1
|
Hu Y, Liao Y, Pan S, Zhou J, Wan C, Huang F, Bai Y, Lin C, Xia Q, Liu Z, Gong J, Nie X, Wang M, Qin R. A Triple-Mismatch Differentiating assay exploiting activation and trans cleavage of CRISPR-Cas12a for mutation detection with ultra specificity and sensitivity. Biosens Bioelectron 2025; 267:116826. [PMID: 39369517 DOI: 10.1016/j.bios.2024.116826] [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: 09/01/2024] [Revised: 09/22/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Liquid biopsy technology is non-invasive and convenient, and is currently an emerging technology for cancer screening. Among them, clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR associated protein 12a (Cas12a) based nucleic acid detection technology has the advantages of high sensitivity, rapidity, and easy operation. However, CRISPR-Cas12a does not discriminate single-base mismatches of targets well enough to meet the needs of clinical detection. Herein, we developed the Triple-Mismatch Differentiating (TMD) assay. This assay amplified the small thermodynamic difference in mismatches at one site at the level of CRISPR-Cas12a activation to a significant thermodynamic difference at three sites at both the level of CRISPR-Cas12a activation and trans-cleavage, which greatly improves the ability of CRISPR-Cas12a to discriminate between base mismatches. Our manipulation greatly improved the specificity of the CRISPR-Cas12a system while maintaining its inherent sensitivity and simplicity, increasing the detection limit to 0.0001%. When testing samples from pancreatic cancer patients, our results were highly consistent with NGS sequencing results. We believe that the TMD assay will provide a new technology for early cancer detection and will be widely used in the clinical practice.
Collapse
Affiliation(s)
- Yibo Hu
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yangwei Liao
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Shutao Pan
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jingcong Zhou
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Changqing Wan
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Feiyang Huang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yu Bai
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Chen Lin
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Qilong Xia
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zixi Liu
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jun Gong
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Xiaoqi Nie
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
2
|
Jiang X, Yuan Z, Ding T, Yu K, Dong J. SMS2 siRNA inhibits pancreatic tumor growth by tumor microenvironment modulation. Int Immunopharmacol 2024; 142:113111. [PMID: 39255679 DOI: 10.1016/j.intimp.2024.113111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 08/12/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024]
Abstract
The massive infiltration of suppressor immune cells within the tumor microenvironment (TME) of pancreatic ductal adenocarcinoma (PDAC) is a major cause of treatment resistance. Reducing this infiltration may represent a potentially effective therapeutic strategy. Sphingomyelin synthase 2 (SMS2) is a crucial enzyme for sphingomyelin synthesis, contributing significantly to the integrity and function of the plasma membrane. In this study, we developed a self-assembling SMS2 siRNA gene expression plasmid for in vivo delivery. The SMS2 siRNA specifically inhibits SMS2 expression while preserving the expression and activity of SMS1. Administration of the self-assembling SMS2 siRNA suppresses tumor growth in a murine model of Panc02 pancreatic carcinoma, modulates the polarization of tumor-associated macrophages (TAMs), and reduces the infiltration of tumor-associated neutrophils (TANs) by regulating the NF-κB/CXCL5 pathway. Consequently, utilizing SMS2 siRNA to improve the local immunosuppressive microenvironment holds promise for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Xin Jiang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, China
| | - Ziqing Yuan
- Experiment & Teaching Center, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Tingbo Ding
- Experiment & Teaching Center, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ker Yu
- Department of Pharmacology, School of Pharmacy, Fudan University, China.
| | - Jibin Dong
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, China.
| |
Collapse
|
3
|
Grabill N, Louis M, Cawthon M, Gherasim C, Chambers J. Diagnostic and therapeutic strategies in pancreatic adenosquamous carcinoma: Molecular and clinical insights in managing metastatic disease. Radiol Case Rep 2024; 19:6016-6026. [PMID: 39345848 PMCID: PMC11439409 DOI: 10.1016/j.radcr.2024.08.120] [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: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024] Open
Abstract
Adenosquamous carcinoma of the pancreas (ASCP) is a rare and aggressive variant of pancreatic cancer, characterized by both adenocarcinoma and squamous cell carcinoma components. It presents significant diagnostic and therapeutic challenges due to its atypical histology and poor prognosis. A 72-year-old male presented with abdominal pain, lighter-colored stools, and intermittent nausea. Initial imaging revealed a complex mass in the distal pancreatic body and tail. Elevated lipase levels and subsequent endoscopic ultrasound-guided fine-needle biopsy (EUS-FNB) suggested an atypical pancreatic lesion with keratinizing squamous cells. Further investigation through fiberoptic bronchoscopy and EBUS-guided transbronchial needle aspiration (TBNA) confirmed carcinoma with squamous differentiation. Genetic testing identified KRAS G12D and PIK3CA mutations. The multidisciplinary tumor board recommended systemic chemotherapy with mFOLFIRINOX and G-CSF support. The patient underwent twelve cycles of mFOLFIRINOX with dose adjustments for thrombocytopenia and effective management of chemotherapy-related side effects. Restaging CT scans showed a decrease in tumor size and stable metastatic nodes. The patient showed a partial biochemical response with decreasing CA 19-9 levels and disease stabilization on imaging. This case demonstrates the critical role of a multidisciplinary approach in managing rare pancreatic malignancies. ASCP requires a comprehensive diagnostic and therapeutic strategy involving advanced imaging, histopathological confirmation, and personalized chemotherapy. Integrating advanced diagnostic techniques, molecular profiling, and a multidisciplinary approach is essential for improving patient outcomes and providing comprehensive care for this challenging malignancy. Addressing the psychological aspects and offering compassionate care are vital for supporting patients through their treatment journey.
Collapse
Affiliation(s)
- Nathaniel Grabill
- Northeast Georgia Medical Center, General Surgery Department, Gainesville, GA 30501, USA
| | - Mena Louis
- Northeast Georgia Medical Center, General Surgery Department, Gainesville, GA 30501, USA
| | - Mariah Cawthon
- Northeast Georgia Medical Center, General Surgery Department, Gainesville, GA 30501, USA
| | - Claudia Gherasim
- Northeast Georgia Medical Center, Pathology Department, Gainesville, GA 30501, USA
| | - James Chambers
- Northeast Georgia Medical Center, General Surgery Department, Braselton, GA 30517, USA
| |
Collapse
|
4
|
Xiao M, Xue J, Jin E. SPOCK: Master regulator of malignant tumors (Review). Mol Med Rep 2024; 30:231. [PMID: 39392048 PMCID: PMC11487499 DOI: 10.3892/mmr.2024.13355] [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: 05/27/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024] Open
Abstract
SPARC/osteonectin, CWCV and Kazal‑like domain proteoglycan (SPOCK) is a family of highly conserved multidomain proteins. In total, three such family members, SPOCK1, SPOCK2 and SPOCK3, constitute the majority of extracellular matrix glycoproteins. The SPOCK gene family has been demonstrated to serve key roles in tumor regulation by affecting MMPs, which accelerates the progression of cancer epithelial‑mesenchymal transition. In addition, they can regulate the cell cycle via overexpression, inhibit tumor cell proliferation by inactivating PI3K/AKT signaling and have been associated with numerous microRNAs that influence the expression of downstream genes. Therefore, the SPOCK gene family are potential cancer‑regulating genes. The present review summarizes the molecular structure, tissue distribution and biological function of the SPOCK family of proteins, in addition to its association with cancer. Furthermore, the present review documents the progress made in investigations into the role of SPOCK, whilst also discussing prospects for the future of SPOCK‑targeted therapy, to provide novel ideas for clinical application and treatment.
Collapse
Affiliation(s)
- Mingyuan Xiao
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110134, P.R. China
| | - Jiancheng Xue
- Department of Otolaryngology, Head and Neck Surgery, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
- Shenzhen Clinical Research Center for Otolaryngology Diseases, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
| | - Enli Jin
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110134, P.R. China
| |
Collapse
|
5
|
Yang W, Hu P, Zuo C. Application of imaging technology for the diagnosis of malignancy in the pancreaticobiliary duodenal junction (Review). Oncol Lett 2024; 28:596. [PMID: 39430731 PMCID: PMC11487531 DOI: 10.3892/ol.2024.14729] [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: 04/15/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
The pancreaticobiliary duodenal junction (PBDJ) is the connecting area of the pancreatic duct, bile duct and duodenum. In a broad sense, it refers to a region formed by the head of the pancreas, the pancreatic segment of the common bile duct and the intraduodenal segment, the descending and the horizontal part of the duodenum, and the soft tissue around the pancreatic head. In a narrow sense, it refers to the anatomical Vater ampulla. Due to its complex and variable anatomical features, and the diversity of pathological changes, it is challenging to make an early diagnosis of malignancy at the PBDJ and define the histological type. The unique anatomical structure of this area may be the basis for the occurrence of malignant tumors. Therefore, understanding and subclassifying the anatomical configuration of the PBDJ is of great significance for the prevention and treatment of malignant tumors at their source. The present review comprehensively discusses commonly used imaging techniques and other new technologies for diagnosing malignancy at the PBDJ, offering evidence for physicians and patients to select appropriate examination methods.
Collapse
Affiliation(s)
- Wanyi Yang
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
- Graduates Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410013, P.R. China
| | - Pingsheng Hu
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
| | - Chaohui Zuo
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
- Graduates Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
6
|
Nopour R. Prediction of 12-month recurrence of pancreatic cancer using machine learning and prognostic factors. BMC Med Inform Decis Mak 2024; 24:339. [PMID: 39543603 PMCID: PMC11566389 DOI: 10.1186/s12911-024-02766-y] [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: 06/19/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND AND AIM Pancreatic cancer is lethal and prevalent among other cancer types. The recurrence of this tumor is high, especially in patients who did not receive adjuvant therapies. Early prediction of PC recurrence has a significant role in enhancing patients' prognosis and survival. So far, machine learning techniques have given us insight into favorable performance efficiency in various medical domains. So, this study aims to establish a prediction model based on machine learning to achieve better prediction on this topic. MATERIALS AND METHODS In this retrospective research, we used data from 585 PC patient cases from January 2019 to November 2023 from three clinical centers in Tehran City. Ten chosen ensemble and non-ensemble algorithms were used to establish prediction models on this topic. RESULTS Random forest and support vector machine with an AU-ROC of approximately 0.9 obtained more performance efficiency regarding PC recurrence. Lymph node metastasis, tumor size, tumor grade, radiotherapy, and chemotherapy were the best factors influencing PC recurrence. CONCLUSION Random forest and support vector machine algorithms demonstrated high-performance ability and clinical usability to improve doctors' decisions in achieving different therapeutic and diagnostic measures.
Collapse
Affiliation(s)
- Raoof Nopour
- Department of Health Information Management, Student Research Committee, School of Health Management and Information Sciences Branch, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Küçükekmekci B, Budak Yıldıran FA. Investigation of the efficacy of siRNA-mediated KRAS gene silencing in pancreatic cancer therapy. PeerJ 2024; 12:e18214. [PMID: 39553720 PMCID: PMC11566511 DOI: 10.7717/peerj.18214] [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: 02/06/2024] [Accepted: 09/11/2024] [Indexed: 11/19/2024] Open
Abstract
Aim Pancreatic carcinoma is an aggressive cancer that progresses without many symptoms. The difficulty of early diagnosis and an inadequate response to traditional treatments also cause the survival rate of pancreatic cancer to be low. Current research is focusing on methods of diagnosis and treatment, such as gene therapy, to increase survival rates. Small interfering ribonucleic acid (siRNA) has emerged as a promising advanced therapeutic strategy for cancer treatment. This study sought to silence the KRAS gene in the human pancreatic carcinoma cell line using a complex of small interfering ribonucleic acid (siRNA) and gold nanoparticles (AuNP). Methods In this study, 25 nM siRNA and gold nanoparticles at 0.5 mg/ml, 0.25 mg/ml, and 0.125 mg/ml concentrations were used to silence the KRAS gene in the CAPAN-1 cell line. Real-time PCR analysis, agarose gel electrophoresis, and double staining were carried out, and xCelligence real-time cell analysis (RTCA) was used to measure proliferation. Results The PCR analysis revealed crossing point (CP) values of actin beta (ACTB) ranging from 33.04 to 35.98, which was in the expected range for all samples. The interaction between the gold nanoparticle/siRNA complex in the double staining analysis revealed that the most effective concentration of gold nanoparticle was 0.125 mg/ml. The WST-1 technique showed that siRNA/AuPEI cells in application groups had a viability rate of over 90%, indicating no toxicity or side effects. The xCELLigence RTCA® showed that at hour 72, there was a significant difference in proliferation in the 0.5 mg/mL PEI/AuNP-siRNA, 0.25 mg/mL PEI/AuNP-siRNA, and 0.125 mg/mL PEI/AuNP-siRNA application groups compared to the control and siRNA groups (p < 0.05). By hour 96, all three groups were statistically different from the control and siRNA groups in terms of proliferation (p < 0.05). Conclusions The results of this analysis suggest that the AuPEI/siRNA complex can be effectively used to silence the target gene, but more studies are needed to verify these results.
Collapse
Affiliation(s)
- Büşra Küçükekmekci
- Institute of Sciences, Department of Biology, Kırıkkale University, Kırıkkale, Yahşihan, Turkey
| | - Fatma Azize Budak Yıldıran
- Vocational High School of Health Care Services, Department of Medical Services and Techniques, Kırıkkale University, Kırıkkale, Yahşihan, Turkey
| |
Collapse
|
8
|
Chen Y, Ye X, Hu M, Hu Y, Ding J. Long non-coding RNAs in pancreatic cancer. Clin Chim Acta 2024:120040. [PMID: 39536894 DOI: 10.1016/j.cca.2024.120040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
This article reviews the recent advances in pathogenesis, diagnosis and treatment of pancreatic cancer, as well as the relationship between long non-coding RNA (lncRNA) in disease progression. Unfortunately, pancreatic cancer has no early symptoms and quickly invades surrounding tissue and organs, making it one of the deadliest. Accordingly, we urgently need to identify high-risk individuals with precancerous lesions through screening methods to identify early disease, provide better prevention strategies and improve overall survival. LncRNAs have a variety of biological functions in both physiologic and pathophysiologic states including tumor growth, differentiation and proliferation. Herein we review the biological functions, expression patterns, clinical significance and targeted therapy potential of lncRNAs to provide new approaches for diagnosis and treatment in pancreatic cancer.
Collapse
Affiliation(s)
- Yuan Chen
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China
| | - Xiaohua Ye
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China
| | - Minli Hu
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China
| | - Yibing Hu
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China
| | - Jin Ding
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China.
| |
Collapse
|
9
|
González Abós C, Lorenzo C, Archilla I, Vidal-Robau N, Cuatrecasas M, Ausania F. Effect of warm and cold ischemia on pancreaticoduodenectomy specimen following robotic pancreaticoduodenectomy. BMC Surg 2024; 24:346. [PMID: 39501227 PMCID: PMC11536913 DOI: 10.1186/s12893-024-02652-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/30/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND The adoption of robotic pancreaticoduodenectomy (PD) has increased in recent years for the treatment of pancreatic head tumors and periampullary lesions. Some potential benefits seem to be demonstrated; however, obtaining specimens through this method can potentially compromise the diagnosis depending on the timing of the specimen retrieval, and the impact of longer perioperative time on ischemia and autolysis of the surgical specimen has not been analyzed. The aim of this study is to evaluate the histological changes associated with timing of specimen retrieval during robotic PD. METHODS A review of histopathology files was performed for all pancreatic specimens collected at our hospital from January 2022 to March 2024. Both warm ischemia time (WIT) and cold ischemia time (CIT) were collected. Histological features related to ischemic damage were evaluated in normal duodenal and pancreatic parenchyma as well as pancreatic tumor, and were graded as: absent, mild, moderate and severe. Univariate and multivariate analyses were performed to determine which variables were associated with moderate and severe ischemic changes. RESULTS Sixty surgical specimens were analyzed: 20 open PD, 17 robotic PD with cold ischemia, and 23 robotic PD. Median total WIT was 182 min (open PD 57 min vs. RPD 190 min vs. RPD-CI 198 min; p < 0.001). Median CIT was 760 min (740-835) in samples stored at 4ºC. Univariate analysis showed that longer intraoperative time, male gender and cold ischemia were associated with pancreatic tissue ischemic changes. In multivariate analysis, cold ischemia was the only independent factor associated with normal pancreatic tissue and tumor tissue moderate and severe ischemic changes. CONCLUSIONS Prolonged ischemia time, especially in the case of cold storage, has a strong effect on the degradation of normal and tumor tissue without affecting pathological evaluation. Operative teams should aim to optimize both the duration and efficiency of the surgical procedure, ensuring minimal ischemic time. Simultaneously, pathology departments must be equipped to process pancreatic specimens promptly, with protocols in place to minimize the time between resection and analysis.
