1
|
Crecca E, Di Giuseppe G, Camplone C, Vigiano Benedetti V, Melaiu O, Mezza T, Cencioni C, Spallotta F. The multifaceted role of agents counteracting metabolic syndrome: A new hope for gastrointestinal cancer therapy. Pharmacol Ther 2025; 270:108847. [PMID: 40216262 DOI: 10.1016/j.pharmthera.2025.108847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/27/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
Metabolic syndrome (MetS) is defined by the presence of at least three of five clinical parameters including abdominal obesity, insulin resistance, elevated triglycerides, reduced high-density lipoprotein (HDL) and hypertension. Major features describing MetS have been recognized risk factors for cancer onset, with an alarming impact on gastrointestinal (GI) tumors. Intriguingly, therapeutic administration of drugs to improve glycemic control and dyslipidemia (including metformin, statins) has been shown to have a preventive role in the development and in prognosis improvement of several cancer types. Overall, these observations highlight the key role of altered metabolism prevalently in cancer risk development and unveil anti-MetS agent repurposing potential beyond their conventional pharmacological action. The objective of this review is to summarize the current knowledge about the antitumor activity of anti-diabetic and anti-lipemic agents in GI cancer onset and progression. Here, pre-clinical evidence of their therapeutic potential and of their integration in novel compelling therapeutic strategies will be discussed. Possible clinical outcomes of these novel therapeutic combined protocols specifically dedicated to GI cancer patients will be put under the spotlight. In the future, these novel therapeutic options should be considered to improve conventional chemotherapy response and prognosis of this group of patients.
Collapse
Affiliation(s)
- Elena Crecca
- Institute of System Analysis and Informatics "Antonio Ruberti", National Research Council (IASI-CNR), 00185 Rome, Italy
| | - Gianfranco Di Giuseppe
- Endocrinology and Diabetology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy; Department of Translational Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Claudia Camplone
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185 Rome, Italy; Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | | | - Ombretta Melaiu
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Teresa Mezza
- Department of Translational Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy; Pancreas Unit, CEMAD Digestive Diseases Center, Internal Medicine and Gastroenterology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Chiara Cencioni
- Institute of System Analysis and Informatics "Antonio Ruberti", National Research Council (IASI-CNR), 00185 Rome, Italy.
| | - Francesco Spallotta
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185 Rome, Italy; Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy.
| |
Collapse
|
2
|
Vesterberg A, Asplund E, Marras G, Vujasinovic M, Haddad Ringborg C, Wengström Y, Löhr M. Exploring the psychological burden in a pancreatic cancer surveillance programme based on high-risk individuals: a Swedish cross-sectional study. BMJ Open 2025; 15:e097814. [PMID: 40306989 PMCID: PMC12049877 DOI: 10.1136/bmjopen-2024-097814] [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: 12/10/2024] [Accepted: 04/08/2025] [Indexed: 05/02/2025] Open
Abstract
INTRODUCTION Pancreatic cancer, an aggressive cancer that presents with few or unspecific symptoms, has a poor prognosis. Thus, diagnosis at an early stage is vital for survival and a chance for curative treatment. Therefore, surveillance programmes for high-risk individuals are of the utmost importance. However, data on the psychological burden among participants in these programmes are limited. AIMS This study aimed to investigate the psychological burden for participants in a pancreatic cancer surveillance programme and explore whether the psychological burden was related to the individual's risk level for pancreatic cancer. METHODS This single-centre cross-sectional study investigated cancer worry, anxiety, coping and perceived physical and mental health using a digital questionnaire, including the following instruments: Cancer Worry Scale (CWS), State-Trait Anxiety Inventory (STAI), 13-Item Sense of Coherence and 12-Item Short-Form Survey. The invited participants (n=413) were healthy individuals with an increased risk of pancreatic cancer enrolled in a pancreatic cancer surveillance programme. RESULTS The results indicated high cancer worry among respondents (n=78) with high scores on CWS (mean, 16.45). The majority (69.3%) had scores indicating high cancer worry (≥14). Anxiety was not equally high among respondents (mean STAI-T, 35.13; STAI-S, 35.9). Female sex and younger age were significantly correlated with higher cancer worry and anxiety (p < 0.001). Outcomes in coping and perceived health were similar to those of the normal population. CONCLUSIONS Cancer worry is particularly high among participants. No correlation was found between the risk level and psychological burden.
Collapse
Affiliation(s)
- Anna Vesterberg
- Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Ebba Asplund
- Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
- Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Giulia Marras
- Division of Nursing, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Miroslav Vujasinovic
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
- Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Cecilia Haddad Ringborg
- Division of Nursing, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Yvonne Wengström
- Division of Nursing, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Matthias Löhr
- Division of Surgery and Oncology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
- Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
- Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| |
Collapse
|
3
|
Yang C, Ge F, Peng M, Cheng L, Wang K, Liu W. Exploring the genetic link between gastroesophageal reflux disease and pancreatic cancer: insights from Mendelian randomization. BMC Cancer 2025; 25:729. [PMID: 40251581 PMCID: PMC12007196 DOI: 10.1186/s12885-025-14128-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/09/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Gastroesophageal reflux disease (GERD) is increasingly recognized for its associations with extragastric diseases, yet its potential role in pancreatic cancer (PC) etiology remains underexplored. This study investigates the genetic causal relationship between GERD and PC using Mendelian randomization (MR), a method designed to reduce confounding factors. METHODS A two-sample MR analysis was conducted using genome-wide association studies (GWAS) data. The inverse variance weighted (IVW) method was applied, with additional sensitivity analyses performed to evaluate pleiotropy and heterogeneity. RESULTS The IVW analysis demonstrated a significant genetic association between the genetic signature predisposing to GERD and an increased risk of PC (OR: 1.36, 95% CI: 1.04-1.80, P = 0.03). There was no evidence of pleiotropy (P = 0.71) or heterogeneity (P = 0.94). CONCLUSIONS Our study provides robust genetic evidence supporting that the genetic predisposition to GERD is associated with an increased risk of PC. These findings emphasize the necessity of integrating GERD into PC risk assessments and encourage further research to elucidate the underlying biological mechanisms. This insight holds potential to inform strategies for early detection, prevention, and personalized management of PC in GERD patients.
Collapse
Affiliation(s)
- Chen Yang
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, China
| | - Fan Ge
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mengye Peng
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, China
| | - Liang Cheng
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, China
| | - Kezheng Wang
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Wei Liu
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
4
|
Shen L, Schaefer A, Huckaby J, Wolf W, Lai SK. Bispecific Siglec-15/T cell antibody (STAB) activates T cells and suppresses pancreatic ductal adenocarcinoma and non-small cell lung tumors in vivo. Theranostics 2025; 15:5529-5542. [PMID: 40365291 PMCID: PMC12068307 DOI: 10.7150/thno.103372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 03/09/2025] [Indexed: 05/15/2025] Open
Abstract
Rationale: Siglec-15 (S15) is a membrane-associated antigen overexpressed across various cancer types, and also induces immunosuppression. We believe this makes S15 a promising target for cellular immunotherapy of solid tumors characterized by an immunosuppressive tumor microenvironment, but this remains underexplored to date. Method: We engineered a bispecific antibody that simultaneously binds S15 on tumor cells and CD3 on T cells in the popular IgG-scFv format; we termed this molecule STAB. Results: In vitro, STAB induced marked proliferation of CD3+ T cells in human PBMCs, and mediated effective killing of Panc-1 pancreatic ductal adenocarcinoma (PDAC) and H460 non-small cell lung cancer (NSCLC) cells in co-culture studies with PBMCs or CD3+ T cells. In NSG mice with human PDAC and NSCLC tumors, STAB effectively suppressed tumor growth and prolonged survival, in sharp contrast to mice receiving either anti-S15 or anti-CD3 mAbs alone. STAB increased activated T cells in both tumor and circulation, as well as reduced the stromal barrier-a key hallmark of PDAC. Conclusion: Our results underscore STAb as a promising therapeutic molecule to be investigated further for PDAC and NSCLC, and potentially other S15-positive solid tumors.
Collapse
MESH Headings
- Animals
- Humans
- Antibodies, Bispecific/pharmacology
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/administration & dosage
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/therapy
- Carcinoma, Pancreatic Ductal/drug therapy
- T-Lymphocytes/immunology
- T-Lymphocytes/drug effects
- Mice
- Cell Line, Tumor
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/therapy
- Lung Neoplasms/drug therapy
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/therapy
- Lectins/immunology
- Lymphocyte Activation/drug effects
- Tumor Microenvironment
- CD3 Complex/immunology
- Xenograft Model Antitumor Assays
- Female
- Membrane Proteins/immunology
- Mice, Inbred NOD
- Immunotherapy/methods
- Immunoglobulins
Collapse
Affiliation(s)
- Limei Shen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA
| | - Alison Schaefer
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, NC, USA
| | - Justin Huckaby
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, NC, USA
| | - Whitney Wolf
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA
| | - Samuel K. Lai
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC, USA
- Department of Immunology and Microbiology, University of North Carolina at Chapel Hill, NC, USA
| |
Collapse
|
5
|
Advani A, Deewan K, Odhwani A, Kumar K. Comment on: "Analysis of exposome and genetic variability suggests stress as a major contributor for development of pancreatic ductal adenocarcinoma". Dig Liver Dis 2025; 57:804-805. [PMID: 39828443 DOI: 10.1016/j.dld.2024.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 12/31/2024] [Indexed: 01/22/2025]
Affiliation(s)
- Aman Advani
- United Medical and Dental College, Karachi, Karachi, Pakistan.
| | - Kuldeep Deewan
- United Medical and Dental College, Karachi, Karachi, Pakistan.
| | - Aadtiya Odhwani
- United Medical and Dental College, Karachi, Karachi, Pakistan.
| | - Kunal Kumar
- United Medical and Dental College, Karachi, Karachi, Pakistan.
| |
Collapse
|
6
|
Lasota M, Jankowski D, Wiśniewska A, Szeleszczuk Ł, Misterka-Kozaka A, Kaczor-Kamińska M, Zarzycka M, Patena M, Brzozowski T. Interaction of Avapritinib with Congo Red in Pancreatic Cancer Cells: Molecular Modeling and Biophysical Studies. Int J Mol Sci 2025; 26:1980. [PMID: 40076604 PMCID: PMC11901030 DOI: 10.3390/ijms26051980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Pancreatic cancer is a malignant tumor with one of the worst prognoses among solid tumors, characterized by resistance to treatment. Therefore, there is an urgent need for new methods of targeted therapy. Previous studies have shown that the overexpression of receptor tyrosine kinases such as c-KIT or PDGFR can increase proliferation, migration, and invasion of cancer cells. The aim of our study was to analyze aggregates between a supramolecular carrier (Congo red, CR) and a tyrosine kinase inhibitor (BLU-258) as well as to investigate the effect of the free inhibitor and its aggregate with Congo red (CR-BLU-258) on selected properties of pancreatic cells, including these cells' viability and three-dimensional cell spheroid cultures. To better understand the interactions between Congo red and BLU-258, we used molecular modeling in addition to biophysical methods. These attempts allowed us to determine the optimal molar ratio, which we used for in vitro studies on pancreatic cancer cell lines. A significantly greater decrease in the viability of the tested 3D cultures was observed after 48 h of incubation with CR-BLU-258, which resulted in a lower IC50 value for the tested co-aggregate compared with BLU-258 alone. Moreover, a higher resistance of PANC-1 and BxPC3 spheroid cells to the tested compounds was noted compared with the 2D culture model. A significantly lower response was observed in 3D cell cultures (BxPC3 and PANC-1) treated with BLU-258 alone compared with the 2D culture. Thus, our results showed that both BLU-258 (alone) and in its co-aggregate with Congo red exhibit anticancer activity, inhibiting the growth of pancreatic cancer cells and reducing their viability, survival, and migration. Both tested compounds also affected the phosphorylation of the selected signaling proteins. We conclude that the selected tyrosine kinase inhibitor (alone) and in its co-aggregate with Congo red exhibit anticancer activity and should be considered as a novel effective therapy against pancreatic cancer.
Collapse
Affiliation(s)
- Małgorzata Lasota
- Center for Biomedicine and Interdisciplinary Sciences, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Daniel Jankowski
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Anna Wiśniewska
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
| | - Łukasz Szeleszczuk
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097 Warsaw, Poland;
| | - Anna Misterka-Kozaka
- Center for Biomedicine and Interdisciplinary Sciences, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Marta Kaczor-Kamińska
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, 31-034 Krakow, Poland; (M.K.-K.); (M.Z.)
| | - Marta Zarzycka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, 31-034 Krakow, Poland; (M.K.-K.); (M.Z.)
| | - Maksym Patena
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Tomasz Brzozowski
- Center for Biomedicine and Interdisciplinary Sciences, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland
| |
Collapse
|
7
|
Cheng Z, Luo X, Liu W, Lu X, Chang H, Wang Y, Zheng W, Yan X, Huang Y. Comprehensive landscape and oncogenic role of extrachromosomal circular DNA in malignant biliary strictures. Cell Biosci 2025; 15:16. [PMID: 39920810 PMCID: PMC11804034 DOI: 10.1186/s13578-025-01361-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Extrachromosomal circular DNA (eccDNA) is crucial for carcinogenesis and bile has direct contact with malignant biliary strictures, yet eccDNA features in bile and its function in malignant biliary strictures remain underexplored. RESULTS We observed the widespread presence of eccDNA in bile and systematically profiled the landscape of bile cell-free eccDNA (bcf-eccDNA). For functional exploration, a simple and efficient workflow was designed to synthesize large eccDNA particularly containing multiple regions. Compared with the noncancer group, bcf-eccDNAs in the cancer group had different origins and larger sizes with six characteristic peaks. These peaks were also identified in the validation cohort (100%). There were more bcf-eccDNA carrying LINC00598 or CELF2 in malignant biliary strictures, showing potential diagnostic performance in training and validation cohorts (all AUCs > 0.9). Bcf-eccDNAs carried cancer-related mutations, which could guide treatment. EccDNA carrying miR-106a/363 cluster or miR-374b/421 cluster were proven to regulate cancer gene expression, accelerate tumor proliferation, and inhibit tumor apoptosis. CONCLUSIONS This study profiles a comprehensive bcf-eccDNA landscape in patients with biliary strictures and offers valuable insights into eccDNA's role in bile liquid biopsy and carcinogenesis.
