1
|
Ponz de Leon Pisani R, Altieri G, Stasio RC, Lazzano P, Reni M, Falconi M, Vanella G, Arcidiacono PG, Capurso G. Gastrointestinal symptoms in the journey of pancreatic cancer patients. Expert Rev Anticancer Ther 2025. [PMID: 40489240 DOI: 10.1080/14737140.2025.2517888] [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: 04/08/2025] [Revised: 06/04/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy frequently arising with nonspecific and overlooked gastrointestinal symptoms. Gastroenterologists are typically the first specialists to encounter these patients, positioning them to play a pivotal role not only in early diagnosis, but also in the ongoing management of the disease's complex symptom burden. AREAS COVERED This review explored gastrointestinal symptoms in patients with PDAC (ranging from pain and diarrhea to anorexia, jaundice, and nausea) and outlined both tumor- and treatment-related causes. A literature review based on non-systematic PubMed search updated to April 2025 was conducted to summarize current diagnostic strategies, medical, endoscopic therapies, and multidisciplinary management approaches. In addition, we present original data from a single-center cohort, suggesting that the involvement of gastroenterologists leads to more comprehensive management of gastrointestinal symptom control and supportive care. EXPERT OPINION Collaboration among specialists is essential for optimizing patient outcomes in the multidisciplinary management of PDAC. Gastroenterologists' 'stewardship' significantly contributes to prompt diagnosis, symptom control, quality of life preservation, and prognosis. Future priorities should focus on strengthening integration within care pathways, fostering interdisciplinary coordination, and implementing shared clinical tools to enhance comprehensive patient care. A well-structured team-based approach is key to advancing holistic PDAC management.
Collapse
Affiliation(s)
- Ruggero Ponz de Leon Pisani
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gabriele Altieri
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rosa Claudia Stasio
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pilar Lazzano
- Gastroenterology and Hepatology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Michele Reni
- Oncology Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- "Vita-Salute" San Raffaele University, Milan, Italy
| | - Massimo Falconi
- "Vita-Salute" San Raffaele University, Milan, Italy
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppe Vanella
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Giorgio Arcidiacono
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- "Vita-Salute" San Raffaele University, Milan, Italy
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- "Vita-Salute" San Raffaele University, Milan, Italy
| |
Collapse
|
2
|
Du X, Ma Y, Wang K, Zhong X, Wang J, Tian X, Wang X, Yang Y. Novel CT radiomics models for the postoperative prediction of early recurrence of resectable pancreatic adenocarcinoma: A single-center retrospective study in China. J Appl Clin Med Phys 2025; 26:e70092. [PMID: 40217563 DOI: 10.1002/acm2.70092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 03/16/2025] [Accepted: 03/25/2025] [Indexed: 06/11/2025] Open
Abstract
PURPOSE To assess the predictive capability of CT radiomics features for early recurrence (ER) of pancreatic ductal adenocarcinoma (PDAC). METHODS Postoperative PDAC patients were retrospectively selected, all of whom had undergone preoperative CT imaging and surgery. Both patients with resectable or borderline-resectable pancreatic cancer met the eligibility criteria in this study. However, owing to the differences in treatment strategies and such, this research mainly focused on patients with resectable pancreatic cancer. All patients were subject to follow-up assessments for a minimum of 9 months. A total of 250 cases meeting the inclusion criteria were included. A clinical model, a conventional radiomics model, and a deep-radiomics model were constructed for ER prediction (defined as occurring within 9 months) in the training set. A model based on the TNM staging was utilized as a baseline for comparison. Assessment of the models' performance was based on the area under the receiver operating characteristic curve (AUC). Additionally, precision-recall (PR) analysis and calibration assessments were conducted for model evaluation. Furthermore, the clinical utility of the models was evaluated through decision curve analysis (DCA), net reclassification improvement (NRI), and improvement of reclassification index (IRI). RESULTS In the test set, the AUC values for ER prediction were as follows: TNM staging, ROC-AUC = 0.673 (95% CI: 0.550, 0.795), PR-AUC = 0.362 (95% CI: 0.493, 0.710); clinical model, ROC-AUC = 0.640 (95% CI: 0.504, 0.775), PR-AUC = 0.481 (95% CI: 0.520, 0.735); radiomics model, ROC-AUC = 0.722 (95% CI: 0.604, 0.839), PR-AUC = 0.575 (95% CI: 0.466, 0.686); and deep-radiomics model, which exhibited the highest ROC-AUC of 0.895 (95% CI: 0.820, 0.970), PR-AUC = 0.834 (95% CI: 0.767, 0.923). The difference in both ROC-AUC and PR-AUC for the deep-radiomics model was statistically significant when compared to the other scores (all p < 0.05). The DCA curve of the deep-radiomics model outperformed the other models. NRI and IRI analyses demonstrated that the deep-radiomics model significantly enhances risk classification compared to the other prediction methods (all p < 0.05). CONCLUSION The predictive performance of deep features based on CT images exhibits favorable outcomes in predicting early recurrence.
Collapse
Affiliation(s)
- Xinze Du
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| | - Yongsu Ma
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| | - Kexin Wang
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Xiejian Zhong
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| | - Jianxin Wang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| | - Xiaodong Tian
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| | - Xiaoying Wang
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Yinmo Yang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
3
|
Hruban C, Bruhm DC, Chen IM, Koul S, Annapragada AV, Vulpescu NA, Short S, Theile S, Boyapati K, Alipanahi B, Skidmore ZL, Leal A, Cristiano S, Adleff V, Johannsen JS, Scharpf RB, Foda ZH, Phallen J, Velculescu VE. Genome-wide analyses of cell-free DNA for therapeutic monitoring of patients with pancreatic cancer. SCIENCE ADVANCES 2025; 11:eads5002. [PMID: 40397745 DOI: 10.1126/sciadv.ads5002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 04/07/2025] [Indexed: 05/23/2025]
Abstract
Determining response to therapy for patients with pancreatic cancer can be challenging. We evaluated methods for assessing therapeutic response using cell-free DNA (cfDNA) in plasma from patients with metastatic pancreatic cancer in the CheckPAC trial (NCT02866383). Patients were evaluated before and after initiation of therapy using tumor-informed plasma whole-genome sequencing (WGMAF) and tumor-independent genome-wide cfDNA fragmentation profiles and repeat landscapes (ARTEMIS-DELFI). Using WGMAF, molecular responders had a median overall survival (OS) of 319 days compared to 126 days for nonresponders [hazard ratio (HR) = 0.29, 95% confidence interval (CI) = 0.11-0.79, P = 0.011]. For ARTEMIS-DELFI, patients with low scores after therapy initiation had longer median OS than patients with high scores (233 versus 172 days, HR = 0.12, 95% CI = 0.046-0.31, P < 0.0001). We validated ARTEMIS-DELFI in patients with pancreatic cancer in the PACTO trial (NCT02767557). These analyses suggest that noninvasive mutation and fragmentation-based cfDNA approaches can identify therapeutic response of individuals with pancreatic cancer.
Collapse
Affiliation(s)
- Carolyn Hruban
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel C Bruhm
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Inna M Chen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Shashikant Koul
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akshaya V Annapragada
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas A Vulpescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah Short
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susann Theile
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Kavya Boyapati
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | - Alessandro Leal
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephen Cristiano
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vilmos Adleff
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julia S Johannsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Medicine, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Robert B Scharpf
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachariah H Foda
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Victor E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Chen Y, Zhang F, Dai S, Zhao J, Cai W, Zhang K, Liao X, Chen L. Lactate-associated gene MCU promotes the proliferation, migration, and invasion of pancreatic ductal adenocarcinoma. BMC Cancer 2025; 25:913. [PMID: 40399795 PMCID: PMC12096505 DOI: 10.1186/s12885-025-14319-1] [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: 01/25/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND The metabolism of lactate and lactylation of proteins are believed to influence tumor development through their effects on the tumor microenvironment and immune escape mechanisms. Nevertheless, its significance in pancreatic ductal adenocarcinoma (PDAC) has yet to be fully understood. This investigation sought to assess the predictive value and treatment implications of lactate-related genes (LRGs) in PDAC. METHODS We analyzed PDAC data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), identifying LRGs. Using weighted gene co-expression network analysis (WGCNA) and consensus clustering, we delineated lactate subtypes and extracted differentially expressed genes. Functional enrichment and gene set enrichment analysis (GSEA) analyses were conducted to explore pathways. A lactate-linked risk signature was constructed using Lasso-Cox regression, and its prognostic value was validated. In vitro experiments were executed to examine the function of MCU in PDAC cells. In vitro experiments were conducted to detect the malignant potential of MCU in PDAC cells and its effect on lactic acid metabolism. RESULTS Two lactate subtypes were identified, with distinct gene expression profiles and clinical outcomes. The risk signature, comprising four LRGs, predicted survival with significant accuracy. In vitro, MCU knockdown reduced cell proliferation, migration, invasion, and stemness, confirming its role in PDAC malignancy. At the same time, it can also inhibit lactate production and glycolysis processes. CONCLUSION Our investigation underscores the importance of LRGs in PDAC, providing a novel prognostic signature and therapeutic target.
Collapse
Affiliation(s)
- Yuhang Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Fenglin Zhang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Graduate School of Anhui, University of Traditional Chinese Medicine, Hefei, 230022, China
| | - Suoyi Dai
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jiangang Zhao
- Department of Oncology, Shaoxing Central Hospital, Shaoxing, 312030, China
- Department of Oncology, The Central Affiliated Hospital, Shaoxing University, Shaoxing, 312030, China
| | - Wenxun Cai
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ke Zhang
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China
| | - Xinghe Liao
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Lianyu Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
5
|
Selvi S, Real CM, Gentiluomo M, Balounova K, Vokacova K, Cumova A, Mohlenikova-Duchonova B, Rizzato C, Halasova E, Vodickova L, Smolkova B, Hemminki K, Campa D, Vodicka P. Genomic instability, DNA damage response and telomere homeostasis in pancreatic cancer. Semin Cancer Biol 2025; 113:59-73. [PMID: 40378535 DOI: 10.1016/j.semcancer.2025.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 04/16/2025] [Accepted: 05/04/2025] [Indexed: 05/19/2025]
Abstract
Pancreatic cancer (PC) is becoming one of the most serious health problems at present, but its causes and risk factors are still unclear. One of the drivers in pancreatic carcinogenesis is altered genomic (DNA) integrity with subsequent genomic instability in cancer cells. The latter comprises a) DNA damage response and DNA repair mechanisms, b) DNA replication and mitosis, c) epigenetic regulation, and d) telomere maintenance. In our review we addressed the above aspects in relation to the most abundant and severe form of PC, pancreatic ductal adenocarcinoma (PDAC). In summary, the interactions between the DNA damage response, telomere homeostasis and mitotic regulation are not comprehensively understood at present, including the epigenetic factors entering the trait of genomic stability maintenance. In addition, the complexity of telomere homeostasis in relation to PDAC risk, prognosis and prediction also warrants further investigations.
Collapse
Affiliation(s)
- Saba Selvi
- Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic; Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague 12800, Czech Republic
| | - Carmen Macías Real
- Cancer Predisposition and Biomarkers Group, Instituto de Investigacion Sanitaria de Santiago, Santiago de Compostela, Spain
| | | | - Katerina Balounova
- Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic; Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Klara Vokacova
- Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Andrea Cumova
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava 84505, Slovakia
| | | | - Cosmeri Rizzato
- Department of Biology, University of Pisa, Pisa 56123, Italy
| | - Erika Halasova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4, Martin 03601, Slovakia
| | - Ludmila Vodickova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4, Martin 03601, Slovakia; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, Pilsen 32300, Czech Republic; Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, Prague 12800, Czech Republic
| | - Bozena Smolkova
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava 84505, Slovakia
| | - Kari Hemminki
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, Pilsen 32300, Czech Republic; Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, FRG 69120, Germany
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa 56123, Italy
| | - Pavel Vodicka
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4, Martin 03601, Slovakia; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, Pilsen 32300, Czech Republic; Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, Prague 12800, Czech Republic.
| |
Collapse
|
6
|
Aboalela MA, Abdelmoneim M, Matsumura S, Eissa IR, Bustos-Villalobos I, Sibal PA, Orikono Y, Takido Y, Naoe Y, Kasuya H. Enhancing mesothelin CAR T cell therapy for pancreatic cancer with an oncolytic herpes virus boosting CAR target antigen expression. Cancer Immunol Immunother 2025; 74:202. [PMID: 40366419 PMCID: PMC12078189 DOI: 10.1007/s00262-025-04039-7] [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: 09/19/2024] [Accepted: 03/27/2025] [Indexed: 05/15/2025]
Abstract
Mesothelin (MSLN) is a prominent target antigen for CAR T cell therapy due to its extensive expression in various solid tumors, including pancreatic cancer. However, the therapeutic efficacy of MSLN-targeted CAR T cell therapy has been limited in clinical trials for pancreatic cancer, often resulting in temporary stable disease as the best response. The heterogeneous expression of MSLN and its loss over time, along with the immunosuppressive tumor microenvironment (TME), are key factors restricting effectiveness. Oncolytic viruses are emerging cancer therapies that replicate in tumor cells and remodel the TME into an immunogenic state. Here, we engineered an oncolytic herpes simplex virus type 1 expressing human MSLN (HSV-MSLN) and evaluated its combination with MSLN-CAR T cells in a murine pancreatic ductal adenocarcinoma model. In vitro, HSV-MSLN effectively induced MSLN expression on murine pancreatic cancer cells, with subsequent cell lysis. In co-culture, HSV-MSLN-infected cancer cells activated MSLN-CAR T cells, which effectively eliminated the infected cells. In vivo, HSV-MSLN delivered MSLN on the tumor cell surface and reprogrammed the TME toward an immunogenic state. The combination therapy significantly enhanced antitumor efficacy, inducing activated, proliferative CD8+ CAR T cells and reducing PD-1+TIM-3+ exhausted endogenous CD8+ T cells and regulatory T cells in tumors. Furthermore, the combination therapy increased migratory XCR1+CD103+ dendritic cells (DCs) in tumors and tumor-draining lymph nodes (TDLNs) while expanding CD44+CD8+ T cells with central and effector memory phenotypes. Taken together, these results demonstrate that HSV-MSLN reprograms immune cells in the TME and TDLNs and synergizes with MSLN-CAR T cells to enhance antitumor responses, leading to a more robust therapeutic effect.
Collapse
Affiliation(s)
- Mona Alhussein Aboalela
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, 466-8550, Japan
- Department of Gastroenterological Surgery, Graduate School of Medicine, Nagoya University, Nagoya, 466-8550, Japan
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Mohamed Abdelmoneim
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, 466-8550, Japan
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Shigeru Matsumura
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, 466-8550, Japan.
| | - Ibrahim Ragab Eissa
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, 466-8550, Japan
- Surgical Oncology Division, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Itzel Bustos-Villalobos
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, 466-8550, Japan
| | - Patricia Angela Sibal
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, 466-8550, Japan
- Department of Gastroenterological Surgery, Graduate School of Medicine, Nagoya University, Nagoya, 466-8550, Japan
| | - Yu Orikono
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, 466-8550, Japan
| | - Yuhei Takido
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, 466-8550, Japan
- Department of Neurosurgery, Graduate School of Medicine, Nagoya University, Nagoya, 466-8550, Japan
| | - Yoshinori Naoe
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, 466-8550, Japan
| | - Hideki Kasuya
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, 466-8550, Japan.
| |
Collapse
|
7
|
Lewellen KA, Cavaghan MK, Fogel EL, Montero AM, House MG, Zyromski NJ. Total Pancreatectomy With Islet Autotransplant: Building a Multidisciplinary Program. Pancreas 2025; 54:e491-e493. [PMID: 40096638 DOI: 10.1097/mpa.0000000000002482] [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: 10/22/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025]
Affiliation(s)
| | | | - Evan L Fogel
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN
| | - Anne M Montero
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN
| | | | | |
Collapse
|
8
|
Wang K, Ni B, Xie Y, Li Z, Yuan L, Meng C, Zhao T, Gao S, Huang C, Wang H, Ma Y, Zhou T, Feng Y, Chang A, Yang C, Yu J, Yu W, Zang F, Zhang Y, Ji RR, Wang X, Hao J. Nociceptor neurons promote PDAC progression and cancer pain by interaction with cancer-associated fibroblasts and suppression of natural killer cells. Cell Res 2025; 35:362-380. [PMID: 40122998 PMCID: PMC12012126 DOI: 10.1038/s41422-025-01098-4] [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: 09/17/2024] [Accepted: 03/05/2025] [Indexed: 03/25/2025] Open
Abstract
The emerging field of cancer neuroscience has demonstrated great progress in revealing the crucial role of the nervous system in cancer initiation and progression. Pancreatic ductal adenocarcinoma (PDAC) is characterized by perineural invasion and modulated by autonomic (sympathetic and parasympathetic) and sensory innervations. Here, we further demonstrated that within the tumor microenvironment of PDAC, nociceptor neurons interacted with cancer-associated fibroblasts (CAFs) through calcitonin gene-related peptide (CGRP) and nerve growth factor (NGF). This interaction led to the inhibition of interleukin-15 expression in CAFs, suppressing the infiltration and cytotoxic function of natural killer (NK) cells and thereby promoting PDAC progression and cancer pain. In PDAC patients, nociceptive innervation of tumor tissue is negatively correlated with the infiltration of NK cells while positively correlated with pain intensity. This association serves as an independent prognostic factor for both overall survival and relapse-free survival for PDAC patients. Our findings highlight the crucial regulation of NK cells by nociceptor neurons through interaction with CAFs in the development of PDAC. We also propose that targeting nociceptor neurons or CGRP signaling may offer a promising therapy for PDAC and cancer pain.