Collapse
Affiliation(s)
- Carolina González Abós
- Department of HBP and Transplant Surgery, Hospital Clinic de Barcelona, C/ Villarroel 170, Barcelona, 08036, Spain.
- University of Barcelona, Barcelona, Spain.
- Gene Therapy and Cancer, IDIBAPS, Barcelona, Spain.
| | - Claudia Lorenzo
- Department of HBP and Transplant Surgery, Hospital Clinic de Barcelona, C/ Villarroel 170, Barcelona, 08036, Spain
| | - Iván Archilla
- Department of Pathology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Nuria Vidal-Robau
- Department of Pathology, Hospital Clinic de Barcelona, Barcelona, Spain
| | | | - Fabio Ausania
- Department of HBP and Transplant Surgery, Hospital Clinic de Barcelona, C/ Villarroel 170, Barcelona, 08036, Spain
- University of Barcelona, Barcelona, Spain
- Gene Therapy and Cancer, IDIBAPS, Barcelona, Spain
| |
Collapse
|
10
|
Limbu KR, Chhetri RB, Kim S, Shrestha J, Oh YS, Baek DJ, Park EY. Targeting sphingosine 1-phosphate and sphingosine kinases in pancreatic cancer: mechanisms and therapeutic potential. Cancer Cell Int 2024; 24:353. [PMID: 39462385 PMCID: PMC11514880 DOI: 10.1186/s12935-024-03535-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
Pancreatic cancer is known to be the most lethal cancer. Fewer new treatments are being developed for pancreatic cancer as compared to other cancers. The bioactive lipid S1P, which is mainly regulated by sphingosine kinase 1 (SK1) and sphingosine kinase 2 (SK2) enzymes, plays significant roles in pancreatic cancer initiation and exacerbation. S1P controls many signaling pathways to modulate the progression of pancreatic cancer through the G-coupled receptor S1PR1-5. Several papers reporting amelioration of pancreatic cancer via modulation of S1P levels or downstream signaling pathways have previously been published. In this paper, for the first time, we have reviewed the results of previous studies to understand how S1P and its receptors contribute to the development of pancreatic cancer, and whether S1P can be a therapeutic target. In addition, we have also reviewed papers dealing with the effects of SK1 and SK2, which are kinases that regulate the level of S1P, on the pathogenesis of pancreatic cancer. We have also listed available drugs that particularly focus on S1P, S1PRs, SK1, and SK2 for the treatment of pancreatic cancer. Through this review, we would like to suggest that the SK/S1P/S1PR signaling system can be an important target for treating pancreatic cancer, where a new treatment target is desperately warranted.
Collapse
Affiliation(s)
- Khem Raj Limbu
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea
| | | | - Subin Kim
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea
| | - Jitendra Shrestha
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA
| | - Yoon Sin Oh
- Department of Food and Nutrition, Eulji University, Seongnam, 13135, South Korea
| | - Dong Jae Baek
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea.
| | - Eun-Young Park
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea.
| |
Collapse
|
11
|
Groß T, Merboth F, Klimowa A, Kahlert C, Distler M, Weitz J, Welsch T, Müssle B. Impact of an enhanced anti-infection prophylaxis strategy for pancreatoduodenectomy: a single centre analysis. Langenbecks Arch Surg 2024; 409:307. [PMID: 39402424 PMCID: PMC11473572 DOI: 10.1007/s00423-024-03465-y] [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: 04/22/2024] [Accepted: 09/01/2024] [Indexed: 10/19/2024]
Abstract
INTRODUCTION : Surgical site infection (SSI) after pancreatoduodenectomy (PD) is a significant concern. Targeted antibiotic prophylaxis (pAP) has been tested to mitigate antibiotic resistance patterns, especially after preoperative bile duct stenting. The aim of this study was to investigate the effect of enhanced anti-infective prophylaxis (EAP) on the incidence of superficial and intraabdominal SSI. METHODS All patients who underwent PD at a single centre between May 2018 and May 2021 were retrospectively analysed. A control cohort of patients who received pAP with intravenous cefuroxime and metronidazole and routine intraoperative abdominal lavage according to the surgeons' preferences. Since March 2020, pAP has been changed to piperacillin/tazobactam according to local resistance patterns and combined with routine intraoperative extended abdominal lavage (EIPL). Preoperative selective decontamination of the digestive tract (SDD) has been applied routinely since Jan 2019. RESULTS In total, 163 patients were included. The standard (n = 100) and EAP (n = 63) groups did not significantly differ with regard to pertinent patient and operative characteristics. In the EAP group, the rates of SSI (14% vs. 37%, p = 0.002, total rate: 28%) and urinary tract infection (24% vs. 8%, p = 0.011, total rate 18%) were significantly lower. Other septic complications were not significantly different. In addition, the risk of developing gastrointestinal bleeding and delayed gastric emptying was significantly lower in the EAP group. Multivariate analysis showed that an age > 67 years was a significant risk factor for SSI. CONCLUSION The results indicate that enhanced anti-infective prophylaxis may significantly decrease the incidence of SSI in patients after PD.
Collapse
Affiliation(s)
- Tina Groß
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Ependorf, Hamburg, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Felix Merboth
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
| | - Anna Klimowa
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
| | - Christoph Kahlert
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidberg, Heidelberg, Germany
| | - Marius Distler
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Thilo Welsch
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Ependorf, Hamburg, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Benjamin Müssle
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- Department of General and Visceral Surgery, University Hospital Ulm, Ulm, Germany.
| |
Collapse
|
12
|
Bahrami A, Mohammadzadeh M, Abdi F, Paydareh A, Khalesi S, Hejazi E. Calcium Intake and the Pancreatic Cancer Risk: A Systematic Review and Meta-Analysis of Observational Studies. Clin Nutr Res 2024; 13:284-294. [PMID: 39526207 PMCID: PMC11543454 DOI: 10.7762/cnr.2024.13.4.284] [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: 05/24/2024] [Revised: 08/27/2024] [Accepted: 09/12/2024] [Indexed: 11/16/2024] Open
Abstract
Calcium plays a major role in apoptosis, cell proliferation, and various cellular mechanisms. It is also essential for the function of the pancreas. However, the association between calcium intake and pancreatic cancer is not clear. This study aims to clarify the links between calcium intake and pancreatic cancer risk using a systematic review and meta-analysis of observational studies. PubMed, Web of Science, Scopus, and Google Scholar were searched for eligible articles published through 31 August 2023. Case-control and cohort studies reporting the association between dietary and/or supplemental calcium intake and risk of pancreatic cancer using relative risk (RR), hazard ratio (HR), or odds ratio (OR) with 95% confidence interval (CI) were included. Meta-analysis using a random effect model was used to estimate the significance of the association. Eight studies were included. An inverse association between total calcium intake (dietary and supplement) and pancreatic cancer risk (RR, 0.83; 95% CI, 0.72-0.97; I2 = 0%) was observed. However, the association between dietary calcium intake alone and pancreatic cancer risk did not reach a statistically significant level (RR, 0.91; 95% CI, 0.78-1.06; I2 = 48%). Higher total calcium intake may reduce the risk of pancreatic cancer but the difference between sources of calcium (dietary vs. supplementation) requires further investigation. Also, due to the heterogeneity between the articles, the results of this study should be interpreted with caution. Trial Registration PROSPERO Identifier: CRD42022331647.
Collapse
Affiliation(s)
- Alireza Bahrami
- Student Research Committee, Department and Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran 4739-19395, Iran
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran 4739-19395, Iran
| | - Milad Mohammadzadeh
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran 4739-19395, Iran
| | - Fatemeh Abdi
- Department of Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz 5166614711, Iran
| | - Amin Paydareh
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran 4739-19395, Iran
| | - Saman Khalesi
- Appleton Institute and School of Health, Medical and Applied Sciences, Central Queensland University, Brisbane 4701, Australia
| | - Ehsan Hejazi
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran 4739-19395, Iran
| |
Collapse
|
13
|
Sung YN, Stojanova M, Shin S, Cho H, Heaphy CM, Hong SM. Gradual telomere shortening in the tumorigenesis of pancreatic and hepatic mucinous cystic neoplasms. Hum Pathol 2024; 152:105653. [PMID: 39214240 DOI: 10.1016/j.humpath.2024.105653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Mucinous cystic neoplasm (MCN) is one of the precursor lesions of pancreatic ductal adenocarcinoma and intrahepatic cholangiocarcinoma. The aim of this study is to examine the presence of short telomeres in promoting the tumorigenesis of MCN by measuring telomere lengths in distinct components of MCN, including the mucinous lining epithelium, non-mucinous lining epithelium, and ovarian-type stroma. A total of 45 patients with MCN (30 pancreatic and 15 hepatic cases) were obtained. Quantitative telomere-specific fluorescent in situ hybridization was performed to measure the telomere length of specific cell types within MCNs, including mucinous lining epithelium, non-mucinous lining epithelium, and ovarian-type stroma, as well as normal ductal epithelium and adenocarcinoma. Relative telomere lengths tended to decrease between normal ductal epithelium, ovarian-type stroma, non-mucinous lining epithelium, mucinous lining epithelium, and adenocarcinoma regardless of the involved organs. Among the analyzed cell types, relative telomere lengths were significantly different between normal ductal epithelium (3.31 ± 0.78), ovarian-type stroma (2.90 ± 0.93), non-mucinous lining epithelium (2.84 ± 0.79), mucinous lining epithelium (2.49 ± 0.93), and adenocarcinoma (1.19 ± 0.59), respectively (P < 0.001, mixed-effects model). As expected, no difference in relative telomere lengths was observed between normal ductal epithelium and ovarian-type stroma; however, significant differences were observed in pair-wise comparisons between ovarian-type stroma vs. non-mucinous lining epithelium (P = 0.001), non-mucinous lining epithelium vs. mucinous lining epithelium (P = 0.005), and mucinous lining epithelium vs. adenocarcinoma (P < 0.001). These findings suggest gradual telomere shortening occurs in the tumorigenesis of MCN, which may have important implications for the progression of this disease.
Collapse
Affiliation(s)
- You-Na Sung
- Department of Pathology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Marija Stojanova
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Seungbeom Shin
- Department of Statistics, Korea University, Seoul, Republic of Korea
| | - HyungJun Cho
- Department of Statistics, Korea University, Seoul, Republic of Korea
| | - Christopher M Heaphy
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
14
|
Son B, Lee W, Kim H, Shin H, Park HH. Targeted therapy of cancer stem cells: inhibition of mTOR in pre-clinical and clinical research. Cell Death Dis 2024; 15:696. [PMID: 39349424 PMCID: PMC11442590 DOI: 10.1038/s41419-024-07077-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 10/02/2024]
Abstract
Cancer stem cells (CSCs) are a type of stem cell that possesses not only the intrinsic abilities of stem cells but also the properties of cancer cells. Therefore, CSCs are known to have self-renewal and outstanding proliferation capacity, along with the potential to differentiate into specific types of tumor cells. Cancers typically originate from CSCs, making them a significant target for tumor treatment. Among the related cascades of the CSCs, mammalian target of rapamycin (mTOR) pathway is regarded as one of the most important signaling pathways because of its association with significant upstream signaling: phosphatidylinositol 3‑kinase/protein kinase B (PI3K/AKT) pathway and mitogen‑activated protein kinase (MAPK) cascade, which influence various activities of stem cells, including CSCs. Recent studies have shown that the mTOR pathway not only affects generation of CSCs but also the maintenance of their pluripotency. Furthermore, the maintenance of pluripotency or differentiation into specific types of cancer cells depends on the regulation of the mTOR signal in CSCs. Consequently, the clinical potential and importance of mTOR in effective cancer therapy are increasing. In this review, we demonstrate the association between the mTOR pathway and cancer, including CSCs. Additionally, we discuss a new concept for anti-cancer drug development aimed at overcoming existing drawbacks, such as drug resistance, by targeting CSCs through mTOR inhibition.
Collapse
Affiliation(s)
- Boram Son
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul, 02707, Republic of Korea
| | - Wonhwa Lee
- Department of Chemistry, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyeonjeong Kim
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea.
| | - Hee Ho Park
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea.
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
15
|
Bhat AA, Afzal M, Moglad E, Thapa R, Ali H, Almalki WH, Kazmi I, Alzarea SI, Gupta G, Subramaniyan V. lncRNAs as prognostic markers and therapeutic targets in cuproptosis-mediated cancer. Clin Exp Med 2024; 24:226. [PMID: 39325172 PMCID: PMC11427524 DOI: 10.1007/s10238-024-01491-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as crucial regulators in various cellular processes, including cancer progression and stress response. Recent studies have demonstrated that copper accumulation induces a unique form of cell death known as cuproptosis, with lncRNAs playing a key role in regulating cuproptosis-associated pathways. These lncRNAs may trigger cell-specific responses to copper stress, presenting new opportunities as prognostic markers and therapeutic targets. This paper delves into the role of lncRNAs in cuproptosis-mediated cancer, underscoring their potential as biomarkers and targets for innovative therapeutic strategies. A thorough review of scientific literature was conducted, utilizing databases such as PubMed, Google Scholar, and ScienceDirect, with search terms like 'lncRNAs,' 'cuproptosis,' and 'cancer.' Studies were selected based on their relevance to lncRNA regulation of cuproptosis pathways and their implications for cancer prognosis and treatment. The review highlights the significant contribution of lncRNAs in regulating cuproptosis-related genes and pathways, impacting copper metabolism, mitochondrial stress responses, and apoptotic signaling. Specific lncRNAs are potential prognostic markers in breast, lung, liver, ovarian, pancreatic, and gastric cancers. The objective of this article is to explore the role of lncRNAs as potential prognostic markers and therapeutic targets in cancers mediated by cuproptosis.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al Kharj, Saudi Arabia
| | - Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
16
|
Lencioni G, Gregori A, Toledo B, Rebelo R, Immordino B, Amrutkar M, Xavier CPR, Kocijančič A, Pandey DP, Perán M, Castaño JP, Walsh N, Giovannetti E. Unravelling the complexities of resistance mechanism in pancreatic cancer: Insights from in vitro and ex-vivo model systems. Semin Cancer Biol 2024; 106-107:217-233. [PMID: 39299411 DOI: 10.1016/j.semcancer.2024.09.002] [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: 07/19/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with poor prognosis and rising global deaths. Late diagnosis, due to absent early symptoms and biomarkers, limits treatment mainly to chemotherapy, which soon encounters resistance. PDAC treatment innovation is hampered by its complex and heterogeneous resistant nature, including mutations in key genes and a stromal-rich, immunosuppressive tumour microenvironment. Recent studies on PDAC resistance stress the need for suitable in vitro and ex vivo models to replicate its complex molecular and microenvironmental landscape. This review summarises advances in these models, which can aid in combating chemoresistance and serve as platforms for discovering new therapeutics. Immortalised cell lines offer homogeneity, unlimited proliferation, and reproducibility, but while many gemcitabine-resistant PDAC cell lines exist, fewer models are available for resistance to other drugs. Organoids from PDAC patients show promise in mimicking tumour heterogeneity and chemosensitivity. Bioreactors, co-culture systems and organotypic slices, incorporating stromal and immune cells, are being developed to understand tumour-stroma interactions and the tumour microenvironment's role in drug resistance. Lastly, another innovative approach is three-dimensional bioprinting, which creates tissue-like structures resembling PDAC architecture, allowing for drug screening. These advanced models can guide researchers in selecting optimal in vitro tests, potentially improving therapeutic strategies and patient outcomes.
