.9%, respectively; P = .023). On multivariate analysis controlling for covariates, smoking history remained independently associated with lower odds of major fibrosis (odds ratio [OR], 0.25; 95% CI, 0.10-0.59; P = .002) and pathologic complete response (OR, 0.21; 95% CI, 0.03-0.84; P = .05). The median overall survival was significantly longer in nonsmokers than in smokers (39.1 vs 26.6 months, respectively; P = .05). CONCLUSION Tobacco use was associated with diminished pathologic responses to NAT. Future research to understand the biology underlying this observation is warranted and may inform differential NAT approaches or counseling among these populations.
Collapse
Affiliation(s)
- Rita C Akumuo
- Department of Surgery, Temple University Hospital, Philadelphia, Pennsylvania, United States; Division of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States.
| | - Sai P Reddy
- Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, United States
| | - Caroline Westwood
- Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, United States
| | - Karthik Devarajan
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States
| | - Dany Barrak
- Division of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States
| | - Sanjay S Reddy
- Division of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States
| | - Anthony M Villano
- Division of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States
| |
Collapse
|
33
|
Vedie A, Laouali N, Gelot A, Severi G, Boutron‐Ruault M, Rebours V. Childhood and adulthood passive and active smoking, and the ABO group as risk factors for pancreatic cancer in women. United European Gastroenterol J 2024; 12:440-450. [PMID: 38064161 PMCID: PMC11091772 DOI: 10.1002/ueg2.12487] [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: 05/16/2023] [Accepted: 09/10/2023] [Indexed: 05/15/2024] Open
Abstract
OBJECTIVES Active smoking and the A blood group are associated with pancreatic adenocarcinoma (PC) risk. However, potential interactions between those risk factors and the role of passive smoking have been little investigated. We aimed to explore specific and joint associations of passive and active smoking, and effect modification by the ABO blood group in French women. METHODS The study included 96,594 women from the E3N prospective cohort, mean age: 49 years (SD 6.7). Information on active and passive smoking was reported at inclusion and throughout follow-up. Cases were classified according to the International Classification of Diseases 10. Associations with passive and active smoking and effect modification by the ABO blood group were investigated with multivariable Cox regression models to estimate hazard ratios (HR) and 95% confidence intervals (CI). RESULTS During a 24-year median follow-up, 346 incident PC cases were identified. Current smoking compared with never and former smoking (HR 1.51 [95% CI 1.08-2.10]), and passive smoking in childhood compared with no childhood exposure (HR 1.47 [95% CI 1.08-2.00]) were associated with increased PC risk, but not passive exposure in adulthood (HR 1.16 [95% CI 0.91-1.47]). Exposure to both passive smoking in childhood and current smoking was associated with a stronger risk (HR 2.80 [95% CI 1.42-5.52]) than exposure to both current smoking and passive smoking only in adulthood (HR 1.68 [95% CI 1.10-2.57]) compared with neither passive nor active smoking. Associations between active smoking and PC risk were strongest in the O or B groups, while associations with passive smoking were strongest in the A or AB blood groups, but the interaction terms were not statistically significant. CONCLUSIONS Both current smoking and passive smoking in childhood were associated with PC risk, with a maximal risk of current smokers exposed to passive smoking during childhood. Possible interactions between blood groups and active or passive smoking must be investigated in a larger series.
Collapse
Affiliation(s)
- Anne‐Laure Vedie
- Pancreatology and Digestive Oncology Department ‐ Beaujon HospitalAPHPClichy et Université Paris‐CitéParisFrance
| | - Nasser Laouali
- Paris‐Saclay UniversityUVSQUniversity Paris‐SudInsermGustave Roussy“Exposome and Heredity” TeamCESPVillejuifFrance
- Department of Biostatistics and EpidemiologySchool of Public Health and Health SciencesUniversity of MassachusettsAmherstMassachusettsUSA
- Scripps Institution of OceanographyUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - Amandine Gelot
- Paris‐Saclay UniversityUVSQUniversity Paris‐SudInsermGustave Roussy“Exposome and Heredity” TeamCESPVillejuifFrance
| | - Gianluca Severi
- Paris‐Saclay UniversityUVSQUniversity Paris‐SudInsermGustave Roussy“Exposome and Heredity” TeamCESPVillejuifFrance
| | | | - Vinciane Rebours
- Pancreatology and Digestive Oncology Department ‐ Beaujon HospitalAPHPClichy et Université Paris‐CitéParisFrance
| |
Collapse
|
34
|
Subramanian SK, Brahmbhatt B, Bailey-Lundberg JM, Thosani NC, Mutha P. Lifestyle Medicine for the Prevention and Treatment of Pancreatitis and Pancreatic Cancer. Diagnostics (Basel) 2024; 14:614. [PMID: 38535034 PMCID: PMC10968821 DOI: 10.3390/diagnostics14060614] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 04/14/2025] Open
Abstract
The incidence of pancreatitis and pancreatic cancer is on the upswing in the USA. These conditions often lead to higher healthcare costs due to the complex nature of diagnosis and the need for specialized medical interventions, surgical procedures, and prolonged medical management. The economic ramification encompasses direct healthcare expenses and indirect costs related to productivity losses, disability, and potential long-term care requirements. Increasing evidence underscores the importance of a healthy lifestyle in preventing and managing these conditions. Lifestyle medicine employs evidence-based interventions to promote health through six key pillars: embracing a whole-food, plant-predominant dietary pattern; regular physical activity; ensuring restorative sleep; managing stress effectively; removing harmful substances; and fostering positive social connections. This review provides a comprehensive overview of lifestyle interventions for managing and preventing the development of pancreatitis and pancreatic cancer.