Collapse
Affiliation(s)
- Kaiyuan Wang
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| | - Bo Ni
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yongjie Xie
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zekun Li
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Limei Yuan
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chenyang Meng
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tiansuo Zhao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Song Gao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chongbiao Huang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Hongwei Wang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ying Ma
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tianxing Zhou
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yukuan Feng
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Antao Chang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chao Yang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jun Yu
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wenwen Yu
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Fenglin Zang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yanhui Zhang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| | - Xiuchao Wang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| | - Jihui Hao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
9
|
Lee JC, Kang SW, Sim EJ, Bae JS, Koo SM, Byoun MS, Kwon S, Hong S, Kim Y, Youn Y, Jung K, Kim J, Jeong HH, Kim J, Hwang JH. Novel mRNA biomarker-based liquid biopsy for the detection of resectable pancreatic cancer. BMC Cancer 2025; 25:762. [PMID: 40269781 PMCID: PMC12016232 DOI: 10.1186/s12885-025-14124-w] [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: 09/19/2024] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies and most often diagnosed at an advanced stage. Identification of markers for the early diagnosis of PDAC is crucial. In this study, we aimed to identify novel mRNA biomarkers for diagnosing PDAC, focusing on early-stage tumorigenesis and associated immunological changes. METHODS Blood samples and clinical information from 1,963 individuals were obtained from a single tertiary hospital between 2015 and 2021. Candidate mRNA biomarkers were identified through literature review, and their expression levels in buffy coat samples were measured using reverse-transcription quantitative polymerase chain reaction. Machine learning-based feature selection confirmed the final biomarker panel, which was tested using an independent dataset for diagnostic performance. RESULTS In total, 1,504 individuals (417 patients with PDAC and 1,087 non-disease controls) were eligible for the study. Among the 55 candidate biomarkers identified, 15 mRNAs (CCL5, CCR5, CLEC7A, CXCL8, CXCR2, CXCR4, FOXP3, IFNA1, IFNL1, PTGES, PTGES2, PTGS2, SLC27A2, TNF, and VEGFA) were selected based on their diagnostic performance in distinguishing PDAC from control groups. The final model, HELP-15 (Human Early Liquid biopsy for PDAC), identified all PDAC stages (area under the curve [AUC] = 0.956) in the test set. For resectable pancreatic cancer (RPC), the AUC was 0.968, compared to 0.910 for carbohydrate antigen 19 - 9 (CA19-9). The combined model of the panel and CA19-9 achieved an AUC of 0.985 in patients with RPC. For all PDAC stages in patients with normal CA19-9 levels, the AUC of the panel was 0.967, whereas CA19-9 alone or in combination with the panel had AUCs of 0.658 and 0.885, respectively. CONCLUSION Compared to CA19-9, the mRNA biomarker panel, HELP-15, improved diagnostic performance in patients with RPC, particularly in those with normal CA19-9 levels.
Collapse
Affiliation(s)
- Jong-Chan Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Won Kang
- Research Center, HuVet bio Inc, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Eun-Jin Sim
- Research Center, HuVet bio Inc, Seoul, Korea
| | - Jin-Sik Bae
- Research Center, HuVet bio Inc, Seoul, Korea
| | | | | | - Serin Kwon
- Department of Life Science, Dongguk University, Seoul, Korea
| | - Seoi Hong
- Department of Computer and Artificial Intelligence, Dongguk University, Seoul, Korea
| | - Yunji Kim
- Department of Computer and Artificial Intelligence, Dongguk University, Seoul, Korea
| | - Yuna Youn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kwangrok Jung
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | | | - Jihie Kim
- Department of Computer and Artificial Intelligence, Dongguk University, Seoul, Korea.
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
10
|
Xu M, Long Y, Chen P, Li A, Xin J, Xu Y. Establishment of a nomogram based on Lasso Cox regression for albumin combined with systemic immune-inflammation index score to predict prognosis in advanced pancreatic carcinoma. Front Oncol 2025; 15:1447055. [PMID: 40265018 PMCID: PMC12011609 DOI: 10.3389/fonc.2025.1447055] [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: 06/11/2024] [Accepted: 03/26/2025] [Indexed: 04/24/2025] Open
Abstract
Purpose The study aims to establish a nomogram to predict advanced pancreatic carcinoma patients' overall survival (OS), incorporating albumin combined with systemic immune-inflammation index (A-SII) score and clinical characteristics. Methods A retrospective study analyzed the clinical data of 205 advanced pancreatic carcinoma patients without antitumor treatment from the Yancheng No.1 People's Hospital between October 2011 and June 2023, and the study divided patients into the training set and the validation set randomly at the proportion of three to one. The A-SII score was divided into scores of 0, 1, and 2 according to the different levels of albumin and SII. Receiver operating characteristic (ROC) curves and time-dependent area under the curve were used to evaluate the predictive ability of the A-SII score. The nomogram1 and nomogram2 were established by the multivariate Cox regression and Lasso Cox regression respectively. The study evaluated the discriminability of nomogram1 and nomogram2 based on C-index and ROC curves to obtain the optimal model. Subsequently, we plotted decision curve analyses (DCA) and calibration curves to estimate the clinical benefit and accuracy of nomogram2. Results Lasso Cox regression showed that A-SII score, number of organ metastases, tumor size, chemotherapy, targeted therapy, Neutrophil-to-albumin ratio, and lactate dehydrogenase were independent prognostic factors for the OS of advanced pancreatic carcinoma patients. The C-index and ROC curve of the nomogram2 are better than the nomogram1. Subsequently, the DCA and calibration curve of the nomogram2 demonstrate excellent performance. Conclusion The nomogram based on the A-SII score and other independent prognostic factors determined by Lasso Cox regression can accurately predict the OS of patients suffering from advanced pancreatic carcinoma.
Collapse
Affiliation(s)
- Min Xu
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, China
- Department of General Surgery, The Affiliated Yancheng First Hospital of Nanjing University Medical School, Yancheng, China
| | - Yu Long
- Department of Clinical Laboratory, The Affiliated Yancheng First Hospital of Nanjing University Medical School, Yancheng, China
| | - Peisheng Chen
- Department of General Surgery, The Affiliated Yancheng First Hospital of Nanjing University Medical School, Yancheng, China
| | - Ang Li
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, China
| | - Jian Xin
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, China
| | - Yonghua Xu
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, China
- Department of General Surgery, The Affiliated Yancheng First Hospital of Nanjing University Medical School, Yancheng, China
| |
Collapse
|
11
|
Ma Y, Jiang Z, Wang Y, Pan L, Liu K, Xia R, Yuan L, Cheng X. Tongue coating microbiota-based machine learning for diagnosing digestive system tumours. J Oral Microbiol 2025; 17:2487645. [PMID: 40206097 PMCID: PMC11980229 DOI: 10.1080/20002297.2025.2487645] [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: 12/14/2024] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
Background Digestive system tumours (DSTs) often diagnosed late due to nonspecific symptoms. Non-invasive biomarkers are crucial for early detection and improved outcomes. Patients and Methods We collected tongue coating samples from 710 patients diagnosed with DST and 489 healthy controls (HC) from April 2023, to December 2023. Microbial composition was analyzed using 16S rRNA sequencing, and five machine learning algorithms were applied to assess the diagnostic potential of tongue coating microbiota. Results Alpha diversity analysis showed that the microbial diversity in the tongue coating was significantly increased in DST patients. LEfSe analysis identified DST-enriched genera Alloprevotella and Prevotella, contrasting with HC-dominant taxa Neisseria, Haemophilus, and Porphyromonas (LDA >4). Notably, when comparing each of the four DST subtypes with the HC group, the proportion of Haemophilus in the HC group was significantly higher, and it was identified as an important feature for distinguishing the HC group. Machine learning validation demonstrated superior diagnostic performance of the Extreme Gradient Boosting (XGBoost) model, achieving an AUC of 0.926 (95% CI: 0.893-0.958) in internal validation, outperforming the other four machine learning models. Conclusion Tongue coating microbiota shows promise as a non-invasive biomarker for DST diagnosis, supported by robust machine learning models.
Collapse
Affiliation(s)
- Yubo Ma
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhengchen Jiang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yanan Wang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Libin Pan
- Department of Pharmacy, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Kang Liu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruihong Xia
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yuan
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Department of Integrated Chinese and Western Medicine, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
12
|
Muaddi H, Glasgow A, Abou Chaar MK, Habermann E, Starlinger P, Warner S, Smoot R, Kendrick M, Truty M, Thiels CA. The Influence of Area Deprivation Index on Surgical Outcomes in Pancreas Cancer. J Surg Oncol 2025; 131:827-833. [PMID: 39628071 DOI: 10.1002/jso.28002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 05/31/2025]
Abstract
BACKGROUND Surgical resection for pancreas ductal adenocarcinoma (PDAC) remains the mainstay of treatment. Regardless of enhanced survival rates, disparities in patient outcomes and experiences driven by the social determinants of health (SDH) persist. The area deprivation index (ADI) comprises 17 indicators related to education, employment, housing-quality, and poverty. We sought to examine the impact of ADI on surgical outcomes of PDAC patients. METHODS Patients who underwent pancreatoduodenectomy or distal pancreatectomy for PDAC between January 2011 and December 2022 were identified. ADI was calculated using patient zip codes and categorized into quartiles, with the highest indicating the most marginalized. The primary outcome was loss to follow-up after surgical resection. To account for confounders, a competing risk multivariable regression analysis was used. RESULTS A total of 1001 patients had a mean age of 66.6 (±9.64), with 46.3% (n = 463) of patients being female. A majority, 94.6% (n = 947), identified as white, and 64.1% (n = 641) had at least a college degree. The median length of follow-up after surgery was 1.8 years (interquartile range: 0.9-3.5). Multivariable analysis adjusting for competing risk of death, showed that patients who are least marginalized are 1.57 times more likely to have their follow-up than those most marginalized (hazard ratio: 1.57, 95% confidence interval: 1.08-2.29, p = 0.017). CONCLUSION SDH impact many aspects of patient's care including a higher risk of loss to follow-up for marginalized patients after surgery for PDAC. Future efforts should seek to identify and lower barriers faced by marginalized patients with system-level changes to ensure equitable access.
Collapse
Affiliation(s)
- Hala Muaddi
- Department of Surgery, Mayo Clinic Rochester, Rochester, Minnesota, USA
| | - Amy Glasgow
- Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Elizabeth Habermann
- Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Susanne Warner
- Department of Surgery, Mayo Clinic Rochester, Rochester, Minnesota, USA
| | - Rory Smoot
- Department of Surgery, Mayo Clinic Rochester, Rochester, Minnesota, USA
| | - Michael Kendrick
- Department of Surgery, Mayo Clinic Rochester, Rochester, Minnesota, USA
| | - Mark Truty
- Department of Surgery, Mayo Clinic Rochester, Rochester, Minnesota, USA
| | | |
Collapse
|
13
|
Hu K, Zhao X, Zhang N, Ma J, Zhang R, Lu Z, Wu W, Ji Y, Li X. Effect of tumor microenvironment in pancreatic cancer on the loss of β-cell mass: implications for type 3c diabetes. J Gastroenterol 2025; 60:512-525. [PMID: 39760782 DOI: 10.1007/s00535-024-02204-w] [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: 08/14/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND To explore the complex interactions between the tumor microenvironment (TME) of pancreatic ductal adenocarcinoma (PDAC) and the loss of β-cell mass, further elucidating the mechanisms of type 3c diabetes mellitus (T3cDM) onset. METHODS Single-cell RNA sequencing was employed to analyze the PDAC TME, identifying cell interactions and gene expression changes of endocrine cells. Pathological changes and paraneoplastic islets were assessed in the proximal paratumor (PP) and distal paratumor (DP). Fractional β-cell area and islet density were compared among normal pancreas from donors and paraneoplastic tissues from non-diabetes mellitus (NDM) and T3cDM patients. TUNEL staining, RT-qPCR and CCK8 assay were applied to demonstrate the β-cell apoptosis. RESULTS Tumor cells, immune cells and fibroblasts could interact with endocrine cells, and apoptotic pathways were activated in endocrine cells of the PP. The PDAC TME was characterized by marked inflammation, sever fibrosis and atrophy. The islets in the PP had lower fractional β-cell area (0.68 ± 0.65% vs. 0.86 ± 1.02%, P = 0.037) and islet density (0.54 ± 0.42 counts/mm2 vs. 0.83 ± 0.90 counts/mm2, P = 0.001) compared to those in the DP. The PDAC TME in T3cDM exerted a more significant impact on the paraneoplastic islets compared to NDM. Moreover, β-cell apoptosis was markedly increased in the PP compared to the DP in PDAC patients without diabetes, particularly in smaller islets. Apoptosis-related genes were highly expressed in INS-1E cells exposed to PANC-1 medium. CONCLUSION Our research revealed that the PDAC TME is usually accompanied by some pathological changes, including inflammation, fibrosis, and atrophy. These pathological changes are related to a reduction in β-cell mass and trigger the development of T3cDM.
Collapse
Affiliation(s)
- Ke Hu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Xuelian Zhao
- Department of Pathology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Na Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Jing Ma
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Ruonan Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Zhiqiang Lu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
| | - Wenchuan Wu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
| | - Xiaomu Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
14
|
Wu Y, Zhang C, Huang J, Chen Q, Zhang Y, Liu F, Xu D, Jiang K, Shi R, Chen M, Yuan H. Integrated analysis of scRNA-seq and bulk RNA-seq data identifies BHLHE40 as a key gene in pancreatic cancer progression and gemcitabine resistance. Semin Oncol 2025; 52:152338. [PMID: 40250076 DOI: 10.1016/j.seminoncol.2025.152338] [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: 02/17/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 04/20/2025]
Abstract
OBJECTIVE Pancreatic cancer is characterized by its high mortality rate and short survival periods, and novel therapeutic targets and tailor personalized strategies are urgently needed. In this study, we aim to investigate the molecular mechanisms underlying pancreatic ductal adenocarcinoma (PDAC) progression and chemoresistance, with a focus on identifying novel therapeutic targets. METHODS Multiomics approaches were integrated to identify novel actionable targets for PDAC. Public datasets such as TCGA and GEO were utilized to investigate the relationship between gene expression and clinical outcomes. Functional enrichment, cell-cell communication, and metabolic pathway analyses were performed to reveal PDAC heterogeneity and therapeutic resistance mechanisms. RESULTS BHLHE40 was identified as a hub gene linked to high-CNV PDAC cells, Gemcitabine resistance, and poor prognosis in PDAC. High BHLHE40 expression is significantly correlated with immunosuppressive tumor microenvironment (TME) features such as reduced CD8+ T infiltration, TCR richness, and lower tumor mutational burden (TMB). ChIP-seq data analysis confirmed BHLHE40 could directly bind to the SAT1 promoter, establishing a transcriptional axis promoting chemoresistance. Single-cell RNA-seq analysis further revealed that the BHLHE40+/SAT1+ subpopulation cells are resistant to Gemcitabine in PDAC. CONCLUSIONS BHLHE40 is significantly correlated with PDAC malignancy and chemoresistance via SAT1 regulation and immune evasion. Targeting BHLHE40 may sensitize PDACs to Gemcitabine and facilitate personalized treatment for BHLHE40+ PDAC patients.