Collapse
Affiliation(s)
- Giulia Lencioni
- Fondazione Pisana per La Scienza, San Giuliano Terme, Italy; Department of Biology, University of Pisa, Pisa, Italy
| | - Alessandro Gregori
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Belén Toledo
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain
| | - Rita Rebelo
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto 4200-135, Portugal; Cancer Drug Resistance Group, Institute of Molecular Pathology and Immunology (IPATIMUP), University of Porto, Porto 4200-135, Portugal; Department of Biological Sciences, Faculty of Pharmacy of the University of Porto (FFUP), Porto, Portugal
| | - Benoît Immordino
- Fondazione Pisana per La Scienza, San Giuliano Terme, Italy; Institute of Life Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Manoj Amrutkar
- Department of Pathology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Cristina P R Xavier
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto 4200-135, Portugal; Cancer Drug Resistance Group, Institute of Molecular Pathology and Immunology (IPATIMUP), University of Porto, Porto 4200-135, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, Gandra, Portugal
| | - Anja Kocijančič
- Centre for Embryology and Healthy Development, Department of Microbiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Deo Prakash Pandey
- Centre for Embryology and Healthy Development, Department of Microbiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| | - Justo P Castaño
- Maimonides Biomedical Research Institute 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
| | - Naomi Walsh
- Life Sciences Institute, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Elisa Giovannetti
- Fondazione Pisana per La Scienza, San Giuliano Terme, Italy; Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
17
|
Fu X, Ma J, Ma F, Guo S, Wang X, Li Y, Tang Y, Qi J, Zhang W, Ye L. MISP-mediated enhancement of pancreatic cancer growth through the Wnt/β-catenin signaling pathway is suppressed by Fisetin. Biochim Biophys Acta Mol Basis Dis 2024:167515. [PMID: 39278512 DOI: 10.1016/j.bbadis.2024.167515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
Pancreatic cancer is a highly malignant tumor characterized by high mortality and low survival rates. The mitotic interactor and substrate of Plk1 (MISP) is a cancer-associated protein that regulates mitotic spindle localization and is highly expressed in several malignant tumors, contributing to tumor development. However, the function and regulatory mechanisms of MISP in pancreatic cancer remain unclear. In this study, we analyzed RNA sequencing data related to pancreatic cancer from the TCGA and GEO databases, identifying MISP as a potential prognostic marker for the disease. MISP was significantly upregulated in pancreatic cancer cells and tissues compared to normal pancreatic cells and tissues. Notably, in pancreatic cancer cells, high MISP protein expression promoted cell proliferation and growth. Mechanistically, the upregulation of MISP facilitated the nuclear accumulation of β-catenin, thereby activating the Wnt/β-catenin signaling pathway and promoting pancreatic cancer growth. In search of effective inhibitors of MISP expression, we screened an FDA-approved drug library and identified Fisetin as a potential suppressor of MISP expression. Fisetin was found to downregulate the transcription factor MYB, thereby reducing MISP expression. Further experiments demonstrated that Fisetin effectively inhibited the in vitro and in vivo growth of pancreatic cancer by suppressing the MISP/Wnt/β-catenin signaling axis. In summary, our research has identified MISP as a novel therapeutic target in pancreatic cancer and uncovered its associated regulatory mechanisms.
Collapse
Affiliation(s)
- Xueli Fu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jiaqi Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Fangyuan Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shiman Guo
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xue Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ye Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yanxin Tang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jingwei Qi
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90001, USA
| | - Weiying Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| | - Lihong Ye
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
18
|
Wu Y, Zhang J, Tian Y, Chi Shing Cho W, Xu HX, Lin ZX, Xian YF. 20(S)-Ginsenoside Rh2 overcomes gemcitabine resistance in pancreatic cancer by inhibiting LAMC2-Modulated ABC transporters. J Adv Res 2024:S2090-1232(24)00390-4. [PMID: 39270979 DOI: 10.1016/j.jare.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/05/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
INTRODUCTION Gemcitabine (GEM) is the first-line drug for pancreatic ductal adenocarcinoma (PDAC), but drug resistance severely restricts its chemotherapeutic efficacy. Laminin subunit γ2 (LAMC2) plays a crucial role in extracellular matrix formation in the development of GEM-resistance. However, the biological function of LAMC2 in GEM resistance and its molecular mechanisms are still unclear. 20(S)-Ginsenoside Rh2 (Rh2), one of the principal active components isolated from Ginseng Radix et Rhizoma, possesses strong anti-tumor effects. However, the effects of Rh2 on overcoming GEM resistance and its action mechanisms remain to be elucidated. OBJECTIVES This study aimed to determine the efficacy of Rh2 on overcoming GEM resistance and to explore its underlying molecular mechanisms. METHODS Clinical study, Western blotting, publicly available databasesand bioinformatic analyses were performed to investigate the protein expression of LAMC2 in the GEM-resistant PDAC patients and the acquired GEM-resistant PDAC cells. Then, the effects of Rh2 on overcoming the GEM resistance in PDAC were evaluated both in vitro and in vivo. Stable silencing or overexpression of LAMC2 in the GEM-resistant PDAC cells were established for validating the role of LAMC2 on Rh2 overcoming the GEM resistance in PDAC. RESULTS The protein expression of LAMC2 was markedly increased in the GEM-resistant PDAC patient biopsies compared to the sensitive cases. The protein expression of LAMC2 was significantly higher in the acquired GEM-resistant PDAC cells than that in their parental cells. Rh2 enhanced the chemosensitivity of GEM in the GEM-resistant PDAC cells, and inhibited the tumor growth of Miapaca-2-GR cell-bearing mice and Krastm4TyjTrp53tm1BrnTg (Pdx1-cre/Esr1*) #Dam/J (KPC) mice. Rh2 effectively reversed the GEM resistance in Miapaca-2-GR and Capan-2-GR cells by inhibiting LAMC2 expression through regulating the ubiquitin-proteasome pathway. Knockdown of LAMC2 enhanced the chemosensitivity of GEM and the effects of Rh2 on overcoming the GEM resistance in PDAC cells and the orthotopic PDAC mouse model. Conversely, LAMC2 overexpression aggravated the chemoresistance of GEM and abolished the effects of Rh2 on overcoming GEM resistance via modulating ATP-binding cassette (ABC) transporters leading to the active GEM efflux. CONCLUSIONS LAMC2 plays an important role in the GEM resistance in PDAC, and Rh2 is a potential adjuvant for overcoming the chemoresistance of GEM in PDAC.
Collapse
Affiliation(s)
- Yulin Wu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Juan Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Yuanyang Tian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - William Chi Shing Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China; Department of Pathology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China
| | - Hong-Xi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China; Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China; The Chinese University of Hong Kong Chinese Medicine Specialty Clinic cum Clinical Teaching and Research Centre, School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| | - Yan-Fang Xian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China; The Chinese University of Hong Kong Chinese Medicine Specialty Clinic cum Clinical Teaching and Research Centre, School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
19
|
Balcer K, Garnier J, Richa Y, Bruneel-Zupanc C, Piessen G, Turrini O, Truant S, El Amrani M. Impact on survival of sarcopenia, systemic inflammatory response and anthropometric factors after pancreatectomy for resectable pancreatic adenocarcinoma. World J Surg Oncol 2024; 22:232. [PMID: 39232731 PMCID: PMC11376042 DOI: 10.1186/s12957-024-03510-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/25/2024] [Indexed: 09/06/2024] Open
Abstract
INTRODUCTION Pancreatic adenocarcinoma (PDAC) is becoming a public health issue with a 5-years survival rate around 10%. Patients with PDAC are often sarcopenic, which impacts postoperative outcome. At the same time, overweight population is increasing and adipose tissue promotes tumor related-inflammation. With several studies supporting independently these data, we aimed to assess if they held an impact on survival when combined. METHODS We included 232 patients from two university hospitals (CHU de Lille, Institut Paoli Calmette), from January 2011 to December 2018, who underwent Pancreaticoduodenectomy (PD) for resectable PDAC. Preoperative CT scan was used to measure sarcopenia and visceral fat according to international cut-offs. Neutrophil to lymphocyte (NLR) and platelet to lymphocyte ratios (PLR) were used to measure inflammation. For univariate and multivariate analyses, the Cox proportional-hazard model was used. P-values below 0.05 were considered significant. RESULTS Sarcopenic patients with visceral obesity were less likely to survive than the others in multivariate analysis (OS, HR 1.65, p= 0.043). Cutaneous obesity did not influence survival. We also observed an influence on survival when we studied sarcopenia with visceral obesity (OS, p= 0.056; PFS, p = 0.014), sarcopenia with cutaneous obesity (PFS, p= 0.005) and sarcopenia with PLR (PFS, p= 0.043). This poor prognosis was also found in sarcopenic obese patients with high PLR (OS, p= 0.05; PFS, p= 0.01). CONCLUSION Sarcopenic obesity was associated with poor prognosis after PD for PDAC, especially in patients with systemic inflammation. Pre operative management of these factors should be addressed in pancreatic cancer patients.
Collapse
Affiliation(s)
- Kaja Balcer
- Digestive Surgery and Transplantation Department, CHU de Lille, University of Lille, Lille, 59000, France.
| | - Jonathan Garnier
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, 13009, France
| | - Yasmina Richa
- School of Medicine, University College Cork, Cork, Ireland
| | | | - Guillaume Piessen
- Department of Digestive and Oncological Surgery, University of Lille, CNRS, INSERM, CHU Lille, University of Lille, Lille, 59000, France
| | - Olivier Turrini
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, 13009, France
| | - Stephanie Truant
- Digestive Surgery and Transplantation Department, CHU de Lille, University of Lille, Lille, 59000, France
| | - Mehdi El Amrani
- Digestive Surgery and Transplantation Department, CHU de Lille, University of Lille, Lille, 59000, France
| |
Collapse
|
20
|
Mahmood S, Sameer HM, Ejaz A, Ahsan SM, Mazhar U, Zulfiqar K. Rising Mortality among Non-Hispanics due to Pancreatic Cancer in the United States. A CDC Database analysis. J Gastrointest Cancer 2024; 55:1229-1238. [PMID: 38888729 DOI: 10.1007/s12029-024-01084-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2024] [Indexed: 06/20/2024]
Abstract
PURPOSE To examine mortality trends among non-Hispanic (NH) adults with pancreatic cancer. METHOD CDC-WONDER database was used to extract death certificate data on pancreatic cancer-related mortality in NH adults aged ≥ 45 from 1999 to 2020. Age-adjusted mortality rates (AAMRs) per 100,000 persons and annual percent changes (APCs) were calculated and stratified by year, age, sex, race, and region. RESULTS From 1999 to 2020, 783,772 deaths occurred among middle-aged (45-64) and older (65-85 +) NH adults. Overall AAMR increased from 31.7 in 1999 to 33.8 in 2020 (APC: 0.35; 95% CI:0.28-0.41). NH older adults had higher AAMRs (67.9) than NH middle-aged adults (12.5). Men consistently had higher AAMRs (37.7) than women (28.4). NH African Americans had the highest AAMRs (40.8) compared to NH Whites (32.1), NH American Indians (23.9), and NH Asians (22.4). Metropolitan areas had a higher AAMR (32.7) than non-metropolitan areas (32.2). The Northeast region had the highest AAMR (34.0) followed by Midwest (33.2), South (32.2), and West (30.1). Delaware, District of Columbia, Louisiana, Michigan, and Mississippi had the highest AAMRs among states. CONCLUSIONS Pancreatic cancer-related mortality among NH adults has increased from 1999 to 2020. Highest AAMRs were reported in older men, NH African Americans, the Northeastern and metropolitan areas.
Collapse
Affiliation(s)
- Samar Mahmood
- Department of Internal Medicine, Dow University of Health Sciences, Mission Rd, New Labour Colony Nanakwara, Karachi, 74200, Pakistan.
| | | | - Arooba Ejaz
- Department of Internal Medicine, Dow University of Health Sciences, Mission Rd, New Labour Colony Nanakwara, Karachi, 74200, Pakistan
| | | | - Urooj Mazhar
- Department of Medicine, Liaquat National Medical College, Karachi, Pakistan
| | - Komal Zulfiqar
- Department of Medicine, Allama Iqbal Medical College, Lahore, Pakistan
| |
Collapse
|
21
|
Lyu SI, Fretter C, Simon AG, Spielmann SM, Damanakis AI, Zhao Y, Bruns CJ, Schmidt T, Popp FC, Waldschmidt D, Quaas A, Knipper K. Extent and clinical significance of the therapy-relevant tight junction protein Claudin 18.2 in pancreatic ductal adenocarcinoma - real-world evidence. Transl Oncol 2024; 47:102044. [PMID: 38917592 PMCID: PMC11255103 DOI: 10.1016/j.tranon.2024.102044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/28/2024] [Accepted: 06/20/2024] [Indexed: 06/27/2024] Open
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is associated with poor prognosis, wherefore targeted therapies have experienced increasing interest. Zolbetuximab is a novel targeted therapy under investigation in patients with PDAC and targets Claudin 18.2 (CLDN18.2), which is a component of tight junctions and is of significance in various solid tumors. As its role in PDAC is not definitively elucidated, this study aims to clarify the significance of CLDN18.2 expression in PDAC in a real-world setting. METHODS All patients (n = 309) were recruited at one of the PANCALYZE study centers and received pancreatic resection with curative intention. Paraffin samples were analyzed using an antibody against CLDN18.2, which is known to be comparable to the antibody used by the SPOTLIGHT and GLOW studies. RESULTS 94 PDACs are positive for CLDN18.2 (30.4 %). Positive CLDN 18.2 expression was associated with significantly better cancer differentiation (p < 0.001). Patients with positive CLDN18.2 expression showed significantly better overall survival when compared to patients with negative expression (median OS: 30 versus 18 months, p = 0.003). Additionally, in multivariable analyses, CLDN18.2 expression was identified as an independent factor for better survival in patients with PDAC (HR = 0.686, 95 %CI = 0.492-0.956, p = 0.026). CONCLUSION Significant improvement in survival could be demonstrated by adding Zolbetuximab to known chemotherapy regimes in patients with gastro-esophageal junction adenocarcinoma with at least 75 % CLDN18.2 positive cancer cells. Our findings demonstrate, that 30.4 % of the included patients with PDAC would potentially be eligible for therapy with Zolbetuximab in a real-world patient cohort. Results of trials targeting Claudin 18.2 are pending in patients with PDAC.