Collapse
Affiliation(s)
- Sruthi Kapliyil Subramanian
- Center for Interventional Gastroenterology at UTHealth (iGUT), Section of Endoluminal Surgery and Interventional Gastroenterology, Division of Elective General Surgery, Department of Surgery, McGovern Medical School at UTHealth, Houston, TX 77030, USA; (S.K.S.); (P.M.)
| | - Bhaumik Brahmbhatt
- Mayo Clinic, Division of Gastroenterology and Hepatology, Jacksonville, FL 32224, USA;
| | - Jennifer M. Bailey-Lundberg
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School at UTHealth, Houston, TX 77030, USA;
| | - Nirav C. Thosani
- Center for Interventional Gastroenterology at UTHealth (iGUT), Section of Endoluminal Surgery and Interventional Gastroenterology, Division of Elective General Surgery, Department of Surgery, McGovern Medical School at UTHealth, Houston, TX 77030, USA; (S.K.S.); (P.M.)
| | - Pritesh Mutha
- Center for Interventional Gastroenterology at UTHealth (iGUT), Section of Endoluminal Surgery and Interventional Gastroenterology, Division of Elective General Surgery, Department of Surgery, McGovern Medical School at UTHealth, Houston, TX 77030, USA; (S.K.S.); (P.M.)
| |
Collapse
|
35
|
Bogumil D, Stram D, Preston DL, Pandol SJ, Wu AH, McKean-Cowdin R, Conti DV, Setiawan VW. Excess pancreatic cancer risk due to smoking and modifying effect of quitting smoking: The Multiethnic Cohort Study. Cancer Causes Control 2024; 35:541-548. [PMID: 37924460 PMCID: PMC10838846 DOI: 10.1007/s10552-023-01811-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: 04/04/2023] [Accepted: 10/02/2023] [Indexed: 11/06/2023]
Abstract
PURPOSE Risk factors for pancreatic cancer include racial/ethnic disparities and smoking. However, risk trajectories by smoking history and race/ethnicity are unknown. We examined the association of smoking with pancreatic cancer by race/ethnicity to generate age-specific incidence estimates by smoking history. METHODS We modeled pancreatic cancer incidence by race/ethnicity, age, pack-years, and years-quit using an excess relative risk model for 182,011 Multiethnic Cohort participants. We tested heterogeneity of smoking variables and pancreatic cancer by race/ethnicity and predicted incidence by smoking history. RESULTS We identified 1,831 incident pancreatic cancer cases over an average 19.3 years of follow-up. Associations of pack-years (p interaction by race/ethnicity = 0.41) and years-quit (p interaction = 0.83) with pancreatic cancer did not differ by race/ethnicity. Fifty pack-years smoked was associated with 91% increased risk (95% CI 54%, 127%) relative to never smokers in the combined sample. Every year quit corresponded to 9% decreased excess risk (95% CI 2%, 15%) from pack-years smoked. Differences in baseline pancreatic cancer risk across racial/ethnic groups (p < 0.001) translated to large differences in risk for smokers at older ages across racial/ethnic groups (65-122 cases per 100,000 at age 70). CONCLUSION Smoking pack-years were positively associated with elevated pancreatic cancer risk. Predicted risk trajectories showed a high impact of smoking cessation at < 65 years. Although we did not identify significant heterogeneity in the association of pack-years or years quit with pancreatic cancer risk, current smoker risk varied greatly by race/ethnicity in later life due to large differences in baseline risk.
Collapse
Affiliation(s)
- David Bogumil
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Room 1517C, Los Angeles, CA, 90033, USA.
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA.
| | - Daniel Stram
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Room 1517C, Los Angeles, CA, 90033, USA
| | | | - Stephen J Pandol
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Veterans Affairs, Los Angeles, CA, USA
| | - Anna H Wu
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Room 1517C, Los Angeles, CA, 90033, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Roberta McKean-Cowdin
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Room 1517C, Los Angeles, CA, 90033, USA
| | - David V Conti
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Room 1517C, Los Angeles, CA, 90033, USA
- Center for Genetic Epidemiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Veronica Wendy Setiawan
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Room 1517C, Los Angeles, CA, 90033, USA
- Center for Genetic Epidemiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
36
|
Zhong H, Liu S, Zhu J, Xu TH, Yu H, Wu L. Elucidating the role of blood metabolites on pancreatic cancer risk using two-sample Mendelian randomization analysis. Int J Cancer 2024; 154:852-862. [PMID: 37860916 PMCID: PMC10843029 DOI: 10.1002/ijc.34771] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/12/2023] [Accepted: 10/02/2023] [Indexed: 10/21/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an uncommon but highly fatal malignancy. Identifying causal metabolite biomarkers offers an opportunity to facilitate effective risk assessment strategies for PDAC. In this study, we performed a two-sample Mendelian randomization (MR) study to characterize the potential causal effects of metabolites in plasma on PDAC risk. Genetic instruments were determined for a total of 506 metabolites from one set of comprehensive genome-wide association studies (GWAS) involving 913 individuals of European ancestry from the INTERVAL/EPIC-Norfolk cohorts. Another set of genetic instruments was developed for 483 metabolites from an independent GWAS conducted with 8299 individuals of European ancestry from the Canadian Longitudinal Study on Aging (CLSA) cohort. We analyzed GWAS data of the Pancreatic Cancer Cohort Consortium (PanScan) and the Pancreatic Cancer Case-Control Consortium (PanC4), comprising 8275 PDAC cases and 6723 controls of European ancestry. The association of metabolites with PDAC risk was assessed using the inverse-variance weighted (IVW) method, and complemented with sensitivity analyses of MR-Egger and MR-PRESSO tests. Potential side effects of targeting the identified metabolites for PDAC intervention were further evaluated by a phenome-wide MR (Phe-MR) analysis. Forty-four unique metabolites were identified to be significantly associated with PDAC risk, of which four top-ranking metabolites (X: 12798, X: 11787, X: 11308 and X: 19141) showed replication evidence when using instruments developed from both two cohorts. Our results highlight novel blood metabolites related to PDAC risk, which may help prioritize metabolic features for PDAC mechanistic research and further evaluation of their potential role in PDAC risk assessment.