Collapse
Affiliation(s)
- Yang Wu
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chun Zhang
- Department of Gastroenterology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiacheng Huang
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qun Chen
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yufeng Zhang
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fengyuan Liu
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dong Xu
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kuirong Jiang
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mengxing Chen
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Yuan
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
15
|
Shen J, Li Q, Li L, Lu T, Han J, Xie Z, Wang P, Cao Z, Zeng M, Zhou J, Yu T, Xu Y, Sun H. Contrast-enhanced MRI-based intratumoral heterogeneity assessment for predicting lymph node metastasis in resectable pancreatic ductal adenocarcinoma. Insights Imaging 2025; 16:76. [PMID: 40159327 PMCID: PMC11955437 DOI: 10.1186/s13244-025-01956-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/17/2025] [Indexed: 04/02/2025] Open
Abstract
OBJECTIVES To develop and validate a contrast-enhanced MRI-based intratumoral heterogeneity (ITH) model for predicting lymph node (LN) metastasis in resectable pancreatic ductal adenocarcinoma (PDAC). METHODS Lesions were encoded into different habitats based on enhancement ratios at arterial, venous, and delayed phases of contrast-enhanced MRI. Habitat models on enhanced ratio mapping and single sequences, radiomic models, and clinical models were developed for evaluating LN metastasis. The performance of the models was evaluated via different metrics. Additionally, patients were stratified into high-risk and low-risk groups based on an ensembled model to assess prognosis after adjuvant therapy. RESULTS We developed an ensembled radiomics-habitat-clinical (RHC) model that integrates radiomics, habitat, and clinical data for precise prediction of LN metastasis in PDAC. The RHC model showed strong predictive performance, with area under the curve (AUC) values of 0.805, 0.779, and 0.615 in the derivation, internal validation, and external validation cohorts, respectively. Using an optimal threshold of 0.46, the model effectively stratified patients, revealing significant differences in recurrence-free survival and overall survival (OS) (p = 0.004 and p < 0.001). Adjuvant therapy improved OS in the high-risk group (p = 0.004), but no significant benefit was observed in the low-risk group (p = 0.069). CONCLUSION We developed an MRI-based ITH model that provides reliable estimates of LN metastasis for resectable PDAC and may offer additional value in guiding clinical decision-making. CRITICAL RELEVANCE STATEMENT This ensemble RHC model facilitates preoperative prediction of LN metastasis in resectable PDAC using contrast-enhanced MRI. This offers a foundation for enhanced prognostic assessment and supports the management of personalized adjuvant treatment strategies. KEY POINTS MRI-based habitat models can predict LN metastasis in PDAC. Both the radiomics model and clinical characteristics were useful for predicting LN metastasis in PDAC. The RHC models have the potential to enhance predictive accuracy and inform personalized therapeutic decisions.
Collapse
Affiliation(s)
- Junjian Shen
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Qing Li
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Lei Li
- Department of Radiology, Fengyang County People's Hospital, Chuzhou, China
| | - Tianyu Lu
- Department of Radiology, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jun Han
- Department of Radiology, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Zongyu Xie
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Peng Wang
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi, P.R. China
| | - Zirui Cao
- Department of Research Center, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Jianjun Zhou
- Department of Radiology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen Municipal Clinical Research Center for Medical Imaging, Fujian Province Key Clinical Specialty for Medical Imaging, Xiamen Key Laboratory of Clinical Transformation of Imaging Big Data and Artificial Intelligence, Xiamen, China
| | - Tianzhu Yu
- Department of Interventional Radiology, Zhongshan Hospital, Shanghai, China
| | - Yaolin Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haitao Sun
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China.
| |
Collapse
|
16
|
Shen J, Wang X, Yu K, Liu K, Wang X, Sun H, Zhou J, Zeng M. Correlation of MRI characteristics with KRAS mutation status in pancreatic ductal adenocarcinoma. Abdom Radiol (NY) 2025:10.1007/s00261-025-04888-x. [PMID: 40156607 DOI: 10.1007/s00261-025-04888-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/26/2025] [Accepted: 03/10/2025] [Indexed: 04/01/2025]
Abstract
PURPOSE To investigate MRI features associated with KRAS mutation status in PDAC and their clinical implications. MATERIALS AND METHODS In our study, 1474 patients pathologically confirmed PDAC patients between January 2016 and December 2023 were evaluated. Patients with genetic testing (KRAS mutation status) and MRI examination were enrolled and grouped as KRAS-mutated PDAC and non-KRAS-mutated PDAC. Contrast-enhanced MRI features, clinicopathologic findings, and prognosis were compared between two groups. RESULTS A total of 308 surgically confirmed PDAC patients (median age, 67 years [IQR, 59, 72]; 183 male and 125 female) with genetic testing data were included, of which 258 had KRAS-mutated PDAC and 50 had non-KRAS-mutated PDAC. KRAS-mutated PDAC demonstrated distinct clinicopathological characteristics, including higher rates of diabetes (OR, 2.450, 95% CI, 1.151-5.212, P = 0.020), pathological peripheral nerve infiltration (OR, 2.296, 95% CI, 1.083-4.867, P = 0.030), and pN stage (OR, 2.006, 95% CI, 1.012-3.976, P = 0.046). The 1-, 3-, 5-year OS rate was worse for KRAS-mutated PDAC (89.9%, 45.4%, 23.2% vs. 95.1%, 60.4% 60.4%, P = 0.045). Rim enhancement (OR = 2.039, 95% CI: 1.053, 3.951, P = 0.035) and larger tumor size (OR = 3.286, 95% CI: 1.523, 7.089, P = 0.002) were identified as distinctive MRI features for KRAS-mutated PDAC. CONCLUSION KRAS-mutated PDAC presents unique clinical and pathological features and is associated with poorer prognosis. Rim enhancement and larger tumor size on MRI were identified as features associated with KRAS-mutated PDAC.
Collapse
Affiliation(s)
- Junjian Shen
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xingxing Wang
- , Department of Pathology, Zhongshan Hospital, Fudan University, PR China
| | - Keqin Yu
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kai Liu
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaolin Wang
- , Department of Interventional Radiology, Zhongshan Hospital, Fudan university, PR China
| | - Haitao Sun
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jianjun Zhou
- Department of Radiology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, PR China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Ohtsubo K, Sato S, Sakaguchi H, Kotani H, Nishiyama A, Yamashita K, Yano S, Toshima F, Inoue D, Gabata T, Ikeda H, Watanabe A, Notohara K, Fujisawa T, Nakamura Y, Yoshino T, Miyake K, Miwa K, Takeuchi S. Case Report: Medullary carcinoma of the pancreas with MLH1 promoter hypermethylation, induced deficient mismatch repair, successfully treated with an immune checkpoint inhibitor. Front Oncol 2025; 15:1551038. [PMID: 40236650 PMCID: PMC11997870 DOI: 10.3389/fonc.2025.1551038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
We report the case of a 75-year-old woman with a pancreatic body mass. Pathological findings from endoscopic ultrasonography-guided fine-needle aspiration revealed medullary carcinoma of the pancreas (MCP). Deficient mismatch repair (dMMR) and high microsatellite instability (MSI-H) were identified through immunohistochemistry and next generation sequencing, respectively. While immunohistochemistry suggested MLH1 abnormality, no MLH1 mutation was; hypermethylation of the MLH1 promoter was later confirmed via bisulfite sequencing. The patient initially received nab-paclitaxel plus gemcitabine, achieving tumor shrinkage. Upon tumor regrowth, she was treated with the anti-programmed cell death-1 immune checkpoint inhibitor (ICI) pembrolizumab, which resulted in significant tumor reduction. This is the first case report of MCP with dMMR/MSI-H due to MLH1 promoter hypermethylation, effectively treated with an ICI.
Collapse
Affiliation(s)
- Koushiro Ohtsubo
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Shigeki Sato
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Hiroyuki Sakaguchi
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Hiroshi Kotani
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Akihiro Nishiyama
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Kaname Yamashita
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Seiji Yano
- Department of Respiratory Medicine, Kanazawa University Hospital, Kanazawa, Japan
| | - Fumihito Toshima
- Department of Radiology, Kanazawa University Hospital, Kanazawa, Japan
| | - Dai Inoue
- Department of Radiology, Kanazawa University Hospital, Kanazawa, Japan
| | - Toshifumi Gabata
- Department of Radiology, Kanazawa University Hospital, Kanazawa, Japan
| | - Hiroko Ikeda
- Division of Human Pathology, Kanazawa University Hospital, Kanazawa, Japan
| | - Atsushi Watanabe
- Division of Clinical Genetics, Kanazawa University Hospital, Kanazawa, Japan
| | - Kenji Notohara
- Department of Anatomic Pathology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Takao Fujisawa
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- International Research Promotion Office, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshiaki Nakamura
- International Research Promotion Office, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- International Research Promotion Office, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kunio Miyake
- Department of Epidemiology and Environmental Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Japan
| | - Kazuhiro Miwa
- Internal Medicine, Komatsu Municipal Hospital, Komatsu, Japan
| | - Shinji Takeuchi
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| |
Collapse
|
18
|
Cui R, Wang G, Hu R, Wang Y, Mu H, Song Y, Chen B, Jiang X. Prognostic and immunotherapeutic potential of disulfidptosis-associated signature in pancreatic cancer. Front Immunol 2025; 16:1568976. [PMID: 40207217 PMCID: PMC11979277 DOI: 10.3389/fimmu.2025.1568976] [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: 01/31/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Disulfidptosis is a newly discovered formation of programmed cell death. However, the significance of disulfidptosis in pancreatic adenocarcinoma remains unclear. Our investigation aims to elucidate the significance of disulfidptosis in pancreatic ductal adenocarcinoma by integrating diverse datasets, including bulk RNA sequencing data, microarray profiles, single-cell transcriptome profiles, spatial transcriptome data, and biospecimens. Utilizing various bioinformatics tools, we screened disulfidptosis-related genes based on single-cell RNA sequencing profiles, subsequently validating them through enrichment analysis. An 8-gene disulfidptosis-related prognostic signature was established by constructing massive LASSO-Cox regression models and validated by multiple external PDAC cohorts. Evaluation methods, such as Kaplan-Meier curves, ROC curves, time-dependent ROC curves, and decision curve analysis, were employed to assess the prognostic signature's reliability. High disulfidptosis-related scores were associated with a poorer prognosis and diminished sensitivity to immune checkpoint blockade. Further investigation uncovered that the potential components of elevated DPS involve malignant tumor hallmarks, extensive interactions between myCAFs and tumor cells, and the exclusion of immune cells. Cell-cell communication analysis highlighted myCAFs' role in signaling, potentially influencing tumor cells towards increased malignancy through collagen, laminin, and FN1 signaling networks. Spatial transcriptome analysis confirmed the crosstalk between myCAFs and tumor cells. Biospecimens including 20 pairs of PDAC samples and adjacent normal tissues further demonstrated the robustness of DPS and its correlation with CAF markers. In conclusion, our study introduces a novel disulfidptosis-related signature with high efficacy in patient risk stratification, which has the ability to predict the sensitivity to immune checkpoint blockade.
Collapse
Affiliation(s)
- Ran Cui
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Gaoming Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Renhao Hu
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yongkun Wang
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huiling Mu
- Department of Biobank, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanxiang Song
- Department of Biobank, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bo Chen
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaohua Jiang
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
19
|
Wang QW, Zou WB, Masson E, Férec C, Liao Z, Chen JM. Genetics and clinical implications of SPINK1 in the pancreatitis continuum and pancreatic cancer. Hum Genomics 2025; 19:32. [PMID: 40140953 PMCID: PMC11948977 DOI: 10.1186/s40246-025-00740-x] [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: 09/02/2024] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Serine peptidase inhibitor, Kazal type 1 (SPINK1), a 56-amino-acid protein in its mature form, was among the first pancreatic enzymes to be extensively characterized biochemically and functionally. Synthesized primarily in pancreatic acinar cells and traditionally known as pancreatic secretory trypsin inhibitor, SPINK1 protects the pancreas by inhibiting prematurely activated trypsin. Since 2000, interest in SPINK1 has resurged following the discovery of genetic variants linked to chronic pancreatitis (CP). This review provides a historical overview of SPINK1's discovery, function, and gene structure before examining key genetic findings. We highlight three variants with well-characterized pathogenic mechanisms: c.-4141G > T, a causative enhancer variant linked to the extensively studied p.Asn34Ser (c.101A > G), which disrupts a PTF1L-binding site within an evolutionarily conserved HNF1A-PTF1L cis-regulatory module; c.194 + 2T > C, a canonical 5' splice site GT > GC variant that retains 10% of wild-type transcript production; and an Alu insertion in the 3'-untranslated region, which causes complete loss of function by forming extended double-stranded RNA structures with pre-existing Alu elements in deep intronic regions. We emphasize the integration of a full-length gene splicing assay (FLGSA) with SpliceAI's predictive capabilities, establishing SPINK1 the first disease gene for which the splicing impact of all possible coding variants was prospectively determined. Findings from both mouse models and genetic association studies support the sentinel acute pancreatitis event (SAPE) model, which explains the progression from acute pancreatitis to CP. Additionally, SPINK1 variants may contribute to an increased risk of pancreatic ductal adenocarcinoma (PDAC). Finally, we discuss the therapeutic potential of SPINK1, particularly through adeno-associated virus type 8 (AAV8)-mediated overexpression of SPINK1 as a strategy for treating and preventing pancreatitis, and highlight key areas for future research.
Collapse
Affiliation(s)
- Qi-Wen Wang
- Department of Gastroenterology, Changhai Hospital; National Key Laboratory of Immunity and Inflammation, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Wen-Bin Zou
- Department of Gastroenterology, Changhai Hospital; National Key Laboratory of Immunity and Inflammation, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Emmanuelle Masson
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 29200, Brest, France
- Service de Génétique Médicale et de Biologie de la Reproduction, CHU Brest, Brest, France
| | - Claude Férec
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 29200, Brest, France
| | - Zhuan Liao
- Department of Gastroenterology, Changhai Hospital; National Key Laboratory of Immunity and Inflammation, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
- Shanghai Institute of Pancreatic Diseases, Shanghai, China.
| | - Jian-Min Chen
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 29200, Brest, France.
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 22 Avenue Camille Desmoulins, 29238, Brest, France.
| |
Collapse
|
20
|
Hao T, Li Y, Ren Q, Zeng Y, Gao L, Zhu W, Liang J, Lin Y, Hu J, Yan G, Sun S, Cai J. circ-1584 selectively promotes the antitumor activity of the oncolytic virus M1 on pancreatic cancer. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200919. [PMID: 39866243 PMCID: PMC11760297 DOI: 10.1016/j.omton.2024.200919] [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: 06/24/2024] [Revised: 10/25/2024] [Accepted: 12/13/2024] [Indexed: 01/28/2025]
Abstract
Pancreatic cancer is among the most challenging tumors to treat, and due to its immune tolerance characteristics, existing immunotherapy methods are not effective in alleviating the disease. Oncolytic virus therapy, a potential new strategy for treating pancreatic cancer, also faces the limitation of being ineffective when used alone. Elucidating the key host endogenous circular RNAs (circRNAs) involved in M1 virus-mediated killing of pancreatic ductal adenocarcinoma (PDAC) cells may help overcome this limitation. Here, we report that the oncolytic virus M1, a nonpathogenic alphavirus, exhibits different cell viability-inhibitory effects on different pancreatic cancer cells in the clinical stage. Through high-throughput circRNA sequencing, we found that circRNA expression varies among these cells. Further gain-of-function and loss-of-function experiments have shown that circ-1584 can selectively enhance the anti-pancreatic cancer effects of the M1 virus in vitro and in vivo. Additionally, circ-1584 may negatively regulate miR-578 to modulate the anti-pancreatic cancer effects of the M1 virus. Our findings lay the foundation for using circRNA as an adjuvant to enhance the M1 virus efficacy against pancreatic cancer.