Collapse
Affiliation(s)
- Su Ir Lyu
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Caroline Fretter
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Adrian Georg Simon
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Sarah-Michele Spielmann
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Alexander I Damanakis
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Yue Zhao
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Christiane J Bruns
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Thomas Schmidt
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Felix C Popp
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Dirk Waldschmidt
- Faculty of Medicine and University Hospital of Cologne, Department of Gastroenterology and Hepatology, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Karl Knipper
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany.
| |
Collapse
|
22
|
Rechner LA, Felter M, Bekke S, Biancardo S, Rønjom MF, Pedersen M, Sjöström D, Chen IM, Sibolt P. Transition of Online Adaptive Stereotactic Radiotherapy for Pancreatic Cancer From Magnetic Resonance-Guided Linear Accelerator (MR-Linac) to State-of-the-Art Cone-Beam Computed Tomography (CBCT). Cureus 2024; 16:e68386. [PMID: 39355470 PMCID: PMC11444803 DOI: 10.7759/cureus.68386] [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] [Accepted: 09/01/2024] [Indexed: 10/03/2024] Open
Abstract
Pancreatic cancer is one of the most challenging tumor sites to treat safely and effectively with radiotherapy due to the anatomical location and aggressiveness of the disease. One modality that has shown promising results, which our institution has been employing, is online adaptive stereotactic radiotherapy using a magnetic resonance-guided linear accelerator (MR-linac). However, due to unforeseen circumstances regarding our MR-linac, it was necessary for our institution to use an alternative treatment technique. In this case report, we describe our experience using our ring-gantry linac equipped with an advanced cone-beam computed tomography (CBCT) system to treat a 61-year-old patient with advanced pancreatic cancer with CBCT-guided online adaptive stereotactic radiotherapy. In a short time period of four weeks, we prepared for this case by training with the surface scanning motion management system and developing procedures for planning and adaptation. The patient was prescribed 24 Gy in three fractions, with a risk-adapted approach using strict organ-at-risk (OAR) constraints. Daily CBCT was used for online adaptation of the plan, and the superior plan (non-adapted or adapted) was selected for treatment. For this patient, the adapted plan was chosen for all three fractions, due to OAR constraints being violated in the non-adapted plan. In summary, we found that for this patient, high-quality CBCT guidance for daily re-contouring, in combination with motion management, enabled the use of daily adaptive radiotherapy to safely deliver stereotactic radiotherapy. The results from this case report are promising, and CBCT-guided online adaptive stereotactic radiotherapy for pancreatic cancer warrants further investigation in more patients.
Collapse
Affiliation(s)
- Laura A Rechner
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, DNK
| | - Mette Felter
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, DNK
| | - Susanne Bekke
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, DNK
| | - Susan Biancardo
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, DNK
| | - Marianne F Rønjom
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, DNK
| | - Mette Pedersen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, DNK
| | - David Sjöström
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, DNK
| | - Inna M Chen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, DNK
| | - Patrik Sibolt
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, DNK
| |
Collapse
|
23
|
Chen BS, Chan SY, Bteich F, Kuang C, Meyerhardt JA, Ma KSK. Safety and efficacy of liposomal irinotecan as the second-line treatment for advanced pancreatic cancer: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2024; 201:104386. [PMID: 38735505 DOI: 10.1016/j.critrevonc.2024.104386] [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: 12/05/2023] [Revised: 04/09/2024] [Accepted: 05/05/2024] [Indexed: 05/14/2024] Open
Abstract
INTRODUCTION Nanoliposomal irinotecan (nal-IRI) is a novel regimen for pancreatic cancer, featuring a longer half-life and an increased area under the concentration-time curve. This study aims to assess the safety and efficacy of nal-IRI as a second-line treatment for advanced pancreatic cancer. METHODS A systemic literature search was conducted based on articles published before September 26th, 2023 in databases, including PubMed, Cochrane Library, EMBASE and Web of Science. The fixed effects model was used to calculate the pooled mean difference for overall survival (OS) and progression-free survival (PFS), as well as the pooled odds ratio for the overall response rate (ORR) and the risk of adverse events. RESULTS A total of 21 studies, including 3044 patients with locally advanced unresectable or metastatic pancreatic cancers, were considered eligible. The use of nal-IRI, combined with 5-fluorouracil and leucovorin, resulted in significantly improved PFS (pooled mean difference=1.01 months, 95 % confidence interval [CI]=0.97-1.05, p<0.01) and OS (pooled mean difference=0.29 months, 95 %CI=0.18-0.39, p<0.01), as well as significantly better ORR (pooled odds ratio=2.06, 95 %CI=1.30-3.27, p=0.002) compared to other second-line regimens. Nonetheless, an increased risk of grade 3 or greater neutropenia, anemia, hypokalemia, diarrhea, and vomiting was also noted. CONCLUSION Second-line treatments based on nal-IRI exhibited significantly improved PFS, OS, and ORR compared to other available treatments in advanced pancreatic cancer. Further research is necessary to corroborate these findings and define the role of nal-IRI in both first and later lines of therapy.
Collapse
Affiliation(s)
- Brian Shiian Chen
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shu-Yen Chan
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fernand Bteich
- Montefiore Einstein Comprehensive Cancer Center, Bronx, NY, USA; Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Chaoyuan Kuang
- Montefiore Einstein Comprehensive Cancer Center, Bronx, NY, USA; Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jeffery A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Kevin Sheng-Kai Ma
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Kaabe S, Amiriani T, Teimoorian M, Besharat S, Salamat F, Hasanpour-Heidari S, Sedaghat S, Sadeghzadeh H, Roshandel G. Incidence Rates and Time Trends of Pancreatic Cancer in the Golestan Province, Northeastern Iran, 2006-2019. ARCHIVES OF IRANIAN MEDICINE 2024; 27:486-493. [PMID: 39465523 PMCID: PMC11496600 DOI: 10.34172/aim.31168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/21/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND Pancreatic cancer (PC) is one of the most malignant cancers with a poor prognosis. Despite advances in the diagnosis and management of PC, the survival rate remains low. In Iran, the incidence of PC is increasing, with mortality rates nearly doubling over the past 25 years. Therefore, this study was designed to assess the temporal variations and incidence of PC in Golestan province, as a prominent hub for gastrointestinal cancers in Iran. METHODS In this cross-sectional study, patient information was obtained from the Golestan Population-Based Cancer Registry (GPCR) from 2006 to 2019. We calculated age-standardized incidence rates (ASRs) using the World standard population and reported the rates per 100000 persons-year. To compare ASRs across sexes and residence areas, incidence rate ratios (IRR) were calculated using Poisson regression models. We calculated the estimated annual percentage changes (EAPC) to assess time trends in incidence rates of PC in Golestan during the study period. RESULTS Among a total of 560 PC new cases (mean age of 63.72 years), 46.61% were diagnosed through clinical or paraclinical methods. The crude incidence rate and ASR were 2.24 and 2.95 (95% CI: 2.70‒3.20) per 100000 persons-year, respectively. The ASR of PC was significantly higher in males (3.78; 95% CI: 3.37‒4.19) than females (2.17; 95% CI: 1.88‒2.46) (IRR=1.71; P<0.01). The ASR was higher in the urban (3.23; 95% CI: 2.88‒3.58) compared to the rural population (2.65; 95% CI: 2.30‒3.00) (IRR=1.23; P=0.02). The ASR of PC increased from 1.97 to 3.53 during 2006 to 2019 with an EAPC of 4.39 (95% CI: -3.56 to 12.75). The EAPCs were 4.85% and 4.37% in women and men, respectively. CONCLUSION Our study showed that the incidence of PC is increasing in the Golestan province. Also, the incidence rate was higher in men, elderly people, and the urban population.
Collapse
Affiliation(s)
- Sajjad Kaabe
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Taghi Amiriani
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehrdad Teimoorian
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sima Besharat
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
- Clinical Research Development Unit (CRDU), Sayad Shirazi Hospital, Golestan University of Medical Sciences, Gorgan, Iran
| | - Faezeh Salamat
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Susan Hasanpour-Heidari
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Hamideh Sadeghzadeh
- Deputy of Public Health, Golestan University of Medical Sciences, Gorgan, Iran
| | - Gholamreza Roshandel
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
25
|
Zhao H, Zhang Y, Liu H, Wang Y, Song Z. Age-period-cohort analysis of global, regional, and national pancreatic cancer incidence, mortality, and disability-adjusted life years, 1990-2019. BMC Cancer 2024; 24:1063. [PMID: 39198814 PMCID: PMC11350939 DOI: 10.1186/s12885-024-12835-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Pancreatic cancer is one of the deadliest cancers in the world. In recent years, the incidence and mortality rates of pancreatic cancer have shown an increasing trend year by year. This study investigates the independent effects of age, period, and cohort on the global incidence, mortality, and disability-adjusted life years (DALYs) of pancreatic cancer from 1990 to 2019, and evaluates the differences in the burden of pancreatic cancer across regions with different Sociodemographic Index (SDI) levels. METHODS Estimating the impact of age, period, and cohort on pancreatic cancer disease burden in different SDI regions using age-period-cohort modeling with data (with 95% uncertainty intervals [UI]) from the Global Burden of Disease (GBD) Study 2019 and net drift of age-standardized incidence rates (ASIR), age-standardized mortality rates (ASMR), and age-standardized DALY rates (ASDR) for pancreatic cancer in 120 countries. RESULTS The number of new cases of pancreatic cancer worldwide increased from 197,348 (95% UI: 188,604,203,971) in 1990 to 530,297 (486,175,573,635) in 2019, the number of deaths increased from 198,051 (189,329 to 204,763) in 1990 to 531,107 (491,948 to 566,537) in 2019, and the number of DALY increased from 4,647,207 (4,465,440 to 4,812,129) in 1990 to 11,549,016 (10,777,405 to 1,238,912) in 2019. The ASIR of the average levels in global pancreatic cancer increased from 5.22 (4.97 to 5.40) per 100,000 population to 6.57 (6.00 to 7.09) per 100,000 population, the ASMR increased from 5.34 (5.07 to 5.52) per 100,000 population to 6.62 (6.11 to 7.06) per 100,000 population, and the ASDR increased from 115.47 (110.82 to 119.60) per 100,000 population to 139.61 (130.18 to 149.14) per 100,000 population. The incidence, mortality, and DALY rates of pancreatic cancer increase with age globally and across all SDI regions, peaking in the 85-89 age group. In high and high-middle SDI regions, the growth rate for males is higher than for females before the age of 85, while females have a higher growth rate after 85. The 75-79 age group exhibits the highest DALY rate in high and high-middle SDI regions, significantly higher than the global and other SDI regions. From 1990 to 2019, the period effects of pancreatic cancer incidence, mortality, and DALY rates have increased significantly worldwide, while remaining almost unchanged in high and high-middle SDI regions. In contrast, period effects have significantly increased in middle, low-middle, and low SDI regions. Cohort effects are more pronounced in middle, low-middle, and low SDI regions. CONCLUSIONS With the aggravation of population aging, the incidence and mortality rates of pancreatic cancer in the world are increasing, and effective prevention and control measures can be achieved by reducing the exposure of risk factors. The APC model used in our analysis provides a novel approach to understanding the complex trends in the incidence, mortality, and disability-adjusted life years of pancreatic cancer. It can inform the development of targeted interventions to reduce the severe disease burden caused by pancreatic cancer.
Collapse
Affiliation(s)
- Haoran Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin City, Heilongjiang Province, People's Republic of China
| | - Yubao Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin City, Heilongjiang Province, People's Republic of China
| | - Haishi Liu
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin City, Heilongjiang Province, People's Republic of China
| | - Yunfeng Wang
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin City, Heilongjiang Province, People's Republic of China
| | - Zengfu Song
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin City, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
26
|
Fang Z, Bunston C, Xu Y, Ruge F, Sui L, Liu M, Al-Sarireh B, Griffiths P, Murphy K, Pugh MR, Hao C, Jiang WG, Ye L. Implication of Capillary Morphogenesis Gene 2 (CMG2) in the Disease Progression and Peritoneal Metastasis of Pancreatic Cancer. Cancers (Basel) 2024; 16:2893. [PMID: 39199664 PMCID: PMC11352480 DOI: 10.3390/cancers16162893] [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: 06/06/2024] [Revised: 08/13/2024] [Accepted: 08/17/2024] [Indexed: 09/01/2024] Open
Abstract
Capillary morphogenesis gene 2 (CMG2) mediates cell-matrix interactions to facilitate cell adhesion and migration. CMG2 has been implicated in the disease progression of breast cancer, prostate cancer and gastric cancer. The present study aims to determine the role of CMG2 in the disease progression and peritoneal metastasis of pancreatic cancer. Pancreatic tumour samples were collected from Peking University Cancer Hospital. CMG2 expression was determined using quantitative PCR. After the creation of knockdown and overexpression of CMG2 in pancreatic cancer cells, the effect of CMG2 on several cell functions and adhesion to the peritoneum was examined. Potential pathways regulated by CMG2 were found via proteomics analysis and drug tests. CMG2 was upregulated in pancreatic cancer tissues and associated with a poor prognosis. CMG2 was increased in metastatic lesions and those primary tumours with distant metastases. CMG2 promotes cell-cell, cell-matrix and cell-hyaluronic acid adhesion, which may be mediated by epidermal growth factor receptor (EGFR) and focal adhesion kinase (FAK) pathway activation.
Collapse
Affiliation(s)
- Ziqian Fang
- Cardiff China Medical Research Collaborative, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (Z.F.); (Y.X.); (F.R.); (L.S.); (M.L.); (W.G.J.)
| | - Carly Bunston
- Cardiff China Medical Research Collaborative, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (Z.F.); (Y.X.); (F.R.); (L.S.); (M.L.); (W.G.J.)
| | - Yali Xu
- Cardiff China Medical Research Collaborative, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (Z.F.); (Y.X.); (F.R.); (L.S.); (M.L.); (W.G.J.)
| | - Fiona Ruge
- Cardiff China Medical Research Collaborative, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (Z.F.); (Y.X.); (F.R.); (L.S.); (M.L.); (W.G.J.)
| | - Laijian Sui
- Cardiff China Medical Research Collaborative, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (Z.F.); (Y.X.); (F.R.); (L.S.); (M.L.); (W.G.J.)
| | - Ming Liu
- Cardiff China Medical Research Collaborative, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (Z.F.); (Y.X.); (F.R.); (L.S.); (M.L.); (W.G.J.)
| | - Bilal Al-Sarireh
- Department of Surgery, Morriston Hospital, ABM University Health Board, Swansea SA6 6NL, UK;
| | - Paul Griffiths
- Department of Pathology, Morriston Hospital, ABM University Health Board, Swansea SA6 6NL, UK; (P.G.); (M.R.P.)
| | - Kate Murphy
- Department of Pathology, Morriston Hospital, ABM University Health Board, Swansea SA6 6NL, UK; (P.G.); (M.R.P.)
| | - Matthew R. Pugh
- Department of Pathology, Morriston Hospital, ABM University Health Board, Swansea SA6 6NL, UK; (P.G.); (M.R.P.)
| | - Chunyi Hao
- Key Laboratory of Carcinogenesis and Translational Research, Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China;
| | - Wen G. Jiang
- Cardiff China Medical Research Collaborative, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (Z.F.); (Y.X.); (F.R.); (L.S.); (M.L.); (W.G.J.)