Collapse
Affiliation(s)
- Zhuo Cheng
- Department of Gastroenterology and Hepatology, Peking University Third Hospital, Beijing, 100191, China
| | - Xuanmei Luo
- Peking University Fifth School of Clinical Medicine, Beijing, China
| | - Wenzheng Liu
- Department of Gastroenterology and Hepatology, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaofang Lu
- Department of Gastroenterology, Beijing Tsinghua Changgung Hospital, Beijing, 102218, China
| | - Hong Chang
- Department of Gastroenterology and Hepatology, Peking University Third Hospital, Beijing, 100191, China
| | - Yingchun Wang
- Department of Gastroenterology and Hepatology, Peking University Third Hospital, Beijing, 100191, China
| | - Wei Zheng
- Department of Gastroenterology and Hepatology, Peking University Third Hospital, Beijing, 100191, China
| | - Xiue Yan
- Department of Gastroenterology, Beijing Tsinghua Changgung Hospital, Beijing, 102218, China.
| | - Yonghui Huang
- Department of Gastroenterology and Hepatology, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
8
|
Pérez-Navarro Y, Salinas-Vera YM, López-Camarillo C, Figueroa-Angulo EE, Alvarez-Sánchez ME. The role of long non-coding RNA NORAD in digestive system tumors. Noncoding RNA Res 2025; 10:55-62. [PMID: 39296642 PMCID: PMC11406672 DOI: 10.1016/j.ncrna.2024.09.002] [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: 07/18/2024] [Revised: 08/28/2024] [Accepted: 09/01/2024] [Indexed: 09/21/2024] Open
Abstract
In recent years, it has been discovered that the expression of long non-coding RNAs is highly deregulated in several types of cancer and contributes to its progression and development. Recently, it has been described that in tumors of the digestive system, such as colorectal cancer, pancreatic cancer, and gastric cancer, DNA damage-activated lncRNA (NORAD) was frequently up-regulated. The purpose of this review is to elucidate the functions of NORAD in tumors of the digestive system, emphasizing its involvement in important cellular processes such as invasion, metastasis, proliferation, and apoptosis. NORAD acts as a ceRNA (competitive endogenous RNA) that sponges microRNAs and regulates the expression of target genes involved in tumorigenesis. Thus, the mechanisms underlying the effects of NORAD are complex and involve multiple signaling pathways. This review consolidates current knowledge on the role of NORAD in digestive cancers and highlights the need for further research to explore its potential as a therapeutic target. Understanding the intricate functions of NORAD could elucidate the way for innovative approaches to cancer treatment.
Collapse
Affiliation(s)
- Yussel Pérez-Navarro
- Posgrado en Ciencias Genómicas, Laboratorio de Patogénesis Celular y Molecular Humana y Veterinaria, Universidad Autónoma de la Ciudad de México, Ciudad de México, CDMX, Mexico
| | - Yarely M. Salinas-Vera
- Centro Nacional de Identificación Humana, Comisión Nacional de Búsqueda, Secretaría de Gobernación, Camino a Santa Teresa No 1679, Jardines del Pedregal, Ciudad de México, Mexico
| | - Cesar López-Camarillo
- Posgrado en Ciencias Genómicas, Laboratorio de Oncogenómica y Proteómica del cáncer, Universidad Autónoma de la Ciudad de México, Ciudad de México, Mexico
| | - Elisa Elvira Figueroa-Angulo
- Licenciatura en Ciencias Genómicas, Laboratorio de Patogénesis Celular y Molecular Humana y Veterinaria, Universidad Autónoma de la Ciudad de México, Mexico
| | - María Elizbeth Alvarez-Sánchez
- Posgrado en Ciencias Genómicas, Laboratorio de Patogénesis Celular y Molecular Humana y Veterinaria, Universidad Autónoma de la Ciudad de México, Ciudad de México, CDMX, Mexico
| |
Collapse
|
9
|
Kar S, Mehrotra S, Prajapati VK. From infection to remedy: Harnessing oncolytic viruses in cancer treatment. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:213-257. [PMID: 39978967 DOI: 10.1016/bs.apcsb.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Oncolytic virus (OV) mediated immunotherapy is one of the recent techniques used to treat higher grade cancers where conventional therapies like chemotherapy, radiation fail. OVs as a therapeutic tool show high efficacy and fewer side effects than conventional methods as supported by multiple preclinical and clinical studies since they are engineered to target tumours. In this chapter, we discuss the modifications in viruses to make them oncolytic, types of strains commonly administered, mechanisms employed by viruses to specifically target and eradicate malignancy and progress achieved as reported in case studies (preclinical and clinical trials). OVs also face some unique challenges with respect to the malignancy being treated and the varied pathogen exposure of the patients, which is also highlighted here. Since pathogen exposure varies according to population dynamics worldwide, chances of generating a non-specific recall response to an OV cannot be negated. Lastly, the future perspectives and ongoing practises of combination therapies are discussed as they provide a leading edge over monotherapies in terms of tumour clearance, blocking metastasis and enhancing patient survival. Efforts undertaken to overcome current challenges are also highlighted.
Collapse
Affiliation(s)
- Sramona Kar
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
10
|
Jain A, Keesari PR, Pulakurthi YS, Katamreddy R, Dhar M, Desai R. Pancreatic Cancer Risk in Prediabetes: A Systematic Meta-analysis Approach. Pancreas 2025; 54:e51-e56. [PMID: 39324961 DOI: 10.1097/mpa.0000000000002391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
OBJECTIVES Pancreatic cancer and prediabetes pose significant public health challenges. Given the lack of strong evidence we performed a meta-analysis to assess the risk of pancreatic cancer in prediabetes. MATERIALS AND METHODS We performed a thorough search of the major databases over the last 10 years to identify relevant articles. The pooled odds ratio (OR) and hazard ratio (HR) were combined to calculate the effect size (ES). RESULTS We analyzed 5 studies including 5,425,111 prediabetic individuals and 16,096,467 normoglycemic population across 5 countries with a median follow-up of 8.5 years. We identified a noteworthy association between prediabetes and pancreatic cancer, reporting an unadjusted ES of 1.36 (95% confidence interval [CI] 1.05-1.77, P = 0.02) and an adjusted ES of 1.40 (1.23-1.59, P < 0.01). Subgroup analyses by age revealed variations in risk, with studies involving participants aged 60 and above exhibiting a higher ES (ES 1.83, 95% CI 1.28-2.62, P < 0.01). Geographical differences were also observed, with Japanese studies reporting a higher risk (ES 1.89, 95% CI 1.15-3.10, P < 0.01) compared with those from the United States (ES 1.32, 95% CI 1.13-1.53, P < 0.01). CONCLUSIONS We identified 40% higher risk of pancreatic cancer in patients with prediabetes than those with normal blood glucose necessitating urgent attention for further research and policy change.
Collapse
Affiliation(s)
- Akhil Jain
- From the University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | - Meekoo Dhar
- Staten Island University Hospital, Staten Island, NY
| | | |
Collapse
|
11
|
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
|
12
|
Goluba K, Parfejevs V, Rostoka E, Jekabsons K, Blake I, Neimane A, Ule AA, Rimsa R, Vangravs R, Pcolkins A, Riekstina U. Personalized PDAC chip with functional endothelial barrier for tumour biomarker detection: A platform for precision medicine applications. Mater Today Bio 2024; 29:101262. [PMID: 39381267 PMCID: PMC11460472 DOI: 10.1016/j.mtbio.2024.101262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/07/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer characterised by poor survival rates and an increasing global incidence. Advances in the staging and categorization of pancreatic tumours, along with the discovery of functional mutations, have made precision treatments possible, which may lead to better clinical results. To further improve customized treatment approaches, in vitro models that can be used for functional drug sensitivity testing and precisely mimic the disease at the organ level are required. In this study, we present a workflow for creating a personalized PDAC chip utilising primary tumour-derived human pancreatic organoids (hPOs) and Human Umbilical Vein Endothelial Cells (HUVECs) to simulate the vascular barrier and tumour interactions within a PDMS-free organ-on-a-chip system. The patient PDAC tissue, expanded as tumour hPOs, could be cultured as adherent cells on the chip for more than 50 days, allowing continuous monitoring of cell viability through outflows from tumour and endothelial channels. Our findings demonstrate a gradual increase in cell density and cell turnover in the pancreatic tumor channel. Tumour-specific biomarkers, including CA-19.9, TIMP-1, Osteopontin, MIC-1, ICAM-1 and sAXL were consistently detected in the PDAC chip outflows. Comparative analyses between tissue culture plates and microfluidic conditions revealed significant differences in biomarker secretion patterns, highlighting the advantages of the microfluidics approach. This PDAC chip provides a stable, reproducible tumour model system with a functional endothelial cell barrier, suitable for drug sensitivity and secretory biomarker studies, thus serving as a platform for functional precision medicine application and multi-organ chip development.
Collapse
Affiliation(s)
- Karina Goluba
- Pharmaceutical Sciences Center, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Riga, Latvia
| | - Vadims Parfejevs
- Pharmaceutical Sciences Center, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Riga, Latvia
| | - Evita Rostoka
- Pharmaceutical Sciences Center, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Riga, Latvia
| | - Kaspars Jekabsons
- Pharmaceutical Sciences Center, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Riga, Latvia
| | - Ilze Blake
- Pharmaceutical Sciences Center, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Riga, Latvia
| | - Anastasija Neimane
- Pharmaceutical Sciences Center, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Riga, Latvia
| | - Annija Anete Ule
- Institute of Solid State Physics, University of Latvia, Kengaraga iela 8, Riga, Latvia
| | - Roberts Rimsa
- Institute of Solid State Physics, University of Latvia, Kengaraga iela 8, Riga, Latvia
| | - Reinis Vangravs
- Latvian Centre of Infectious Diseases, Laboratory Service, Riga East University Hospital, Linezera iela 3, LV-1006, Riga, Latvia
| | - Andrejs Pcolkins
- Department of Abdominal and Soft Tissue Surgery, Riga East Clinical University Hospital, Hipokrata iela 2, Riga, Latvia
| | - Una Riekstina
- Pharmaceutical Sciences Center, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Riga, Latvia
| |
Collapse
|
13
|
Graham S, Dmitrieva M, Vendramini-Costa DB, Francescone R, Trujillo MA, Cukierman E, Wood LD. From precursor to cancer: decoding the intrinsic and extrinsic pathways of pancreatic intraepithelial neoplasia progression. Carcinogenesis 2024; 45:801-816. [PMID: 39514554 DOI: 10.1093/carcin/bgae064] [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: 08/09/2024] [Revised: 09/04/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
This review explores the progression of pancreatic intraepithelial neoplasia (PanIN) to pancreatic ductal adenocarcinoma through a dual lens of intrinsic molecular alterations and extrinsic microenvironmental influences. PanIN development begins with Kirsten rat sarcoma viral oncogene (KRAS) mutations driving PanIN initiation. Key additional mutations in cyclin-dependent kinase inhibitor 2A (CDKN2A), tumor protein p53 (TP53), and mothers against decapentaplegic homolog 4 (SMAD4) disrupt cell cycle control and genomic stability, crucial for PanIN progression from low-grade to high-grade dysplasia. Additional molecular alterations in neoplastic cells, including epigenetic modifications and chromosomal alterations, can further contribute to neoplastic progression. In parallel with these alterations in neoplastic cells, the microenvironment, including fibroblast activation, extracellular matrix remodeling, and immune modulation, plays a pivotal role in PanIN initiation and progression. Crosstalk between neoplastic and stromal cells influences nutrient support and immune evasion, contributing to tumor development, growth, and survival. This review underscores the intricate interplay between cell-intrinsic molecular drivers and cell-extrinsic microenvironmental factors, shaping PanIN predisposition, initiation, and progression. Future research aims to unravel these interactions to develop targeted therapeutic strategies and early detection techniques, aiming to alleviate the severe impact of pancreatic cancer by addressing both genetic predispositions and environmental influences.