Collapse
Affiliation(s)
- Hua Zhong
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Shuai Liu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Jingjing Zhu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Teddy H. Xu
- Torrey Pines High School, San Diego, CA, USA
| | - Herbert Yu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Lang Wu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| |
Collapse
|
37
|
Sbeit W, Gershovitz G, Shahin A, Shhadeh S, Salman M, Basheer M, Khoury T. Obesity Is Associated with Distal Migration of Pancreatic Adenocarcinoma to Body and Tail: A Multi-Center Study. Cancers (Basel) 2024; 16:359. [PMID: 38254848 PMCID: PMC10814908 DOI: 10.3390/cancers16020359] [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: 12/25/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
(1) Background: Pancreatic adenocarcinoma (PAC) is one of the most lethal types of cancer. Most cases of PAC occur in the head of the pancreas. Given the proximity of the pancreatic head to the bile duct, most patients present clinically during early stages of the disease, while distally located PAC could have delayed clinical presentation. (2) Aims: To assess predictors of non-head PAC. (3) Methods: A retrospective multicenter study was conducted, including all patients who had endoscopic ultrasound (EUS) for pancreatic masses and who had histologic confirmation of PAC. (4) Results: Of the 151 patients included, 92 (60.9%) had pancreatic head cancer, and 59 (39.1%) had distal pancreatic cancer. PAC at body was the most common location in the distal PAC group (31 patients (52.5%)). Logistic regression analysis demonstrated a significant association of obesity with distal migration of PAC (OR 4.44, 95% CI 1.15-17.19, p = 0.03), while none of the other assessed parameters showed a significant association. Notably, abdominal pain was more significantly associated with distal PAC vs. head location (OR 2.85, 95% CI 1.32-6.16, p = 0.008). (5) Conclusions: Obesity shows a significant association as a clinical predictor of distal PAC. Further studies are needed to better explore this association.
Collapse
Affiliation(s)
- Wisam Sbeit
- Gastroenterology Department, Galilee Medical Center, Nahariya 22100, Israel; (W.S.); (G.G.); (A.S.); (S.S.); (M.B.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Gil Gershovitz
- Gastroenterology Department, Galilee Medical Center, Nahariya 22100, Israel; (W.S.); (G.G.); (A.S.); (S.S.); (M.B.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Amir Shahin
- Gastroenterology Department, Galilee Medical Center, Nahariya 22100, Israel; (W.S.); (G.G.); (A.S.); (S.S.); (M.B.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Shhady Shhadeh
- Gastroenterology Department, Galilee Medical Center, Nahariya 22100, Israel; (W.S.); (G.G.); (A.S.); (S.S.); (M.B.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Mahmoud Salman
- Department of Surgery, Shaare Zedek Medical Center, Jerusalem 91120, Israel;
| | - Maamoun Basheer
- Gastroenterology Department, Galilee Medical Center, Nahariya 22100, Israel; (W.S.); (G.G.); (A.S.); (S.S.); (M.B.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Tawfik Khoury
- Gastroenterology Department, Galilee Medical Center, Nahariya 22100, Israel; (W.S.); (G.G.); (A.S.); (S.S.); (M.B.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| |
Collapse
|
38
|
Buckley CW, O’Reilly EM. Next-generation therapies for pancreatic cancer. Expert Rev Gastroenterol Hepatol 2024; 18:55-72. [PMID: 38415709 PMCID: PMC10960610 DOI: 10.1080/17474124.2024.2322648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/20/2024] [Indexed: 02/29/2024]
Abstract
INTRODUCTION Pancreas ductal adenocarcinoma (PDAC) is a frequently lethal malignancy that poses unique therapeutic challenges. The current mainstay of therapy for metastatic PDAC (mPDAC) is cytotoxic chemotherapy. NALIRIFOX (liposomal irinotecan, fluorouracil, leucovorin, oxaliplatin) is an emerging standard of care in the metastatic setting. An evolving understanding of PDAC pathogenesis is driving a shift toward targeted therapy. Olaparib, a poly-ADP-ribose polymerase (PARP) inhibitor, has regulatory approval for maintenance therapy in BRCA-mutated mPDAC along with other targeted agents receiving disease-agnostic approvals including for PDAC with rare fusions and mismatch repair deficiency. Ongoing research continues to identify and evaluate an expanding array of targeted therapies for PDAC. AREAS COVERED This review provides a brief overview of standard therapies for PDAC and an emphasis on current and emerging targeted therapies. EXPERT OPINION There is notable potential for targeted therapies for KRAS-mutated PDAC with opportunity for meaningful benefit for a sizable portion of patients with this disease. Further, emerging approaches are focused on novel immune, tumor microenvironment, and synthetic lethality strategies.