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (Z.F.); (Y.X.); (F.R.); (L.S.); (M.L.); (W.G.J.)
| |
Collapse
|
27
|
Dinagde TA, Abubeker Z. Surgical management of pancreatic cancer in Tikur Anbessa Specialized Hospital, Addis Ababa, Ethiopia: a 5 years retrospective descriptive study. BMC Surg 2024; 24:223. [PMID: 39103810 DOI: 10.1186/s12893-024-02503-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 02/12/2024] [Indexed: 08/07/2024] Open
Abstract
INTRODUCTION The incidence of Pancreatic cancer is different in different parts of the world. It is a cancer with the worst prognosis of all malignancies. Pancreatic cancer is predominantly a disease of an older population. There are different environmental (modifiable) and non-modifiable risk factors associated with the development of pancreatic cancer. At present, surgical resection is the only potential cure for pancreatic cancer. However, as only 10-20% of the patients have resectable disease at the time of diagnosis. The morbidities associated with surgeries for pancreatic cancers remain high though the post-operative mortality has shown significant reduction in the past few decades. So far, no study has been conducted to investigate pancreatic cancer in Ethiopia. OBJECTIVES To assess the clinico-pathologic profile, associated factors, surgical management and short-term outcome of patients with pancreatic cancer in Tikur Anbessa Specialized hospital. METHODS A 5 years retrospective hospital-based cross-sectional study was conducted on 52 patients operated with the diagnosis of pancreatic cancer with either curative or palliative intents. The study period was from April 2016 to July 2021. The data collected includes demographic profile, associated risk factors and comorbidities, clinical presentations, biochemical parameters, pathologic features of the tumors as well as type of treatment offered and short term treatment outcome. The data was analyzed using SPSS version 25. RESULT The mean and median age of patients was 54.1 and 54.5% respectively. Males constitute about 52% the patients. 21% of the patients have potential risk factors; whereas only 10 (19.2%) of the patients had medical comorbidities. Median duration of symptoms at diagnosis was 12 weeks. Abdominal pain (88.5%) was the most common presenting symptom followed by anorexia (80.8%) and significant weight loss (78.8%), while 71.2% of the patients have jaundice. On clinical evaluation, 69.2% were jaundiced, while 34.6% had a palpable gallbladder. More than two third of patients presented with advanced disease. 76.9% of the tumors are located in the head of pancreas. More than three quarters (77%) of the surgeries performed were palliative. Postoperative morbidity and mortality were 19.2% and 3.8% respectively. CONCLUSION Age at first diagnosis of pancreatic cancer is relatively earlier in our setup. Most patients present with advanced condition, only amenable for palliative measures. The post-operative morbidity and mortality are more or less comparable with similar studies. The need for adjuvant therapy in pancreatic cancer should be emphasized.
Collapse
Affiliation(s)
- Tesfaye Aga Dinagde
- Department of Surgery, Tikur Anbessa Specialized Hospital, Addis Ababa University, Addis Ababa, Ethiopia.
| | - Zeki Abubeker
- Department of Surgery, Tikur Anbessa Specialized Hospital, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
28
|
Luo R, Li S, Yang C, Tang B, Li L, Luo C. Curcumin Inhibits the Development of Pancreatic Cancer by Targeting the circ_0079440/miR-522-3p/EIF4A1 Pathway. Cell Biochem Biophys 2024:10.1007/s12013-024-01466-8. [PMID: 39102088 DOI: 10.1007/s12013-024-01466-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 08/06/2024]
Abstract
Pancreatic cancer (PC) is a common gastrointestinal cancer with high invasiveness and high mortality. Curcumin is a natural polyphenol with anti-tumor activity against different cancers, including PC. Curcumin has been verified to mediate the expression of circular RNAs (circRNAs) to inhibit tumor development. This study aimed to explore the function and regulatory mechanism of curcumin on circ_0079440 in PC. PC cells were treated with different concentrations of curcumin (0, 5, 10 or 15 μM) for 24 h. Gene expression in PC cells and tissues was detected using RT-qPCR. Cell malignant phenotypes were determined by functional assays. The levels of EMT-related proteins were tested using western blot. RNA interaction was determined using RNA pulldown assay, luciferase reporter assay and RIP assay. The results showed that curcumin suppressed cell proliferative, migratory, and invasive capabilities, and weakened epithelial-mesenchymal transition (EMT) in a concentration-dependent way. Circ_0079440 was expressed at a high level in PC and its level was reduced via curcumin administration in PC cells. Rescue assays showed that circ_0079440 overexpression reversed the suppressive effects of curcumin on PC cell malignant phenotypes. Furthermore, in the xenograft mouse models, curcumin treatment inhibited tumor growth and metastasis, and circ_0079440 upregulation reversed the function of curcumin. Additionally, circ_0079440 was revealed to bind to miR-522-3p to upregulate eukaryotic initiation factor 4A1 (EIF4A1) expression in PC cells. EIF4A1 expression was also downregulated by curcumin, and EIF4A1 overexpression abolished the suppressive functions of curcumin. Moreover, EIF4A overexpression or miR-522-3p inhibition counteracted the anti-tumor effects of circ_0079440 depletion on PC development. To sum up, curcumin suppresses PC development by targeting the circ_0079440/miR-522-3p/EIF4A1 pathway, which might provide novel therapeutic targets for treatment of PC.
Collapse
Affiliation(s)
- Ruiying Luo
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Shuang Li
- Department of respiratory medicine, The Third People's Hospital of Gansu Province, Lanzhou, 730000, Gansu, China
| | - Chi Yang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Baoyuan Tang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Long Li
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Changjiang Luo
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
29
|
Noguchi A, Numata Y, Sugawara T, Miura H, Konno K, Adachi Y, Yamaguchi R, Ishida M, Kokumai T, Douchi D, Miura T, Ariake K, Nakayama S, Maeda S, Ohtsuka H, Mizuma M, Nakagawa K, Morikawa H, Akatsuka J, Maeda I, Unno M, Yamamoto Y, Furukawa T. Deep learning predicts the 1-year prognosis of pancreatic cancer patients using positive peritoneal washing cytology. Sci Rep 2024; 14:17059. [PMID: 39095474 PMCID: PMC11297136 DOI: 10.1038/s41598-024-67757-5] [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: 10/06/2023] [Accepted: 07/15/2024] [Indexed: 08/04/2024] Open
Abstract
Peritoneal washing cytology (CY) in patients with pancreatic cancer is mainly used for staging; however, it may also be used to evaluate the intraperitoneal status to predict a more accurate prognosis. Here, we investigated the potential of deep learning of CY specimen images for predicting the 1-year prognosis of pancreatic cancer in CY-positive patients. CY specimens from 88 patients with prognostic information were retrospectively analyzed. CY specimens scanned by the whole slide imaging device were segmented and subjected to deep learning with a Vision Transformer (ViT) and a Convolutional Neural Network (CNN). The results indicated that ViT and CNN predicted the 1-year prognosis from scanned images with accuracies of 0.8056 and 0.8009 in the area under the curve of the receiver operating characteristic curves, respectively. Patients predicted to survive 1 year or more by ViT showed significantly longer survivals by Kaplan-Meier analyses. The cell nuclei found to have a negative prognostic impact by ViT appeared to be neutrophils. Our results indicate that AI-mediated analysis of CY specimens can successfully predict the 1-year prognosis of patients with pancreatic cancer positive for CY. Intraperitoneal neutrophils may be a novel prognostic marker and therapeutic target for CY-positive patients with pancreatic cancer.
Collapse
Affiliation(s)
- Aya Noguchi
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yasushi Numata
- Pathology Informatics Team, RIKEN Center for Advanced Intelligence Project, Nihonbashi 1-chome Mitsui Bldg. 15F, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
| | - Takanori Sugawara
- Department of Pathology, Tohoku University Hospital, Sendai, 980-8574, Japan
| | - Hiroshu Miura
- Department of Pathology, Tohoku University Hospital, Sendai, 980-8574, Japan
| | - Kaori Konno
- Department of Pathology, Tohoku University Hospital, Sendai, 980-8574, Japan
| | - Yuzu Adachi
- Department of Pathology, Tohoku University Hospital, Sendai, 980-8574, Japan
| | - Ruri Yamaguchi
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Masaharu Ishida
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Takashi Kokumai
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Daisuke Douchi
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Takayuki Miura
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Kyohei Ariake
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Shun Nakayama
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Shimpei Maeda
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Hideo Ohtsuka
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Masamichi Mizuma
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Kei Nakagawa
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Hiromu Morikawa
- Pathology Informatics Team, RIKEN Center for Advanced Intelligence Project, Nihonbashi 1-chome Mitsui Bldg. 15F, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
| | - Jun Akatsuka
- Pathology Informatics Team, RIKEN Center for Advanced Intelligence Project, Nihonbashi 1-chome Mitsui Bldg. 15F, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
- Department of Urology, Nippon Medical School Hospital, Tokyo, 113-8603, Japan
| | - Ichiro Maeda
- Pathology Informatics Team, RIKEN Center for Advanced Intelligence Project, Nihonbashi 1-chome Mitsui Bldg. 15F, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
- Department of Pathology, Kitasato University Kitasato Institute Hospital, Tokyo, 108-0072, Japan
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, 252-0373, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Yoichiro Yamamoto
- Pathology Informatics Team, RIKEN Center for Advanced Intelligence Project, Nihonbashi 1-chome Mitsui Bldg. 15F, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan.
- Mathematical Intelligence for Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan.
| | - Toru Furukawa
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| |
Collapse
|
30
|
Dai S, Kong H, Ja Y, Bao L, Wang C, Qin L. Expression of the laminin genes family and its relationship to prognosis in pancreatic carcinoma. Arab J Gastroenterol 2024; 25:306-314. [PMID: 39039002 DOI: 10.1016/j.ajg.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/16/2024] [Accepted: 07/05/2024] [Indexed: 07/24/2024]
Abstract
BACKGROUND AND STUDY AIMS Laminin is an extracellular matrix molecule that is the major component of the basement membrane and plays a key role in regulating various processes. However, the association between the laminin gene family and the prognosis of pancreatic carcinoma has not been systematically investigated. PATIENTS AND METHODS The role of the laminin gene family in pancreatic cancer was evaluated using data from the TCGA database. The effects of different expressions of members of the laminin gene family on pancreatic cancer survival were compared, and their primary cellular roles were examined. The effects of different expressions of positive family genes on proliferation, metastasis, and invasion, as well as EMT and ferroptosis in pancreatic cancer, were also examined. RESULTS Based on univariate and multifactorial analysis of pancreatic cancer patients, LAMA3 was identified as an independent prognostic factor for overall survival in pancreatic cancer. LAMA3 was found to be enriched in the actin cytoskeleton, P53 signaling pathway, adhesion molecule junctions, pentose phosphate pathway, and regulatory differences in the cell cycle and focal adhesion. Additionally, high expression of LAMA3 was found to promote cancer proliferation, invasion, and metastasis, facilitate the EMT process, and inhibit ferroptosis. CONCLUSIONS Our results identified LAMA3 was associated with the prognosis of patients with pancreatic cancer and may serve as a prognostic biomarker for pancreatic cancer.
Collapse
Affiliation(s)
- ShengJie Dai
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongru Kong
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yang Ja
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liqi Bao
- Renji College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chengchao Wang
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lei Qin
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
31
|
Mahadiuzzaman ASM, Dain Md Opo FA, Alkarim S. Stem cell-based targeted therapy in pancreatic cancer: Current approaches and future prospects. Tissue Cell 2024; 89:102449. [PMID: 38924893 DOI: 10.1016/j.tice.2024.102449] [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: 01/18/2024] [Revised: 05/22/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Despite recent improvements in oncology, diagnosis, and therapy, pancreatic cancer remains extremely difficult to cure due to its aggressive growth pattern with early invasion and distant metastases, chemoresistance, and a lack of effective screening modalities for early detection. Here, novel therapeutic approaches for treating pancreatic cancer are urgently needed. Recently, stem cells have drawn a lot of interest as a possible treatment for pancreatic cancer due to their ability to locate tumors. Though research over the last few decades has revealed some very exciting and promising new treatment approaches, the clinical success of these stem-cell based anti-cancer medicines has been quite limited. The most effective stem cell-mediated therapeutic options will only be available with a deeper understanding of the intricate molecular biology underlying pancreatic cancer and the subsequent identification of cancer stem cells as a novel target that promotes the growth of the cancer and resistance to chemotherapy. This review will highlight the stem cell based anti-cancer therapy targeting pancreatic cancer stem cells and different molecular signaling pathways. A particular focus will be on the therapeutic potential of naïve Stem cells, anti-cancer drug loaded stem cells, genetically engineered stem cells and exosomal miRNA released by stem cells in pancreatic cancer treatment. Similarly, the role of nanotechnology in stem cell based anticancer therapy will be further discussed to better implementation of these cell-based cancer therapy.
Collapse
Affiliation(s)
- A S M Mahadiuzzaman
- Department of Biological Science, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Embryonic Stem Cell Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - F A Dain Md Opo
- Department of Biological Science, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Embryonic Stem Cell Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Saleh Alkarim
- Department of Biological Science, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Embryonic Stem Cell Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Embryonic and Cancer Stem Cell Research Group, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
32
|
Okuno M, Tanaka T, Iwata K, Mukai T, Watanabe N, Shimojo K, Iwasa Y, Tezuka R, Iwashita T, Tomita E, Shimizu M. Diagnosis of pancreatic malignancies using an overnight-stored pancreatic juice cell block specimen. Pancreatology 2024; 24:732-739. [PMID: 38879435 DOI: 10.1016/j.pan.2024.06.005] [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: 04/09/2024] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND AND AIMS Pancreatic juice cytology is useful for diagnosing pancreatic duct strictures and cystic lesions. However, some cases cannot be diagnosed using cytology. This study aimed to evaluate the utility of the overnight-stored pancreatic juice cell block (CB) method for diagnosing pancreatic disease. METHODS This retrospective study included 32 patients who presented with pancreatic duct strictures or cystic lesions between 2018 and 2024. The sensitivity, specificity, and accuracy of the CB method and single/multiple pancreatic juice cytology were compared to evaluate the utility of the CB. RESULT An endoscopic nasopancreatic drainage tube was placed in the main pancreatic duct, and pancreatic juice was collected to create a CB specimen. The median amount of pancreatic juice collected was 180(30-200) mL, and the median number of cytological examinations was three(2-8). Of the 32 cases, 13 were malignant, and 19 were benign (non-malignant). The sensitivity was significantly higher for the CB method (62 %) than for single cytology(15 %, P = 0.0414), and there was no significant difference between CB and multiple cytology(54 %, P = 1.0). The specificity and accuracy were not significantly different between the CB method and single or multiple cytology. When multiple cytology and CB were combined, sensitivity improved to 77 %. The pathological findings of the CB specimens were similar to the surgical specimens, including immunohistochemistry. CONCLUSION The overnight-stored pancreatic juice CB method was more effective than single cytology, with similar sensitivities to multiple cytology and can also be used for immunohistochemistry. The pancreatic juice CB method is useful for pancreatic juice assessment.