Collapse
Affiliation(s)
- Sarah Graham
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - Mariia Dmitrieva
- Cancer Signaling & Microenvironment Program, M&C Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz School of Medicine, Temple Health, Philadelphia, PA 19111, United States
| | - Debora Barbosa Vendramini-Costa
- Henry Ford Pancreatic Cancer Center, Henry Ford Health, Henry Ford Health + Michigan State University Health Sciences, Detroit, MI 48202, United States
| | - Ralph Francescone
- Henry Ford Pancreatic Cancer Center, Henry Ford Health, Henry Ford Health + Michigan State University Health Sciences, Detroit, MI 48202, United States
| | - Maria A Trujillo
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - Edna Cukierman
- Cancer Signaling & Microenvironment Program, M&C Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz School of Medicine, Temple Health, Philadelphia, PA 19111, United States
| | - Laura D Wood
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD 21231, United States
| |
Collapse
|
14
|
Pergolizzi RG, Brower ST. Molecular Targets for the Diagnosis and Treatment of Pancreatic Cancer. Int J Mol Sci 2024; 25:10843. [PMID: 39409171 PMCID: PMC11476914 DOI: 10.3390/ijms251910843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Pancreatic cancer is one of the most aggressive and lethal forms of cancer, with a five-year survival rate of less than 10%. Despite advances in treatment modalities, the prognosis for pancreatic cancer patients remains poor, highlighting the urgent need for innovative approaches for early diagnosis and targeted therapies. In recent years, there has been significant progress in understanding the molecular mechanisms underlying pancreatic cancer development and progression. This paper reviews the current knowledge of molecular targets for the diagnosis and treatment of pancreatic cancer.
Collapse
Affiliation(s)
| | - Steven T. Brower
- Department of Surgical Oncology and HPB Surgery, Englewood Health, Englewood, NJ 07631, USA
| |
Collapse
|
15
|
Balcer K, Garnier J, Richa Y, Bruneel-Zupanc C, Piessen G, Turrini O, Truant S, El Amrani M. Impact on survival of sarcopenia, systemic inflammatory response and anthropometric factors after pancreatectomy for resectable pancreatic adenocarcinoma. World J Surg Oncol 2024; 22:232. [PMID: 39232731 PMCID: PMC11376042 DOI: 10.1186/s12957-024-03510-6] [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: 04/26/2024] [Accepted: 08/25/2024] [Indexed: 09/06/2024] Open
Abstract
INTRODUCTION Pancreatic adenocarcinoma (PDAC) is becoming a public health issue with a 5-years survival rate around 10%. Patients with PDAC are often sarcopenic, which impacts postoperative outcome. At the same time, overweight population is increasing and adipose tissue promotes tumor related-inflammation. With several studies supporting independently these data, we aimed to assess if they held an impact on survival when combined. METHODS We included 232 patients from two university hospitals (CHU de Lille, Institut Paoli Calmette), from January 2011 to December 2018, who underwent Pancreaticoduodenectomy (PD) for resectable PDAC. Preoperative CT scan was used to measure sarcopenia and visceral fat according to international cut-offs. Neutrophil to lymphocyte (NLR) and platelet to lymphocyte ratios (PLR) were used to measure inflammation. For univariate and multivariate analyses, the Cox proportional-hazard model was used. P-values below 0.05 were considered significant. RESULTS Sarcopenic patients with visceral obesity were less likely to survive than the others in multivariate analysis (OS, HR 1.65, p= 0.043). Cutaneous obesity did not influence survival. We also observed an influence on survival when we studied sarcopenia with visceral obesity (OS, p= 0.056; PFS, p = 0.014), sarcopenia with cutaneous obesity (PFS, p= 0.005) and sarcopenia with PLR (PFS, p= 0.043). This poor prognosis was also found in sarcopenic obese patients with high PLR (OS, p= 0.05; PFS, p= 0.01). CONCLUSION Sarcopenic obesity was associated with poor prognosis after PD for PDAC, especially in patients with systemic inflammation. Pre operative management of these factors should be addressed in pancreatic cancer patients.
Collapse
Affiliation(s)
- Kaja Balcer
- Digestive Surgery and Transplantation Department, CHU de Lille, University of Lille, Lille, 59000, France.
| | - Jonathan Garnier
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, 13009, France
| | - Yasmina Richa
- School of Medicine, University College Cork, Cork, Ireland
| | | | - Guillaume Piessen
- Department of Digestive and Oncological Surgery, University of Lille, CNRS, INSERM, CHU Lille, University of Lille, Lille, 59000, France
| | - Olivier Turrini
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, 13009, France
| | - Stephanie Truant
- Digestive Surgery and Transplantation Department, CHU de Lille, University of Lille, Lille, 59000, France
| | - Mehdi El Amrani
- Digestive Surgery and Transplantation Department, CHU de Lille, University of Lille, Lille, 59000, France
| |
Collapse
|
16
|
Kane WJ, Haden KR, Martin EN, Shami VM, Wang AY, Strand DS, Adair SJ, Nagdas S, Tsung A, Zaydfudim VM, Adams RB, Bauer TW. Survival benefit associated with screening of patients at elevated risk for pancreatic cancer. J Surg Oncol 2024; 130:485-492. [PMID: 39016067 DOI: 10.1002/jso.27784] [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: 06/21/2024] [Accepted: 06/30/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND & OBJECTIVES: Screening for pancreatic cancer is recommended for individuals with a strong family history, certain genetic syndromes, or a neoplastic cyst of the pancreas. However, limited data supports a survival benefit attributable to screening these higher-risk individuals. METHODS All patients enrolled in screening at a High-Risk Pancreatic Cancer Clinic (HRC) from July 2013 to June 2020 were identified from a prospectively maintained institutional database and compared to patients evaluated at a Surgical Oncology Clinic (SOC) at the same institution during the same period. Clinical outcomes of patients selected for surgical resection, particularly clinicopathologic stage and overall survival, were compared. RESULTS Among 826 HRC patients followed for a median (IQR) of 2.3 (0.8-4.2) years, 128 were selected for surgical resection and compared to 402 SOC patients selected for resection. Overall survival was significantly longer among HRC patients (median survival: not reached vs. 2.6 years, p < 0.001). Among 31 HRC and 217 SOC patients with a diagnosis of pancreatic ductal adenocarcinoma (PDAC), the majority of HRC patients were diagnosed with stage 0 disease (carcinoma in situ), while the majority of SOC patients were diagnosed with stage II disease (p < 0.001). Overall survival after resection of invasive PDAC was also significantly longer among HRC patients compared to SOC patients (median survival 5.5 vs. 1.6 years, p = 0.002). CONCLUSION Patients at increased risk for PDAC and followed with guideline-based screening exhibited downstaging of disease and improved survival from PDAC in comparison to patients who were not screened.
Collapse
Affiliation(s)
- William J Kane
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Kathleen R Haden
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Elizabeth N Martin
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Vanessa M Shami
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia, USA
| | - Andrew Y Wang
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel S Strand
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia, USA
| | - Sara J Adair
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Sarbajeet Nagdas
- School of Medicine, University of Virginia, Charlottesville, University of Virginia, USA
| | - Allan Tsung
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Victor M Zaydfudim
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Reid B Adams
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Todd W Bauer
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
17
|
Khan I, Kamal A, Akhtar S. Diabetes Driven Oncogenesis and Anticancer Potential of Repurposed Antidiabetic Drug: A Systemic Review. Cell Biochem Biophys 2024; 82:1907-1929. [PMID: 38954353 DOI: 10.1007/s12013-024-01387-6] [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] [Accepted: 06/22/2024] [Indexed: 07/04/2024]
Abstract
Diabetes and cancer are two prevalent disorders, pose significant public health challenges and contribute substantially to global mortality rates, with solely 10 million reported cancer-related deaths in 2020. This review explores the pathological association between diabetes and diverse cancer progressions, examining molecular mechanisms and potential therapeutic intersections. From altered metabolic landscapes to dysregulated signaling pathways, the intricate links are delineated, offering a comprehensive understanding of diabetes as a modulator of tumorigenesis. Cancer cells develop drug resistance through mechanisms like enhanced drug efflux, genetic mutations, and altered drug metabolism, allowing them to survive despite chemotherapeutic agent. Glucose emerges as a pivotal player in diabetes progression, and serving as a crucial energy source for cancer cells, supporting their biosynthetic needs and adaptation to diverse microenvironments. Glycation, a non-enzymatic process that produces advanced glycation end products (AGEs), has been linked to the etiology of cancer and has been shown in a number of tumor forms, such as leiomyosarcomas, adenocarcinomas, and squamous cell carcinomas. Furthermore, in aggressive and metastatic breast cancer, the receptor for AGEs (RAGE) is increased, which may increase the malignancy of the tumor. Reprogramming glucose metabolism manifests as hallmark cancer features, including accelerated cell proliferation, angiogenesis, metastasis, and evasion of apoptosis. This manuscript encapsulates the dual narrative of diabetes as a driver of cancer progression and the potential of repurposed antidiabetic drugs as formidable countermeasures. The amalgamation of mechanistic understanding and clinical trial outcomes establishes a robust foundation for further translational research and therapeutic advancements in the dynamic intersection of diabetes and cancer.
Collapse
Affiliation(s)
- Iqra Khan
- Department of Bioengineering, Integral University, Lucknow, 226026, Uttar Pradesh, India
| | - Aisha Kamal
- Department of Bioengineering, Integral University, Lucknow, 226026, Uttar Pradesh, India.
| | - Salman Akhtar
- Department of Bioengineering, Integral University, Lucknow, 226026, Uttar Pradesh, India
| |
Collapse
|
18
|
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: 0] [Impact Index Per Article: 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
|
19
|
Rodriguez-Tirado C, Sosa MS. How much do we know about the metastatic process? Clin Exp Metastasis 2024; 41:275-299. [PMID: 38520475 PMCID: PMC11374507 DOI: 10.1007/s10585-023-10248-0] [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: 08/13/2023] [Accepted: 11/17/2023] [Indexed: 03/25/2024]
Abstract
Cancer cells can leave their primary sites and travel through the circulation to distant sites, where they lodge as disseminated cancer cells (DCCs), even during the early and asymptomatic stages of tumor progression. In experimental models and clinical samples, DCCs can be detected in a non-proliferative state, defined as cellular dormancy. This state can persist for extended periods until DCCs reawaken, usually in response to niche-derived reactivation signals. Therefore, their clinical detection in sites like lymph nodes and bone marrow is linked to poor survival. Current cancer therapy designs are based on the biology of the primary tumor and do not target the biology of the dormant DCC population and thus fail to eradicate the initial or subsequent waves of metastasis. In this brief review, we discuss the current methods for detecting DCCs and highlight new strategies that aim to target DCCs that constitute minimal residual disease to reduce or prevent metastasis formation. Furthermore, we present current evidence on the relevance of DCCs derived from early stages of tumor progression in metastatic disease and describe the animal models available for their study. We also discuss our current understanding of the dissemination mechanisms utilized by genetically less- and more-advanced cancer cells, which include the functional analysis of intermediate or hybrid states of epithelial-mesenchymal transition (EMT). Finally, we raise some intriguing questions regarding the clinical impact of studying the crosstalk between evolutionary waves of DCCs and the initiation of metastatic disease.
Collapse
Affiliation(s)
- Carolina Rodriguez-Tirado
- Department of Microbiology and Immunology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
- Department of Oncology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
- Cancer Dormancy and Tumor Microenvironment Institute/Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
| | - Maria Soledad Sosa
- Department of Microbiology and Immunology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
- Department of Oncology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
- Cancer Dormancy and Tumor Microenvironment Institute/Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, 10461, USA.
| |
Collapse
|
20
|
Chattopadhyay S, Hazra R, Mallick A, Gayen S, Roy S. A review exploring the fusion of oncolytic viruses and cancer immunotherapy: An innovative strategy in the realm of cancer treatment. Biochim Biophys Acta Rev Cancer 2024; 1879:189110. [PMID: 38754793 DOI: 10.1016/j.bbcan.2024.189110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
Oncolytic viruses (OVs) are increasingly recognized as potent tools in cancer therapy, effectively targeting and eradicating oncogenic conditions while sparing healthy cells. They enhance antitumor immunity by triggering various immune responses throughout the cancer cycle. Genetically engineered OVs swiftly destroy cancerous tissues and activate the immune system by releasing soluble antigens like danger signals and interferons. Their ability to stimulate both innate and adaptive immunity makes them particularly attractive in cancer immunotherapy. Recent advancements involve combining OVs with other immune therapies, yielding promising results. Transgenic OVs, designed to enhance immunostimulation and specifically target cancer cells, further improve immune responses. This review highlights the intrinsic mechanisms of OVs and underscores their synergistic potential with other immunotherapies. It also proposes strategies for optimizing armed OVs to bolster immunity against tumors.