Collapse
Affiliation(s)
- Taofang Hao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yuanyuan Li
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qianyao Ren
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ying Zeng
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Leyi Gao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wenbo Zhu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jiankai Liang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yuan Lin
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital-Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Human Microbiome and Elderly Chronic Diseases, Ministry of Education, Beijing, China
| | - Jun Hu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Guangmei Yan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shuxin Sun
- Pancreatic Center, Guangdong Provincial People’s Hospital, Guangzhou, China
| | - Jing Cai
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Molecular Biology and Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
21
|
Xu K, Wang X, Zhou C, Zuo J, Zeng C, Zhou P, Zhang L, Gao X, Wang X. Synergic value of 3D CT-derived body composition and triglyceride glucose body mass for survival prognostic modeling in unresectable pancreatic cancer. Front Nutr 2025; 12:1499188. [PMID: 40177184 PMCID: PMC11961436 DOI: 10.3389/fnut.2025.1499188] [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: 09/23/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Background Personalized and accurate survival risk prognostication remains a significant challenge in advanced pancreatic ductal adenocarcinoma (PDAC), despite extensive research on prognostic and predictive markers. Patients with PDAC are prone to muscle loss, fat consumption, and malnutrition, which is associated with inferior outcomes. This study investigated the use of three-dimensional (3D) anthropometric parameters derived from computed tomography (CT) scans and triglyceride glucose-body mass index (TyG-BMI) in relation to overall survival (OS) outcomes in advanced PDAC patients. Additionally, a predictive model for 1 year OS was developed based on body components and hematological indicators. Methods A retrospective analysis was conducted on 303 patients with locally advanced PDAC or synchronous metastases undergoing first-line chemotherapy, all of whom had undergone pretreatment abdomen-pelvis CT scans. Automatic 3D measurements of subcutaneous and visceral fat volume, skeletal muscle volume, and skeletal muscle density (SMD) were assessed at the L3 vertebral level by an artificial intelligence assisted diagnosis system (HY Medical). Various indicators including TyG-BMI, nutritional indicators [geriatric nutritional risk index (GNRI) and prealbumin], and inflammation indicators [(C-reactive protein (CRP) and neutrophil to lymphocyte ratio (NLR)] were also recorded. All patients underwent follow-up for at least 1 year and a dynamic nomogram for personalized survival prediction was constructed. Results We included 211 advanced PDAC patients [mean (standard deviation) age, 63.4 ± 11.2 years; 89 women (42.2) %)]. Factors such as low skeletal muscle index (SMI) (P = 0.011), high visceral to subcutaneous adipose tissue area ratio (VSR) (P < 0.001), high visceral fat index (VFI) (P < 0.001), low TyG-BMI (P = 0.004), and low prealbumin (P = 0.001) were identified as independent risk factors associated with 1 year OS. The area under the curve of the established dynamic nomogram was 0.846 and the calibration curve showed good consistency. High-risk patients (> 211.9 points calculated using the nomogram) had significantly reduced survival rates. Conclusion In this study, the proposed nomogram model (with web-based tool) enabled individualized prognostication of OS and could help to guide risk-adapted nutritional treatment for patients with unresectable PDAC or synchronous metastases.
Collapse
Affiliation(s)
- Kangjing Xu
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinbo Wang
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Changsheng Zhou
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Junbo Zuo
- Department of General Surgery, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Chenghao Zeng
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Pinwen Zhou
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Li Zhang
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xuejin Gao
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinying Wang
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
22
|
Breaza GM, Closca RM, Cindrea AC, Hut FE, Cretu O, Sima LV, Rakitovan M, Zara F. Immunohistochemical Evaluation of the Tumor Immune Microenvironment in Pancreatic Ductal Adenocarcinoma. Diagnostics (Basel) 2025; 15:646. [PMID: 40075893 PMCID: PMC11899021 DOI: 10.3390/diagnostics15050646] [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: 02/07/2025] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma is an aggressive neoplasm with a complex carcinogenesis process that must be understood through the interactions between tumor cells and tumor microenvironment cells. Methods: This study was retrospective with a chronological extension period of 16 years and included 56 cases of pancreatic ductal adenocarcinoma. This study identified, quantified, and correlated the cells of the tumor immune microenvironment in pancreatic ductal adenocarcinoma with major prognostic factors as well as overall survival, using an extensive panel of immunohistochemical markers. Results: Three tumor immunotypes were identified: subtype A (hot immunotype), subtype B (intermediate immunotype), and subtype C (cold immunotype). Patients with immunotype C exhibit considerably higher rates of both pancreatic fistulas and acute pancreatitis. Immunotypes B and C significantly increased the risk of this complication by factors of 3.68 (p = 0.002) and 3.94 (p = 0.001), respectively. The estimated probabilities of fistula formation for each immunotype are as follows: 2.5% for immunotype A, 25% for immunotype B, and 28% for immunotype C. There was a statistically significant difference in median survival times according to tumor immunotype (p < 0.001). Specifically, patients with immunotype C tumors had a median survival time of only 120.5 days, compared to 553.5 days for those with immunotype A and 331.5 for immunotype B tumors. Conclusions: The identification of the immunotype of pancreatic ductal adenocarcinoma can be a predictive factor for the occurrence of complications such as pancreatic fistula as well as for overall survival.
Collapse
Affiliation(s)
- Gelu Mihai Breaza
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Raluca Maria Closca
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Department of Pathology, Emergency City Hospital, 300254 Timisoara, Romania
| | - Alexandru Cristian Cindrea
- Department of Surgery, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania;
- Emergency Department, Emergency Clinical Municipal Hospital, 300079 Timisoara, Romania
| | - Florin Emil Hut
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
- Center for Hepato-Bilio-Pancreatic Surgery, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania
| | - Octavian Cretu
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Laurentiu Vasile Sima
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Marina Rakitovan
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Oro-Maxillo-Facial Surgery Clinic, Emergency City Hospital, 300062 Timisoara, Romania
| | - Flavia Zara
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Department of Pathology, Emergency City Hospital, 300254 Timisoara, Romania
| |
Collapse
|
23
|
Guo Y, Li Y, Li J, Cai H, Liu K, Duan D, Zhang W, Han G, Zhao Y. Controlled Inflammation Drives Neutrophil-Mediated Precision Drug Delivery in Heterogeneous Tumors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411307. [PMID: 39799561 PMCID: PMC11923894 DOI: 10.1002/advs.202411307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/05/2024] [Indexed: 01/15/2025]
Abstract
Tumor heterogeneity remains a formidable obstacle in targeted cancer therapy, often leading to suboptimal treatment outcomes. This study presents an innovative approach that harnesses controlled inflammation to guide neutrophil-mediated drug delivery, effectively overcoming the limitations imposed by tumor heterogeneity. By inducing localized inflammation within tumors using lipopolysaccharide, it significantly amplify the recruitment of drug-laden neutrophils to tumor sites, irrespective of specific tumor markers. This strategy not only enhances targeted drug delivery but also triggers the release of neutrophil extracellular traps, further potentiating the anti-tumor effect. Crucially, this study demonstrates that potential systemic inflammatory responses can be effectively mitigated through neutrophil transfusion, ensuring the safety and clinical viability of this approach. In a murine breast cancer model, the method significantly impedes tumor growth compared to conventional treatments. This work offers a versatile strategy for precise drug delivery across diverse tumor types. The findings pave the way for more effective and broadly applicable cancer treatments, potentially addressing the long-standing challenge of tumor heterogeneity.
Collapse
Affiliation(s)
- Yunfei Guo
- Department of RadiologyThe Second Hospital of Tianjin Medical UniversityTianjin300211P. R. China
| | - Yiming Li
- Department of RadiologyThe Second Hospital of Tianjin Medical UniversityTianjin300211P. R. China
| | - Jianmin Li
- Tianjin Institute of UrologyThe Second Hospital of Tianjin Medical UniversityTianjin300211P. R. China
| | - Haoran Cai
- Department of RadiologyThe Second Hospital of Tianjin Medical UniversityTianjin300211P. R. China
| | - Kangkang Liu
- Tianjin Institute of UrologyThe Second Hospital of Tianjin Medical UniversityTianjin300211P. R. China
| | - Dengyi Duan
- Department of RadiologyThe Second Hospital of Tianjin Medical UniversityTianjin300211P. R. China
| | - Wenyi Zhang
- Department of RadiologyThe Second Hospital of Tianjin Medical UniversityTianjin300211P. R. China
| | - Gang Han
- Biochemistry and Molecular BiotechnologyUniversity of Massachusetts Chan Medical SchoolWorcesterMA01605USA
| | - Yang Zhao
- Department of RadiologyThe Second Hospital of Tianjin Medical UniversityTianjin300211P. R. China
- Tianjin Institute of UrologyThe Second Hospital of Tianjin Medical UniversityTianjin300211P. R. China
- Biochemistry and Molecular BiotechnologyUniversity of Massachusetts Chan Medical SchoolWorcesterMA01605USA
| |
Collapse
|
24
|
Tsilimigras DI, Woldesenbet S, Chatzipanagiotou OP, Iyer S, Pawlik TM. Long-term lorazepam use may be associated with worse long-term outcomes among patients with pancreatic adenocarcinoma. Surgery 2025; 179:108794. [PMID: 39304446 DOI: 10.1016/j.surg.2024.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Lorazepam recently has been reported to alter the tumor microenvironment of pancreatic adenocarcinoma in a murine model. We sought to evaluate whether the use of lorazepam was associated with worse outcomes among patients with pancreatic adenocarcinoma. METHODS Medicare beneficiaries diagnosed with stage I-IV pancreatic adenocarcinoma between 2013 and 2019 were identified from the Surveillance, Epidemiology and End Results-Medicare database. The association of lorazepam prescription relative to overall survival and recurrence-free survival was examined. RESULTS Among 2,810 patients with stage I-III and 10,181 patients with stage IV pancreatic adenocarcinoma, a total of 133 (4.7%) and 444 individuals (4.4%) had a lorazepam prescription before disease diagnosis, respectively. Although the overall lorazepam group had comparable 5-year overall survival (15.0% vs 14.2%, P = .20) and recurrence-free survival (12.7% vs 10.9%, P = .42) with the no-lorazepam group after pancreatic adenocarcinoma resection, individuals with long-term lorazepam prescription (>30 days) had worse 5-year overall survival (9.0% vs 21.0%, P = .02) and recurrence-free survival (6.4% vs 17.1%, P = .009) compared with short-term lorazepam users (≤30 days). Similarly, among patients with metastatic pancreatic adenocarcinoma, individuals with a long-term lorazepam prescription had worse 1-year overall survival (9.7% vs 15.9%, P = .02) compared with patients who had short-term lorazepam prescriptions. On multivariable analysis, long-term lorazepam prescription was independently associated with overall survival among patients with resectable (hazard ratio, 1.82; 95% confidence interval, 1.22-2.74) and metastatic pancreatic adenocarcinoma (hazard ratio, 1.24; 95% confidence interval, 1.02-1.51). CONCLUSION Long-term lorazepam prescription was associated with worse long-term outcomes among patients who underwent resection for pancreatic adenocarcinoma and patients with metastatic pancreatic adenocarcinoma. These data support the need for further large scale studies to confirm a potential harmful effect of lorazepam among patients with pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Diamantis I Tsilimigras
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH.
| | - Selamawit Woldesenbet
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH
| | - Odysseas P Chatzipanagiotou
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH
| | - Sidharth Iyer
- College of Medicine, The Ohio State University, Columbus, OH
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH.
| |
Collapse
|
25
|
Yay F, Yıldırım HÇ, Kuş F, Yalçın Ş. Dynamine 3 as a diagnostic and prognostic biomarker in pancreatic cancer: Implications for early detection and targeted therapy. Biomarkers 2025; 30:147-166. [PMID: 39847517 DOI: 10.1080/1354750x.2025.2458104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Dynamins are defined as a group of molecules with GTPase activity. Among them, DNM3 has gained recognition in oncology for its tumor suppressor role. Based on this, the aim of this study is to investigate the effects of the DNM3 gene in patients diagnosed with pancreatic cancer using bioinformatics databases. MATERIALS AND METHODS For differential gene expression analysis, TCGA TARGET GTEx study on the UCSC Xena and GEO datasets were utilized; for the analysis of changes in gene expression according to clinical and pathological characteristics, UALCAN was employed; for Overall Survival (OS) analysis, Kaplan-Meier Plotter was used; for gene alteration analysis, cBioPortal was utilized; for immune cell infiltration analysis, Tumor Immune Estimation Resource (TIMER) and TIMER2.0 were employed; for enrichment analyses Enrichr was used; for Gene Set Correlation Enrichment Analysis Gscore was used on GSE15471; for essentiality of DNM3 gene in pancratic cancer cell lines DepMap was used; and for the detection of miRNAs, miRDB was utilized; ENCORI was used for gene-miRNA correlation and miRNA prognosis analyses. RESULTS In the pancreatic adenocarcinoma (PAAD) cohort, DNM3 gene expression was higher in tumor samples, and there was no significant difference in expression among cancer stages. High levels of DNM3 gene expression were associated with longer OS in PAAD. A weak positive correlation was observed between DNM3 gene expression and B-Cell and CD4+ T Cell infiltrations, while a moderate positive correlation was found with CD8+ T Cell, Macrophage, Neutrophil, and Dendritic Cell infiltrations in TIMER. NK cell by QUANTISEQ, CD 4+ T Cell by TIMER, T cell regulatory (Tregs) by CIBERSORT-ABS infiltrations were positively associated with DNM3 gene expression and decreased risk in prognosis. Common lymphoid progenitor by XCELL and MDSC by TIDE infiltrations were negatively associated with DNM3 gene expression and increased risk of prognosis. Macrophage M1 by QUANTISEQ was positively associated with DNM3 gene expression and increased risk in prognosis. DNM3 gene appears to be associated with various pathways related to inflammation and the immune system. Amplification of the DNM3 gene was detected in 5 out of 175 patients. Enrichment was observed in pathways such as bacterial invasion of epithelial cells, endocytosis, endocrine and other factor-regulated calcium reabsorption, synaptic vesicle cycle, and phospholipase D signaling pathway. According to Gscore, DNM3 gene was associated with Fc epsilon RI signaling pathway, HALLMARK MTORC1 SIGNALING, HALLMARK EPITHELIAL MESENCHYMAL TRANSITION gene sets. According to ENCORI, DNM3 gene was negatively correlated with hsa-miR-203a-3p and increased expression of this miRNA was associated with adverse prognosis in PAAD. CONCLUSIONS The DNM3 gene may play a tumor suppressor role in pancreatic cancer, similar to its role in other malignancies. The contribution of immune cells may also be significant in this effect. However, in vitro studies are needed to elucidate the mechanisms triggered in pancreatic cancer.