Collapse
Affiliation(s)
- Mitsuru Okuno
- Department of Gastroenterology, Matsunami General Hospital, Gifu, Japan; Department of Gastroenterology, Gifu Municipal Hospital, Gifu, Japan.
| | - Takuji Tanaka
- Department of Diagnostic Pathology, Gifu Municipal Hospital, Gifu, Japan
| | - Keisuke Iwata
- Department of Gastroenterology, Gifu Municipal Hospital, Gifu, Japan
| | - Tsuyoshi Mukai
- Department of Gastroenterology, Gifu Municipal Hospital, Gifu, Japan; Department of Gastroenterological Endoscopy, Kanazawa Medical University, Ishikawa, Japan
| | - Naoki Watanabe
- Department of Diagnostic Pathology, Gifu Municipal Hospital, Gifu, Japan
| | - Kota Shimojo
- Department of Gastroenterology, Gifu Municipal Hospital, Gifu, Japan
| | - Yuhei Iwasa
- Department of Gastroenterology, Gifu Municipal Hospital, Gifu, Japan
| | - Ryuichi Tezuka
- Department of Gastroenterology, Gifu Municipal Hospital, Gifu, Japan
| | - Takuji Iwashita
- First Department of Internal Medicine, Gifu University Hospital, Gifu, Japan
| | - Eiichi Tomita
- Department of Gastroenterology, Matsunami General Hospital, Gifu, Japan; Department of Gastroenterology, Gifu Municipal Hospital, Gifu, Japan
| | - Masahito Shimizu
- First Department of Internal Medicine, Gifu University Hospital, Gifu, Japan
| |
Collapse
|
33
|
Maurer E, Bartsch DK. Surgical aspects related to hereditary pancreatic cancer. Fam Cancer 2024; 23:341-350. [PMID: 38662263 PMCID: PMC11254980 DOI: 10.1007/s10689-024-00384-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/29/2024] [Indexed: 04/26/2024]
Abstract
The goal of surveillance programs for individuals at risk (IAR) from familial pancreatic cancer (FPC) families or families with other inherited tumor syndromes predisposing to the development of pancreatic adenocarcinoma (PDAC), such as hereditary pancreatitis or Peutz-Jeghers syndrome, is the dectection and consecutive curative resection of early PDAC or even better its high-grade precursor lesions. Although the indication for surgery is quite established, the extent of surgery is not well defined due to the lack of evidence-based data. In addition, multiple factors have to be taken into account to determine an optimal personalized surgical strategy. This holds especially true since pancreatic surgery is associated with a relatively high morbidity and might impair the quality of life significantly. In this article the surgical aspects in the setting of hereditary PDAC are discussed.
Collapse
Affiliation(s)
- Elisabeth Maurer
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University Marburg, 35043, Baldingerstrasse, Marburg, Germany.
| | - Detlef K Bartsch
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University Marburg, 35043, Baldingerstrasse, Marburg, Germany
| |
Collapse
|
34
|
Tang Y, Su YX, Zheng JM, Zhuo ML, Qian QF, Shen QL, Lin P, Chen ZK. Radiogenomic analysis for predicting lymph node metastasis and molecular annotation of radiomic features in pancreatic cancer. J Transl Med 2024; 22:690. [PMID: 39075486 PMCID: PMC11288107 DOI: 10.1186/s12967-024-05479-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 07/03/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND To provide a preoperative prediction model for lymph node metastasis in pancreatic cancer patients and provide molecular information of key radiomic features. METHODS Two cohorts comprising 151 and 54 pancreatic cancer patients were included in the analysis. Radiomic features from the tumor region of interests were extracted by using PyRadiomics software. We used a framework that incorporated 10 machine learning algorithms and generated 77 combinations to construct radiomics-based models for lymph node metastasis prediction. Weighted gene coexpression network analysis (WGCNA) was subsequently performed to determine the relationships between gene expression levels and radiomic features. Molecular pathways enrichment analysis was performed to uncover the underlying molecular features. RESULTS Patients in the in-house cohort (mean age, 61.3 years ± 9.6 [SD]; 91 men [60%]) were separated into training (n = 105, 70%) and validation (n = 46, 30%) cohorts. A total of 1,239 features were extracted and subjected to machine learning algorithms. The 77 radiomic models showed moderate performance for predicting lymph node metastasis, and the combination of the StepGBM and Enet algorithms had the best performance in the training (AUC = 0.84, 95% CI = 0.77-0.91) and validation (AUC = 0.85, 95% CI = 0.73-0.98) cohorts. We determined that 15 features were core variables for lymph node metastasis. Proliferation-related processes may respond to the main molecular alterations underlying these features. CONCLUSIONS Machine learning-based radiomics could predict the status of lymph node metastasis in pancreatic cancer, which is associated with proliferation-related alterations.
Collapse
Affiliation(s)
- Yi Tang
- Department of Medical Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan road, Fuzhou, China
| | - Yi-Xi Su
- Department of Medical Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan road, Fuzhou, China
| | - Jin-Mei Zheng
- Department of Radiology, Fujian Medical University Union Hospital, 29 Xinquan road, Fuzhou, China
| | - Min-Ling Zhuo
- Department of Medical Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan road, Fuzhou, China
| | - Qing-Fu Qian
- Department of Medical Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan road, Fuzhou, China
| | - Qing-Ling Shen
- Department of Medical Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan road, Fuzhou, China
| | - Peng Lin
- Department of Medical Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan road, Fuzhou, China.
| | - Zhi-Kui Chen
- Department of Medical Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan road, Fuzhou, China.
| |
Collapse
|
35
|
Barbagallo D, Ponti D, Bassani B, Bruno A, Pulze L, Akkihal SA, George-William JN, Gundamaraju R, Campomenosi P. MiR-223-3p in Cancer Development and Cancer Drug Resistance: Same Coin, Different Faces. Int J Mol Sci 2024; 25:8191. [PMID: 39125761 PMCID: PMC11311375 DOI: 10.3390/ijms25158191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
MicroRNAs (miRNAs) are mighty post-transcriptional regulators in cell physiology and pathophysiology. In this review, we focus on the role of miR-223-3p (henceforth miR-223) in various cancer types. MiR-223 has established roles in hematopoiesis, inflammation, and most cancers, where it can act as either an oncogenic or oncosuppressive miRNA, depending on specific molecular landscapes. MiR-223 has also been linked to either the sensitivity or resistance of cancer cells to treatments in a context-dependent way. Through this detailed review, we highlight that for some cancers (i.e., breast, non-small cell lung carcinoma, and glioblastoma), the oncosuppressive role of miR-223 is consistently reported in the literature, while for others (i.e., colorectal, ovarian, and pancreatic cancers, and acute lymphocytic leukemia), an oncogenic role prevails. In prostate cancer and other hematological malignancies, although an oncosuppressive role is frequently described, there is less of a consensus. Intriguingly, NLRP3 and FBXW7 are consistently identified as miR-223 targets when the miRNA acts as an oncosuppressor or an oncogene, respectively, in different cancers. Our review also describes that miR-223 was increased in biological fluids or their extracellular vesicles in most of the cancers analyzed, as compared to healthy or lower-risk conditions, confirming the potential application of this miRNA as a diagnostic and prognostic biomarker in the clinic.
Collapse
Affiliation(s)
- Davide Barbagallo
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics “Giovanni Sichel”, University of Catania, Via Santa Sofia 89, 95123 Catania, Italy
- Interdisciplinary Research Centre on the Diagnosis and Therapy of Brain Tumors, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
| | - Donatella Ponti
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Corso della Repubblica 79, 04100 Latina, Italy;
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Via Fantoli 16/15, 20138 Milano, Italy; (B.B.); (A.B.)
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Via Fantoli 16/15, 20138 Milano, Italy; (B.B.); (A.B.)
- Department of Biotechnology and Life Sciences, DBSV, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy;
| | - Laura Pulze
- Department of Biotechnology and Life Sciences, DBSV, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy;
| | - Shreya A. Akkihal
- Independent Researcher, 35004 SE Swenson St, Snoqualmie, WA 98065, USA;
| | - Jonahunnatha N. George-William
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi, 93, 20054 Segrate, Italy;
| | - Rohit Gundamaraju
- Department of Laboratory Medicine, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA;
- ER Stress and Mucosal Immunology Team, School of Health Sciences, University of Tasmania, Launceston, TAS 7248, Australia
| | - Paola Campomenosi
- Department of Biotechnology and Life Sciences, DBSV, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy;
| |
Collapse
|
36
|
Buchholz M, Majchrzak-Stiller B, Peters I, Hahn S, Skrzypczyk L, Beule L, Uhl W, Braumann C, Strotmann J, Höhn P. Maintenance Therapy for Pancreatic Cancer, a New Approach Based on the Synergy between the Novel Agent GP-2250 (Misetionamide) and Gemcitabine. Cancers (Basel) 2024; 16:2612. [PMID: 39061250 PMCID: PMC11275110 DOI: 10.3390/cancers16142612] [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: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
The novel Oxathiazinane derivative GP-2250 (Misetionamide) displays antineoplastic activity in vitro and in vivo, as previously shown in pancreatic cancer cells and in patient-derived mouse xenografts (PDX). Currently, GP 2250 is under phase I clinical trial in pancreatic ductal adenocarcinoma (PDAC). GP-2250 in combination with Gemcitabine displays a high synergistic capacity in various primary and established pancreatic cancer cell lines. Additionally, in the eight PDX models tested, the drug combination was superior in reducing tumor volume with an aggregate tumor regression (ATR) of 74% compared to Gemcitabine alone (ATR: 10%). Similarly, in a PDX maintenance setting following two weeks of treatment with nab-Paclitaxel plus Gemcitabine, the combination of GP-2250 plus Gemcitabine resulted in outstanding tumor control (ATR: 79%) compared to treatment with Gemcitabine alone (ATR: 60%). Furthermore, GP-2250 reduced the ratio of tumor-initiating CD133+ markers on the surface of PDAC cells in spheroid cultures, indicating a possible mechanism for the synergistic effect of both substances. Considering the high tolerability of GP 2250, these results may open up a new approach to maintenance therapy with GP-2250/Gemcitabine combination following nab-Paclitaxel plus Gemcitabine as first-line treatment.
Collapse
Affiliation(s)
- Marie Buchholz
- Department of General and Visceral Surgery, Division of Molecular and Clinical Research, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany; (B.M.-S.); (I.P.); (L.S.); (L.B.); (W.U.); (C.B.); (J.S.); (P.H.)
| | - Britta Majchrzak-Stiller
- Department of General and Visceral Surgery, Division of Molecular and Clinical Research, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany; (B.M.-S.); (I.P.); (L.S.); (L.B.); (W.U.); (C.B.); (J.S.); (P.H.)
| | - Ilka Peters
- Department of General and Visceral Surgery, Division of Molecular and Clinical Research, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany; (B.M.-S.); (I.P.); (L.S.); (L.B.); (W.U.); (C.B.); (J.S.); (P.H.)
| | - Stephan Hahn
- Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, 44780 Bochum, Germany;
| | - Lea Skrzypczyk
- Department of General and Visceral Surgery, Division of Molecular and Clinical Research, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany; (B.M.-S.); (I.P.); (L.S.); (L.B.); (W.U.); (C.B.); (J.S.); (P.H.)
| | - Lena Beule
- Department of General and Visceral Surgery, Division of Molecular and Clinical Research, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany; (B.M.-S.); (I.P.); (L.S.); (L.B.); (W.U.); (C.B.); (J.S.); (P.H.)
| | - Waldemar Uhl
- Department of General and Visceral Surgery, Division of Molecular and Clinical Research, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany; (B.M.-S.); (I.P.); (L.S.); (L.B.); (W.U.); (C.B.); (J.S.); (P.H.)
| | - Chris Braumann
- Department of General and Visceral Surgery, Division of Molecular and Clinical Research, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany; (B.M.-S.); (I.P.); (L.S.); (L.B.); (W.U.); (C.B.); (J.S.); (P.H.)
- Department of General, Visceral and Vascular Surgery, Evangelische Kliniken Gelsenkirchen, Akademisches Lehrkrankenhaus der Universität Duisburg-Essen, 45878 Gelsenkirchen, Germany
| | - Johanna Strotmann
- Department of General and Visceral Surgery, Division of Molecular and Clinical Research, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany; (B.M.-S.); (I.P.); (L.S.); (L.B.); (W.U.); (C.B.); (J.S.); (P.H.)
| | - Philipp Höhn
- Department of General and Visceral Surgery, Division of Molecular and Clinical Research, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany; (B.M.-S.); (I.P.); (L.S.); (L.B.); (W.U.); (C.B.); (J.S.); (P.H.)
| |
Collapse
|
37
|
Maurer E, Lehman B, Matthäi E, Denzer U, Figiel J, Jesinghaus M, Slater EP, Stefenelli U, Gress TM, Bartsch DK. Pancreatic cancer screening is effective in individuals at risk with predisposing germline gene variants, but not in gene variant-negative familial pancreatic cancer families. United European Gastroenterol J 2024. [PMID: 39031472 DOI: 10.1002/ueg2.12631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 06/20/2024] [Indexed: 07/22/2024] Open
Abstract
OBJECTIVE To evaluate the diagnostic yield of pancreatic cancer screening in individuals at risk (IAR) from familial pancreatic cancer (FPC) families with respect to the presence or absence of pathogenic germline variants predisposing to pancreatic adenocarcinoma (PDAC). DESIGN In a 20 years period, IAR from FPC families were enrolled in a prospective screening program of the national case collection for FPC of Germany, including magnet resonance imaging (MRI) and endoscopic ultrasound (EUS). The diagnostic yield was analyzed regarding significant pancreatic lesions such as PDAC, high-grade pancreatic-intraepithelial-neoplasia (PanIN3) and intraductal-papillary-mucinous-neoplasia (IPMN) with high-grade dysplasia. Screening results were compared between carriers of pathogenic variants and variant-negative IAR. RESULTS 337 IAR, including 74 (22%) variant-carriers and 263 IAR of variant-negative FPC families (mean age 49; standard deviation [SD] + 8.9) were followed 64 (SD + 55) months. IAR underwent 5.1 (SD + 3.9) screening visits with 1733 MRI (5.1,SD + 3.9 per IAR) and 728 EUS (2.2,SD + 1.7 per IAR). In 12 (4%) cases, significant pancreatic lesions were detected, including 4 PDAC, 3 PanIN3 and 5 high-grade IPMN. Three of 4 IAR with PDAC died after a mean of 27 months postoperatively, and one IAR is alive without evidence of disease after 31 months. The diagnostic yield for significant lesions was 13.5% (10/74) for variant carriers compared to 0.8% (2/263) for IAR of variant-negative FPC families (p < 0.001). Logistic regression analysis revealed that a negative variant status was almost always accompanied by the absence of a significant lesion over time with a negative predictive value of 99.2% (95% CI 97.3%-99.9%). CONCLUSION The diagnostic yield seems to justify PDAC screening in IAR of FPC-families with pathogenic germline variants in PDAC predisposing genes, not in IAR of variant-negative families.
Collapse
Affiliation(s)
- Elisabeth Maurer
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | - Bettina Lehman
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | - Elvira Matthäi
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | - Ulrike Denzer
- Department of Gastroenterology, Endocrinology, Metabolism and Infectiology, Philipps University Marburg, Marburg, Germany
| | - Jens Figiel
- Department of Diagnostic and Interventional Radiology, Philipps University Marburg, Marburg, Germany
| | - Moritz Jesinghaus
- Institute of Pathology, Philipps University Marburg, Marburg, Germany
| | - Emily P Slater
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | | | - Thomas M Gress
- Department of Gastroenterology, Endocrinology, Metabolism and Infectiology, Philipps University Marburg, Marburg, Germany
| | - Detlef K Bartsch
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| |
Collapse
|
38
|
Topkan E, Senyurek S, Kılic Durankus N, Ozturk D, Selek U. Novel Somay's GLUCAR Index Efficiently Predicts Survival Outcomes in Locally Advanced Pancreas Cancer Patients Receiving Definitive Chemoradiotherapy: A Propensity-Score-Matched Cohort Analysis. J Pers Med 2024; 14:746. [PMID: 39064000 PMCID: PMC11278407 DOI: 10.3390/jpm14070746] [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: 04/30/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Propensity score matching (PSM) was used to investigate the prognostic value of a novel GLUCAR index [Glucose × (C-reactive protein ÷ albumin)] in unresectable locally advanced pancreatic cancer (LA-NPC) patients who received definitive concurrent chemoradiotherapy (CCRT). METHODS The PSM analysis comprised 142 LA-PAC patients subjected to definitive CCRT. Receiver operating characteristic (ROC) curve analysis was utilized to identify relevant pre-CCRT cutoffs that could effectively stratify survival results. The primary and secondary objectives were the correlations between the pre-CCRT GLUCAR measures and overall survival (OS) and progression-free survival (PFS). RESULTS The ROC analysis revealed significance at 43.3 for PFS [area under the curve (AUC): 85.1%; sensitivity: 76.8%; specificity: 74.2%; J-index: 0.510)] and 42.8 for OS (AUC: 81.8%; sensitivity: 74.2%; specificity: 71.7%; J-index: 0.459). Given that these cutoff points were close, the standard cutoff point, 42.8, was selected for further analysis. Comparative survival analyses showed that pre-CCRT GLUCAR ≥ 42.8 (n = 71) measures were associated with significantly shorter median PFS (4.7 vs. 15.8 months; p < 0.001) and OS (10.1 vs. 25.4 months; p < 0.001) durations compared to GLUCAR < 42.8 measures (n = 71). The multivariate analysis results confirmed the independent significance of the GLUCAR index on PFS (p < 0.001) and OS (p < 0.001) outcomes. CONCLUSIONS Elevated pre-CCRT GLUCAR levels are robustly and independently linked to significantly poorer PFS and OS outcomes in unresectable LA-PAC patients treated with definitive CCRT.