Collapse
Affiliation(s)
- Soumyadeep Chattopadhyay
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India
| | - Rudradeep Hazra
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India
| | - Arijit Mallick
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India
| | - Sakuntala Gayen
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India.
| |
Collapse
|
21
|
Chen C, Demirkhanyan L, Gondi CS. The Multifaceted Role of miR-21 in Pancreatic Cancers. Cells 2024; 13:948. [PMID: 38891080 PMCID: PMC11172074 DOI: 10.3390/cells13110948] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
With the lack of specific signs and symptoms, pancreatic ductal adenocarcinoma (PDAC) is often diagnosed at late metastatic stages, resulting in poor survival outcomes. Among various biomarkers, microRNA-21 (miR-21), a small non-coding RNA, is highly expressed in PDAC. By inhibiting regulatory proteins at the 3' untranslated regions (UTR), miR-21 holds significant roles in PDAC cell proliferation, epithelial-mesenchymal transition, angiogenesis, as well as cancer invasion, metastasis, and resistance therapy. We conducted a systematic search across major databases for articles on miR-21 and pancreatic cancer mainly published within the last decade, focusing on their diagnostic, prognostic, therapeutic, and biological roles. This rigorous approach ensured a comprehensive review of miR-21's multifaceted role in pancreatic cancers. In this review, we explore the current understandings and future directions regarding the regulation, diagnostic, prognostic, and therapeutic potential of targeting miR-21 in PDAC. This exhaustive review discusses the involvement of miR-21 in proliferation, epithelial-mesenchymal transition (EMT), apoptosis modulation, angiogenesis, and its role in therapy resistance. Also discussed in the review is the interplay between various molecular pathways that contribute to tumor progression, with specific reference to pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Clare Chen
- Department of Internal Medicine, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Lusine Demirkhanyan
- Department of Internal Medicine, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Departments of Internal Medicine and Surgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Christopher S. Gondi
- Department of Internal Medicine, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Departments of Internal Medicine and Surgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Departments of Internal Medicine, Surgery, and Health Science Education and Pathology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Health Care Engineering Systems Center, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
22
|
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
|
23
|
Saleh O, Shihadeh H, Yousef A, Erekat H, Abdallh F, Al-Leimon A, Elsalhy R, Altiti A, Dajani M, AlBarakat MM. The Effect of Intratumor Heterogeneity in Pancreatic Ductal Adenocarcinoma Progression and Treatment. Pancreas 2024; 53:e450-e465. [PMID: 38728212 DOI: 10.1097/mpa.0000000000002342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
BACKGROUND AND OBJECTIVES Pancreatic cancer is one of the most lethal malignancies. Even though many substantial improvements in the survival rates for other major cancer forms were made, pancreatic cancer survival rates have remained relatively unchanged since the 1960s. Even more, no standard classification system for pancreatic cancer is based on cellular biomarkers. This review will discuss and provide updates about the role of stem cells in the progression of PC, the genetic changes associated with it, and the promising biomarkers for diagnosis. MATERIALS AND METHODS The search process used PubMed, Cochrane Library, and Scopus databases to identify the relevant and related articles. Articles had to be published in English to be considered. RESULTS The increasing number of studies in recent years has revealed that the diversity of cancer-associated fibroblasts is far greater than previously acknowledged, which highlights the need for further research to better understand the various cancer-associated fibroblast subpopulations. Despite the huge diversity in pancreatic cancer, some common features can be noted to be shared among patients. Mutations involving CDKN2, P53, and K-RAS can be seen in a big number of patients, for example. Similarly, some patterns of genes and biomarkers expression and the level of their expression can help in predicting cancer behavior such as metastasis and drug resistance. The current trend in cancer research, especially with the advancement in technology, is to sequence everything in hopes of finding disease-related mutations. CONCLUSION Optimizing pancreatic cancer treatment requires clear classification, understanding CAF roles, and exploring stroma reshaping approaches.
Collapse
Affiliation(s)
- Othman Saleh
- From the Faculty of Medicine, The Hashemite University, Zarqa
| | | | | | - Hana Erekat
- School of medicine, University of Jordan, Amman
| | - Fatima Abdallh
- From the Faculty of Medicine, The Hashemite University, Zarqa
| | | | | | | | - Majd Dajani
- From the Faculty of Medicine, The Hashemite University, Zarqa
| | - Majd M AlBarakat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
24
|
Jiang Z, Ni J, Zhou S, Yang L, Huang X, Bao J, Liu J. NiWo4- RGO composite exerts cytotoxic effects on pancreatic carcinoma cells via a cross-talk between reactive oxygen species-independent canonical autophagy of the mitochondria and epithelial-mesenchymal transition. J Drug Deliv Sci Technol 2024; 95:105584. [DOI: 10.1016/j.jddst.2024.105584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2024]
|
25
|
Li Z, Mo F, Guo K, Ren S, Wang Y, Chen Y, Schwartz PB, Richmond N, Liu F, Ronnekleiv-Kelly SM, Hu Q. Nanodrug-bacteria conjugates-mediated oncogenic collagen depletion enhances immune checkpoint blockade therapy against pancreatic cancer. MED 2024; 5:348-367.e7. [PMID: 38521069 DOI: 10.1016/j.medj.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 11/15/2023] [Accepted: 02/27/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) cancer cells specifically produce abnormal oncogenic collagen to bind with integrin α3β1 receptor and activate the downstream focal adhesion kinase (FAK), protein kinase B (AKT), and mitogen-activated protein kinase (MAPK) signaling pathway. Collectively, this promotes immunosuppression and tumor proliferation and restricts the response rate of clinical cancer immunotherapies. METHODS Here, by leveraging the hypoxia tropism and excellent motility of the probiotic Escherichia coli strain Nissle 1917 (ECN), we developed nanodrug-bacteria conjugates to penetrate the extracellular matrix (ECM) and shuttle the surface-conjugated protein cages composed of collagenases and anti-programmed death-ligand 1 (PD-L1) antibodies to PDAC tumor parenchyma. FINDINGS We found the oncogenic collagen expression in human pancreatic cancer patients and demonstrated its interaction with integrin α3β1. We proved that reactive oxygen species (ROS) in the microenvironment of PDAC triggered collagenase release to degrade oncogenic collagen and block integrin α3β1-FAK signaling pathway, thus overcoming the immunosuppression and synergizing with anti-PD-L1 immunotherapy. CONCLUSIONS Collectively, our study highlights the significance of oncogenic collagen in PDAC immunotherapy, and consequently, we developed a therapeutic strategy that can deplete oncogenic collagen to synergize with immune checkpoint blockade for enhanced PDAC treatment efficacy. FUNDING This work was supported by the University of Wisconsin Carbone Cancer Center Research Collaborative and Pancreas Cancer Research Task Force, UWCCC Transdisciplinary Cancer Immunology-Immunotherapy Pilot Project, and the start-up package from the University of Wisconsin-Madison (to Q.H.).
Collapse
Affiliation(s)
- Zhaoting Li
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Fanyi Mo
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kai Guo
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Shuai Ren
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Yixin Wang
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yu Chen
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Patrick B Schwartz
- Department of Surgery, Division of Surgical Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Nathaniel Richmond
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Fengyuan Liu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sean M Ronnekleiv-Kelly
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Surgery, Division of Surgical Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
26
|
Singh S, Sawal A. Comprehensive Review on Pancreatic Head Cancer: Pathogenesis, Diagnosis, and Treatment Challenges in the Quest for Improved Survival. Cureus 2024; 16:e54290. [PMID: 38500905 PMCID: PMC10945288 DOI: 10.7759/cureus.54290] [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: 07/25/2023] [Accepted: 02/16/2024] [Indexed: 03/20/2024] Open
Abstract
This comprehensive review explores the complexities surrounding pancreatic head cancer, a highly fatal and challenging-to-treat illness with a survival rate of less than five years. Despite being a major contributor to cancer-related deaths, pancreatic head malignancy often eludes early detection due to its posterior location and high metastatic potential. The review delves into the associated symptoms, including gastric outlet obstruction and obstructive jaundice, highlighting the impact on the patient's eligibility for surgery. Examining recent advancements, the article discusses fast-track surgery recovery programs and emerging immunotherapeutic approaches, acknowledging the unique challenges posed by the immunosuppressive environment of pancreatic head cancer. Additionally, the review elucidates the intricate relationship between pancreatic cancer and glucose levels, emphasizing the role of islets of Langerhans in insulin production. The pathogenesis section explores lifestyle and genetic factors contributing to pancreatic head carcinoma, shedding light on risk factors such as smoking, obesity, diabetes, and hereditary predispositions. The extensive analysis of pancreatic cancer diagnosis methods encompasses imaging techniques, biopsies, and biomarkers, emphasizing the challenges posed by late-stage diagnoses. Addressing treatment modalities, the review emphasizes the significance of surgery, chemotherapy, radiotherapy, and targeted therapy. The intricate details of neoadjuvant, immunotherapy, and microbial therapy provide a comprehensive understanding of evolving treatment strategies. The review concludes by highlighting promising areas of research, including oncolytic viral therapy and gene editing technology, aiming to enhance the limited treatment options for this devastating disease.
Collapse
Affiliation(s)
- Shreya Singh
- Anatomy, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Anupama Sawal
- Anatomy, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
27
|
Jain M, Atayan D, Rakhmatullin T, Dakhtler T, Popov P, Kim P, Viborniy M, Gontareva I, Samokhodskaya L, Egorov V. Cell-Free Tumor DNA Detection-Based Liquid Biopsy of Plasma and Bile in Patients with Various Pancreatic Neoplasms. Biomedicines 2024; 12:220. [PMID: 38255325 PMCID: PMC10813046 DOI: 10.3390/biomedicines12010220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
The key challenge of cell-free tumor DNA (cftDNA) analysis in pancreatic ductal adenocarcinoma (PDAC) is overcoming its low detection rate, which is mainly explained by the overall scarcity of this biomarker in plasma. Obstructive jaundice is a frequent event in PDAC, which enables bile collection as a part of routine treatment. The aim of this study was to evaluate the performance of KRAS-mutated cftDNA detection-based liquid biopsy of plasma and bile in patients with pancreatic neoplasms using digital droplet PCR. The study included healthy volunteers (n = 38), patients with PDAC (n = 95, of which 20 had obstructive jaundice) and other pancreatic neoplasms (OPN) (n = 18). The sensitivity and specificity compared to the control group were 61% and 100% (AUC-ROC-0.805), and compared to the OPN group, they were 61% and 94% (AUC-ROC-0.794), respectively. Bile exhibited higher cftDNA levels than plasma (248.6 [6.743; 1068] vs. 3.26 [0; 19.225] copies/mL) and a two-fold higher detection rate (p < 0.01). Plasma cftDNA levels were associated with distant metastases, tumor size, and CA 19-9 (p < 0.05). The probability of survival was worse in patients with higher levels of cftDNA in plasma (hazard ratio-2.4; 95% CI: 1.3-4.6; p = 0.005) but not in bile (p > 0.05). Bile is a promising alternative to plasma in patients with obstructive jaundice, at least for the diagnostic purposes of liquid biopsy.
Collapse
Affiliation(s)
- Mark Jain
- Medical Research and Educational Center, Lomonosov Moscow State University, 119992 Moscow, Russia;
| | - David Atayan
- Joint Stock Company “Ilyinsky Hospital”, 143421 Moscow, Russia; (D.A.); (T.D.); (P.P.); (P.K.); (M.V.); (I.G.); (V.E.)
| | - Tagir Rakhmatullin
- Department of Fundamental Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Tatyana Dakhtler
- Joint Stock Company “Ilyinsky Hospital”, 143421 Moscow, Russia; (D.A.); (T.D.); (P.P.); (P.K.); (M.V.); (I.G.); (V.E.)
| | - Pavel Popov
- Joint Stock Company “Ilyinsky Hospital”, 143421 Moscow, Russia; (D.A.); (T.D.); (P.P.); (P.K.); (M.V.); (I.G.); (V.E.)
| | - Pavel Kim
- Joint Stock Company “Ilyinsky Hospital”, 143421 Moscow, Russia; (D.A.); (T.D.); (P.P.); (P.K.); (M.V.); (I.G.); (V.E.)
| | - Mikhail Viborniy
- Joint Stock Company “Ilyinsky Hospital”, 143421 Moscow, Russia; (D.A.); (T.D.); (P.P.); (P.K.); (M.V.); (I.G.); (V.E.)
| | - Iuliia Gontareva
- Joint Stock Company “Ilyinsky Hospital”, 143421 Moscow, Russia; (D.A.); (T.D.); (P.P.); (P.K.); (M.V.); (I.G.); (V.E.)
| | - Larisa Samokhodskaya
- Medical Research and Educational Center, Lomonosov Moscow State University, 119992 Moscow, Russia;
| | - Vyacheslav Egorov
- Joint Stock Company “Ilyinsky Hospital”, 143421 Moscow, Russia; (D.A.); (T.D.); (P.P.); (P.K.); (M.V.); (I.G.); (V.E.)
| |
Collapse
|
28
|
Berbís MÁ, Godino FP, Rodríguez-Comas J, Nava E, García-Figueiras R, Baleato-González S, Luna A. Radiomics in CT and MR imaging of the liver and pancreas: tools with potential for clinical application. Abdom Radiol (NY) 2024; 49:322-340. [PMID: 37889265 DOI: 10.1007/s00261-023-04071-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/28/2023]
Abstract
Radiomics allows the extraction of quantitative imaging features from clinical magnetic resonance imaging (MRI) and computerized tomography (CT) studies. The advantages of radiomics have primarily been exploited in oncological applications, including better characterization and staging of oncological lesions and prediction of patient outcomes and treatment response. The potential introduction of radiomics in the clinical setting requires the establishment of a standardized radiomics pipeline and a quality assurance program. Radiomics and texture analysis of the liver have improved the differentiation of hypervascular lesions such as adenomas, focal nodular hyperplasia, and hepatocellular carcinoma (HCC) during the arterial phase, and in the pretreatment determination of HCC prognostic factors (e.g., tumor grade, microvascular invasion, Ki-67 proliferation index). Radiomics of pancreatic CT and MR images has enhanced pancreatic ductal adenocarcinoma detection and its differentiation from pancreatic neuroendocrine tumors, mass-forming chronic pancreatitis, or autoimmune pancreatitis. Radiomics can further help to better characterize incidental pancreatic cystic lesions, accurately discriminating benign from malignant intrapancreatic mucinous neoplasms. Nonetheless, despite their encouraging results and exciting potential, these tools have yet to be implemented in the clinical setting. This non-systematic review will describe the essential steps in the implementation of the radiomics and feature extraction workflow from liver and pancreas CT and MRI studies for their potential clinical application. A succinct overview of reported radiomics applications in the liver and pancreas and the challenges and limitations of their implementation in the clinical setting is also discussed, concluding with a brief exploration of the future perspectives of radiomics in the gastroenterology field.