Collapse
Affiliation(s)
- Fatih Yay
- Clinical Biochemistry Laboratory, Nigde Omer Halisdemir University Training and Research Hospital, Nigde, Turkey
| | - Hasan Çağrı Yıldırım
- Department of Medical Oncology, Nigde Omer Halisdemir University Training and Research Hospital, Nigde, Turkey
| | - Fatih Kuş
- Faculty of Medicine, Deparment of Medical Oncology, Hacettepe University, Ankara, Turkey
| | - Şuayib Yalçın
- Faculty of Medicine, Deparment of Medical Oncology, Hacettepe University, Ankara, Turkey
| |
Collapse
|
26
|
Zhou SQ, Wan P, Zhang S, Ren Y, Li HT, Ke QH. Programmed cell death 1 inhibitor sintilimab plus S-1 and gemcitabine for liver metastatic pancreatic ductal adenocarcinoma. World J Clin Oncol 2025; 16:98079. [PMID: 39995563 PMCID: PMC11686555 DOI: 10.5306/wjco.v16.i2.98079] [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: 06/17/2024] [Revised: 10/21/2024] [Accepted: 11/25/2024] [Indexed: 12/11/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with poor prognosis. When it metastasizes to the liver, treatment options become particularly limited and challenging. Current treatment options for liver metastatic PDAC are limited, and chemotherapy alone often proves insufficient. Immunotherapy, particularly programmed cell death 1 (PD-1) inhibitors like sintilimab, shows potential efficacy for various cancers but has limited reports on PDAC. This study compares the efficacy and safety of sintilimab plus S-1 and gemcitabine vs S-1 and gemcitabine alone in liver metastatic PDAC. AIM To explore the feasibility and effectiveness of combined PD-1 inhibitor sintilimab and S-1 and gemcitabine (combination group) vs S-1 and gemcitabine used alone (chemotherapy group) for treating liver metastatic pancreatic adenocarcinoma. METHODS Eligible patients were those with only liver metastatic PDAC, an Eastern Cooperative Oncology Group performance status of 0-1, adequate organ and marrow functions, and no prior anticancer therapy. Participants in the combination group received intravenous sintilimab 200 mg every 3 weeks, oral S-1 40 mg/m² twice daily on days 1-14 of a 21-day cycle, and intravenous gemcitabine 1000 mg/m² on days 1 and 8 of the same cycle for up to eight cycles or until disease progression, death, or unacceptable toxicity. Participants in the chemotherapy group received oral S-1 40 mg/m² twice daily on days 1-14 of a 21-day cycle and intravenous gemcitabine 1000 mg/m² on days 1 and 8 of the same cycle for up to eight cycles. Between June 2020 and December 2021, 66 participants were enrolled, with 32 receiving the combination treatment and 34 receiving chemotherapy alone. RESULTS The group receiving the combined therapy exhibited a markedly prolonged median overall survival (18.8 months compared to 10.3 months, P < 0.05) and progression-free survival (9.6 months vs 5.4 months, P < 0.05). compared to the chemotherapy group. The incidence of severe adverse events did not differ significantly between the two groups (P > 0.05). CONCLUSION The combination of PD-1 inhibitor sintilimab with S-1 and gemcitabine demonstrated effectiveness and safety for treating liver metastatic PDAC, meriting further investigation.
Collapse
Affiliation(s)
- Shi-Qiong Zhou
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Peng Wan
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Seng Zhang
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Yuan Ren
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Hong-Tao Li
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| | - Qing-Hua Ke
- Department of Chemoradiotherapy, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei Province, China
| |
Collapse
|
27
|
Leiphrakpam PD, Chowdhury S, Zhang M, Bajaj V, Dhir M, Are C. Trends in the Global Incidence of Pancreatic Cancer and a Brief Review of its Histologic and Molecular Subtypes. J Gastrointest Cancer 2025; 56:71. [PMID: 39992560 DOI: 10.1007/s12029-025-01183-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2025] [Indexed: 02/25/2025]
Abstract
The global burden of pancreatic cancer has more than doubled in recent decades. It is now the sixth leading cause of cancer-related death worldwide, with an estimated 510,922 new cases and 467,409 deaths in 2022. The incidence of the disease continues to rise annually, with projections indicating a 95.4% increase in new cases by 2050, potentially reaching a total of 998,663 new cases globally. The overall five-year survival rate for pancreatic cancer is 10% worldwide, showing only a modest improvement compared to the past decade. The rising trends in the incidence rates are likely to continue as the global population ages and access to healthcare improves. The relatively low survival rate is primarily attributed to late-stage diagnoses and the lack of an effective screening method. Currently, population-based screening for asymptomatic individuals is not recommended, highlighting the importance of identifying and monitoring individuals at high risk for pancreatic cancer. Numerous studies have highlighted the differences in the molecular pathology of pancreatic cancer, underscoring the need for continued research to better understand these differences. The silent progression of the disease, poor prognosis, lack of screening options, and the necessity to improve our comprehension of its molecular characteristics emphasize the critical need for ongoing monitoring of disease trends at the population level. This review article analyses trends in the incidence of pancreatic cancer and its histological subtypes and provides an update on its molecular subtypes.
Collapse
Affiliation(s)
- Premila Devi Leiphrakpam
- Graduate Medical Education, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Surgical Oncology, Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sanjib Chowdhury
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michelle Zhang
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Varnica Bajaj
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mashaal Dhir
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Chandrakanth Are
- Graduate Medical Education, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
- Division of Surgical Oncology, Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
28
|
Frimpong E, Annor E, Bulusu R, Okoro J, Kiros GE, Reams R, Agyare E. Sociodemographic characteristics associated with pancreatic cancer incidence and mortality among Blacks in the United States: a SEER-based study. Am J Cancer Res 2025; 15:705-722. [PMID: 40084357 PMCID: PMC11897636 DOI: 10.62347/gjcx1238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/03/2025] [Indexed: 03/16/2025] Open
Abstract
Pancreatic cancer (PC) is the third leading cause of all cancer-related fatalities and accounts for approximately 3% of cancer cases in the United States. PC survival rates are lower in Blacks compared to other races, and this has been attributed to socioeconomic and genetic factors. In this study, we evaluated sociodemographic and genetic characteristics associated with PC incidence and mortality among Blacks. Data from the SEER 22 registries (2000-2020) were used to calculate the incidence rates and relative survival. County mortality rates from 2017 to 2021 were analyzed. Incidence rate ratios based on gender, age, primary disease site, stage, level of education, and poverty were calculated. Survival analysis was conducted using the Kaplan-Meier method. Mutant gene expression was obtained from the MSK-CHORD tumor registry. Overall, 48,606 Black patients were diagnosed with malignant PC between 2000 and 2020: females (53.53%) and males (46.47%). Both males and females experienced a slight increase in Annual Percent Change (APC) of PC incidence (0.24, 95% CI, -0.02-0.53) and (0.22, 95% CI, -0.05-0.51), respectively, from 2000 to 2020. Males aged 55 to 75 years were most frequently affected. Overall incidence risk from 2000-2020 by age was higher in Black males IRR > 1 (1.18, 95% CI, 1.16-1.21). The most common primary PC site for Black males and females was the head of the pancreas, 49.06% and 49.88%, respectively. By staging, distant PC had the highest frequency in Blacks. Poverty level was associated with PC incidence among females and PC mortality among both males and females. Stage was associated with survival among males with localized and regional PC. The 5-year relative survival was less than 11% across combined PC stages for both sexes. Black males had a relatively lower 5-year survival than Black females in localized (31.7 vs. 37.2%) and distant PC (2.6% vs. 2.90%). Mutant KRAS expression was higher in Black males. PC incidence and mortality were significantly higher in Black males. Our analysis points to the importance of poverty alleviation programs that target females are likely to reduce PC incidence. Furthermore, receiving recommended screening for PC and early-stage diagnostics is important to lower PC mortality.
Collapse
Affiliation(s)
- Esther Frimpong
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, The United States
| | - Eugene Annor
- Department of Internal Medicine, University of Illinois College of Medicine at PeoriaPeoria, Illinois, The United States
| | - Raviteja Bulusu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, The United States
| | - Joy Okoro
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, The United States
| | - Gebre-Egziabher Kiros
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, The United States
| | - Renee Reams
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, The United States
| | - Edward Agyare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, The United States
| |
Collapse
|
29
|
Láinez Ramos-Bossini AJ, Gámez Martínez A, Luengo Gómez D, Valverde-López F, Morillo Gil AJ, González Flores E, Salmerón Ruiz Á, Jiménez Gutiérrez PM, Melguizo C, Prados J. Computed Tomography-Based Sarcopenia and Pancreatic Cancer Survival-A Comprehensive Meta-Analysis Exploring the Influence of Definition Criteria, Prevalence, and Treatment Intention. Cancers (Basel) 2025; 17:607. [PMID: 40002202 PMCID: PMC11853262 DOI: 10.3390/cancers17040607] [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: 11/12/2024] [Revised: 01/16/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Sarcopenia has been associated with poor outcomes in pancreatic cancer (PC). However, published results are heterogeneous in terms of study design, oncological outcomes, and sarcopenia measurements. This meta-analysis aims to evaluate the impact of computed tomography (CT)-based sarcopenia on overall survival (OS) and progression-free survival (PFS) in patients with PC, considering potential confounders such as the CT-based method and thresholds used to define sarcopenia, as well as treatment intention. Methods: We systematically searched databases for observational studies reporting hazard ratios (HRs) for OS and PFS in PC patients stratified by CT-based sarcopenia status. Random-effects models were used to calculate pooled crude and adjusted HRs (cHRs and aHRs, respectively), with subgroup analyses based on sarcopenia measurement methods, cutoff values, sarcopenia prevalence, and treatment intention. Heterogeneity was assessed using the I2 and τ2 statistics, and publication bias was evaluated using funnel plots and Egger's test. Results: Data from 48 studies were included. Sarcopenia was significantly associated with worse OS (pooled cHR = 1.58, 95% CI: 1.38-1.82; pooled aHR = 1.39, 95% CI: 1.16-1.66) and worse PFS (pooled cHR = 1.55, 95% CI: 1.29-1.86; pooled aHR = 1.31, 95% CI: 1.11-1.55). Subgroup analyses revealed significantly different, stronger associations in studies using stricter sarcopenia cutoffs (<50 cm2/m2 for males) and in patients undergoing curative treatments. Heterogeneity was substantial across analyses (I2 > 67%), but with generally low τ2 values (0.01-0.25). Egger's test indicated potential publication bias for OS (p < 0.001), but no significant bias was observed for PFS (p = 0.576). Conclusions: Sarcopenia determined by CT is an independent predictor of poor OS and PFS in PC, but this association varies depending on the cutoff used for its definition as well as on the treatment intention. Therefore, its routine assessment in clinical practice could provide valuable prognostic information, but future research should focus on standardizing sarcopenia assessment methods.
Collapse
Affiliation(s)
- Antonio Jesús Láinez Ramos-Bossini
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
- Advanced Medical Imaging Group (TeCe-22), Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; (C.M.); (J.P.)
| | - Antonio Gámez Martínez
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
| | - David Luengo Gómez
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
- Advanced Medical Imaging Group (TeCe-22), Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
| | - Francisco Valverde-López
- Department of Gastroenterology and Hepatology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain;
| | - Antonio Jesús Morillo Gil
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
| | | | - Ángela Salmerón Ruiz
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
- Advanced Medical Imaging Group (TeCe-22), Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
| | | | - Consolación Melguizo
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; (C.M.); (J.P.)
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - José Prados
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; (C.M.); (J.P.)
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| |
Collapse
|
30
|
Min JH, Yu JI, Kim SH, Kim YK, Kim K, Park HC, Park JO, Hong JY, Lee KT, Lee KH, Lee JK, Park JK, Choi JH, Heo JS, Han IW, Kim H, Shin SH, Yoon SJ, Woo SY. Skeletal Muscle Index Changes on Locoregional Treatment Application After FOLFIRINOX and Survival in Pancreatic Cancer. J Cachexia Sarcopenia Muscle 2025; 16:e16343. [PMID: 39578950 DOI: 10.1002/jcsm.13643] [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: 01/23/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Patients with borderline resectable (BR) or locally advanced pancreatic cancer (LAPC) require complex management strategies. This study evaluated the prognostic significance of the perichemotherapy skeletal muscle index (SMI) and carbohydrate antigen 19-9 (CA 19-9) in patients with BRPC or LAPC treated with FOLFIRINOX. METHODS We retrospectively evaluated 227 patients with BR or LAPC who received at least four cycles of chemotherapy between 2015 and 2020. We analysed chemotherapy response, changes in SMI (ΔSMI, %) on computed tomography (CT) and CA19-9 to determine their impact on progression-free survival (PFS) and overall survival (OS). After the early application of loco-regional treatments (LRT) within 3 months after completing four cycles of chemotherapy, the outcomes were compared between ΔSMI and CA19-9 subgroups. RESULTS Among 227 patients (median age, 60 years; 124 [54.6%] male) with 97 BR and 130 LAPC, 50.7% showed partial response (PR) to chemotherapy, 44.5% showed stable disease and 4.8% showed progressive disease (PD). Post-chemotherapy CA19-9 levels were normalized in 41.0% of patients. The high and low ΔSMI groups (based on the gender-specific cut-off of -8.6% for males and -2.9% for females) comprised 114 (50.2%) and 113 (49.8%) patients, respectively. The high ΔSMI group had poorer survival rates than the low ΔSMI group in both PFS (HR = 1.32, p = 0.05) and OS (HR = 1.74, p = 0.001). Multivariable analysis showed that ΔSMI (high vs. low; PFS, HR = 1.39, p = 0.03; OS, HR = 1.82, p < 0.001) and post-chemotherapy response (PD vs. PR/SD; PFS, HR = 18.69, p < 0.001; OS, HR = 6.19, p < 0.001) were independently associated with both PFS and OS. Additionally, the post-chemotherapy CA19-9 (≥ 37 vs. < 37; HR = 1.48, p = 0.01) was an independent predictor for PFS. Early application of LRT after chemotherapy significantly improved PFS and OS in both ΔSMI groups (all p < 0.05). However, it was not beneficial in the group with high ΔSMI and post-chemotherapy CA19-9 ≥ 37 (PFS, p = 0.39 and OS, p = 0.33). CONCLUSIONS Progressive sarcopenic deterioration after four cycles of chemotherapy was associated with poor survival outcomes in patients with BR or LAPC after FOLFIRINOX. We also investigated the optimal clinical setting for the early application LRTs using the ΔSMI and post-chemotherapy CA 19-9.
Collapse
Affiliation(s)
- Ji Hye Min
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jeong Il Yu
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Seong Hyun Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Young Kon Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kangpyo Kim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hee Chul Park
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Joon Oh Park
- Divisions of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jung Yong Hong
- Divisions of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyu Taek Lee
- Divisions of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kwang Hyuck Lee
- Divisions of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jong Kyun Lee
- Divisions of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Joo Kyung Park
- Divisions of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jin Ho Choi
- Divisions of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jin Seok Heo
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - In Woong Han
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hongbeom Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sang Hyun Shin
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - So Jung Yoon
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sook-Young Woo
- Biomedical Statistics Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea
| |
Collapse
|
31
|
Shibamoto J, Ohgi K, Ashida R, Yamada M, Otsuka S, Kato Y, Yamazaki K, Uesaka K, Sugiura T. Clinical significance of resection and adjuvant chemotherapy for pancreatic ductal adenocarcinoma with occult para-aortic lymph node metastasis. Surgery 2025; 178:108925. [PMID: 39627914 DOI: 10.1016/j.surg.2024.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 10/07/2024] [Accepted: 10/24/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND This study aimed to determine the clinical significance of resection of pancreatic ductal adenocarcinoma diagnosed with occult para-aortic lymph node metastasis using intraoperative para-aortic lymph node sampling. METHODS Between January 2005 and May 2021, a total of 606 patients who underwent surgery for pancreatic ductal adenocarcinoma with intraoperative para-aortic lymph node sampling were retrospectively investigated and divided into the resected para-aortic lymph node-negative (n = 543), resected para-aortic lymph node-positive (n = 44), and unresected para-aortic lymph node-positive (n = 19) groups. Overall survival, clinicopathologic characteristics, and prognostic factors were analyzed. RESULTS The overall survival in the resected para-aortic lymph node-positive group was significantly worse than that in the resected para-aortic lymph node-negative group (3-year overall survival, 29.8% vs 48.4%, P < .001) and significantly better than that in the unresected para-aortic lymph node-positive group (3-year overall survival, 29.8% vs 0.0%, P = .008). In the resected para-aortic lymph node-positive group, adjuvant chemotherapy was an independent prognostic factor (hazard ratio = 2.689, P = .033). The overall survival of patients in the resected para-aortic lymph node-positive group who received adjuvant chemotherapy was comparable to that of patients in the resected para-aortic lymph node-negative group who had 4 or more regional lymph node metastases and received adjuvant chemotherapy (3-year overall survival, 33.9% vs 34.1%, P = .343). A logistic regression analysis showed that neoadjuvant therapy, age <65 years, creatinine clearance >60 mL/min, pancreatic body or tail tumor, and serum albumin level >3.5 g/dL were significant predictive factors for induction of adjuvant chemotherapy in 587 resected patients. CONCLUSIONS Resection may be acceptable for patients with para-aortic lymph node-positive pancreatic ductal adenocarcinoma who are likely to tolerate adjuvant chemotherapy.