Collapse
Affiliation(s)
- Conor W. Buckley
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Eileen M. O’Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
- Weill Cornell Medicine, New York, USA
| |
Collapse
|
39
|
Mohammadi M, Tadger P, Sadeghi A, Salehi N, Rajabnia M, Paraandavaji E, Shafiei S, Pirani A, Hatamnejad MR, Taherifard E, Kheshti F, Naderilordejani A, Honarfar F, Rahmani K, Soruri M, Kord Varkaneh H, Dadras O, Jahanian A, Rasta S, Zali MR. Opium use and gastrointestinal cancers: a systematic review and meta-analysis study. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2024; 17:104-120. [PMID: 38994503 PMCID: PMC11234493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/02/2024] [Indexed: 07/13/2024]
Abstract
Aim The current systematic review and meta-analysis aimed to assess the association between Gastrointestinal (GI) cancers and opium use. Background GI malignancies are a global public health issue and are associated with many risk factors including genetic and lifestyle factors. Methods PubMed, Web of Science, Embase and Scopus and the Google Scholar search engine in addition to Persian databases including Magiran and SID were searched using relevant keywords. The associations of opium use, long duration of opium use, high daily amount opium use and high cumulative opium use and GI cancer and various subtypes of GI cancers were estimated and pooled in format of odds ratios (OR) and their corresponding 95% confidence intervals (CI) with a random effects model. Results 22 articles that were published between 1983 and 2022 entered the analyses. There were significant relationships between opium use based on crude effect sizes (OR: 2.53, 1.95-3.29) and adjusted effect sizes (OR: 2.64, 1.99-3.51), high daily opium use (or: 3.41, 1.92-6.06), long duration of opium use (OR: 3.03, 1.90-4.84) and high cumulative opium use (OR: 3.88, 2.35-6.41), all compared to never opium use, and GI cancer. The results were not sensitive to sensitivity analyses and no influential publication biases were found in these analyses. Conclusion Our meta-analysis showed that opium use could be associated with increased risk of overall and some particular GI cancers including oropharyngeal, gastric, pancreatic and colorectal cancers. Opium use as a potentially modifiable factor, therefore, should be more emphasized.
Collapse
Affiliation(s)
- Mahsa Mohammadi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Guilan, Iran
| | | | - Amir Sadeghi
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Niloufar Salehi
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Rajabnia
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Elham Paraandavaji
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sasan Shafiei
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Pirani
- Mental Health Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Hatamnejad
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Erfan Taherifard
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Kheshti
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Arman Naderilordejani
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Forough Honarfar
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khaled Rahmani
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Soruri
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Kord Varkaneh
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Omid Dadras
- Department of Global Public Health and Primary Care, University of Bergen, Norway
- Section Global Health and Rehabilitation, Western Norway University of Applied Science, Norway
| | - Ali Jahanian
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Rasta
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Schlueter DJ, Sulieman L, Mo H, Keaton JM, Ferrara TM, Williams A, Qian J, Stubblefield O, Zeng C, Tran TC, Bastarache L, Dai J, Babbar A, Ramirez A, Goleva SB, Denny JC. Systematic replication of smoking disease associations using survey responses and EHR data in the All of Us Research Program. J Am Med Inform Assoc 2023; 31:139-153. [PMID: 37885303 PMCID: PMC10746325 DOI: 10.1093/jamia/ocad205] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/04/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023] Open
Abstract
OBJECTIVE The All of Us Research Program (All of Us) aims to recruit over a million participants to further precision medicine. Essential to the verification of biobanks is a replication of known associations to establish validity. Here, we evaluated how well All of Us data replicated known cigarette smoking associations. MATERIALS AND METHODS We defined smoking exposure as follows: (1) an EHR Smoking exposure that used International Classification of Disease codes; (2) participant provided information (PPI) Ever Smoking; and, (3) PPI Current Smoking, both from the lifestyle survey. We performed a phenome-wide association study (PheWAS) for each smoking exposure measurement type. For each, we compared the effect sizes derived from the PheWAS to published meta-analyses that studied cigarette smoking from PubMed. We defined two levels of replication of meta-analyses: (1) nominally replicated: which required agreement of direction of effect size, and (2) fully replicated: which required overlap of confidence intervals. RESULTS PheWASes with EHR Smoking, PPI Ever Smoking, and PPI Current Smoking revealed 736, 492, and 639 phenome-wide significant associations, respectively. We identified 165 meta-analyses representing 99 distinct phenotypes that could be matched to EHR phenotypes. At P < .05, 74 were nominally replicated and 55 were fully replicated. At P < 2.68 × 10-5 (Bonferroni threshold), 58 were nominally replicated and 40 were fully replicated. DISCUSSION Most phenotypes found in published meta-analyses associated with smoking were nominally replicated in All of Us. Both survey and EHR definitions for smoking produced similar results. CONCLUSION This study demonstrated the feasibility of studying common exposures using All of Us data.
Collapse
Affiliation(s)
- David J Schlueter
- Precision Health Informatics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
- Department of Health and Society, University of Toronto, Scarborough, Toronto, ON, Canada
| | - Lina Sulieman
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Huan Mo
- Precision Health Informatics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
- The Cohort Analytics Core (CAC), Center for Precision Health Research, National Human Genome Research Institute, Bethesda, MD, USA
| | - Jacob M Keaton
- Precision Health Informatics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Tracey M Ferrara
- Precision Health Informatics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ariel Williams
- Precision Health Informatics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jun Qian
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Onajia Stubblefield
- Precision Health Informatics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Chenjie Zeng
- Precision Health Informatics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Tam C Tran
- Precision Health Informatics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
- The Cohort Analytics Core (CAC), Center for Precision Health Research, National Human Genome Research Institute, Bethesda, MD, USA
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jian Dai
- Precision Health Informatics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Anav Babbar
- Precision Health Informatics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Andrea Ramirez
- Precision Health Informatics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Slavina B Goleva
- Precision Health Informatics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Joshua C Denny
- Precision Health Informatics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
41
|
Su Y, Wang F, Lei Z, Li J, Ma M, Yan Y, Zhang W, Chen X, Xu B, Hu T. An Integrated Multi-Omics Analysis Identifying Immune Subtypes of Pancreatic Cancer. Int J Mol Sci 2023; 25:142. [PMID: 38203311 PMCID: PMC10779306 DOI: 10.3390/ijms25010142] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Limited studies have explored novel pancreatic cancer (PC) subtypes or prognostic biomarkers based on the altered activity of relevant signaling pathway gene sets. Here, we employed non-negative matrix factorization (NMF) to identify three immune subtypes of PC based on C7 immunologic signature gene set activity in PC and normal samples. Cluster 1, the immune-inflamed subtype, showed a higher response rate to immune checkpoint blockade (ICB) and had the lowest tumor immune dysfunction and exclusion (TIDE) scores. Cluster 2, the immune-excluded subtype, exhibited strong associations with stromal activation, characterized by elevated expression levels of transforming growth factor (TGF)-β, cell adhesion, extracellular matrix remodeling, and epithelial-to-mesenchymal transition (EMT) related genes. Cluster 3, the immune-desert subtype, displayed limited immune activity. For prognostic prediction, we developed an immune-related prognostic risk model (IRPM) based on four immune-related prognostic genes in pancreatic cancer, RHOF, CEP250, TSC1, and KIF20B. The IRPM demonstrated excellent prognostic efficacy and successful validation in an external cohort. Notably, the key gene in the prognostic model, RHOF, exerted significant influence on the proliferation, migration, and invasion of pancreatic cancer cells through in vitro experiments. Furthermore, we conducted a comprehensive analysis of somatic mutational landscapes and immune landscapes in PC patients with different IRPM risk scores. Our findings accurately stratified patients based on their immune microenvironment and predicted immunotherapy responses, offering valuable insights for clinicians in developing more targeted clinical strategies.