Collapse
Affiliation(s)
- M Álvaro Berbís
- Department of Radiology, HT Médica, San Juan de Dios Hospital, 14960, Córdoba, Spain.
- Department of Radiology, HT Médica, San Juan de Dios Hospital, Av. del Brillante, 106, 14012, Córdoba, Spain.
| | | | | | - Enrique Nava
- Department of Communications Engineering, University of Málaga, 29016, Málaga, Spain
| | - Roberto García-Figueiras
- Abdominal Imaging Section, University Clinical Hospital of Santiago, 15706, Santiago de Compostela, A Coruña, Spain
| | - Sandra Baleato-González
- Abdominal Imaging Section, University Clinical Hospital of Santiago, 15706, Santiago de Compostela, A Coruña, Spain
| | - Antonio Luna
- Department of Radiology, HT Médica, Clínica las Nieves, 23007, Jaén, Spain
| |
Collapse
|
29
|
Srilatha M, Malla R, Adem MP, Foote JB, Nagaraju GP. Obesity associated pancreatic ductal adenocarcinoma: Therapeutic challenges. Semin Cancer Biol 2023; 97:12-20. [PMID: 37926347 DOI: 10.1016/j.semcancer.2023.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/07/2023]
Abstract
Obesity is a prominent health issue worldwide and directly impacts pancreatic health, with obese individuals exhibiting a significant risk for increasing pancreatic ductal adenocarcinoma (PDAC). Several factors potentially explain the increased risk for the development of PDAC, including obesity-induced chronic inflammation within and outside of the pancreas, development of insulin resistance and metabolic dysfunction, promotion of immune suppression within the pancreas during inflammation, pre- and malignant stages, variations in hormones levels (adiponectin, ghrelin, and leptin) produced from the adipose tissue, and acquisition of somatic mutations in tumor once- and suppressor proteins critical for pancreatic tumorigenesis. In this manuscript, we will explore the broad impact of these obesity-induced risk factors on the development and progression of PDAC, focusing on changes within the tumor microenvironment (TME) as they pertain to prevention, current therapeutic strategies, and future directions for targeting obesity management as they relate to the prevention of pancreatic tumorigenesis.
Collapse
Affiliation(s)
- Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati, Andhra Pradesh 517502, India
| | - Ramarao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh 530045, India
| | - Megha Priya Adem
- Department of Biotechnology, Sri Padmavati Mahila Visvavidyalayam (Women's University), Tirupati, Andhra Pradesh 517502, India
| | - Jeremy B Foote
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | | |
Collapse
|
30
|
Kitamura H, Morizane C, Tanabe N, Go I, Maruki Y, Ohba A, Nagashio Y, Kondo S, Hijioka S, Ueno H, Yoshida T, Okusaka T. Clinical features of germline BRCA1/2 or ATM pathogenic variant positive pancreatic cancer in Japan. Pancreatology 2023; 23:964-969. [PMID: 37914629 DOI: 10.1016/j.pan.2023.10.017] [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: 07/03/2023] [Revised: 10/08/2023] [Accepted: 10/22/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND There has been increasing interest into the role of germline BRCA1/2 pathogenic variants (gBRCA PV) and gATM PV and likely PV (PV and LPV; PV + LPV) in the carcinogenesis and treatment of pancreatic cancer (PC), but the clinical features have not been well described. METHODS Patients with confirmed gBRCA PV and gATM PV + LPV PC treated at our hospital between April 2016 and December 2021, were retrospectively evaluated for clinical characteristics and outcomes. RESULTS Twenty-two patients harbored gBRCA PV and three patients harbored gATM PV + LPV. Of the gBRCA PV patients, 81.8 % received platinum-based chemotherapy with favorable treatment outcomes with an objective response rate of 50.0 % (95 % CI: 23.0-77.0), median progression free survival (PFS) of 334 days, and median overall survival (OS) of 926 days from the initiation of first-line chemotherapy. The annual number of patients with gBRCA PV was two patients per year before January 2021 (when BRACAnalysis became available in Japan), and ten patients during the 10 months thereafter. Four patients (20 %) with gBRCA PV developed soft-tissue metastasis with progression. Two patients with gATM PV + LPV received platinum-based chemotherapy and the best response of those patients was partial response and stable disease and their OS from the initiation of first-line chemotherapy was 1192 and 989 days, and PFS was 579 and 140 days, respectively. CONCLUSION The diagnosis of gBRCA PV-positive PC has increased revealed in recent years. These tumors appear to be sensitive to platinum-based chemotherapy, with long term survival observed in gATM PV + LPV-positive patients.
Collapse
Affiliation(s)
- Hidetoshi Kitamura
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan.
| | - Noriko Tanabe
- Department of Genetic Medicine and Services, National Cancer Center Hospital, Japan
| | - Ikeda Go
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Yuta Maruki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Akihiro Ohba
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Yoshikuni Nagashio
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Shunsuke Kondo
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Susumu Hijioka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Hideki Ueno
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| | - Teruhiko Yoshida
- Department of Genetic Medicine and Services, National Cancer Center Hospital, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Japan
| |
Collapse
|
31
|
Hinestrosa JP, Sears RC, Dhani H, Lewis JM, Schroeder G, Balcer HI, Keith D, Sheppard BC, Kurzrock R, Billings PR. Development of a blood-based extracellular vesicle classifier for detection of early-stage pancreatic ductal adenocarcinoma. COMMUNICATIONS MEDICINE 2023; 3:146. [PMID: 37857666 PMCID: PMC10587093 DOI: 10.1038/s43856-023-00351-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/24/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has an overall 5-year survival rate of just 12.5% and thus is among the leading causes of cancer deaths. When detected at early stages, PDAC survival rates improve substantially. Testing high-risk patients can increase early-stage cancer detection; however, currently available liquid biopsy approaches lack high sensitivity and may not be easily accessible. METHODS Extracellular vesicles (EVs) were isolated from blood plasma that was collected from a training set of 650 patients (105 PDAC stages I and II, 545 controls). EV proteins were analyzed using a machine learning approach to determine which were the most informative to develop a classifier for early-stage PDAC. The classifier was tested on a validation cohort of 113 patients (30 PDAC stages I and II, 83 controls). RESULTS The training set demonstrates an AUC of 0.971 (95% CI = 0.953-0.986) with 93.3% sensitivity (95% CI: 86.9-96.7) at 91.0% specificity (95% CI: 88.3-93.1). The trained classifier is validated using an independent cohort (30 stage I and II cases, 83 controls) and achieves a sensitivity of 90.0% and a specificity of 92.8%. CONCLUSIONS Liquid biopsy using EVs may provide unique or complementary information that improves early PDAC and other cancer detection. EV protein determinations herein demonstrate that the AC Electrokinetics (ACE) method of EV enrichment provides early-stage detection of cancer distinct from normal or pancreatitis controls.
Collapse
Affiliation(s)
| | - Rosalie C Sears
- Department of Molecular and Medical Genetics, Brenden-Colson Center for Pancreatic Cancer, Knight Cancer Institute, Oregon Health and Sciences University, Portland, OR, USA
| | | | | | | | | | - Dove Keith
- Brenden-Colson Center for Pancreatic Cancer, Knight Cancer Institute, Oregon Health and Sciences University, Portland, OR, USA
| | - Brett C Sheppard
- Brenden-Colson Center for Pancreatic Cancer, Knight Cancer Institute, Oregon Health and Sciences University, Portland, OR, USA
| | - Razelle Kurzrock
- Medical College of Wisconsin, Milwaukee, WI, USA
- Worldwide Innovative Network for Personalized Cancer Medicine, Chevilly-Larue, France
| | | |
Collapse
|
32
|
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: 0] [Impact Index Per Article: 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
|
33
|
Bastos N, Castaldo SA, Adem B, Machado JC, Melo CA, Melo SA. SMC3 epigenetic silencing regulates Rab27a expression and drives pancreatic cancer progression. J Transl Med 2023; 21:578. [PMID: 37641131 PMCID: PMC10463307 DOI: 10.1186/s12967-023-04448-1] [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: 05/10/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is expected to soon surpass colorectal cancer as a leading cause of cancer mortality in both males and females in the US, only lagging behind lung cancer. The lethality of PDAC is driven by late diagnosis and inefficient therapies. The complex biology of PDAC involves various cellular components, including exosomes that carry molecular information between cells. Thus, recipient cells can be reprogrammed, impacting tumorigenesis. Rab27a is a GTPase responsible for the last step of exosomes biogenesis. Hence, dissecting the mechanisms that regulate the expression of Rab27a and that control exosomes biogenesis can provide fundamental insights into the molecular underpinnings regulating PDAC progression. METHODS To assess the mechanism that regulates Rab27a expression in PDAC, we used PDAC cell lines. The biological significance of these findings was validated in PDAC genetically engineered mouse models (GEMMs) and human samples. RESULTS In this work we demonstrate in human PDAC samples and GEMMs that Rab27a expression decreases throughout the development of the disease, and that Rab27a knockout promotes disease progression. What is more, we demonstrate that Rab27a expression is epigenetically regulated in PDAC. Treatment with demethylating agents increases Rab27a expression specifically in human PDAC cell lines. We found that SMC3, a component of the cohesin complex, regulates Rab27a expression in PDAC. SMC3 methylation is present in human PDAC specimens and treatment with demethylating agents increases SMC3 expression in human PDAC cell lines. Most importantly, high levels of SMC3 methylation are associated with a worse prognosis in PDAC. Mechanistically, we identified an enhancer region within the Rab27a gene that recruits SMC3, and modulates Rab27a expression. CONCLUSION Overall, we dissected a mechanism that regulates Rab27a expression during PDAC progression and impacts disease prognosis.
Collapse
Affiliation(s)
- Nuno Bastos
- i3S - Instituto de Investigação e Inovação Em Saúde, University of Porto, 4200-135, Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313, Porto, Portugal
| | - Stéphanie A Castaldo
- i3S - Instituto de Investigação e Inovação Em Saúde, University of Porto, 4200-135, Porto, Portugal
| | - Bárbara Adem
- i3S - Instituto de Investigação e Inovação Em Saúde, University of Porto, 4200-135, Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313, Porto, Portugal
| | - José C Machado
- i3S - Instituto de Investigação e Inovação Em Saúde, University of Porto, 4200-135, Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- Porto Comprehensive Cancer Center (P.CCC) Raquel Seruca, Porto, Portugal
| | - Carlos A Melo
- The Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge, UK
| | - Sonia A Melo
- i3S - Instituto de Investigação e Inovação Em Saúde, University of Porto, 4200-135, Porto, Portugal.
- Department of Pathology, Faculty of Medicine, University of Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.
- Porto Comprehensive Cancer Center (P.CCC) Raquel Seruca, Porto, Portugal.
| |
Collapse
|
34
|
Yang Y, Shi J, Huang J, Cheng M, Geng S, Yu W, Chen N, Chen C, Wang Z. Case-Control Trials on Risk Factors for Pancreatic Cancer: A Systematic Review and Meta-Analysis. IRANIAN JOURNAL OF PUBLIC HEALTH 2023; 52:1578-1588. [PMID: 37744539 PMCID: PMC10512132 DOI: 10.18502/ijph.v52i8.13397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 04/19/2022] [Indexed: 09/26/2023]
Abstract
Background The single risk factors of pancreatic cancer (PC) has been extensively studied. We aimed to synthesize results from such studies to identify and estimate multiple independent risk factors of PC. Methods Articles published up to Feb 28, 2020 in English or Chinese reporting risk factors of PC were reviewed. The fixed-effects model with 95% confidence interval (CI) were used to calculate the pooled Odds Ratio (OR). Data were analyzed using RevMan 5.3. Results PC was significantly associated with smoking (OR: 1.76, 95% CI: 1.61-1.92, P < 0.00001, I2 = 6%), diabetes (OR: 2.69, 95% CI: 2.52-2.88, P < 0.00001, I2 = 0%), family history of PC (OR: 2.58, 95% CI: 2.13-3.11, P < 0.00001, I2 = 0%), and chronic pancreatitis (OR: 5.84, 95% CI: 3.63-9.41, P < 0.00001, I2 = 0%). Conclusion Smoking, diabetes, family history of PC, and chronic pancreatitis were independent risk factors for PC. These independent risk factors have an important role in identifying high-risk groups, which is of great significance to reduce the incidence of PC and improve the quality of life and prognosis of patients.
