1
|
Heller M, Mann DA, Katona BW. Current Approaches of Pancreatic Cancer Surveillance in High-Risk Individuals. J Gastrointest Cancer 2025; 56:61. [PMID: 39932614 PMCID: PMC11814005 DOI: 10.1007/s12029-025-01184-1] [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: 02/14/2025]
Abstract
Currently, those recommended to undergo pancreatic cancer (PC) surveillance include appropriately aged individuals at high risk of PC due to an identifiable genetic susceptibility or those without identifiable genetic susceptibility who nonetheless have a strong family history of PC. With increases in identification of individuals at high risk for PC and increased use of PC surveillance in clinical practice, there has been increasing debate about who should undergo surveillance as well as how surveillance should be performed including use of imaging and blood-based testing. Furthermore, there is increasing interest in the outcomes of PC surveillance in high-risk individuals with some studies demonstrating that surveillance leads to downstaging of PC and improvements in survival. In this review, we summarize the current state of PC surveillance in high-risk individuals, providing an overview of the risk factors associated with PC, selection of high-risk individuals for PC surveillance, and the current, but non-uniform, recommendations for performing PC surveillance. Additionally, we review approaches to apply various imaging and blood-based tests to surveillance and the outcomes of PC surveillance.
Collapse
Affiliation(s)
- Melissa Heller
- Division of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Derek A Mann
- Division of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bryson W Katona
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd., 751 South Pavilion, Philadelphia, PA, 19104, USA.
| |
Collapse
|
2
|
Zhan T, Betge J, Schulte N, Dreikhausen L, Hirth M, Li M, Weidner P, Leipertz A, Teufel A, Ebert MP. Digestive cancers: mechanisms, therapeutics and management. Signal Transduct Target Ther 2025; 10:24. [PMID: 39809756 PMCID: PMC11733248 DOI: 10.1038/s41392-024-02097-4] [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: 06/29/2024] [Revised: 10/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Cancers of the digestive system are major contributors to global cancer-associated morbidity and mortality, accounting for 35% of annual cases of cancer deaths. The etiologies, molecular features, and therapeutic management of these cancer entities are highly heterogeneous and complex. Over the last decade, genomic and functional studies have provided unprecedented insights into the biology of digestive cancers, identifying genetic drivers of tumor progression and key interaction points of tumor cells with the immune system. This knowledge is continuously translated into novel treatment concepts and targets, which are dynamically reshaping the therapeutic landscape of these tumors. In this review, we provide a concise overview of the etiology and molecular pathology of the six most common cancers of the digestive system, including esophageal, gastric, biliary tract, pancreatic, hepatocellular, and colorectal cancers. We comprehensively describe the current stage-dependent pharmacological management of these malignancies, including chemo-, targeted, and immunotherapy. For each cancer entity, we provide an overview of recent therapeutic advancements and research progress. Finally, we describe how novel insights into tumor heterogeneity and immune evasion deepen our understanding of therapy resistance and provide an outlook on innovative therapeutic strategies that will shape the future management of digestive cancers, including CAR-T cell therapy, novel antibody-drug conjugates and targeted therapies.
Collapse
Affiliation(s)
- Tianzuo Zhan
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johannes Betge
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Schulte
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Dreikhausen
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Hirth
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Moying Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philip Weidner
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Antonia Leipertz
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Teufel
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany.
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
3
|
Zhang JX, Shen YB, Ma DD, Li ZH, Zhang ZY, Jin WD. LINC01857 promotes cell proliferation and migration while dampening cell apoptosis in pancreatic cancer by upregulating CDC42EP3 via miR-450b-5p. Heliyon 2024; 10:e38427. [PMID: 39524859 PMCID: PMC11547965 DOI: 10.1016/j.heliyon.2024.e38427] [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: 05/28/2024] [Revised: 09/14/2024] [Accepted: 09/24/2024] [Indexed: 11/16/2024] Open
Abstract
Background Pancreatic cancer (PC) is a devastating human malignancy with a poor survival outcome (5-year survival less than 10 %). In recent years, the regulatory roles of long non-coding RNAs (lncRNAs) in various types of cancers have been widely reported. Based on bioinformatics analysis, LINC01857 is shown to be highly expressed in PC tissue. Nevertheless, the role of LINC01857 in PC is limitedly reported. Hence, this study aimed to explore the effects of lncRNA LINC01857 on PC cell process and the related mechanism. Methods RT-qPCR and fluorescence in situ hybridization (FISH) assay were conducted to measure LINC01857 expression and distribution in PANC-1 and MIA PaCa-2 cells. Colony formation and wound healing assays as well as flow cytometry analyses were employed to estimate the proliferation, migration, and apoptosis of PC cells transfected with pcDNA3.1-LINC01857 or si-LINC01857 compared with the behavior of PC cells transfected with empty pcDNA3.1 vector (control) or si-negative control (NC). Furthermore, RNA pulldown and luciferase reporter assays were utilized to demonstrate the interaction of LINC01857 and miR-450b-5p or to validate the binding of miR-450b-5p and cell division cycle 42 effector protein 3 (CDC42EP3). Results LINC01857 was highly expressed in PANC-1 and MIA PaCa-2 cells in contrast to its expression in pancreatic ductal epithelial cells (8.9 folds and 7.1 folds, p < 0.001). Silencing LINC01857 significantly reduced cell proliferation and migration while enhancing apoptosis (p < 0.0005). In contrast, overexpression of LINC01857 markedly (p < 0.05) accelerated these malignant behavior of PC cells. MiR-450b-5p was targeted and inversely regulated by LINC01857. Moreover, CDC42EP3 was verified to be targeted by miR-450b-5p, and CDC42EP3 was correlated to LINC01857 in a positive manner (p < 0.001). Rescue experiments manifested that silencing CDC42EP3 effectively (p < 0.05) reversed the promoting effect of LINC01857 on malignant behavior of PC cells. Conclusion LINC01857 promotes PC cell proliferation and migration while obstructing cell apoptosis by binding to miR-450b-5p and thus regulating CDC42EP3 expression. The study presents a novel and promising regulatory axis, which holds potential for the identification of biomarkers and development of therapeutic strategies for PC treatment.