Collapse
Affiliation(s)
- Jun Shibamoto
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Katsuhisa Ohgi
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan.
| | - Ryo Ashida
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Mihoko Yamada
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Shimpei Otsuka
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Yoshiyasu Kato
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Katsuhiko Uesaka
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Teiichi Sugiura
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
32
|
Zhao Q, Xie H, Wang X, Xie J, Liu J, Bai Y, Liu B, Ding H, Kuang S, Zhang B. Comprehensive bioinformatics analysis of the prognostic value and immune infiltration of CAPN2 in pancreatic adenocarcinoma. Gene 2025; 934:149035. [PMID: 39454972 DOI: 10.1016/j.gene.2024.149035] [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: 06/04/2024] [Revised: 09/26/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAAD) is a highly aggressive cancer with a poor prognosis, highlighting an urgent requirement for effective biomarkers for its early diagnosis and prognosis prediction. CAPN2, a calcium-dependent protease, has been implicated in various cancers, but its role in PAAD remains unclear. METHODS In this study, we utilized multiple bioinformatics methods, including differential expression, survival, correlation, and enrichment analyses, to investigate the prognostic value of CAPN2 in PAAD using data from the TCGA and GEO databases. Additionally, the correlation between CAPN2 expression and the tumor microenvironment (TME), immunotherapy potential, and drug sensitivity was also explored. RESULTS CAPN2 was upregulated in PAAD tissues and was correlated with higher tumor grade. And high expression of CAPN2 was significantly associated with reduced overall survival, establishing it as an independent prognostic biomarker for PAAD. Enrichment analysis implicated that CAPN2 was involved in multiple biological processes and pathways associated with tumor immunity. Furthermore, CAPN2 expression had a negative correlation with immune cell infiltration and a positive association with tumor mutational burden, which may have potential implications for immunotherapy strategies. CONCLUSIONS CAPN2 is a promising biomarker for PAAD prognosis and a potential therapeutic target. Its association with the TME and immunotherapy response highlights its importance in PAAD progression and patient outcomes, warranting further investigation into its role and potential clinical applications.
Collapse
Affiliation(s)
- Qiuyan Zhao
- Department of Gastroenterology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Haoran Xie
- Hepatobiliary Pancreatic Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Xing Wang
- Centre for Medical Research and Translation, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China
| | - Jiabei Xie
- Department of Gastroenterology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Jin Liu
- Department of Gastroenterology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Yangqiu Bai
- Department of Gastroenterology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Bowei Liu
- Department of Gastroenterology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Hui Ding
- Department of Gastroenterology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Shengli Kuang
- Department of Gastroenterology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China.
| | - Bingyong Zhang
- Department of Gastroenterology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China.
| |
Collapse
|
33
|
Mere Del Aguila E, Tang XH, Gudas LJ. Retinoic acid receptor-β deletion in a model of early pancreatic ductal adenocarcinoma (PDAC) tumorigenesis. Am J Cancer Res 2025; 15:127-140. [PMID: 39949931 PMCID: PMC11815370 DOI: 10.62347/xfot8509] [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: 09/13/2024] [Accepted: 11/28/2024] [Indexed: 02/16/2025] Open
Abstract
Vitamin A (VA, retinol) and its metabolites, including retinoic acid (RA), play a major role in the maintenance of cell populations in the adult pancreas. Pancreatic ductal adenocarcinomas (PDACs) contain lower amounts of VA and express lower levels of retinoic acid receptors (RARs) compared to normal human pancreatic tissues. Our goal was to determine if VA signaling directly impacts molecular events underlying pancreatic carcinogenesis using cell-type specific genetic approaches in mice. We knocked out retinoic acid receptor beta (RAR-β) selectively in pancreatic cells by tamoxifen treatment after crossing these adult RAR-βfl/fl mice with Pdx1/CreER (PCer) and lox-stop-lox KRasG12D transgenic mice. Our data show that the rounds of tamoxifen we used were able to induce the knockout of the RAR-β gene in pancreatic cells in this PCer;KRas;RAR-βfl/fl transgenic model. We detected increases in proteins involved in RA metabolism (CYP26A1, RBP1, and ALDH1A2) in the PCer;RAR-βD/wt pancreata, but the levels of RBP1 and ALDH1A2 were decreased in PCer;RAR-βD (both RAR-β alleles deleted) compared to PCer;KRas;RAR-βD and wild-type pancreata. Ki67 and vimentin proteins exhibited lower levels in the PCer;KRas;RAR-βD and PCer;RAR-βD pancreata compared to wild-type, indicating that deletion of RAR-β reduced cell proliferation in acinar cells. Expression of SOX9, a key protein required for formation and maintenance of PDAC, was higher in PCer;RAR-βD/wt and PCer;RAR-βD pancreata compared to wild-type, indicating that deletion of RAR-β increases SOX9 levels even without the KRas activating mutation. In summary, lack of RAR-β in pancreatic acinar cells reduced cell proliferation and increased SOX9 protein levels in this transgenic model.
Collapse
Affiliation(s)
- Eduardo Mere Del Aguila
- Department of Pharmacology and Meyer Cancer Center, Weill Cornell Medicine 1300 York Avenue, New York, NY 10065, USA
| | - Xiao-Han Tang
- Department of Pharmacology and Meyer Cancer Center, Weill Cornell Medicine 1300 York Avenue, New York, NY 10065, USA
| | - Lorraine J Gudas
- Department of Pharmacology and Meyer Cancer Center, Weill Cornell Medicine 1300 York Avenue, New York, NY 10065, USA
| |
Collapse
|
34
|
Madani SP, Mohseni A, Mirza-Aghazadeh-Attari M, Shahbazian H, Afyouni S, Borhani A, Zandieh G, Laheru D, Kamel IR. Role of volumetric tumor enhancement on CT in predicting overall survival in patients with unresectable pancreatic ductal adenocarcinoma. Clin Imaging 2025; 117:110365. [PMID: 39613522 DOI: 10.1016/j.clinimag.2024.110365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/06/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024]
Abstract
PURPOSE To assess the utility of volumetric tumor enhancement on CT to predict tumor treatment response and the overall survival (OS) of patients with PDAC undergoing FOLFIRINOX-based systemic chemotherapy. Additionally, we aim to explore the performance of a novel model that incorporates relevant volumetric CT-derived parameters to the established RECIST 1.1 in predicting both treatment response and OS. MATERIAL AND METHODS In this retrospective single-institution study, 127 patients with PDAC who received FOLFIRINOX neoadjuvant chemotherapy between December 2012 and November 2021 were included. Manual volumetric segmentation of the single largest tumor was performed on portal venous phase images. Total and enhancing tumor volumes were calculated. Response by RECIST 1.1 was compared to response by tumor volume and enhancing tumor volume on follow-up CT. RESULTS There was no association between overall survival and RECIST 1.1 (p-value = 0.284), volumetric RECIST (p-value = 0.402), and other volumetric CT variables, except for a percentage reduction in enhancing tumor volume (p-value = 0.043). Using univariate survival analysis for categorical thresholds defined by CART, the percentage change in enhancing tumor volume was associated with OS (p-value = 0.018). There was also a significant association between baseline enhancing tumor volume and OS (p-value <0.0001). Using these two categories, we defined a multivariable model associated with OS (p-value <0.0001). CONCLUSION Percentage reduction in enhancing tumor volume was related to OS in non-surgical PDAC patients treated with FOLFIRINOX chemotherapy and could potentially be incorporated into patient survival prediction models.
Collapse
Affiliation(s)
- Seyedeh Panid Madani
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Alireza Mohseni
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | | | - Haneyeh Shahbazian
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Shadi Afyouni
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Ali Borhani
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Ghazal Zandieh
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Daniel Laheru
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Ihab R Kamel
- Department of Radiology, University of Colorado, Aurora, CO, USA.
| |
Collapse
|
35
|
Chen X, Gong R, Wang L, Lei K, Liu X, Wang J, Sun M, Saluja AK, Yu Q, Ren H. hnRNPLL regulates MYOF alternative splicing and correlates with early metastasis in pancreatic ductal adenocarcinoma. Cancer Lett 2024; 611:217436. [PMID: 39742990 DOI: 10.1016/j.canlet.2024.217436] [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: 10/01/2024] [Revised: 11/30/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly cancer known for its high rate of early metastasis, necessitating the discovery of the underlying mechanisms. Herein, we report that heterogeneous nuclear ribonucleoprotein L-like (hnRNPLL) expression significantly increases at the invasion forefront in PDAC and is associated with early metastasis and poor prognosis. Our findings revealed that hnRNPLL knockdown resulted in extensive exon skipping (ES) events. In particular, we identified myoferlin (MYOF), a member of the ferlin family involved in membrane processes, as a functional splicing target of hnRNPLL. hnRNPLL depletion stimulates MYOF exon 17 retention to reduce the short isoform of MYOF (MYOFb) and inhibit metastasis. In contrast, hnRNPLL or MYOFb overexpression promoted pancreatic cancer cell migration and invasion. These results suggest that hnRNPLL is a critical factor for early PDAC metastasis. hnRNPLL and MYOFb may be potential therapeutic targets for PDAC.
Collapse
Affiliation(s)
- Xianghan Chen
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China; State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University (Air Force Medical University), Xi'an, 710032, China
| | - Ruining Gong
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China; Gastrointestinal Cancer Institute/Pancreatic Disease Institute, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Lili Wang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Ke Lei
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Xiaolan Liu
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Jigang Wang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Mingyue Sun
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Ashok Kumar Saluja
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Qian Yu
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - He Ren
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China; Gastrointestinal Cancer Institute/Pancreatic Disease Institute, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
36
|
Li Z, Wang C, Li J, Wang X, Li X, Yu T, Zhou J, Wang X, Zeng M, Sun H. Identification of SMAD4-mutated pancreatic ductal adenocarcinoma using preoperative contrast-enhanced MRI and clinical characteristics. BMC Med Imaging 2024; 24:349. [PMID: 39716095 DOI: 10.1186/s12880-024-01539-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024] Open
Abstract
AIM To assess the value of preoperatively contrast-enhanced MRI and clinical characteristics for identification of SMAD4-mutated pancreatic ductal adenocarcinoma (PDAC) patients. MATERIALS AND METHODS This retrospective study included patients with surgically confirmed PDAC from January 2016 to December 2022. Based on immunostaining results indicating the mutation of SMAD4, the enrolled participants were grouped into SMAD4-mutated PDAC and non-SMAD4-mutated PDAC. Contrast-enhanced MRI findings, clinical-pathological characteristics, and prognosis were recorded and reviewed. The pathological findings and clinical prognosis were compared between the two groups. Uni- and multivariable logistic regression analyses were further performed to determine the radiological and clinical predictive factors for the mutation of SMAD4. RESULTS In total, 428 PDAC patients were enrolled and analyzed, who were grouped as SMAD4-mutated PDAC (n = 224) and non-SMAD4-mutated PDAC (n = 204). SMAD4-mutated PDAC demonstrated higher frequency of pathological fatty infiltration (83.4% vs. 74.2%, P = 0.016), peripheral nerve infiltration (84.4% vs. 76.5%, P = 0.039). and higher recurrence rates (43.6% vs. 58.9%, P = 0.045) than non-SMAD4-mutated PDAC. The 3-year recurrence-free survival rates were worse for SMAD4-mutated PDAC (28.7% vs. 39.1%). In multivariable logistic regression analyses, CA19-9 > 100 U/mL (odds ratio [OR] = 1.519, P = 0.041), CBD dilation (OR = 1.564, P = 0.036), and rim enhancement (OR = 1.631, P = 0.025) were independent predictive factors. CONCLUSION Rim enhancement, CBD dilation on contrast-enhanced MRI and higher CA19-9 level are promising radiological and clinical factors for identifying SMAD4-mutated PDAC.
Collapse
Affiliation(s)
- Zhina Li
- Department of Radiology, Zhongshan Hospital, Shanghai Institute of Medical Imaging, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
- Department of Radiology, Linyi Centra Hospital, No.17 Jiankang Road, Linyi City, Shandong Province, 276400, China
| | - Cheng Wang
- Department of Radiology, Zhongshan Hospital, Shanghai Institute of Medical Imaging, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Jianbo Li
- Department of Radiology, Zhongshan Hospital, Shanghai Institute of Medical Imaging, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Xingxing Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Xiang Li
- Department of Pathology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Tianzhu Yu
- Department of Interventional Radiology, Zhongshan Hospital, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China.
| | - Jianjun Zhou
- Department of Radiology, Zhongshan Hospital (Xiamen), Xiamen Municipal Clinical Research Center for Medical Imaging, Fujian Province Key Clinical Specialty for Medical Imaging, Xiamen Key Laboratory of Clinical Transformation of Imaging Big Data and Artificial Intelligence, Fudan University, Xiamen, 361015, China
| | - Xiaolin Wang
- Department of Interventional Radiology, Zhongshan Hospital, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Shanghai Institute of Medical Imaging, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Haitao Sun
- Department of Radiology, Zhongshan Hospital, Shanghai Institute of Medical Imaging, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China.
| |
Collapse
|
37
|
Muscella A, Cossa LG, Stefàno E, Rovito G, Benedetti M, Fanizzi FP, Marsigliante S. Different Cytotoxic Effects of Cisplatin on Pancreatic Ductal Adenocarcinoma Cell Lines. Int J Mol Sci 2024; 25:13662. [PMID: 39769425 PMCID: PMC11727771 DOI: 10.3390/ijms252413662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
This study examined the response to cisplatin in BxPC-3, Mia-Paca-2, PANC-1, and YAPC pancreatic cancer lines with different genotypic and phenotypic characteristics, and the mechanisms associated with their resistance. BxPC-3 and MIA-PaCa-2 cell lines were the most sensitive to cisplatin, while YAPC and PANC-1 were more resistant. Consistently, in cisplatin-treated BxPC-3 cells, the cleavage patterns of pro-caspase-9, -7, -3, and PARP-1 demonstrated that they were more sensitive than YAPC cells. The autophagic pathway, promoting cisplatin resistance, was active in BxPC-3 cells, as demonstrated by the time-dependent conversion of LC3-I to LC3-II, whereas it was not activated in YAPC cells. In cisplatin-treated BxPC-3 cells, Bcl-2 decreased, while Beclin-1, Atg-3, and Atg-5 increased along with JNK1/2 phosphorylation. Basal levels of phosphorylated ERK1/2 in each cell line were positively correlated with cisplatin IC50 values, and cisplatin caused the activation of ERK1/2 in BxPC-3 and YAPC cells. Furthermore, ERK1/2 pharmacological inactivation increased cisplatin lethality in both BxPC-3 and YAPC cells, suggesting that p-ERK1/2 may be related to cisplatin resistance of PDAC cells. Different mechanisms and strategies are generally required to acquire drug resistance. Here, we partially explain the other response to cisplatin of BxPC-3 and YAPC cell lines by relating it to the role of ERK pathway.
Collapse
Affiliation(s)
- Antonella Muscella
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Università del Salento, Via Provinciale per Monteroni, 73100 Lecce, Italy; (L.G.C.); (E.S.); (G.R.); (M.B.); (F.P.F.); (S.M.)
| | | | | | | | | | | | | |
Collapse
|
38
|
Lv K, He T. Cancer-associated fibroblasts: heterogeneity, tumorigenicity and therapeutic targets. MOLECULAR BIOMEDICINE 2024; 5:70. [PMID: 39680287 PMCID: PMC11649616 DOI: 10.1186/s43556-024-00233-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/04/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
Cancer, characterized by its immune evasion, active metabolism, and heightened proliferation, comprises both stroma and cells. Although the research has always focused on parenchymal cells, the non-parenchymal components must not be overlooked. Targeting cancer parenchymal cells has proven to be a formidable challenge, yielding limited success on a broad scale. The tumor microenvironment(TME), a critical niche for cancer cell survival, presents a novel way for cancer treatment. Cancer-associated fibroblast (CAF), as a main component of TME, is a dynamically evolving, dual-functioning stromal cell. Furthermore, their biological activities span the entire spectrum of tumor development, metastasis, drug resistance, and prognosis. A thorough understanding of CAFs functions and therapeutic advances holds significant clinical implications. In this review, we underscore the heterogeneity of CAFs by elaborating on their origins, types and function. Most importantly, by elucidating the direct or indirect crosstalk between CAFs and immune cells, the extracellular matrix, and cancer cells, we emphasize the tumorigenicity of CAFs in cancer. Finally, we highlight the challenges encountered in the exploration of CAFs and list targeted therapies for CAF, which have implications for clinical treatment.