Collapse
Affiliation(s)
- Yan Yang
- School of Economics & Management, Tongji University, Shanghai, China
| | - Jianwei Shi
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaoling Huang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingwang Cheng
- School of Economics & Management, Tongji University, Shanghai, China
| | - Shasha Geng
- General Practice Medicine, Dongfang Hospital of Tongji University, Shanghai, China
| | - Wenya Yu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Chen
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Chen
- Shanghai Jing’an District Jiangning Road Community Health Service Centre, Shanghai, China
| | - Zhaoxin Wang
- Department of Dermatology, The Fifth People’s Hospital of Hainan Province, Hainan Medical University, Haikou, China
- School of Management, Hainan Medical University, Haikou, China
| |
Collapse
|
35
|
Morgan A, Griffin M, Kameni L, Wan DC, Longaker MT, Norton JA. Medical Biology of Cancer-Associated Fibroblasts in Pancreatic Cancer. BIOLOGY 2023; 12:1044. [PMID: 37626931 PMCID: PMC10451924 DOI: 10.3390/biology12081044] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023]
Abstract
Pancreatic cancer is one of the deadliest forms of cancer with one of the lowest 5-year survival rates of all cancer types. A defining characteristic of pancreatic cancer is the existence of dense desmoplastic stroma that, when exposed to stimuli such as cytokines, growth factors, and chemokines, generate a tumor-promoting environment. Cancer-associated fibroblasts (CAFs) are activated during the progression of pancreatic cancer and are a crucial component of the tumor microenvironment (TME). CAFs are primarily pro-tumorigenic in their activated state and function as promoters of cancer invasion, proliferation, metastasis, and immune modulation. Aided by many signaling pathways, cytokines, and chemokines in the tumor microenvironment, CAFs can originate from many cell types including resident fibroblasts, mesenchymal stem cells, pancreatic stellate cells, adipocytes, epithelial cells, endothelial cells, and other cell types. CAFs are a highly heterogeneous cell type expressing a variety of surface markers and performing a wide range of tumor promoting and inhibiting functions. Single-cell transcriptomic analyses have revealed a high degree of specialization among CAFs. Some examples of CAF subpopulations include myofibrotic CAFs (myCAFs), which exhibit a matrix-producing contractile phenotype; inflammatory CAFs (iCAF) that are classified by their immunomodulating, secretory phenotype; and antigen-presenting CAFs (apCAFs), which have antigen-presenting capabilities and express Major Histocompatibility Complex II (MHC II). Over the last several years, various attempts have been undertaken to describe the mechanisms of CAF-tumor cell interaction, as well as CAF-immune cell interaction, that contribute to tumor proliferation, invasion, and metastasis. Although our understanding of CAF biology in cancer has steadily increased, the extent of CAFs heterogeneity and their role in the pathobiology of pancreatic cancer remains elusive. In this regard, it becomes increasingly evident that further research on CAFs in pancreatic cancer is necessary.
Collapse
Affiliation(s)
- Annah Morgan
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.M.); (M.G.); (L.K.); (D.C.W.); (M.T.L.)
| | - Michelle Griffin
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.M.); (M.G.); (L.K.); (D.C.W.); (M.T.L.)
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lionel Kameni
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.M.); (M.G.); (L.K.); (D.C.W.); (M.T.L.)
| | - Derrick C. Wan
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.M.); (M.G.); (L.K.); (D.C.W.); (M.T.L.)
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael T. Longaker
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.M.); (M.G.); (L.K.); (D.C.W.); (M.T.L.)
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jeffrey A. Norton
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.M.); (M.G.); (L.K.); (D.C.W.); (M.T.L.)
- Division of General Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
36
|
Partyka O, Pajewska M, Kwaśniewska D, Czerw A, Deptała A, Budzik M, Cipora E, Gąska I, Gazdowicz L, Mielnik A, Sygit K, Sygit M, Krzych-Fałta E, Schneider-Matyka D, Grochans S, Cybulska AM, Drobnik J, Bandurska E, Ciećko W, Ratajczak P, Kamecka K, Marczak M, Kozłowski R. Overview of Pancreatic Cancer Epidemiology in Europe and Recommendations for Screening in High-Risk Populations. Cancers (Basel) 2023; 15:3634. [PMID: 37509296 PMCID: PMC10377815 DOI: 10.3390/cancers15143634] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Pancreatic cancer is the seventh most common cause of death in the group of oncological diseases. Due to the asymptomatic course, early diagnosis is difficult. Currently, early detection methods are only used in high-risk groups. A literature review based on the available results of observational studies on patients with pancreatic cancer and people from high-risk groups was used to summarize the knowledge on risk factors. The GLOBOCAN 2020 data were used to assess the epidemiological situation in Europe. A summary of screening recommendations was prepared based on the available documents from medical organizations and associations. Pancreatic cancer risk factors are divided into two main groups: non-modifiable factors, e.g., hereditary factors and age, which increase the risk of developing this disease, and modifiable factors-BMI, smoking, and alcohol consumption. Hereditary factors account for 10% of pancreatic cancer cases. The highly specialized methods of early detection, (MRI, CT, or EUS) are used for screening high-risk populations. Of all the imaging methods, EUS is considered the most sensitive for pancreatic cancer and allows an accurate assessment of the size of even small lesions (<30 mm) and the extent of tumour infiltration into blood vessels. The available studies vary on the level of sensitivity and specificity of these methods for the diagnosis of pancreatic cancer. EUS, MRI, and CT are also expensive procedures and in some patients can be invasive, which is one of the arguments against the introduction of population screening programs based on imaging methods. Therefore, it is important to look for viable solutions that would improve early detection. This is important from the point of view of healthcare systems in Europe, where almost 29% of all global pancreatic cancer cases are reported.
Collapse
Affiliation(s)
- Olga Partyka
- Department of Health Economics and Medical Law, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Monika Pajewska
- Department of Health Economics and Medical Law, Medical University of Warsaw, 01-445 Warsaw, Poland
- Department of Economic and System Analyses, National Institute of Public Health NIH-National Research Institute, 00-791 Warsaw, Poland
| | - Daria Kwaśniewska
- Department of Oncology, The National Institute of Medicine of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | - Aleksandra Czerw
- Department of Health Economics and Medical Law, Medical University of Warsaw, 01-445 Warsaw, Poland
- 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
| | - Michał Budzik
- 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
| | - Lucyna Gazdowicz
- Medical Institute, Jan Grodek State University in Sanok, 38-500 Sanok, Poland
| | - Aneta Mielnik
- 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
| | - Szymon Grochans
- Department of Specialised 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
| | - Jarosław Drobnik
- Department of Family Medicine, Faculty of Medicine, Wrocław Medical University, 51-141 Wrocław, 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
| | - Piotr Ratajczak
- Department of Pharmacoeconomics and Social Pharmacy, Poznan University of Medical Sciences, 60-806 Poznań, Poland
| | - Karolina Kamecka
- Department of Management and Logistics in Healthcare, Medical University of Lodz, 90-131 Lodz, Poland
| | - Michał Marczak
- Collegium Management, WSB Merito University in Warsaw, 03-204 Warszawa, Poland
| | - Remigiusz Kozłowski
- Department of Management and Logistics in Healthcare, Medical University of Lodz, 90-131 Lodz, Poland
| |
Collapse
|
37
|
Tornel-Avelar AI, Velarde Ruiz-Velasco JA, Pelaez-Luna M. Pancreatic cancer, autoimmune or chronic pancreatitis, beyond tissue diagnosis: Collateral imaging and clinical characteristics may differentiate them. World J Gastrointest Oncol 2023; 15:925-942. [PMID: 37389107 PMCID: PMC10302998 DOI: 10.4251/wjgo.v15.i6.925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/21/2023] [Accepted: 04/28/2023] [Indexed: 06/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies and is developing into the 2nd leading cause of cancer-related death. Often, the clinical and radiological presentation of PDAC may be mirrored by other inflammatory pancreatic masses, such as autoimmune pancreatitis (AIP) and mass-forming chronic pancreatitis (MFCP), making its diagnosis challenging. Differentiating AIP and MFCP from PDAC is vital due to significant therapeutic and prognostic implications. Current diagnostic criteria and tools allow the precise differentiation of benign from malignant masses; however, the diagnostic accuracy is imperfect. Major pancreatic resections have been performed in AIP cases under initial suspicion of PDAC after a diagnostic approach failed to provide an accurate diagnosis. It is not unusual that after a thorough diagnostic evaluation, the clinician is confronted with a pancreatic mass with uncertain diagnosis. In those cases, a re-evaluation must be entertained, preferably by an experienced multispecialty team including radiologists, pathologists, gastroenterologists, and surgeons, looking for disease-specific clinical, imaging, and histological hallmarks or collateral evidence that could favor a specific diagnosis. Our aim is to describe current diagnostic limitations that hinder our ability to reach an accurate diagnosis among AIP, PDAC, and MFCP and to highlight those disease-specific clinical, radiological, serological, and histological characteristics that could support the presence of any of these three disorders when facing a pancreatic mass with uncertain diagnosis after an initial diagnostic approach has been unsuccessful.
Collapse
Affiliation(s)
- Ana I Tornel-Avelar
- Department of Gastroenterology, Hospital Civil of Guadalajara “Fray Antonio Alcalde”, Guadalajara 44340, Jalisco, Mexico
| | | | - Mario Pelaez-Luna
- Research Division School of Medicine/Department of Gastroenterology, Universidad Nacional Autonoma de México/National Institute of Medical Sciences and Nutrition “Salvador Zubiran”, Tlalpan 14000, Mexico City, Mexico
| |
Collapse
|
38
|
Liou GY, Byrd CJ. Diagnostic Bioliquid Markers for Pancreatic Cancer: What We Have vs. What We Need. Cancers (Basel) 2023; 15:2446. [PMID: 37173913 PMCID: PMC10177101 DOI: 10.3390/cancers15092446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer, currently has a dismal five-year survival rate of approximately 10% due to late diagnosis and a lack of efficient treatment options such as surgery. Furthermore, the majority of PDAC patients have surgically unresectable cancer, meaning cancer cells have either reached the surrounding blood vessels or metastasized to other organs distant from the pancreas area, resulting in low survival rates as compared to other types of cancers. In contrast, the five-year survival rate of surgically resectable PDAC patients is currently 44%. The late diagnosis of PDAC is a result of little or no symptoms in its early stage of development and a lack of specific biomarkers that may be utilized in routine examinations in the clinic. Although healthcare professionals understand the importance of early detection of PDAC, the research on the subject has lagged and no significant changes in the death toll of PDAC patients has been observed. This review is focused on understanding potential biomarkers that may increase the early diagnosis of PDAC patients at its surgically resectable stage. Here, we summarize the currently available biomarkers used in the clinic as well as those being developed with the hope of providing insight into the future of liquid biomarkers to be used in routine examinations for the early diagnosis of PDAC.
Collapse
Affiliation(s)
- Geou-Yarh Liou
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Crystal J. Byrd
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA
| |
Collapse
|
39
|
Anderson MA, Knipp DE, Noda Y, Kamran SC, Baliyan V, Kordbacheh H, Hong TS, Kambadakone A. MRI-Based Tumor Necrosis Depiction in Pancreatic Ductal Adenocarcinoma: Can It Predict Tumor Aggressiveness? Cancers (Basel) 2023; 15:cancers15082313. [PMID: 37190241 DOI: 10.3390/cancers15082313] [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: 03/30/2023] [Revised: 04/13/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
The purpose of this study was to investigate whether tumor necrosis depicted on contrast-enhanced abdominal MRI can predict tumor aggressiveness in pancreatic ductal adenocarcinoma (PDAC). In this retrospective analysis, we included 71 patients with pathology-proven PDAC who underwent contrast-enhanced MRI from 2006 to 2020. Assessment for the presence/absence of imaging detected necrosis was performed on T2-weighted and contrast-enhanced T1-weighted images. Primary tumor characteristics, regional lymphadenopathy, metastases, stage, and overall survival were analyzed. Fisher's exact and Mann-Whitney U tests were used for statistical analysis. Of the 72 primary tumors, necrosis was identified on MRI in 58.3% (42/72). Necrotic PDACs were larger (44.6 vs. 34.5 mm, p = 0.0016), had higher rates of regional lymphadenopathy (69.0% vs. 26.7%, p = 0.0007), and more frequent metastases (78.6% vs. 40.0%, p = 0.0010) than those without MRI-evident necrosis. A non-statistically significant reduction in median overall survival was observed in patients with versus without MRI-evident necrosis (15.8 vs. 38.0 months, p = 0.23). PDAC tumor necrosis depicted on MRI was associated with larger tumors and higher frequency of regional lymphadenopathy and metastases.
Collapse
Affiliation(s)
- Mark A Anderson
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - David E Knipp
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Yoshifumi Noda
- Department of Radiology, Gifu University, 1-1-1 Yanagido Street, Gifu City 501-1194, Japan
| | - Sophia C Kamran
- Department of Radiation Oncology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Vinit Baliyan
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Hamed Kordbacheh
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Avinash Kambadakone
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| |
Collapse
|
40
|
Agrawal R, Natarajan KN. Oncogenic signaling pathways in pancreatic ductal adenocarcinoma. Adv Cancer Res 2023; 159:251-283. [PMID: 37268398 DOI: 10.1016/bs.acr.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common (∼90% cases) pancreatic neoplasm and one of the most lethal cancer among all malignances. PDAC harbor aberrant oncogenic signaling that may result from the multiple genetic and epigenetic alterations such as the mutation in driver genes (KRAS, CDKN2A, p53), genomic amplification of regulatory genes (MYC, IGF2BP2, ROIK3), deregulation of chromatin-modifying proteins (HDAC, WDR5) among others. A key event is the formation of Pancreatic Intraepithelial Neoplasia (PanIN) that often results from the activating mutation in KRAS. Mutated KRAS can direct a variety of signaling pathways and modulate downstream targets including MYC, which play an important role in cancer progression. In this review, we discuss recent literature shedding light on the origins of PDAC from the perspective of major oncogenic signaling pathways. We highlight how MYC directly and indirectly, with cooperation with KRAS, affect epigenetic reprogramming and metastasis. Additionally, we summarize the recent findings from single cell genomic approaches that highlight heterogeneity in PDAC and tumor microenvironment, and provide molecular avenues for PDAC treatment in the future.