Collapse
Affiliation(s)
| | | | - Dan-Dan Ma
- Department of General Surgery, The Central Theater Hospital of the Chinese People's Liberation Army, Wuhan, 430070, China
| | - Zhong-Hu Li
- Department of General Surgery, The Central Theater Hospital of the Chinese People's Liberation Army, Wuhan, 430070, China
| | - Zhi-Yong Zhang
- Department of General Surgery, The Central Theater Hospital of the Chinese People's Liberation Army, Wuhan, 430070, China
| | - Wei-Dong Jin
- Department of General Surgery, The Central Theater Hospital of the Chinese People's Liberation Army, Wuhan, 430070, China
| |
Collapse
|
4
|
Luo X, Ye Z, Xu C, Chen H, Dai S, Chen W, Bao G. Corosolic acid enhances oxidative stress-induced apoptosis and senescence in pancreatic cancer cells by inhibiting the JAK2/STAT3 pathway. Mol Biol Rep 2024; 51:176. [PMID: 38252208 DOI: 10.1007/s11033-023-09105-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: 09/26/2023] [Accepted: 12/05/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Pancreatic cancer (PC) is a fatal human malignancy with a poor prognosis. Corosolic acid (CRA) is a triterpenoid, has been reported to have inhibitory effects on tumor growth. However, the role of CRA on PC has not been explored. Here, we aimed to uncover the molecular mechanisms of CRA in PC progression. METHODS Cell viability, lactate dehydrogenase (LDH) release, cell apoptosis and senescence were detected by cell counting kit-8 (CCK-8), LDH, flow cytometry and senescence associated-β-galactosidase (SA-β-gal) assay. Levels of relevant proteins and oxidative stress (OS) markers were evaluated by Western blot and enzyme-linked immunosorbent assay (ELISA). A xenograft tumor model was established to explore the in vivo effects of CRA on PC. RESULTS We found that CRA inhibited PC cell viability and promoted LDH release in a dose-dependent manner, but had no significant effect on human normal pancreatic ductal epithelial cells HPDE6C7. CRA increased OS-induced cell apoptosis and senescence in HAPC and SW1990 cells. And CRA decreased the levels of anti-apoptotic protein Bcl-2, and elevated the expression of pro-apoptotic protein Bax and senescence-associated proteins P21 and P53. Besides, CRA decreased tumor growth in xenograft models. Furthermore, CRA inactivated the Janus kinase-2 (JAK2)/Signal Transducer and Activator of Transcription 3 (STAT3) signaling pathway in HAPC and SW1990 cells. Functional experiments demonstrated that activation of the JAK2/STAT3 pathway by the JAK2 activator coumermycin A1 (C-A1) or the STAT3 activator colivelin (col) reduced the contribution effect of OS, apoptosis and senescence by CRA. CONCLUSION Taken together, our findings indicated that CRA exerted anti-cancer effects in PC by inhibiting the JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Xu Luo
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Kunming, 650032, Yunnan Province, China
| | - Zhengchen Ye
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Kunming, 650032, Yunnan Province, China
| | - Chenglei Xu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Kunming, 650032, Yunnan Province, China
| | - Huan Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Kunming, 650032, Yunnan Province, China
| | - Shupeng Dai
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Kunming, 650032, Yunnan Province, China
| | - Weihong Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Kunming, 650032, Yunnan Province, China
| | - Guoqing Bao
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Kunming, 650032, Yunnan Province, China.
| |
Collapse
|
5
|
Wall NR, Fuller RN, Morcos A, De Leon M. Pancreatic Cancer Health Disparity: Pharmacologic Anthropology. Cancers (Basel) 2023; 15:5070. [PMID: 37894437 PMCID: PMC10605341 DOI: 10.3390/cancers15205070] [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: 09/20/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Pancreatic cancer (PCa) remains a formidable global health challenge, with high mortality rates and limited treatment options. While advancements in pharmacology have led to improved outcomes for various cancers, PCa continues to exhibit significant health disparities, disproportionately affecting certain populations. This paper explores the intersection of pharmacology and anthropology in understanding the health disparities associated with PCa. By considering the socio-cultural, economic, and behavioral factors that influence the development, diagnosis, treatment, and outcomes of PCa, pharmacologic anthropology provides a comprehensive framework to address these disparities and improve patient care.
Collapse
Affiliation(s)
- Nathan R. Wall
- Division of Biochemistry, Department of Basic Science, Center for Health Disparities and Molecular Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (R.N.F.); (A.M.)
| | - Ryan N. Fuller
- Division of Biochemistry, Department of Basic Science, Center for Health Disparities and Molecular Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (R.N.F.); (A.M.)
| | - Ann Morcos
- Division of Biochemistry, Department of Basic Science, Center for Health Disparities and Molecular Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (R.N.F.); (A.M.)
| | - Marino De Leon
- Division of Physiology, Department of Basic Science, Center for Health Disparities and Molecular Medicine, Loma Linda University, Loma Linda, CA 92350, USA;
| |
Collapse
|