Collapse
Affiliation(s)
- Keke Lv
- Department of Hepatopanreatobiliary Surgery, Changhai Hospital, 168 Changhai Road, Yangpu District, Shanghai, 200433, China
| | - Tianlin He
- Department of Hepatopanreatobiliary Surgery, Changhai Hospital, 168 Changhai Road, Yangpu District, Shanghai, 200433, China.
| |
Collapse
|
39
|
Rong Z, Xu J, Yang J, Wang W, Tang R, Zhang Z, Tan Z, Meng Q, Hua J, Liu J, Zhang B, Liang C, Yu X, Shi S. CircRREB1 Mediates Metabolic Reprogramming and Stemness Maintenance to Facilitate Pancreatic Ductal Adenocarcinoma Progression. Cancer Res 2024; 84:4246-4263. [PMID: 39288082 DOI: 10.1158/0008-5472.can-23-3596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 05/24/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal tumor with limited treatment options and poor patient survival. Circular RNAs (circRNA) play crucial regulatory roles in the occurrence and development of various cancers, including PDAC. In this study, using circRNA sequencing of diverse PDAC samples, we identified circRREB1 as an oncogenic circRNA that is significantly upregulated in PDAC and is correlated with an unfavorable patient prognosis. Functionally, loss of circRREB1 markedly inhibited glycolysis and stemness, whereas elevated circRREB1 elicited the opposite effects. Mechanistically, circRREB1 interacted with PGK1, disrupting the association between PTEN and PGK1 and increasing PGK1 phosphorylation to activate glycolytic flux. Moreover, circRREB1 promoted WNT7B transcription by directly interacting with YBX1 and facilitating its nuclear translocation, consequently activating the Wnt/β-catenin signaling pathway to maintain PDAC stemness. Overall, these results highlight circRREB1 as a key regulator of metabolic and stemness properties of PDAC. Significance: CircRREB1 stimulates PGK1 to induce glycolysis and activates the Wnt/β-catenin signaling pathway to maintain stemness in pancreatic cancer, indicating the potential of circRREB1 as a biomarker and therapeutic target.
Collapse
MESH Headings
- Humans
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/genetics
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/genetics
- Mice
- Animals
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Glycolysis
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Wnt Signaling Pathway
- Phosphoglycerate Kinase/metabolism
- Phosphoglycerate Kinase/genetics
- Disease Progression
- Prognosis
- Gene Expression Regulation, Neoplastic
- Cell Line, Tumor
- Mice, Nude
- Male
- Female
- Cell Proliferation
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
- PTEN Phosphohydrolase/metabolism
- PTEN Phosphohydrolase/genetics
- Mice, Inbred BALB C
- Wnt Proteins/metabolism
- Wnt Proteins/genetics
- Metabolic Reprogramming
- Y-Box-Binding Protein 1
Collapse
Affiliation(s)
- Zeyin Rong
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Jianhui Yang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Rong Tang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Zifeng Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Zhen Tan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Department of Hepatobiliary Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qingcai Meng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Jie Hua
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Chen Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| |
Collapse
|
40
|
Wang D, Lv L, Du J, Tian K, Chen Y, Chen M. TRIM16 and PRC1 Are Involved in Pancreatic Cancer Progression and Targeted by Delphinidin. Chem Biol Drug Des 2024; 104:e70026. [PMID: 39635962 DOI: 10.1111/cbdd.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/03/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
Pancreatic cancer (PC) is the leading cause of cancer-related death worldwide, and new biomarkers, therapeutic targets, and candidate drugs are needed. In this work, three PC-related microarray datasets (GSE183795, GSE28735, and GSE62452) were analyzed. The differentially expressed genes (DEGs) of PC were obtained with the limma package in R. Weighted gene co-expression network analysis (WGCNA) and machine learning approaches were used to screen the hub genes. Kaplan-Meier plotter and receiver operating characteristic (ROC) curve analysis were utilized to assess the diagnostic efficacy of the hub genes. The binding ability between natural bioactive ingredients and hub proteins was evaluated by molecular docking and molecular dynamics simulation. CCK-8, flow cytometry, transwell, and western blot assays were used to analyze the viability, apoptosis, cell cycle progression, invasion, and pathway change of PC cells. Additionally, a nude mice model was used to evaluate the aggressive properties of PC cells in vivo. In this study, a total of 988 DEGs were identified, which were mainly enriched in cell adhesion and PI3K-Akt signaling pathway, and two core genes TRIM16 and PRC1 were further identified. The overall survival of patients with high expression of TRIM16 and PRC1 was shorter. Delphinidin (Del) had good binding affinity with both TRIM16 and PRC1, and Del could inhibit the viability, invasion, and metastasis of PC cells and induce cell apoptosis and G0/G1 phase arrest. In addition, Del could promote the activation of p53 and inhibit the activation of the PI3K/AKT signaling pathway in PC cells. In summary, TRIM16 and PRC1 are identified as prognostic biomarkers and therapeutic targets for PC, and Del has good binding affinity with them and may be a potential therapeutic agent for PC.
Collapse
Affiliation(s)
- Donghua Wang
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Long Lv
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Jinghu Du
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Kui Tian
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Yu Chen
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Manyu Chen
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| |
Collapse
|
41
|
Zhu X, Olson B, Keith D, Norgard MA, Levasseur PR, Diba P, Protzek S, Li J, Li X, Korzun T, Sattler AL, Buenafe AC, Grossberg AJ, Marks DL. GDF15 and LCN2 for early detection and prognosis of pancreatic cancer. Transl Oncol 2024; 50:102129. [PMID: 39353236 PMCID: PMC11474189 DOI: 10.1016/j.tranon.2024.102129] [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: 02/18/2024] [Revised: 08/20/2024] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND The prognosis of pancreatic ductal adenocarcinomas (PDAC) remains very poor, emphasizing the critical importance of early detection, where biomarkers offer unique potential. Although growth differentiation factor 15 (GDF15) and Lipocalin 2 (LCN2) have been linked to PDAC, their precise roles as biomarkers are uncertain. METHODS Circulating levels of GDF15 and LCN2 were examined in human PDAC patients, heathy controls, and individuals with benign pancreatic diseases. Circulating levels of IL-6, CA19-9, and neutrophil-to-lymphocyte ratio (NLR) were measured for comparisons. Correlations between PDAC progression and overall survival were assessed. A mouse PDAC model was employed for comprehensive analyses, complementing the human studies by exploring associations with various metabolic and inflammatory parameters. Sensitivity and specificity of the biomarkers were evaluated. FINDINGS Our results demonstrated elevated levels of circulating GDF15 and LCN2 in PDAC patients compared to both healthy controls and individuals with benign pancreatic diseases, with higher GDF15 levels associated with disease progression and increased mortality. In PDAC mice, circulating GDF15 and LCN2 progressively increased, correlating with tumor growth, behavioral manifestations, tissue and molecular pathology, and cachexia development. GDF15 exhibited highly sensitive and specific for PDAC patients compared to CA19-9, IL-6, or NLR, while LCN2 showed even greater sensitivity and specificity in PDAC mice. Combining GDF15 and LCN2, or GDF15 and CA19-9, enhanced sensitivity and specificity. INTERPRETATION Our findings indicate that GDF15 holds promise as a biomarker for early detection and prognosis of PDAC, while LCN2 could strengthen diagnostic panels.
Collapse
Affiliation(s)
- Xinxia Zhu
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, USA; Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA
| | - Brennan Olson
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, USA; Medical Scientist Training program, Oregon Health & Science University, Portland, Oregon, USA; Department of Otolaryngology-Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Dove Keith
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA
| | - Mason A Norgard
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Peter R Levasseur
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, USA; Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA
| | - Parham Diba
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, USA; Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA; Medical Scientist Training program, Oregon Health & Science University, Portland, Oregon, USA
| | - Sara Protzek
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA
| | - Ju Li
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Xiaolin Li
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, USA; Nutritional Biology, Division of Human Nutrition, Wageningen University, Wageningen, Netherlands
| | - Tetiana Korzun
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, USA; Medical Scientist Training program, Oregon Health & Science University, Portland, Oregon, USA
| | - Ariana L Sattler
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, USA; Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Abigail C Buenafe
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Aaron J Grossberg
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA; Department of Radiation Medicine, Oregon Health & Science University, Portland, Oregon, USA; Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, USA; Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA.
| |
Collapse
|
42
|
Millastre J, Hermoso-Durán S, de Solórzano MO, Fraunhoffer N, García-Rayado G, Vega S, Bujanda L, Sostres C, Lanas Á, Velázquez-Campoy A, Abian O. Thermal Liquid Biopsy: A Promising Tool for the Differential Diagnosis of Pancreatic Cystic Lesions and Malignancy Detection. Cancers (Basel) 2024; 16:4024. [PMID: 39682210 PMCID: PMC11640424 DOI: 10.3390/cancers16234024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Pancreatic cystic lesions (PCLs) are a heterogeneous group of lesions with increasing incidence, usually identified incidentally on imaging studies (multidetector computed tomography (MDCT), magnetic resonance imaging (MRI), or endoscopic ultrasound (EUS)) [...].
Collapse
Affiliation(s)
- Judith Millastre
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain; (J.M.); (M.O.d.S.); (G.G.-R.); (C.S.); (Á.L.)
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Sonia Hermoso-Durán
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain;
| | - María Ortiz de Solórzano
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain; (J.M.); (M.O.d.S.); (G.G.-R.); (C.S.); (Á.L.)
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Nicolas Fraunhoffer
- Programa Franco-Argentino de Estudio del Cáncer de Páncreas, Buenos Aires, Argentina;
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, CNRS UMR, Aix-Marseille Université, 13009 Marseille, France
- Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Equipe Labellisée La Ligue, 13288 Marseille, France
| | - Guillermo García-Rayado
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain; (J.M.); (M.O.d.S.); (G.G.-R.); (C.S.); (Á.L.)
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Sonia Vega
- Institute for Biocomputation and Physics of Complex Systems, University of Zaragoza, 50018 Zaragoza, Spain;
| | - Luis Bujanda
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain;
- Donostia University Hospital, University of the Basque Country (UPV/EHU), 20014 San Sebastian, Spain
| | - Carlos Sostres
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain; (J.M.); (M.O.d.S.); (G.G.-R.); (C.S.); (Á.L.)
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Ángel Lanas
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain; (J.M.); (M.O.d.S.); (G.G.-R.); (C.S.); (Á.L.)
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Adrián Velázquez-Campoy
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain;
- Institute for Biocomputation and Physics of Complex Systems, University of Zaragoza, 50018 Zaragoza, Spain;
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, 50009 Zaragoza, Spain
| | - Olga Abian
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain;
- Institute for Biocomputation and Physics of Complex Systems, University of Zaragoza, 50018 Zaragoza, Spain;
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, 50009 Zaragoza, Spain
| |
Collapse
|
43
|
Lin XJ, Zhu S, Wang D, Chen JY, Wei SX, Chen SY, Luo HC. Correlation of dynamic contrast-enhanced ultrasonography and the Ki-67 labelling index in pancreatic ductal adenocarcinoma. World J Gastroenterol 2024; 30:4697-4708. [PMID: 39610780 PMCID: PMC11580603 DOI: 10.3748/wjg.v30.i44.4697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/22/2024] [Accepted: 10/23/2024] [Indexed: 11/12/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant and aggressive tumor, and high Ki-67 expression indicates poor histological differentiation and prognosis. Therefore, one of the challenges in diagnosing preoperatively patients with PDAC is predicting the degree of malignancy. Dynamic contrast-enhanced ultrasonography (DCE-US) plays a crucial role in abdominal tumor diagnosis, and can adequately show the microvascular composition within the tumors. However, the relationship between DCE-US and the Ki-67 labelling index remains unclear at the present time. AIM To predict the correlation between Ki-67 expression and the parameters of DCE-US. METHODS Patients with PDAC who underwent DCE-US were retrospectively analyzed. Patients who had received any treatment (radiotherapy or chemotherapy) prior to DCE-US; had incomplete clinical, imaging, or pathologic information; and had poor-quality image analysis were excluded. Correlations between Ki-67 expression and the parameters of DCE-US in patients with PDAC were assessed using Spearman's rank correlation analysis. The diagnostic performances of these parameters in high Ki-67 expression group were evaluated according to receiver operating characteristic curve. RESULTS Based on the Ki-67 labelling index, 30 patients were divided into two groups, i.e., the high expression group and the low expression group. Among the relative quantitative parameters between the two groups, relative half-decrease time (rHDT), relative peak enhancement, relative wash-in perfusion index and relative wash-in rate were significantly different between two groups (P = 0.018, P = 0.025, P = 0.028, P = 0.035, respectively). The DCE-US parameter rHDT was moderately correlated with Ki-67 expression, and rHDT ≥ 1.07 was more helpful in accurately diagnosing high Ki-67 expression, exhibiting a sensitivity and specificity of 53.8% and 94.1%, respectively. CONCLUSION One parameter of DCE-US, rHDT, correlates with high Ki-67 expression. It demonstrates that parameters obtained noninvasively by DCE-US could better predict Ki-67 expression in PDAC preoperatively.
Collapse
Affiliation(s)
- Xiao-Jing Lin
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Shu Zhu
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Dan Wang
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Jing-Yuan Chen
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Su-Xian Wei
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Shi-Yun Chen
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Hong-Chang Luo
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| |
Collapse
|
44
|
Jiang L, Lu X, Dai Y, Jiang K, Miao Y, Yu J, Yin L, Wei J. Establishment and analysis of a prognostic model of pancreatic ductal adenocarcinomas based on nerve-cancer crosstalk-related genes. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2024; 17:396-410. [PMID: 39660330 PMCID: PMC11626290 DOI: 10.62347/ghum8504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 10/26/2024] [Indexed: 12/12/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor with a five-year survival rate of 13%, the lowest among all malignant tumors. The work aims to use bioinformatics methods to mine Nerve-cancer crosstalk-related genes (NCCGs) in pancreatic cancer and evaluate their correlation with tumor stage and prognosis, thereby providing a new direction of development and experimental basis for pancreatic cancer treatment. This study included 185 individuals with PDAC from the TCGA database, together with clinical and RNA sequencing data. A review of prior studies revealed the mechanism of neural-cancer crosstalk and identified 42 neural-cancer crosstalk-related genes (NCCGs). Multivariate logistic regression analysis showed that NGFR (OR=39.076, 95% CI; P<0.05), CHRNB2 (OR=41.076, 95% CI; P<0.05), and CHRNA10 (OR=39.038, 95% CI; P<0.05) were identified as independent risk factors for PNI development. Pearson correlation analysis revealed that CHRNA10 was negatively connected with PDAC microsatellite instability, whereas CHRNA10, CHRNB2, and NGFR were negatively correlated with PDAC tumor mutation burden. The GEPIA database revealed that CHRNB2 expression was higher in stage I PDAC. The pancreatic cancer single-cell dataset PAAD_CRA001160 revealed that malignant tumor cells, ductal cells, endothelial cells and fibroblasts accounted for a large proportion in the tumor microenvironment of pancreatic cancer. Furthermore, the NGFR gene was shown to be more significantly expressed in various pancreatic cancer cells. Bioinformatics analysis was used to create a validated prognostic model of pancreatic cancer, which explored the critical mechanisms of neural-tumor interactions and revealed the potential of cancer-neural crosstalk-related genes as prognostic biomarkers and anti-tumor therapy targets.