Collapse
Affiliation(s)
- Rahul Agrawal
- DTU Bioengineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | | |
Collapse
|
41
|
Tovar DR, Rosenthal MH, Maitra A, Koay EJ. Potential of artificial intelligence in the risk stratification for and early detection of pancreatic cancer. ARTIFICIAL INTELLIGENCE SURGERY 2023; 3:14-26. [PMID: 37124705 PMCID: PMC10141523 DOI: 10.20517/ais.2022.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third most lethal cancer in the United States, with a 5-year life expectancy of 11%. Most symptoms manifest at an advanced stage of the disease when surgery is no longer appropriate. The dire prognosis of PDAC warrants new strategies to improve the outcomes of patients, and early detection has garnered significant attention. However, early detection of PDAC is most often incidental, emphasizing the importance of developing new early detection screening strategies. Due to the low incidence of the disease in the general population, much of the focus for screening has turned to individuals at high risk of PDAC. This enriches the screening population and balances the risks associated with pancreas interventions. The cancers that are found in these high-risk individuals by MRI and/or EUS screening show favorable 73% 5-year overall survival. Even with the emphasis on screening in enriched high-risk populations, only a minority of incident cancers are detected this way. One strategy to improve early detection outcomes is to integrate artificial intelligence (AI) into biomarker discovery and risk models. This expert review summarizes recent publications that have developed AI algorithms for the applications of risk stratification of PDAC using radiomics and electronic health records. Furthermore, this review illustrates the current uses of radiomics and biomarkers in AI for early detection of PDAC. Finally, various challenges and potential solutions are highlighted regarding the use of AI in medicine for early detection purposes.
Collapse
Affiliation(s)
- Daniela R. Tovar
- Department of Gastrointestinal Radiation Oncology, The University of Texas, Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Anirban Maitra
- Department of Radiology, The University of Texas, Anderson Cancer Center, Houston, TX 77030, USA
| | - Eugene J. Koay
- Department of Gastrointestinal Radiation Oncology, The University of Texas, Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
42
|
Llach J, Aguilera P, Sánchez A, Ginès A, Fernández-Esparrach G, Soy G, Sendino O, Vaquero E, Carballal S, Ausania F, Ayuso JR, Darnell A, Pellisé M, Castellví-Bel S, Puig S, Balaguer F, Moreira L. Pancreatic Cancer Surveillance in Carriers of a Germline Pathogenic Variant in CDKN2A. Cancers (Basel) 2023; 15:1690. [PMID: 36980574 PMCID: PMC10046865 DOI: 10.3390/cancers15061690] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/07/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Three percent of patients with pancreatic ductal adenocarcinoma (PDAC) present a germline pathogenic variant (GPV) associated with an increased risk of this tumor, CDKN2A being one of the genes associated with the highest risk. There is no clear consensus on the recommendations for surveillance in CDKN2A GPV carriers, although the latest guidelines from the International Cancer of the Pancreas Screening Consortium recommend annual endoscopic ultrasound (EUS) or magnetic resonance imaging (MRI) regardless of family history. Our aim is to describe the findings of the PDAC surveillance program in a cohort of healthy CDKN2A GPV heterozygotes. This is an observational analysis of prospectively collected data from all CDKN2A carriers who underwent screening for PDAC at the high-risk digestive cancer clinic of the "Hospital Clínic de Barcelona" between 2013 and 2021. A total of 78 subjects were included. EUS or MRI was performed annually with a median follow-up of 66 months. Up to 17 pancreatic findings were described in 16 (20.5%) individuals under surveillance, although most of them were benign. No significant precursor lesions were identified, but an early PDAC was detected and treated. While better preventive strategies are developed, we believe that annual surveillance with EUS and/or MRI in CDKN2A GPV heterozygotes may be beneficial.
Collapse
Affiliation(s)
- Joan Llach
- Department of Gastroenterology, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain; (J.L.); (A.S.); (A.G.); (G.F.-E.); (G.S.); (O.S.); (E.V.); (S.C.); (M.P.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain;
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
| | - Paula Aguilera
- Dermatology Department, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain; (P.A.); (S.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras Instituto de Salud Carlos III, 08036 Barcelona, Spain
| | - Ariadna Sánchez
- Department of Gastroenterology, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain; (J.L.); (A.S.); (A.G.); (G.F.-E.); (G.S.); (O.S.); (E.V.); (S.C.); (M.P.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain;
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
| | - Angels Ginès
- Department of Gastroenterology, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain; (J.L.); (A.S.); (A.G.); (G.F.-E.); (G.S.); (O.S.); (E.V.); (S.C.); (M.P.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain;
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
- Facultat de Medicina I Ciències de la Salut, Universitat de Barcelona (UB), 08036 Barcelona, Spain
| | - Glòria Fernández-Esparrach
- Department of Gastroenterology, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain; (J.L.); (A.S.); (A.G.); (G.F.-E.); (G.S.); (O.S.); (E.V.); (S.C.); (M.P.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain;
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
- Facultat de Medicina I Ciències de la Salut, Universitat de Barcelona (UB), 08036 Barcelona, Spain
| | - Guillem Soy
- Department of Gastroenterology, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain; (J.L.); (A.S.); (A.G.); (G.F.-E.); (G.S.); (O.S.); (E.V.); (S.C.); (M.P.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain;
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
| | - Oriol Sendino
- Department of Gastroenterology, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain; (J.L.); (A.S.); (A.G.); (G.F.-E.); (G.S.); (O.S.); (E.V.); (S.C.); (M.P.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain;
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
| | - Eva Vaquero
- Department of Gastroenterology, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain; (J.L.); (A.S.); (A.G.); (G.F.-E.); (G.S.); (O.S.); (E.V.); (S.C.); (M.P.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain;
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
| | - Sabela Carballal
- Department of Gastroenterology, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain; (J.L.); (A.S.); (A.G.); (G.F.-E.); (G.S.); (O.S.); (E.V.); (S.C.); (M.P.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain;
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
| | - Fabio Ausania
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
- Facultat de Medicina I Ciències de la Salut, Universitat de Barcelona (UB), 08036 Barcelona, Spain
- Department of General and Digestive Surgery, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain
| | - Juan Ramón Ayuso
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
- Department of Radiology, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain
| | - Anna Darnell
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
- Department of Radiology, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain
| | - María Pellisé
- Department of Gastroenterology, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain; (J.L.); (A.S.); (A.G.); (G.F.-E.); (G.S.); (O.S.); (E.V.); (S.C.); (M.P.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain;
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
- Facultat de Medicina I Ciències de la Salut, Universitat de Barcelona (UB), 08036 Barcelona, Spain
| | - Sergi Castellví-Bel
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain;
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
| | - Susana Puig
- Dermatology Department, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain; (P.A.); (S.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras Instituto de Salud Carlos III, 08036 Barcelona, Spain
| | - Francesc Balaguer
- Department of Gastroenterology, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain; (J.L.); (A.S.); (A.G.); (G.F.-E.); (G.S.); (O.S.); (E.V.); (S.C.); (M.P.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain;
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
- Facultat de Medicina I Ciències de la Salut, Universitat de Barcelona (UB), 08036 Barcelona, Spain
| | - Leticia Moreira
- Department of Gastroenterology, Hospital Clínic Barcelona, University of Barcelona, 08036 Barcelona, Spain; (J.L.); (A.S.); (A.G.); (G.F.-E.); (G.S.); (O.S.); (E.V.); (S.C.); (M.P.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain;
- IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona (UB), 08036 Barcelona, Spain; (F.A.); (J.R.A.); (A.D.)
- Facultat de Medicina I Ciències de la Salut, Universitat de Barcelona (UB), 08036 Barcelona, Spain
| |
Collapse
|
43
|
Kim B, Gwak J, Kim M, Yang S, Hwang S, Shin S, Kim JH, Son J, Jeong SM. Suppression of fatty acid oxidation supports pancreatic cancer growth and survival under hypoxic conditions through autophagy induction. Cancer Gene Ther 2023:10.1038/s41417-023-00598-y. [PMID: 36807391 DOI: 10.1038/s41417-023-00598-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/18/2023] [Accepted: 02/03/2023] [Indexed: 02/19/2023]
Abstract
Hypoxia, one of the key features of solid tumors, induces autophagy, which acts as an important adaptive mechanism for tumor progression under hypoxic environment. Cellular metabolic reprogramming has been correlated with hypoxia, but the molecular connection to the induction of autophagy remains obscure. Here, we show that suppression of fatty acid oxidation (FAO) by hypoxia induces autophagy in human pancreatic ductal adenocarcinoma (PDAC) cells that is required for their growth and survival. Reduced cellular acetyl-CoA levels caused by FAO inhibition decreases LC3 acetylation, resulting in autophagosome formation. Importantly, PDAC cells are significantly dependent on this metabolic reprogramming, as improving FAO leads to a reduction in hypoxia-induced autophagy and an increase in cell death after chemotherapy. Thus, our study supports that suppression of FAO is an important metabolic response to hypoxia and indicates that targeting this pathway in PDAC may be an effective therapeutic approach.
Collapse
Affiliation(s)
- Byungjoo Kim
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, South Korea
| | - Jihye Gwak
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, South Korea
| | - Minjoong Kim
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, South Korea
| | - Seungyeon Yang
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, South Korea
| | - Sunsook Hwang
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, South Korea
| | - Seungmin Shin
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, South Korea
| | - Ji Hye Kim
- Department of Biomedical Sciences, Asan Medical Center, AMIST, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Jaekyoung Son
- Department of Biomedical Sciences, Asan Medical Center, AMIST, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Seung Min Jeong
- Department of Biochemistry, Institute for Aging and Metabolic Diseases, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, South Korea.
| |
Collapse
|
44
|
Hořejší K, Jin C, Vaňková Z, Jirásko R, Strouhal O, Melichar B, Teneberg S, Holčapek M. Comprehensive characterization of complex glycosphingolipids in human pancreatic cancer tissues. J Biol Chem 2023; 299:102923. [PMID: 36681125 PMCID: PMC9976472 DOI: 10.1016/j.jbc.2023.102923] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/20/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most common causes of cancer-related deaths worldwide, accounting for 90% of primary pancreatic tumors with an average 5-year survival rate of less than 10%. PDAC exhibits aggressive biology, which, together with late detection, results in most PDAC patients presenting with unresectable, locally advanced, or metastatic disease. In-depth lipid profiling and screening of potential biomarkers currently appear to be a promising approach for early detection of PDAC or other cancers. Here, we isolated and characterized complex glycosphingolipids (GSL) from normal and tumor pancreatic tissues of patients with PDAC using a combination of TLC, chemical staining, carbohydrate-recognized ligand-binding assay, and LC/ESI-MS2. The major neutral GSL identified were GSL with the terminal blood groups A, B, H, Lea, Leb, Lex, Ley, P1, and PX2 determinants together with globo- (Gb3 and Gb4) and neolacto-series GSL (nLc4 and nLc6). We also revealed that the neutral GSL profiles and their relative amounts differ between normal and tumor tissues. Additionally, the normal and tumor pancreatic tissues differ in type 1/2 core chains. Sulfatides and GM3 gangliosides were the predominant acidic GSL along with the minor sialyl-nLc4/nLc6 and sialyl-Lea/Lex. The comprehensive analysis of GSL in human PDAC tissues extends the GSL coverage and provides an important platform for further studies of GSL alterations; therefore, it could contribute to the development of new biomarkers and therapeutic approaches.