Collapse
Affiliation(s)
- Erkan Topkan
- Department of Radiation Oncology, Faculty of Medicine, Baskent University, Adana 01120, Turkey
| | - Sukran Senyurek
- Department of Radiation Oncology, Koc University School of Medicine, Istanbul 34450, Turkey; (S.S.); (N.K.D.); (U.S.)
| | - Nulifer Kılic Durankus
- Department of Radiation Oncology, Koc University School of Medicine, Istanbul 34450, Turkey; (S.S.); (N.K.D.); (U.S.)
| | - Duriye Ozturk
- Department of Radiation Oncology, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar 03030, Turkey;
| | - Ugur Selek
- Department of Radiation Oncology, Koc University School of Medicine, Istanbul 34450, Turkey; (S.S.); (N.K.D.); (U.S.)
| |
Collapse
|
39
|
Kobayashi K, Kishi Y, Tsunenari T, Yonamine N, Takihata Y, Nakazawa A, Takao M, Einama T, Tsujimoto H, Ueno H. Risk of hepatic steatosis with the preoperative treatment of pancreatic cancer and the short-term postoperative outcomes. Surg Today 2024:10.1007/s00595-024-02895-x. [PMID: 38980333 DOI: 10.1007/s00595-024-02895-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/19/2024] [Indexed: 07/10/2024]
Abstract
PURPOSE We investigated whether the preoperative treatment of patients with pancreatic cancer is a risk factor for hepatic steatosis (HS), and whether preoperative HS affects the short-term postoperative outcomes. METHODS Patients who underwent radical surgery for pancreatic cancer between 2010 and 2023 were enrolled. The patients' medical records were reviewed. Albumin and carbohydrate antigen 19-9 were measured before and after chemotherapy in the patients who received preoperative chemotherapy. A logistic regression univariate analysis was performed to analyze the factors associated with new-onset HS. RESULTS A total of 230 patients who underwent surgery were included. HS was observed on the date of surgery in 11 (10%) and two (2%) patients with and without preoperative chemotherapy, respectively. Female sex, initially borderline resectable or unresectable disease, history of cholangitis, presence of PEI, long-term (≥ 3 months) biliary drainage, preoperative chemotherapy, and serum albumin ≥ 3.9 mg/dl before chemotherapy were identified as risk factors for HS. The incidence of postoperative morbidity did not differ between the patients with and without preoperative steatosis. CONCLUSIONS Preoperative chemotherapy, a history of cholangitis, the presence of PEI, and ≥ 3 months' duration of biliary drainage were risk factors for the development of HS before surgery for pancreatic cancer. However, preoperative HS did not affect the short-term postoperative outcomes.
Collapse
Affiliation(s)
- Kazuki Kobayashi
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Yoji Kishi
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| | - Takazumi Tsunenari
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Naoto Yonamine
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Yasuhiro Takihata
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Akiko Nakazawa
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Mikiya Takao
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Takahiro Einama
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Hironori Tsujimoto
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| |
Collapse
|
40
|
Schreiber T, Koopmann I, Brandstetter J, Talbot SR, Goldstein L, Hoffmann L, Schildt A, Joksch M, Krause B, Jaster R, Palme R, Zechner D, Vollmar B, Kumstel S. Evidence-Based Severity Assessment of Animal Models for Pancreatic Cancer. Biomedicines 2024; 12:1494. [PMID: 39062067 PMCID: PMC11275077 DOI: 10.3390/biomedicines12071494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Animal models are crucial to preclinical oncological research and drug development. Animal experiments must be performed in accordance with the 3R principles of replacement and reduction, if possible, and refinement where these procedures remain crucial. In addition, European Union legislations demand a continuous refinement approach, as well as pro- and retrospective severity assessment. In this study, an objective databased severity assessment was performed in murine models for pancreatic cancer induced by orthotopic, subcutaneous, or intravenous injection of Panc02 cells. Parameters such as body weight change, distress score, perianal temperature, mouse grimace scale, burrowing, nesting behavior, and the concentration of corticosterone in plasma and its metabolites in feces were monitored during tumor progression. The most important parameters were combined into a score and mapped against a reference data set by the Relative Severity Assessment procedure (RELSA) to obtain the maximum achieved severity for each animal (RELSAmax). This scoring revealed a significantly higher RELSAmax for the orthotopic model than for the subcutaneous and intravenous models. However, compared to animal models such as pancreatitis and bile duct ligation, the pancreatic cancer models are shown to be less severe. Data-based animal welfare assessment proved to be a valuable tool for comparing the severity of differently induced cancer models.
Collapse
Affiliation(s)
- Tim Schreiber
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| | - Ingo Koopmann
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| | - Jakob Brandstetter
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| | - Steven R. Talbot
- Institute for Laboratory Animal Science, Preclinical Data Science, Hannover Medical School, 30625 Hannover, Germany;
| | - Lea Goldstein
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| | - Lisa Hoffmann
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| | - Anna Schildt
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Markus Joksch
- Department of Nuclear Medicine, Rostock University Medical Center, 18057 Rostock, Germany; (M.J.); (B.K.)
| | - Bernd Krause
- Department of Nuclear Medicine, Rostock University Medical Center, 18057 Rostock, Germany; (M.J.); (B.K.)
| | - Robert Jaster
- Division of Gastroenterology, Department of Medicine II, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Rupert Palme
- Unit of Experimental Endocrinology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Dietmar Zechner
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| | - Simone Kumstel
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| |
Collapse
|
41
|
Okuno M, Mukai T, Iwata K, Takagi A, Ito Y, Ohashi Y, Tezuka R, Iwasa Y, Iwata S, Tomita E. A phase-I study of second-line S-IROX for unresectable pancreatic cancer after gemcitabine plus nab-paclitaxel failure. Med Oncol 2024; 41:195. [PMID: 38967720 DOI: 10.1007/s12032-024-02438-x] [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: 05/09/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024]
Abstract
Gemcitabine plus nab-paclitaxel (GnP) and FOLFIRINOX are widely used as first-line regimens for unresectable pancreatic cancer (PC). When GnP therapy is selected, considering patient age or condition, second-line FOLFIRINOX is sometimes difficult to administer owing to its toxicity. This study aimed to determine the recommended dose (RD) of S-IROX (S-1, oxaliplatin, and irinotecan combination) regimens in patients with unresectable PC after first-line GnP failure. This phase-I study used the "3 + 3" dose-escalation design with two dose levels. Patients who failed first-line GnP therapy for unresectable PC were enrolled. Oxaliplatin and irinotecan were administered on day 1, and S-1 was administered orally twice daily on days 1-7, followed by 7 days of rest. The primary endpoints were dose-limiting toxicities (DLTs) and determination of RD. The secondary endpoint was the evaluation of potential antitumor activity. Nine patients received the second-line S-IROX regimen. In level-0 (S-1, 80 mg/m2; oxaliplatin, 85 mg/m2; and irinotecan, 120 mg/m2), no patient experienced DLT; however, one patient experienced grade 3 neutropenia. At level-1 (irinotecan increased to 150 mg/m2), one of six patients experienced DLTs, including G3 diarrhea. The RD was confirmed at the level-1 dose. The response rate, disease control rate, median progression-free survival, and median overall survival were 33.3%, 77.8%, 172 (range:77-422) days, and 414 (101-685) days, respectively. One patient underwent surgery after the second-line S-IROX therapy. Second-line S-IROX treatment was deemed acceptable. The RD was set at level-1 dose (S-1, 80 mg/m2; oxaliplatin, 85 mg/m2; and irinotecan, 150 mg/m2).
Collapse
Affiliation(s)
- Mitsuru Okuno
- Department of Gastroenterology, Gifu Municipal Hospital, Japan 7-1 Kashimacho, Gifu City, 500-8513, Japan.
- Department of Gastroenterology, Matsunami General Hospital, Kanazawa, Japan.
| | - Tsuyoshi Mukai
- Department of Gastroenterology, Gifu Municipal Hospital, Japan 7-1 Kashimacho, Gifu City, 500-8513, Japan
- Department of Gastroenterological Endoscopy, Kanazawa Medical University, Kanazawa, Japan
| | - Keisuke Iwata
- Department of Gastroenterology, Gifu Municipal Hospital, Japan 7-1 Kashimacho, Gifu City, 500-8513, Japan
| | - Akihiro Takagi
- Department of Gastroenterology, Gifu Municipal Hospital, Japan 7-1 Kashimacho, Gifu City, 500-8513, Japan
| | - Yuki Ito
- Department of Gastroenterology, Gifu Municipal Hospital, Japan 7-1 Kashimacho, Gifu City, 500-8513, Japan
| | - Yosuke Ohashi
- Department of Gastroenterology, Gifu Municipal Hospital, Japan 7-1 Kashimacho, Gifu City, 500-8513, Japan
| | - Ryuichi Tezuka
- Department of Gastroenterology, Gifu Municipal Hospital, Japan 7-1 Kashimacho, Gifu City, 500-8513, Japan
| | - Yuhei Iwasa
- Department of Gastroenterology, Gifu Municipal Hospital, Japan 7-1 Kashimacho, Gifu City, 500-8513, Japan
| | - Shota Iwata
- Department of Gastroenterology, Gifu Municipal Hospital, Japan 7-1 Kashimacho, Gifu City, 500-8513, Japan
| | - Eiichi Tomita
- Department of Gastroenterology, Gifu Municipal Hospital, Japan 7-1 Kashimacho, Gifu City, 500-8513, Japan
- Department of Gastroenterology, Matsunami General Hospital, Kanazawa, Japan
| |
Collapse
|
42
|
Inoue T, Mandai K, Uno K. Multiple Carcinomas In Situ Scattered Throughout the Pancreas Diagnosed by Assessing Focal Pancreatic Parenchymal Atrophy. Pancreas 2024; 53:e544-e545. [PMID: 38888843 DOI: 10.1097/mpa.0000000000002332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Affiliation(s)
- Takato Inoue
- Department of Gastroenterology, Kyoto Second Red Cross Hospital, Kyoto City, Kyoto, Japan
| | | | | |
Collapse
|
43
|
Ren M, Zhang J, Zong R, Sun H. A Novel Pancreatic Cancer Hypoxia Status Related Gene Signature for Prognosis and Therapeutic Responses. Mol Biotechnol 2024; 66:1684-1703. [PMID: 37405638 DOI: 10.1007/s12033-023-00807-x] [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: 02/16/2023] [Accepted: 06/26/2023] [Indexed: 07/06/2023]
Abstract
Pancreatic cancer (PAC) is a highly fatal and aggressive type of cancer. Hypoxia is a common feature of PAC. The aim of this study was to develop a hypoxia status-related prognostic model for predicting the survival outcomes in PAC. The data sets of PAC from The Cancer Genome Atlas and the International Cancer Genome Consortium were used to construct and validate the signature. A 6 hypoxia status-related differential expression genes prognostic model for predicting the survival outcomes was established. The Kaplan-Meier analysis and Received operating characteristic curve indicated the good performance of the signature at predicting overall survival. Univariate and Multivariate Cox regression revealed that the signature was an independent prognostic factor in PAC. Weighted Gene Co-expression Network Analysis and immune infiltration analysis indicated that Immune-related pathways and immune cell infiltration was mostly enriched in the low-risk group, which presented a better prognosis. We also evaluated the predictive of the signature for immunotherapy and chemoradiotherapy. Risk gene LY6D may be a potential prognostic predictor of PAC. This model can be used as an independent prognostic factor for predicting clinical outcomes and a possible classifier for response to chemotherapy.
Collapse
Affiliation(s)
- Min Ren
- College of Life Science, Yan'an University, Yan'an, 716000, China.
| | - Jianing Zhang
- College of Life Science, Yan'an University, Yan'an, 716000, China
| | - Rongrong Zong
- College of Life Science, Yan'an University, Yan'an, 716000, China
| | - Huiru Sun
- College of Life Science, Yan'an University, Yan'an, 716000, China.
| |
Collapse
|
44
|
Askari N, Pirouz MS, Mafikandi V, Hadizadeh M, Mousavi SZ. The lncRNA UCA1 Enhances Pancreatic Cancer EMT by Regulating miR-708-5p and miR-135b-5p: A Bioinformatics Approach. IRANIAN JOURNAL OF PUBLIC HEALTH 2024; 53:1659-1669. [PMID: 39086404 PMCID: PMC11287582 DOI: 10.18502/ijph.v53i7.16060] [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: 08/24/2023] [Accepted: 11/11/2023] [Indexed: 08/02/2024]
Abstract
Background Pancreatic cancer (PC) is an exceedingly malignant ailment that is not only characterized by its insidious onset and rapid progression but also by its poor therapeutic effects. Recently, emerging evidence has shed light on the significant role that non-coding RNAs (ncRNAs), particularly long ncRNAs (lncRNAs) and microRNAs (miRNAs), play in the pathogenesis of PC. This investigation aimed to construct a network of interactions between miRNAs, lncRNAs, and mRNAs, as well as to perform correlation analyses in the context of PC. Methods This study carried out in Kerman City, southeastern Iran in 2023. We utilized the GSE119794 dataset from the Gene Expression Omnibus (GEO) to analyze differentially expressed lncRNAs (DE-lncRNAs), miRNAs (DE-miRNAs), and mRNAs (DE-mRNAs). Following the identification of the DE-lncRNAs, DE-mRNAs, and DE-miRNAs, we proceeded to examine differentially expressed epithelialmesenchymal transition (EMT) genes. Subsequently, we utilized the RNAInter database to predict interactions among lncRNAs, miRNAs, and mRNAs. Finally, we employed Cytoscape to visualize and analyze the constructed network. Results 14 DE-lncRNAs, 14 DE-miRNAs, 545 DE-mRNAs, and 65 DE-EMT from pancreatic cancer and its adjacent tissue RNA-Seq data were identified. 1184 EMT genes from dbEMT were obtained, among which 65 DE-EMT were assigned as EMT genes and correlated with tumor progression. One functional lncRNA (UCA1) was identified as a key functional lncRNA. The area under the ROC curve (AUC) of UCA1 and miR-708-5p were 0.79 and 0.86, respectively. Thus, it is reasonable to believe that this prognostic risk model helps predict PC metastasis. Conclusion UCA1 is a new lncRNA linked with EMT in PC and contributes to a better knowledge of the regulatory mechanisms related to lncRNAs in PC.