Collapse
Affiliation(s)
- Yongcheng Su
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (F.W.)
| | - Fen Wang
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (F.W.)
| | - Ziyu Lei
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (F.W.)
| | - Jiangquan Li
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (F.W.)
| | - Miaomiao Ma
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (F.W.)
| | - Ying Yan
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (F.W.)
| | - Wenqing Zhang
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (F.W.)
| | - Xiaolei Chen
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (F.W.)
| | - Beibei Xu
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Tianhui Hu
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (F.W.)
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
| |
Collapse
|
42
|
Jiang Z, Zheng X, Li M, Liu M. Improving the prognosis of pancreatic cancer: insights from epidemiology, genomic alterations, and therapeutic challenges. Front Med 2023; 17:1135-1169. [PMID: 38151666 DOI: 10.1007/s11684-023-1050-6] [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: 08/30/2023] [Accepted: 11/15/2023] [Indexed: 12/29/2023]
Abstract
Pancreatic cancer, notorious for its late diagnosis and aggressive progression, poses a substantial challenge owing to scarce treatment alternatives. This review endeavors to furnish a holistic insight into pancreatic cancer, encompassing its epidemiology, genomic characterization, risk factors, diagnosis, therapeutic strategies, and treatment resistance mechanisms. We delve into identifying risk factors, including genetic predisposition and environmental exposures, and explore recent research advancements in precursor lesions and molecular subtypes of pancreatic cancer. Additionally, we highlight the development and application of multi-omics approaches in pancreatic cancer research and discuss the latest combinations of pancreatic cancer biomarkers and their efficacy. We also dissect the primary mechanisms underlying treatment resistance in this malignancy, illustrating the latest therapeutic options and advancements in the field. Conclusively, we accentuate the urgent demand for more extensive research to enhance the prognosis for pancreatic cancer patients.
Collapse
Affiliation(s)
- Zhichen Jiang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of General Surgery, Division of Gastroenterology and Pancreas, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Xiaohao Zheng
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| | - Mingyang Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
43
|
Huang FF, Cui WH, Ma LY, Chen Q, Liu Y. Crosstalk of nervous and immune systems in pancreatic cancer. Front Cell Dev Biol 2023; 11:1309738. [PMID: 38099290 PMCID: PMC10720593 DOI: 10.3389/fcell.2023.1309738] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
Pancreatic cancer is a highly malignant tumor known for its extremely low survival rate. The combination of genetic disorders within pancreatic cells and the tumor microenvironment contributes to the emergence and progression of this devastating disease. Extensive research has shed light on the nature of the microenvironmental cells surrounding the pancreatic cancer, including peripheral nerves and immune cells. Peripheral nerves release neuropeptides that directly target pancreatic cancer cells in a paracrine manner, while immune cells play a crucial role in eliminating cancer cells that have not evaded the immune response. Recent studies have revealed the intricate interplay between the nervous and immune systems in homeostatic condition as well as in cancer development. In this review, we aim to summarize the function of nerves in pancreatic cancer, emphasizing the significance to investigate the neural-immune crosstalk during the advancement of this malignant cancer.
Collapse
Affiliation(s)
- Fei-Fei Huang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Wen-Hui Cui
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Lan-Yue Ma
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qi Chen
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
| | - Yang Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
44
|
Chen Z, He Y, Ding C, Chen J, Gu Y, Xiao M, Li Q. Safety and Efficacy Analysis of PD-1 Inhibitors in Combination with Gemcitabine Plus Nab-Paclitaxel for Advanced Pancreatic Cancer: A Real-World, Single-Center Study. Onco Targets Ther 2023; 16:923-935. [PMID: 37965584 PMCID: PMC10642393 DOI: 10.2147/ott.s427942] [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: 06/28/2023] [Accepted: 10/20/2023] [Indexed: 11/16/2023] Open
Abstract
Background Pancreatic cancer is a deadly disease with a low five years survival rate, and chemotherapy remains the standard treatment for advanced cases. However, the efficacy of chemotherapy alone is limited, and there is a need for new treatment options. Recently, immune checkpoint inhibitors (ICIs), particularly programmed death-1 (PD-1) inhibitors, have shown promising results in various cancers, including pancreatic cancer. In this study, we explore the safety and efficacy of PD-1 inhibitors in combination with chemotherapy for advanced pancreatic cancer. Materials and Methods A retrospective analysis was conducted on clinical data from 27 patients with advanced pancreatic cancer who were administered a combination of anti-PD-1 antibody and gemcitabine plus nab-paclitaxel (GnP) regimen. The study evaluated the safety of the treatment as well as the objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). Results In this study, treatment with a combination of anti-PD-1 antibody and GnP regimen for pancreatic cancer resulted in partial response (PR) for 10 out of 27 (37.04%) patients, stable disease (SD) for 10 (37.04%) patients, and progressive disease (PD) for 7 (25.92%) patients. The study found that the median OS (mOS) for these patients was 16.4 months [standard error (SE) = 1.117, 95% confidence interval (CI) 14.211-18.589], while the median PFS (mPFS) was 6.4 months (SE = 1.217, 95% CI 3.981-8.752). Subgroup analysis revealed that pancreatic cancer patients' Eastern Cooperative Oncology Group (ECOG) performance status (PS) (0 vs 1) and treatment cycles (≤6 cycles vs >6 cycles) significantly affected OS and PFS. Patients experienced mostly grade 1-2 adverse events (AEs), which were relieved through clinical treatment. Conclusion The combination of GnP with anti-PD-1 antibodies shows promise as a potential treatment option for advanced pancreatic cancer.