Collapse
Affiliation(s)
- Lei Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Xiaozhi Lu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Yuran Dai
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Kuirong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Yi Miao
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Jun Yu
- Department of Surgery, Johns Hopkins University School of MedicineBaltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence, Johns Hopkins University School of MedicineBaltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Lingdi Yin
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Jishu Wei
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| |
Collapse
|
45
|
Huang Y, Zhang H, Ding Q, Chen D, Zhang X, Weng S, Liu G. Comparison of multiple machine learning models for predicting prognosis of pancreatic ductal adenocarcinoma based on contrast-enhanced CT radiomics and clinical features. Front Oncol 2024; 14:1419297. [PMID: 39605884 PMCID: PMC11598923 DOI: 10.3389/fonc.2024.1419297] [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: 04/18/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Objective The aim of this study was to evaluate the prognostic potential of combining clinical features and radiomics with multiple machine learning (ML) algorithms in pancreatic ductal adenocarcinoma (PDAC). Methods A total of 116 patients with PDAC who met the eligibility criteria were randomly assigned to a training or validation cohort. Seven ML algorithms, including Supervised Principal Components, stepwise Cox, Random Survival Forest, CoxBoost, Least absolute shrinkage and selection operation (Lasso), Ridge, and Elastic network, were integrated into 43 algorithm combinations. Forty-three radiomics models were constructed separately using radiomics features extracted from arterial phase (AP), venous phase (VP), and combined arterial and venous phase (AP+VP) images. The concordance index (C-index) of each model was calculated. The model with the highest mean C-index was identified as the best model for calculating the radiomics score (Radscore). Univariate and multivariate Cox analyses were used to identify independent prognostic indicators and create a clinical model for prognosis prediction. The multivariable Cox regression was used to combine Radscore with clinical features to create a combined model. The efficacy of the model was evaluated using the C-index, calibration curves, and decision curve analysis (DCA). Results The model based on the Lasso+StepCox[both] algorithm constructed using AP+VP radiomics features showed the best predictive ability among the 114 radiomics models. The C-indices of the model in the training and validation cohorts were 0.742 and 0.722, respectively. Based on the results of the univariate and multivariate Cox regression analyses, sex, Tumor-Node-Metastasis (TNM) stage, and systemic inflammation response index were included to build the clinical model. The combined model, incorporating three clinical factors and AP+VP-Radscore, achieved the highest C-indices of 0.764 and 0.746 in the training and validation cohorts, respectively. In terms of preoperative prognosis prediction for PDAC, the calibration curve and DCA showed that the combined model had a good consistency and greatest net benefit. Conclusion A combined model of clinical features and AP+VP-Radscore screened using multiple ML algorithms has an excellent ability to predict the prognosis of PDAC and may provide a noninvasive and effective method for clinical decision-making.
Collapse
Affiliation(s)
- Yue Huang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Han Zhang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Qingzhu Ding
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Dehua Chen
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xiang Zhang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Shangeng Weng
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Clinical Research Center for Hepatobiliary Pancreatic and Gastrointestinal Malignant Tumors Precise Treatment of Fujian, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Guozhong Liu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
46
|
Philip PA, Sahai V, Bahary N, Mahipal A, Kasi A, Rocha Lima CMS, Alistar AT, Oberstein PE, Golan T, Metges JP, Lacy J, Fountzilas C, Lopez CD, Ducreux M, Hammel P, Salem M, Bajor D, Benson AB, Luther S, Pardee T, Van Cutsem E. Devimistat (CPI-613) With Modified Fluorouarcil, Oxaliplatin, Irinotecan, and Leucovorin (FFX) Versus FFX for Patients With Metastatic Adenocarcinoma of the Pancreas: The Phase III AVENGER 500 Study. J Clin Oncol 2024; 42:3692-3701. [PMID: 39088774 DOI: 10.1200/jco.23.02659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/27/2024] [Accepted: 05/07/2024] [Indexed: 08/03/2024] Open
Abstract
PURPOSE Metastatic pancreatic adenocarcinoma (mPC) remains a difficult-to-treat disease. Fluorouarcil, oxaliplatin, irinotecan, and leucovorin (FFX) is a standard first-line therapy for mPC for patients with a favorable performance status and good organ function. In a phase I study, devimistat (CPI-613) in combination with modified FFX (mFFX) was deemed safe and exhibited promising efficacy in mPC. METHODS The AVENGER 500 trial (ClinicalTrials.gov identifier: NCT03504423) is a global, randomized phase III trial conducted at 74 sites across six countries to investigate the efficacy and safety of devimistat in combination with mFFX (experimental arm) compared with standard-dose FFX (control arm) in treatment-naïve patients with mPC. Treatment, administered in once-every-2-weeks cycles until disease progression or intolerable toxicity, included intravenous devimistat at 500 mg/m2 total per day on days 1 and 3 in the experimental arm. The primary end point of the study was overall survival (OS). RESULTS Five hundred and twenty-eight patients were randomly assigned (266 in the experimental arm and 262 in the control arm). The median OS was 11.10 months for devimistat plus mFFX versus 11.73 months for FFX (hazard ratio [HR], 0.95 [95% CI, 0.77 to 1.18]; P = .655) and median progression-free survival was 7.8 months versus 8.0 months, respectively (HR, 0.99 [95% CI, 0.76 to 1.29]; P = .94). Grade ≥3 treatment-emergent adverse events with >10% frequency in the devimistat plus mFFX arm versus the FFX arm were neutropenia (29.0% v 34.5%), diarrhea (11.2% v 19.6%), hypokalemia (13.1% v 14.9%), anemia (13.9% v 13.6%), thrombocytopenia (11.6% v 13.6%), and fatigue (10.8% v 11.5%), respectively. CONCLUSION Devimistat in combination with mFFX did not improve long- and short-term mPC patient outcomes compared with standard FFX. There were no new toxicity signals with the addition of devimistat.
Collapse
Affiliation(s)
- Philip A Philip
- Department of Oncology and Department of Pharmacology, Henry Ford Cancer Institute, Wayne State University School of Medicine, Detroit, MI
| | - Vaibhav Sahai
- University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Nathan Bahary
- Allegheny Health Network Cancer Institute, Pittsburgh, PA
| | - Amit Mahipal
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Anup Kasi
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS
| | - Caio Max S Rocha Lima
- Division of Hematology and Oncology, Atrium Wake Forest Baptist Medical Center, Winston-Salem, NC
| | | | | | - Talia Golan
- Institute of Oncology, Sheba Medical Center, Faculty of Medicine Tel Avi University, Ramat Gan, Israel
| | - Jean-Philippe Metges
- Institut de Cancérologie et d'Hématologie, ARPEGO Network CHU Morvan, Brest, France
| | - Jill Lacy
- Department of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - Christos Fountzilas
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | | | - Michel Ducreux
- Gustave Roussy, University Paris Saclay, Inserm, U1279, Villejuif, France
| | - Pascal Hammel
- Department of Digestive and Medical Oncology, University Paris-Saclay, Paul Brousse Hospital (AP-HP), Villejuif, France
| | - Mohamed Salem
- Department of Medical Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - David Bajor
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Al B Benson
- Division of Hematology & Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | | | - Timothy Pardee
- Division of Hematology and Oncology, Atrium Wake Forest Baptist Medical Center, Winston-Salem, NC
| | - Eric Van Cutsem
- University Hospitals Gasthuisberg Leuven and KU Leuven, Leuven, Belgium
| |
Collapse
|
47
|
Liu W, Wang X, Wu W. Role and functional mechanisms of IL‑17/IL‑17R signaling in pancreatic cancer (Review). Oncol Rep 2024; 52:144. [PMID: 39219271 PMCID: PMC11378154 DOI: 10.3892/or.2024.8803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Interleukin‑17 (IL‑17), an inflammatory cytokine primarily secreted by T helper 17 cells, serves a crucial role in numerous inflammatory diseases and malignancies via its receptor, IL‑17R. In addition to stimulating inflammatory responses, IL‑17 exhibits dual functions in tumors, exerting both pro‑ and antitumor effects. Pancreatic ductal adenocarcinoma (PDAC) is the most common pancreatic malignancy and accounts for >90% of pancreatic cancer cases. PDAC is characterized by a prominent stromal microenvironment with significant heterogeneity, which contributes to treatment resistance. IL‑17/IL‑17R signaling has a notable effect on tumorigenesis, the tumor microenvironment and treatment efficacy in various cancer types, including PDAC. However, the specific mechanisms of IL‑17/IL‑17R signaling in pancreatic cancer remain uncertain. This review presents a brief overview of the current knowledge and recent advances in the role and functional mechanisms of IL‑17/IL‑17R signaling in pancreatic cancer. Furthermore, the potential of IL‑17‑targeted therapeutic strategies for PDAC treatment is also discussed.
Collapse
Affiliation(s)
- Wanli Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Xianze Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Wenming Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
48
|
Machicado JD, Sheth SG, Chalhoub JM, Forbes N, Desai M, Ngamruengphong S, Papachristou GI, Sahai V, Nassour I, Abidi W, Alipour O, Amateau SK, Coelho-Prabhu N, Cosgrove N, Elhanafi SE, Fujii-Lau LL, Kohli DR, Marya NB, Pawa S, Ruan W, Thiruvengadam NR, Thosani NC, Qumseya BJ. American Society for Gastrointestinal Endoscopy guideline on role of endoscopy in the diagnosis and management of solid pancreatic masses: methodology and review of evidence. Gastrointest Endosc 2024; 100:e1-e78. [PMID: 39269378 DOI: 10.1016/j.gie.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 06/02/2024] [Indexed: 09/15/2024]
Affiliation(s)
- Jorge D Machicado
- Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sunil G Sheth
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jean M Chalhoub
- Division of Gastroenterology and Hepatology, Staten Island University Hospital, Northwell Health, Staten Island, New York, USA
| | - Nauzer Forbes
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Madhav Desai
- Center for Interventional Gastroenterology at UTHealth, McGovern Medical School, Houston, Texas, USA
| | - Saowanee Ngamruengphong
- Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Georgios I Papachristou
- Division of Gastroenterology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Vaibhav Sahai
- Division of Hematology and Oncology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ibrahim Nassour
- Division of Surgical Oncology, University of Florida, Gainesville, Florida, USA
| | - Wasif Abidi
- Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| | - Omeed Alipour
- Division of Gastroenterology, University of Washington Medical Center, Seattle, Washington, USA
| | - Stuart K Amateau
- Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| | | | - Natalie Cosgrove
- Center for Interventional Endoscopy, AdventHealth, Orlando, Florida, USA
| | - Sherif E Elhanafi
- Division of Gastroenterology, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | | | - Divyanshoo R Kohli
- Pancreas and Liver Clinic, Providence Sacred Medical Center, Elon Floyd School of Medicine, Washington State University, Spokane, Washington, USA
| | - Neil B Marya
- Division of Gastroenterology, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Swati Pawa
- Department of Gastroenterology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Wenly Ruan
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Nikhil R Thiruvengadam
- Division of Gastroenterology and Hepatology, Loma Linda University, Loma Linda, California, USA
| | - Nirav C Thosani
- Center for Interventional Gastroenterology at UTHealth, McGovern Medical School, Houston, Texas, USA
| | - Bashar J Qumseya
- Department of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
49
|
Mibelli N, Oehme F, Radulova-Mauersberger O, Selbmann AC, Merboth F, Hempel S, Distler M, Weitz J, Teske C. Bacterial shift and resistance pattern in pancreatic head resections after selective decontamination of the digestive tract - a propensity score-matched analysis. J Gastrointest Surg 2024; 28:1844-1852. [PMID: 39241947 DOI: 10.1016/j.gassur.2024.08.030] [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: 07/22/2024] [Revised: 08/21/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Pancreatic head resection is associated with postoperative morbidity, primarily because of infectious complications. The microbiota in these infections is crucial, and selective decontamination of the digestive tract (SDD) aims to mitigate this risk by targeting pathogenic organisms while preserving beneficial flora. This study aimed to determine the effect of SDD on bacterial shifts and resistance patterns in pancreatic head resection. METHODS All patients who underwent pancreatic head resection either between January 2012 and August 2018 (non-SDD group) or between January 2019 and December 2021 (SDD group) were included. Propensity score-matched analysis was performed to compare the bacterial presence and resistance patterns in bile duct smear tests and postoperative complications. RESULTS Positive bile duct smear tests were observed more often in the non-SDD group (63.5%) than in the SDD group (51.0%). Moreover, the SDD group exhibited a significant reduction in the median number of bacterial species in the bile ducts compared with the non-SDD group (P = .04). However, a notable increase in gram-negative species was observed in the SDD group. The SDD group experienced higher rates of postoperative complications, including relevant pancreatic fistulas (24.8% in the SDD group vs 11.6% in the non-SDD group; P < .01) and delayed gastric emptying (33.8% in the SDD group vs 21.9% in the non-SDD group; P < .01). No significant difference in antibiotic resistance patterns was observed. CONCLUSION SDD in pancreatic head resection reduces bacterial load in the biliary tract, but it is associated with a shift toward more gram-negative species and higher rates of severe postoperative complications. Our findings suggest that SDD may negatively affect postoperative outcomes and should be carefully considered in clinical practice.
Collapse
Affiliation(s)
- Nicolas Mibelli
- Department of Visceral, Thoracic, and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases, Dresden, Germany: German Cancer Research Center, Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; and Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
| | - Florian Oehme
- Department of Visceral, Thoracic, and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases, Dresden, Germany: German Cancer Research Center, Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; and Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
| | - Olga Radulova-Mauersberger
- Department of Visceral, Thoracic, and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases, Dresden, Germany: German Cancer Research Center, Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; and Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
| | - Anne-Christin Selbmann
- Department of Visceral, Thoracic, and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases, Dresden, Germany: German Cancer Research Center, Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; and Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
| | - Felix Merboth
- Department of Visceral, Thoracic, and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases, Dresden, Germany: German Cancer Research Center, Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; and Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
| | - Sebastian Hempel
- Department of Visceral, Thoracic, and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases, Dresden, Germany: German Cancer Research Center, Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; and Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
| | - Marius Distler
- Department of Visceral, Thoracic, and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases, Dresden, Germany: German Cancer Research Center, Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; and Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic, and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases, Dresden, Germany: German Cancer Research Center, Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; and Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
| | - Christian Teske
- Department of Visceral, Thoracic, and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases, Dresden, Germany: German Cancer Research Center, Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; and Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany.
| |
Collapse
|
50
|
Blázquez-Encinas R, Alors-Pérez E, Moreno-Montilla MT, García-Vioque V, Sánchez-Frías ME, Mafficini A, López-Cánovas JL, Bousquet C, Gahete MD, Lawlor RT, Luque RM, Scarpa A, Arjona-Sánchez Á, Pedraza-Arevalo S, Ibáñez-Costa A, Castaño JP. The Exon Junction Complex component EIF4A3 plays a splicing-linked oncogenic role in pancreatic ductal adenocarcinoma. Cancer Gene Ther 2024; 31:1646-1657. [PMID: 39232176 PMCID: PMC11567885 DOI: 10.1038/s41417-024-00814-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 09/06/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers, underscoring the urgent need for in-depth biological research. The phenomenon of alternative RNA splicing dysregulation is a common hallmark in cancer, including PDAC, presenting new avenues for understanding and developing diagnostic and therapeutic tools. Our research focuses on EIF4A3, a core component of the Exon Junction Complex intimately linked to RNA splicing, and its role in PDAC. EIF4A3 is overexpressed in PDAC tissue and associated to clinical parameters of malignancy and poorer patient survival. Mechanistically, exploration of PDAC RNA-seq data unveiled the link of EIF4A3 to diverse malignancy processes, consistent with its association to key molecular pathways. EIF4A3 targeting in vitro decreased essential functional tumor features such as proliferation, migration, colony formation and sphere formation, while its in vivo targeting reduced tumor growth. EIF4A3 silencing in PDAC cell lines severely altered its transcriptional and spliceosomic landscapes, as shown by RNA-seq analyses, suggesting a role for EIF4A3 in maintaining RNA homeostasis. Our results indicate that EIF4A3 dysregulation in PDAC has a pleiotropic regulatory role on RNA biology, influencing key cellular functions. This paves the way to explore its potential as novel biomarker and actionable target candidate for this lethal cancer.
Collapse
Affiliation(s)
- Ricardo Blázquez-Encinas
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Emilia Alors-Pérez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - María Trinidad Moreno-Montilla
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Víctor García-Vioque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Marina Esther Sánchez-Frías
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- Pathology Service, Reina Sofía University Hospital, Córdoba, Spain
| | - Andrea Mafficini
- Department of Engineering for Innovation Medicine, Section of Innovation Biomedicine, University and Hospital Trust of Verona, Verona, Italy
- ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Juan L López-Cánovas
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Corinne Bousquet
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Team 'Labellisée Ligue Contre le Cancer', University of Toulouse, Toulouse, France
| | - Manuel D Gahete
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Rita T Lawlor
- Department of Engineering for Innovation Medicine, Section of Innovation Biomedicine, University and Hospital Trust of Verona, Verona, Italy
- ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Raúl M Luque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Aldo Scarpa
- ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Álvaro Arjona-Sánchez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- Surgery Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Sergio Pedraza-Arevalo
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Alejandro Ibáñez-Costa
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.
- Reina Sofia University Hospital, Córdoba, Spain.
| | - Justo P Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.
- Reina Sofia University Hospital, Córdoba, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.
| |
Collapse
|