Collapse
Affiliation(s)
- Karel Hořejší
- University of Pardubice, Faculty of Chemical Technology, Department of Analytical Chemistry, , Pardubice, Czech Republic; University of South Bohemia in České Budějovice, Faculty of Science, Department of Chemistry, České Budějovice, Czech Republic
| | - Chunsheng Jin
- University of Gothenburg, Sahlgrenska Academy, Proteomics Core Facility, Göteborg, Sweden
| | - Zuzana Vaňková
- University of Pardubice, Faculty of Chemical Technology, Department of Analytical Chemistry, , Pardubice, Czech Republic
| | - Robert Jirásko
- University of Pardubice, Faculty of Chemical Technology, Department of Analytical Chemistry, , Pardubice, Czech Republic
| | - Ondřej Strouhal
- Palacký University Olomouc, Faculty of Medicine and Dentistryand University Hospital, Department of Oncology, Olomouc, Czech Republic
| | - Bohuslav Melichar
- Palacký University Olomouc, Faculty of Medicine and Dentistryand University Hospital, Department of Oncology, Olomouc, Czech Republic
| | - Susann Teneberg
- University of Gothenburg, Sahlgrenska Academy, Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, Göteborg, Sweden.
| | - Michal Holčapek
- University of Pardubice, Faculty of Chemical Technology, Department of Analytical Chemistry, , Pardubice, Czech Republic.
| |
Collapse
|
45
|
Zhong L, Liu J, Liu S, Tan G. Correlation between pancreatic cancer and metabolic syndrome: A systematic review and meta-analysis. Front Endocrinol (Lausanne) 2023; 14:1116582. [PMID: 37113491 PMCID: PMC10126301 DOI: 10.3389/fendo.2023.1116582] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/16/2023] [Indexed: 04/29/2023] Open
Abstract
Objective Pancreatic cancer is a globally frequent cause of death, which can be caused by many factors. This meta-analysis was performed to assess the correlation between pancreatic cancer and metabolic syndrome (MetS). Methods Publications were identified by searching PubMed, EMBASE, and the Cochrane Library for studies published until November 2022. Case-control and cohort studies published in English that provided information on the odds ratio (OR), relative risk (RR), or hazard ratio (HR) of metabolic syndrome and pancreatic cancer were included in the meta-analysis. Two researchers separately retrieved the core data from the included Random effects meta-analysis was conducted to summarize the findings. Results were presented as relative risk (RR) and 95% confidence interval (CI). Results MetS showed a strong association with an increased risk of developing pancreatic cancer (RR1.34, 95% CI1.23-1.46, P<0.001), and gender differences were also observed (men: RR 1.26, 95% CI 1.03-1.54, P=0.022; women: RR 1.64, 95% CI 1.41-1.90, P< 0.001). Moreover, an increased risk of developing pancreatic cancer was strongly linked to hypertension, poor high-density lipoprotein cholesterol, and hyperglycemia (hypertension: RR 1.10 CI 1.01-1.19, P=0.027; low high-density lipoprotein cholesterol: RR 1.24 CI 1.11-1.38, P<0.001; hyperglycemia: RR 1.55, CI 1.42-1.70, P< 0.001). However, pancreatic cancer was independent of obesity and hypertriglyceridemia (obesity: RR 1.13 CI 0.96-1.32, P=0.151, hypertriglyceridemia: RR 0.96, CI 0.87-1.07, P=0.486). Conclusions Although further prospective studies are required for confirmation, this meta-analysis indicated a strong relationship between MetS and pancreatic cancer. Regardless of gender, a greater risk of pancreatic cancer existed in people with MetS. Patients with MetS were more likely to develop pancreatic cancer, regardless of gender. Hypertension, hyperglycemia, and low HDL-c levels may largely account for this association. Further, the prevalence of pancreatic cancer was independent of obesity and hypertriglyceridemia. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42022368980.
Collapse
Affiliation(s)
- Lei Zhong
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jifeng Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shuo Liu
- Department of Endocrinology and Metabolic Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guang Tan
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- *Correspondence: Guang Tan,
| |
Collapse
|
46
|
Raut P, Nimmakayala RK, Batra SK, Ponnusamy MP. Clinical and Molecular Attributes and Evaluation of Pancreatic Cystic Neoplasm. Biochim Biophys Acta Rev Cancer 2023; 1878:188851. [PMID: 36535512 PMCID: PMC9898173 DOI: 10.1016/j.bbcan.2022.188851] [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: 08/29/2022] [Revised: 11/08/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Intraductal papillary mucinous neoplasms (IPMNs) and mucinous cystic neoplasms (MCNs) are all considered "Pancreatic cystic neoplasms (PCNs)" and show a varying risk of developing into pancreatic ductal adenocarcinoma (PDAC). These lesions display different molecular characteristics, mutations, and clinical manifestations. A lack of detailed understanding of PCN subtype characteristics and their molecular mechanisms limits the development of efficient diagnostic tools and therapeutic strategies for these lesions. Proper in vivo mouse models that mimic human PCNs are also needed to study the molecular mechanisms and for therapeutic testing. A comprehensive understanding of the current status of PCN biology, mechanisms, current diagnostic methods, and therapies will help in the early detection and proper management of patients with these lesions and PDAC. This review aims to describe all these aspects of PCNs, specifically IPMNs, by describing the future perspectives.
Collapse
Affiliation(s)
- Pratima Raut
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| |
Collapse
|
47
|
Topham JT, Renouf DJ, Schaeffer DF. Circulating tumor DNA: toward evolving the clinical paradigm of pancreatic ductal adenocarcinoma. Ther Adv Med Oncol 2023; 15:17588359231157651. [PMID: 36895849 PMCID: PMC9989430 DOI: 10.1177/17588359231157651] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 01/30/2023] [Indexed: 03/06/2023] Open
Abstract
Over a decade of sequencing-based genomics research has unveiled a diverse somatic mutation landscape across patients with pancreatic ductal adenocarcinoma (PDAC), and the identification of druggable mutations has aligned with the development of novel targeted therapeutics. However, despite these advances, direct translation of years of PDAC genomics research into the clinical care of patients remains a critical and unmet need. Technologies that enabled the initial mapping of the PDAC mutation landscape, namely whole-genome and transcriptome sequencing, remain overly expensive in terms of both time and financial resources. Consequentially, dependence on these technologies to identify the relatively small subset of patients with actionable PDAC alterations has greatly impeded enrollment for clinical trials testing novel targeted therapies. Liquid biopsy tumor profiling using circulating tumor DNA (ctDNA) generates new opportunities by overcoming these challenges while further addressing issues particularly relevant to PDAC, namely, difficulty of obtaining tumor tissue via fine-needle biopsy and the need for faster turnaround time due to rapid disease progression. Meanwhile, ctDNA-based approaches for tracking disease kinetics with respect to surgical and therapeutic interventions offer a means to elevate the current clinical management of PDAC toward higher granularity and accuracy. This review provides a clinically focused summary of ctDNA advances, limitations, and opportunities in PDAC and postulates ctDNA sequencing technology as a catalyst for evolving the clinical decision-making paradigm of this disease.
Collapse
Affiliation(s)
| | - Daniel J Renouf
- Pancreas Centre BC, Vancouver, BC, Canada.,Division of Medical Oncology, BC Cancer, Vancouver, BC, Canada.,Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - David F Schaeffer
- Division of Anatomic Pathology, Vancouver General Hospital, 910 West 10th Avenue, Vancouver, BC V5Z 1M9, Canada.,Pancreas Centre BC, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, UBC, Vancouver, BC, Canada
| |
Collapse
|
48
|
Rouette J, McDonald EG, Schuster T, Brophy JM, Azoulay L. Dihydropyridine Calcium Channel Blockers and Risk of Pancreatic Cancer: A Population-Based Cohort Study. J Am Heart Assoc 2022; 11:e026789. [PMID: 36515246 PMCID: PMC9798809 DOI: 10.1161/jaha.122.026789] [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: 07/13/2022] [Accepted: 10/28/2022] [Indexed: 12/15/2022]
Abstract
Background Recent studies have reported that dihydropyridine calcium channel blockers (dCCBs) may increase the risk of pancreatic cancer, but these studies had methodological limitations. We thus aimed to determine whether dCCBs are associated with an increased risk of pancreatic cancer compared with thiazide diuretics, a clinically relevant comparator. Methods and Results We conducted a new user, active comparator, population-based cohort study using the UK Clinical Practice Research Datalink. We identified new users of dCCBs and new users of thiazide diuretics between 1990 and 2018, with follow-up until 2019. Cox proportional hazards models were used to estimate hazard ratios (HRs) with 95% CIs for pancreatic cancer, comparing dCCBs with thiazide diuretics. Models were weighted using standardized morbidity ratio weights based on calendar time-specific propensity scores. We also conducted secondary analyses by cumulative duration of use, time since initiation, and individual drugs and assessed for the presence of effect modification by age, sex, smoking status, body mass index, history of chronic pancreatitis, and diabetes. The cohort included 344 480 initiators of dCCBs and 357 968 initiators of thiazide diuretics, generating 3 360 745 person-years of follow-up. After a median follow-up of 4.5 years, the weighted incidence rate per 100 000 person-years was 37.2 (95% CI, 34.1-40.4) for dCCBs and 39.4 (95% CI, 36.1-42.9) for thiazide diuretics. Overall, dCCBs were not associated with an increased risk of pancreatic cancer (weighted HR, 0.93; 95% CI, 0.80-1.09). Similar results were observed in secondary analyses. Conclusions In this large, population-based cohort study, dCCBs were not associated with an increased risk of pancreatic cancer compared with thiazide diuretics. These findings provide reassurance regarding the long-term pancreatic cancer safety of these drugs.
Collapse
Affiliation(s)
- Julie Rouette
- Centre for Clinical EpidemiologyLady Davis Institute, Jewish General HospitalMontrealCanada
- Department of Epidemiology, Biostatistics, and Occupational HealthMcGill UniversityMontrealCanada
| | - Emily G. McDonald
- Division of General Internal Medicine, Department of MedicineMcGill University Health CentreMontrealCanada
- Division of Experimental MedicineMcGill UniversityMontrealCanada
| | - Tibor Schuster
- Department of Epidemiology, Biostatistics, and Occupational HealthMcGill UniversityMontrealCanada
- Department of Family MedicineMcGill UniversityMontrealCanada
| | - James M. Brophy
- Department of Epidemiology, Biostatistics, and Occupational HealthMcGill UniversityMontrealCanada
- Division of Clinical EpidemiologyMcGill University Health Centre–Research InstituteMontrealCanada
- Department of MedicineMcGill UniversityMontrealCanada
| | - Laurent Azoulay
- Centre for Clinical EpidemiologyLady Davis Institute, Jewish General HospitalMontrealCanada
- Department of Epidemiology, Biostatistics, and Occupational HealthMcGill UniversityMontrealCanada
- Gerald Bronfman Department of OncologyMcGill UniversityMontrealCanada
| |
Collapse
|
49
|
Tang Z, Xu W, Zhang M. Association between type 2 diabetes and 5-year overall survival in early-stage pancreatic cancer: a retrospective cohort study. PeerJ 2022; 10:e14538. [PMID: 36530401 PMCID: PMC9753753 DOI: 10.7717/peerj.14538] [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: 08/01/2022] [Accepted: 11/17/2022] [Indexed: 12/14/2022] Open
Abstract
Background This study examined the association between type 2 diabetes mellitus (T2DM) and 5-year overall survival (OS) in patients with pancreatic cancer (PC). Methods This retrospective cohort study included patients diagnosed with stage I/II PC at Shengjing Hospital of China Medical University from January 2012 to December 2017. All patients had pancreatic ductal adenocarcinoma or its subtypes. The outcome was the 5-year OS rate based on data from the patient charts. Data analysis was performed using SPSS 22.0. Results A total of 238 patients were included: 72 with T2DM and 166 without T2DM. There were significant differences in blood glucose levels and OS between the two groups (all P < 0.05). The median OS was 11.4 (95% confidence interval CI [8.49-14.31]) months in the T2DM group and 16.3 (95% CI [12.44-20.16], P = 0.023) months in the non-T2DM group. After adjustment for confounders, T2DM was an independent factor affecting 5-year OS (P = 0.010). Compared with non-T2DM patients, T2DM patients had a higher risk of death (HR = 1.475, 95% CI [1.096-1.985]). Conclusions T2DM is associated with 5-year OS in patients with PC.
Collapse
Affiliation(s)
- Zhiyin Tang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wanfeng Xu
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mingming Zhang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
50
|
Waleleng BJ, Adiwinata R, Wenas NT, Haroen H, Rotty L, Gosal F, Rotty L, Winarta J, Waleleng A, Simadibrata M. Screening of pancreatic cancer: Target population, optimal timing and how? Ann Med Surg (Lond) 2022; 84:104814. [PMID: 36582884 PMCID: PMC9793126 DOI: 10.1016/j.amsu.2022.104814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/26/2022] [Accepted: 10/30/2022] [Indexed: 11/08/2022] Open
Abstract
Pancreatic cancer patients usually present at a late stage due to subtle clinical manifestations. One of the most predictive prognostic factors in pancreatic cancer is the pancreatic cancer stage at diagnosis; therefore, early diagnosis is essential. Until now, pancreatic cancer screening has not become a standard practice for the general population due to the low incidence. In current circumstances, targeting individuals with a high risk of pancreatic cancer may be more rational. Several screening modalities for pancreatic cancer have also become debatable topics. Therefore, this article will review current evidence and recommendations regarding pancreatic screening cancer protocol in general and in high-risk populations.
Collapse
Affiliation(s)
- Bradley Jimmy Waleleng
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi/Prof. dr. R. D. Kandou Hospital, Manado, Indonesia
| | - Randy Adiwinata
- Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi/Prof. dr. R. D. Kandou Hospital, Manado, Indonesia
| | - Nelly Tendean Wenas
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi/Prof. dr. R. D. Kandou Hospital, Manado, Indonesia
| | - Harlinda Haroen
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi/Prof. dr. R. D. Kandou Hospital, Manado, Indonesia
| | - Linda Rotty
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi/Prof. dr. R. D. Kandou Hospital, Manado, Indonesia
| | - Fandy Gosal
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi/Prof. dr. R. D. Kandou Hospital, Manado, Indonesia
| | - Luciana Rotty
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi/Prof. dr. R. D. Kandou Hospital, Manado, Indonesia
| | - Jeanne Winarta
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi/Prof. dr. R. D. Kandou Hospital, Manado, Indonesia
| | - Andrew Waleleng
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi/Prof. dr. R. D. Kandou Hospital, Manado, Indonesia
| | - Marcellus Simadibrata
- Division of Gastroenterology, Pancreatobiliary and Digestive Endoscopy, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| |
Collapse
|