Collapse
Affiliation(s)
- Zhitao Chen
- Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310003, People’s Republic of China
| | - Yahui He
- School of Medicine, Zhejiang Chinese Medical University Zhejiang Shuren College, Hangzhou, 310003, People’s Republic of China
| | - Chenchen Ding
- Child and Adolescent Psychology, Affiliated Mental Health Centre & Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310013, People’s Republic of China
| | - Jun Chen
- School of Medicine, Zhejiang Chinese Medical University Zhejiang Shuren College, Hangzhou, 310003, People’s Republic of China
| | - Yangjun Gu
- Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310003, People’s Republic of China
| | - Min Xiao
- Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310003, People’s Republic of China
| | - Qiyong Li
- Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310003, People’s Republic of China
| |
Collapse
|
45
|
Wang L, Grimshaw AA, Mezzacappa C, Larki NR, Yang YX, Justice AC. Do Polygenic Risk Scores Add to Clinical Data in Predicting Pancreatic Cancer? A Scoping Review. Cancer Epidemiol Biomarkers Prev 2023; 32:1490-1497. [PMID: 37610426 PMCID: PMC10873036 DOI: 10.1158/1055-9965.epi-23-0468] [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: 05/02/2023] [Revised: 07/21/2023] [Accepted: 08/21/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Polygenic risk scores (PRS) summarize an individual's germline genetic risk, but it is unclear whether PRS offer independent information for pancreatic cancer risk prediction beyond routine clinical data. METHODS We searched 8 databases from database inception to March 10, 2023 to identify studies evaluating the independent performance of pancreatic cancer-specific PRS for pancreatic cancer beyond clinical risk factors. RESULTS Twenty-one studies examined associations between a pancreatic cancer-specific PRS and pancreatic cancer. Seven studies evaluated risk factors beyond age and sex. Three studies evaluated the change in discrimination associated with the addition of PRS to routine risk factors and reported improvements (AUCs: 0.715 to 0.745; AUC 0.791 to 0.830; AUC from 0.694 to 0.711). Limitations to clinical applicability included using source populations younger/healthier than those at risk for pancreatic cancer (n = 10), exclusively of European ancestry (n = 13), or controls without relevant exposures (n = 1). CONCLUSIONS While most studies of pancreatic cancer-specific PRS did not evaluate the independent discrimination of PRS for pancreatic cancer beyond routine risk factors, three that did showed improvements in discrimination. IMPACT For pancreatic cancer PRS to be clinically useful, they must demonstrate substantial improvements in discrimination beyond established risk factors, apply to diverse ancestral populations representative of those at risk for pancreatic cancer, and use appropriate controls.
Collapse
Affiliation(s)
- Louise Wang
- VA Connecticut Healthcare System, West Haven, CT, USA
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Catherine Mezzacappa
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Navid Rahimi Larki
- VA Connecticut Healthcare System, West Haven, CT, USA
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Yu-Xiao Yang
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA USA
| | - Amy C. Justice
- VA Connecticut Healthcare System, West Haven, CT, USA
- Section of General Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- School of Public Health, Yale University, New Haven, CT, USA
| |
Collapse
|
46
|
Leonhardt CS, Pils D, Qadan M, Gustorff C, Sahora K, Klaiber U, Warshaw AL, Prager G, Ferrone CR, Lillemoe KD, Schindl M, Strobel O, Castillo CFD, Hank T. Smoking impairs the effect of neoadjuvant FOLFIRINOX on postresection survival in pancreatic cancer. Eur J Cancer 2023; 193:113293. [PMID: 37713740 DOI: 10.1016/j.ejca.2023.113293] [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: 08/05/2023] [Accepted: 08/10/2023] [Indexed: 09/17/2023]
Abstract
INTRODUCTION Smoking plays an important role in carcinogenesis, including pancreatic ductal adenocarcinoma (PDAC). However, little is known about the association between smoking status and prognosis in resected PDAC. METHODS All patients who underwent resection for PDAC were identified from two prospective institutional databases. Clinicopathologic data as well as demographics including smoking status were extracted. Survival analysis and multivariable Cox regression modelling was performed. Restricted cubic splines were used for linear data to define cut-off points. RESULTS Out of 848 patients, 357 (42.1%) received neoadjuvant treatment (NAT), 491 upfront resection (57.9%), and 475 (56%) adjuvant therapy. The median overall survival (OS) was 27.8 months, 36.1 months, and 23.0 months for the entire cohort, after NAT and upfront resection. 464 patients were never smokers (54.7%), 250 former smokers (29.5%), and 134 active smokers (15.8%). In the multivariable model, the interaction of neoadjuvant FOLFIRINOX and active smoking was associated with the highest risk for decreased OS (harzard ratio (HR) 2.35; 95% confidence interval 1.13-4.90) and strongly mitigated the benefit of FOLFIRNOX (HR 0.40; 95% CI 0.25-0.63). Adjusted median OS in NAT patients with FOLFIRINOX was not reached for never and former smokers, compared to 26.2 months in active smokers. Based on the model, a nomogram was generated to illustrate the probability of 5-year survival after PDAC resection. CONCLUSION The present study confirms that neoadjuvant FOLFIRINOX is associated with excellent survival and demonstrates that active smoking reduces its benefit. The nomogram can assist in daily clinical practice and emphasises the importance of smoking cessation in patients with PDAC, especially prior to NAT with FOLFIRINOX.
Collapse
Affiliation(s)
- Carl-Stephan Leonhardt
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Dietmar Pils
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Motaz Qadan
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Charlotte Gustorff
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Klaus Sahora
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Ulla Klaiber
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Andrew L Warshaw
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gerald Prager
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Cristina R Ferrone
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Keith D Lillemoe
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Martin Schindl
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Oliver Strobel
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | | | - Thomas Hank
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria; Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
47
|
Luo W, Wang J, Chen H, Ye L, Qiu J, Liu Y, Wang R, Weng G, Liu T, Su D, Tao J, Ding C, You L, Zhang T. Epidemiology of pancreatic cancer: New version, new vision. Chin J Cancer Res 2023; 35:438-450. [PMID: 37969957 PMCID: PMC10643340 DOI: 10.21147/j.issn.1000-9604.2023.05.03] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 10/16/2023] [Indexed: 11/17/2023] Open
Abstract
Pancreatic cancer (PC) is a devastating malignancy with an extremely high mortality rate and poses significant challenges to healthcare systems worldwide. The prevalence of PC risk factors spiked over the years, leading to a global increase in PC incidence rates. The contribution of different risk factors, however, varied from region to region due to genetic predisposition, environmental, social, and political factors underlying disease prevalence in addition to public health strategies. This comprehensive review aims to provide a thorough analysis of the epidemiology of PC, discussing its incidence, risk factors, screening strategies and socioeconomic burden. We compiled a wide range of seminal studies as well as epidemiological investigations to serve this review as a comprehensive guide for researchers, healthcare professionals, and policymakers keen for a more profound understanding of PC epidemiology. This review highlights the essentiality of persistent research efforts, interdisciplinary collaboration, and public health initiatives to address the expanding burden of this malignancy.
Collapse
Affiliation(s)
- Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jun Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Hao Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Liyuan Ye
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yueze Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Ruobing Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Guihu Weng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Tao Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Dan Su
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jinxin Tao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Chen Ding
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
48
|
Xu Y, Liu W, Long Z, Wang L, Zhou M, Yin P. Mortality and years of life lost due to pancreatic cancer in China, its provinces, urban and rural areas from 2005 to 2020: results from the national mortality surveillance system. BMC Cancer 2023; 23:893. [PMID: 37735368 PMCID: PMC10512506 DOI: 10.1186/s12885-023-11258-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 08/05/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Pancreatic cancer is a growing public health concern in China, and depicting it from different perspectives would provide a comprehensive understanding of its epidemiological characteristics. METHODS Data from the National Mortality Surveillance System (NMSS) in China was used to estimate the number of deaths, years of life lost (YLL), age-standardized mortality rate (ASMR) and age-standardized YLL rate in China, its provinces and urban-rural areas from 2005 to 2020. Joinpoint regression analysis was employed to explore the temporal trends of ASMR and age-standardized YLL rate. Decomposition analysis was conducted to assess the contribution of population growth, population aging and cause-specific mortality rate to the increment of pancreatic cancer deaths. RESULTS A total of 100,427 pancreatic cancer deaths and 2,166,355 pancreatic cancer related YLL were estimated in China in 2020. The overall ASMR significantly increased from 6.6/100 000 in 2005 to 7.4/100 000 in 2020, and was higher in men than that in women. Age-standardized YLL rate showed a similar trend. The mortality rates of pancreatic cancer were generally higher in northeast China than in southwest China. The highest ASMRs were found in Jilin, Zhejiang, Inner Mongolia and Anhui, and the lowest ones in Guangxi, Yunnan, Tibet, and Hainan. The disease burden due to pancreatic cancer presented a significant upward trend in rural areas and a downward trend in urban areas. CONCLUSIONS The burden associated with pancreatic cancer had been increasing in China from 2005 to 2020. The escalating disease burden of pancreatic cancer in rural areas necessitates the implementation of effective control and prevention measures. Relevant provinces should pay greater attention to the prevailing of pancreatic cancer, particularly those exhibiting higher mortality rates.
Collapse
Affiliation(s)
- Yangyang Xu
- National Center for Chronic and Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 27 Nanwei Road, Xicheng District, Beijing, 10050, China
| | - Wei Liu
- National Center for Chronic and Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 27 Nanwei Road, Xicheng District, Beijing, 10050, China
| | - Zheng Long
- National Center for Chronic and Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 27 Nanwei Road, Xicheng District, Beijing, 10050, China
| | - Lijun Wang
- National Center for Chronic and Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 27 Nanwei Road, Xicheng District, Beijing, 10050, China
| | - Maigeng Zhou
- National Center for Chronic and Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 27 Nanwei Road, Xicheng District, Beijing, 10050, China
| | - Peng Yin
- National Center for Chronic and Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 27 Nanwei Road, Xicheng District, Beijing, 10050, China.
| |
Collapse
|
49
|
Koltai T. Earlier Diagnosis of Pancreatic Cancer: Is It Possible? Cancers (Basel) 2023; 15:4430. [PMID: 37760400 PMCID: PMC10526520 DOI: 10.3390/cancers15184430] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/31/2023] [Accepted: 08/06/2023] [Indexed: 09/29/2023] Open
Abstract
Pancreatic ductal adenocarcinoma has a very high mortality rate which has been only minimally improved in the last 30 years. This high mortality is closely related to late diagnosis, which is usually made when the tumor is large and has extensively infiltrated neighboring tissues or distant metastases are already present. This is a paradoxical situation for a tumor that requires nearly 15 years to develop since the first founding mutation. Response to chemotherapy under such late circumstances is poor, resistance is frequent, and prolongation of survival is almost negligible. Early surgery has been, and still is, the only approach with a slightly better outcome. Unfortunately, the relapse percentage after surgery is still very high. In fact, early surgery clearly requires early diagnosis. Despite all the advances in diagnostic methods, the available tools for improving these results are scarce. Serum tumor markers permit a late diagnosis, but their contribution to an improved therapeutic result is very limited. On the other hand, effective screening methods for high-risk populations have not been fully developed as yet. This paper discusses the difficulties of early diagnosis, evaluates whether the available diagnostic tools are adequate, and proposes some simple and not-so-simple measures to improve it.
Collapse
Affiliation(s)
- Tomas Koltai
- Hospital del Centro Gallego de Buenos Aires, Buenos Aires C1094, Argentina
| |
Collapse
|
50
|
Pajewska M, Partyka O, Czerw A, Deptała A, Cipora E, Gąska I, Wojtaszek M, Sygit K, Sygit M, Krzych-Fałta E, Schneider-Matyka D, Cybulska AM, Grochans E, Asendrych-Woźniak A, Romanowicz A, Drobnik J, Bandurska E, Ciećko W, Maciuszek-Bartkowska B, Curyło M, Wróbel K, Kozłowski R, Marczak M. Management of Metastatic Pancreatic Cancer-Comparison of Global Guidelines over the Last 5 Years. Cancers (Basel) 2023; 15:4400. [PMID: 37686675 PMCID: PMC10486352 DOI: 10.3390/cancers15174400] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Pancreatic cancer (PC) is usually diagnosed at an advanced stage of its development, which results in lower overall survival (OS). Prognosis is also poor even with curative-intent surgery. Approximately 80% of patients with localized PDAC have micrometastases at the time of diagnosis, which leads to a worse prognosis than in other cancers. The objective of this study is to present the progress in the treatment of metastatic pancreatic cancer based on the recommendations of oncological scientific societies, such as ESMO, NCCN, ASCO, NICE and SEOM, over the last 5 years. Combined FOLFIRINOX therapy is mostly a recommended therapy among patients with good performance statuses, while gemcitabine is recommended for more fragile patients as a first-line treatment. The newest guidelines suggest that molecular profiling of the tumor should be the first step in determining the course of treatment. The use of modern molecular therapies in patients with specific gene mutations should extend the survival of patients with this disease.
Collapse
Affiliation(s)
- Monika Pajewska
- Department of Health Economics and Medical Law, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.P.)
- Department of Economic and System Analyses, National Institute of Public Health NIH-National Research Institute, 00-791 Warsaw, Poland
| | - Olga Partyka
- Department of Health Economics and Medical Law, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.P.)
- Department of Economic and System Analyses, National Institute of Public Health NIH-National Research Institute, 00-791 Warsaw, Poland
| | - Aleksandra Czerw
- Department of Health Economics and Medical Law, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.P.)
- Department of Economic and System Analyses, National Institute of Public Health NIH-National Research Institute, 00-791 Warsaw, Poland
| | - Andrzej Deptała
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Elżbieta Cipora
- Medical Institute, Jan Grodek State University in Sanok, 38-500 Sanok, Poland
| | - Izabela Gąska
- Medical Institute, Jan Grodek State University in Sanok, 38-500 Sanok, Poland
| | - Marek Wojtaszek
- Medical Institute, Jan Grodek State University in Sanok, 38-500 Sanok, Poland
| | - Katarzyna Sygit
- Faculty of Health Sciences, Calisia University, 62-800 Kalisz, Poland
| | - Marian Sygit
- Faculty of Health Sciences, Calisia University, 62-800 Kalisz, Poland
| | - Edyta Krzych-Fałta
- Department of Basic of Nursing, Faculty of Health Sciences, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Daria Schneider-Matyka
- Department of Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 71-210 Szczecin, Poland
| | - Anna M. Cybulska
- Department of Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 71-210 Szczecin, Poland
| | - Elżbieta Grochans
- Department of Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 71-210 Szczecin, Poland
| | - Alicja Asendrych-Woźniak
- Clinical Department of Oncology, The National Institute of Medicine of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | - Agnieszka Romanowicz
- Clinical Department of Oncology, The National Institute of Medicine of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | - Jarosław Drobnik
- Department of Family Medicine, Faculty of Medicine, Wroclaw Medical University, 51-141 Wroclaw, Poland
| | - Ewa Bandurska
- Center for Competence Development, Integrated Care and e-Health, Medical University of Gdansk, 80-204 Gdansk, Poland
| | - Weronika Ciećko
- Center for Competence Development, Integrated Care and e-Health, Medical University of Gdansk, 80-204 Gdansk, Poland
| | | | - Mateusz Curyło
- Department of Internal Medicine, Rehabilitation and Physical Medicine, Medical University of Lodz, 90-647 Lodz, Poland
- Medical Rehabilitation Department, The Ministry of the Interior and Administration Hospital, 30-053 Cracow, Poland
| | - Kacper Wróbel
- Department of Management and Logistics in Healthcare, Medical University of Lodz, 90-131 Lodz, Poland
| | - Remigiusz Kozłowski
- Center for Security Technologies in Logistics, Faculty of Management, University of Lodz, 90-237 Lodz, Poland
| | - Michał Marczak
- Collegium of Management, WSB Merito University in Warsaw, 03-204 Warszawa, Poland
| |
Collapse
|