Collapse
Affiliation(s)
- Nahid Askari
- Department of Biotechnology, Institute of Sciences and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Marziye Shad Pirouz
- Laboratory of Complex Biological Systems and Bioinformatics (CBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Vida Mafikandi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Hadizadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyedeh Zahra Mousavi
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
45
|
Du Q, Yu Z, Zhang Z, Yang J, Jonckheere N, Shi S, Wang W, Xu J, Liu J, Yu X. Identification of pancreatic adenocarcinoma immune subtype associated with tumor neoantigen from aberrant alternative splicing. J Gastrointest Oncol 2024; 15:1179-1197. [PMID: 38989416 PMCID: PMC11231849 DOI: 10.21037/jgo-24-340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024] Open
Abstract
Background Pancreatic adenocarcinoma (PAAD) is referred to as an immunologically "cold" tumor that responds poorly to immunotherapy. A fundamental theory that explains the low immunogenicity of PAAD is the dramatically low tumor mutation burden (TMB) of PAAD tumors, which fails to induce sufficient immune response. Alternative splicing of pre-mRNA, which could alter the proteomic diversity of many cancers, has been reported to be involved in neoantigen production. Therefore, we aim to identify novel PAAD antigens and immune subtypes through systematic bioinformatics research. Methods Data for splicing analysis were downloaded from The Cancer Genome Atlas (TCGA) SpliceSeq database. Among the available algorithms, we chose CIBERSORT to evaluate the immune cell distribution among PAADs. The TCGA-PAAD expression matrix was used to construct a co-expression network. Single-cell analysis was performed based on the Seurat workflow. Results Integrated analysis of aberrantly upregulated genes, alternatively spliced genes, genes associated with nonsense-mediated RNA decay (NMD) factors, antigen presentation and overall survival (OS) in TCGA-PAAD revealed that PLEC is a promising neoantigen for PAAD-targeted therapy. We identified a C2 TCGA-PAAD subtype that had better prognosis and more CD8+ T-cell infiltration. We propose a novel immune subtyping system for PAAD to indicate patient prognosis and opportunities for immunotherapy, such as immune checkpoint (ICP) inhibitors. Conclusions In conclusion, the present study used a transcriptome-guided approach to screen neoantigen candidates based on alternative splicing, NMD factors, and antigen-presenting signatures for PAAD. A prognosis model with guidance of immunotherapy will aid in patient selection for appropriate treatment.
Collapse
Affiliation(s)
- Qiong Du
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhan Yu
- State Key Laboratory of Radiation Medicine and Protection/Proton & Heavy Ion Medical Research Center, Soochow University, Suzhou, China
- Radiation Oncology Department, Shanghai Concord Medical Cancer Center, Shanghai, China
| | - Zifeng Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jianhui Yang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Nicolas Jonckheere
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, France
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jiyong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| |
Collapse
|
46
|
Kim BH, Kwon M, Lee D, Park SW, Shin E. K-ras mutation detected by peptide nucleic acid-clamping polymerase chain reaction, Ki-67, S100P, and SMAD4 expression can improve the diagnostic accuracy of inconclusive pancreatic EUS-FNB specimens. Pancreatology 2024; 24:584-591. [PMID: 38693041 DOI: 10.1016/j.pan.2024.04.005] [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: 01/06/2024] [Revised: 04/11/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024]
Abstract
OBJECTIVES We aimed to assess the diagnostic utility of an immunohistochemical panel including calcium-binding protein P, p53, Ki-67, and SMAD family member 4 and K-ras mutation for diagnosing pancreatic solid lesion specimens obtained by endoscopic ultrasound-guided fine-needle biopsy and to confirm their usefulness in histologically inconclusive cases. METHODS Immunohistochemistry and peptide nucleic acid-clamping polymerase chain reaction for K-ras mutation were performed on 96 endoscopic ultrasound-guided fine-needle biopsy specimens. The diagnostic efficacy of each marker and the combination of markers was calculated. The diagnostic performances of these markers were evaluated in 27 endoscopic ultrasound-guided fine-needle biopsy specimens with histologically inconclusive diagnoses. A classification tree was constructed. RESULTS K-ras mutation showed the highest accuracy and consistency. Positivity in more than two or three of the five markers showed high diagnostic accuracy (94.6 % and 93.6 %, respectively), and positivity for more than three markers showed the highest accuracy for inconclusive cases (92.0 %). A classification tree using K-ras mutation, Ki-67, S100P, and SMAD4 showed high diagnostic performance, with only two misclassifications in inconclusive cases. CONCLUSIONS K-ras mutation detection via peptide nucleic acid-clamping polymerase chain reaction is a stable and accurate method for distinguishing between pancreatic ductal adenocarcinoma and non-pancreatic ductal adenocarcinoma lesions. A classification tree using K-ras mutation, Ki-67, S100P, and SMAD4 helps increase the diagnostic accuracy of cases that are histologically difficult to diagnose.
Collapse
Affiliation(s)
- Bo-Hyung Kim
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University Hospital, Seoul, South Korea; East-West Medical Research Institute, Kyung Hee University, Seoul, South Korea.
| | - Minji Kwon
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University Hospital, Seoul, South Korea.
| | - Donghwan Lee
- Department of Statistics, Ewha Womans University, Seoul, South Korea.
| | - Se Woo Park
- Division of Gastroenterology, Department of Internal Medicine, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Gyeonggi-do, South Korea.
| | - Eun Shin
- Department of Pathology, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Gyeonggi-do, South Korea.
| |
Collapse
|
47
|
Bhat AA, Gupta G, Dahiya R, Thapa R, Gahtori A, Shahwan M, Jakhmola V, Tiwari A, Kumar M, Dureja H, Singh SK, Dua K, Kumarasamy V, Subramaniyan V. CircRNAs: Pivotal modulators of TGF-β signalling in cancer pathogenesis. Noncoding RNA Res 2024; 9:277-287. [PMID: 38505309 PMCID: PMC10945146 DOI: 10.1016/j.ncrna.2024.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 03/21/2024] Open
Abstract
The intricate molecular landscape of cancer pathogenesis continues to captivate researchers worldwide, with Circular RNAs (circRNAs) emerging as pivotal players in the dynamic regulation of biological functions. The study investigates the elusive link between circRNAs and the Transforming Growth Factor-β (TGF-β) signalling pathway, exploring their collective influence on cancer progression and metastasis. Our comprehensive investigation begins by profiling circRNA expression patterns in diverse cancer types, revealing a repertoire of circRNAs intricately linked to the TGF-β pathway. Through integrated bioinformatics analyses and functional experiments, we elucidate the specific circRNA-mRNA interactions that modulate TGF-β signalling, unveiling the regulatory controls governing this crucial pathway. Furthermore, we provide compelling evidence of the impact of circRNA-mediated TGF-β modulation on key cellular processes, including epithelial-mesenchymal transition (EMT), migration, and cell proliferation. In addition to their mechanistic roles, circRNAs have shown promise as diagnostic and prognostic biomarkers, as well as potential molecular targets for cancer therapy. Their ability to modulate critical pathways, such as the TGF-β signalling axis, underscores their significance in cancer biology and clinical applications. The intricate interplay between circRNAs and TGF-β is dissected, uncovering novel regulatory circuits that contribute to the complexity of cancer biology. This review unravels a previously unexplored dimension of carcinogenesis, emphasizing the crucial role of circRNAs in shaping the TGF-β signalling landscape.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman, 346, United Arab Emirates
| | - Rajiv Dahiya
- School of Pharmacy, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad & Tobago
| | - Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Archana Gahtori
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, 248001, Uttarakhand, India
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman, 346, United Arab Emirates
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, 346, United Arab Emirates
| | - Vikas Jakhmola
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, 248007, India
| | - Abhishek Tiwari
- Pharmacy Academy, IFTM University, Lodhipur-Rajput, Moradabad, (U.P.), 244102, India
| | - Mahish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo, NSW, 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo, NSW, 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology, Sydney, Ultimo, NSW, 2007, Australia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
48
|
Dorma MPF, Giuliani G, Guerra F, Santelli F, Esposito A, De Pastena M, Turri G, Pedrazzani C, Kauffmann EF, Boggi U, Solaini L, Ercolani G, Mastrangelo L, Jovine E, Di Franco G, Morelli L, Mazzola M, Ferrari G, Langella S, Ferrero A, La Mendola R, Abu Hilal M, Depalma N, D'Ugo S, Spampinato MG, Frisini M, Brolese A, Palaia R, Belli A, Cillara N, Deserra A, Cannavera A, Sagnotta A, Mancini S, Pinotti E, Montuori M, Coppola A, Di Benedetto F, Coratti A. The pan - COVID - AGICT study. The impact of COVID-19 pandemic on surgically treated pancreatic cancer patients. A multicentric Italian study. Surg Oncol 2024; 54:102081. [PMID: 38729088 DOI: 10.1016/j.suronc.2024.102081] [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: 12/28/2023] [Revised: 03/28/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND In this article we aimed to perform a subgroup analysis using data from the COVID-AGICT study, to investigate the perioperative outcomes of patients undergoing surgery for pancreatic cancers (PC) during the COVID-19 pandemic. METHODS The primary endpoint of the study was to find out any difference in the tumoral stage of surgically treated PC patients between 2019 and 2020. Surgical and oncological outcomes of the entire cohort of patients were also appraised dividing the entire peri-pandemic period into six three-month timeframes to balance out the comparison between 2019 and 2020. RESULTS Overall, a total of 1815 patients were surgically treated during 2019 and 2020 in 14 Italian surgical Units. In 2020, the rate of patients treated with an advanced pathological stage was not different compared to 2019 (p = 0.846). During the pandemic, neoadjuvant chemotherapy (NCT) has dropped significantly (6.2% vs 21.4%, p < 0.001) and, for patients who didn't undergo NCT, the latency between diagnosis and surgery was shortened (49.58 ± 37 days vs 77.40 ± 83 days, p < 0.001). During 2020 there was a significant increase in minimally invasive procedures (p < 0.001). The rate of postoperative complication was the same in the two years but during 2020 there was an increase of the medical ones (19% vs 16.1%, p = 0.001). CONCLUSIONS The post-pandemic dramatic modifications in healthcare provision, in Italy, did not significantly impair the clinical history of PC patients receiving surgical resection. The present study is one of the largest reports available on the argument and may provide the basis for long-term analyses.
Collapse
Affiliation(s)
- Maria Pia Federica Dorma
- Department of General and Emergency Surgery, Misericordia Hospital, Azienda Usl Toscana Sud Est, School of Robotic Surgery, Grosseto, Italy; Hepato-Pancreato-Biliary Surgery and Liver Transplantation Unit, University of Modena and Reggio Emilia, Modena, Italy.
| | - Giuseppe Giuliani
- Department of General and Emergency Surgery, Misericordia Hospital, Azienda Usl Toscana Sud Est, School of Robotic Surgery, Grosseto, Italy
| | - Francesco Guerra
- Department of General and Emergency Surgery, Misericordia Hospital, Azienda Usl Toscana Sud Est, School of Robotic Surgery, Grosseto, Italy
| | - Francesco Santelli
- Department of Economics, Business, Mathematics and Statistics (DEAMS), University of Trieste, Trieste, Italy
| | - Alessandro Esposito
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona Hospital Trust, Piazzale L.A. Scuro, 10, 37134, Verona, Italy
| | - Matteo De Pastena
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona Hospital Trust, Piazzale L.A. Scuro, 10, 37134, Verona, Italy
| | - Giulia Turri
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, Unit of General and Hepatobiliary Surgery, University and Hospital Trust of Verona, 37134, Verona, Italy
| | - Corrado Pedrazzani
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, Unit of General and Hepatobiliary Surgery, University and Hospital Trust of Verona, 37134, Verona, Italy
| | | | - Ugo Boggi
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | - Leonardo Solaini
- Department of Medical and Surgical Sciences, University of Bologna, Morgagni-Pierantoni Hospital, Forlì, Italy
| | - Giorgio Ercolani
- Department of Medical and Surgical Sciences, University of Bologna, Morgagni-Pierantoni Hospital, Forlì, Italy
| | - Laura Mastrangelo
- Division of General and Emergency Surgery, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Elio Jovine
- Division of General and Emergency Surgery, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Gregorio Di Franco
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56125, Pisa, Italy
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56125, Pisa, Italy
| | - Michele Mazzola
- Division of Minimally-Invasive Surgical Oncology, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Giovanni Ferrari
- Division of Minimally-Invasive Surgical Oncology, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Serena Langella
- Department of General and Oncological Surgery, Mauriziano Hospital, Largo Turati 62, 10128, Turin, Italy
| | - Alessandro Ferrero
- Department of General and Oncological Surgery, Mauriziano Hospital, Largo Turati 62, 10128, Turin, Italy
| | - Roberta La Mendola
- Hepato-Bilio-Pancreatic Minimally Invasive Surgery, Poliambulanza Foundation Hospital, Brescia, Italy
| | - Mohamnad Abu Hilal
- Hepato-Bilio-Pancreatic Minimally Invasive Surgery, Poliambulanza Foundation Hospital, Brescia, Italy
| | - Norma Depalma
- Department of General Surgery, "Vito Fazzi" Hospital, Piazza Muratore 1-73100, Lecce, Italy
| | - Stefano D'Ugo
- Department of General Surgery, "Vito Fazzi" Hospital, Piazza Muratore 1-73100, Lecce, Italy
| | | | - Marco Frisini
- APSS, Department of General Surgery & HPB Unit, Largo Medaglie d'oro 9, 38122, Trento, Italy
| | - Alberto Brolese
- APSS, Department of General Surgery & HPB Unit, Largo Medaglie d'oro 9, 38122, Trento, Italy
| | - Raffaele Palaia
- Department of Abdominal Oncology, Division of Gastro-esophageal and Pancreatic Surgical Oncology, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, Naples, 80131, Italy
| | - Andrea Belli
- Department of Abdominal Oncology, Division of Gastro-esophageal and Pancreatic Surgical Oncology, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, Naples, 80131, Italy
| | - Nicola Cillara
- UOC Chirurgia Generale PO Santissima Trinità ASL Cagliari, Cagliari, Italy
| | - Antonello Deserra
- UOC Chirurgia Generale PO Santissima Trinità ASL Cagliari, Cagliari, Italy
| | | | - Andrea Sagnotta
- General and Oncology Surgery - San Filippo Neri Hospital - ASL Roma 1, Italy
| | - Stefano Mancini
- General and Oncology Surgery - San Filippo Neri Hospital - ASL Roma 1, Italy
| | - Enrico Pinotti
- Department of Surgery, Ponte San Pietro Hospital, Bergamo, Italy
| | - Mauro Montuori
- Department of Surgery, Ponte San Pietro Hospital, Bergamo, Italy
| | | | - Fabrizio Di Benedetto
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Andrea Coratti
- Department of General and Emergency Surgery, Misericordia Hospital, Azienda Usl Toscana Sud Est, School of Robotic Surgery, Grosseto, Italy
| |
Collapse
|
49
|
Wang Z, Wu D, Zhang Y, Chen W, Yang Y, Yang Y, Zu G, An Y, Yu X, Qin Y, Xu X, Chen X. PITX2 functions as a transcription factor for GPX4 and protects pancreatic cancer cells from ferroptosis. Exp Cell Res 2024; 439:114074. [PMID: 38710403 DOI: 10.1016/j.yexcr.2024.114074] [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: 12/12/2023] [Revised: 04/21/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024]
Abstract
Ferroptosis inhibits tumor progression in pancreatic cancer cells, while PITX2 is known to function as a pro-oncogenic factor in various tumor types, protecting them from ferroptosis and thereby promoting tumor progression. In this study, we sought to investigate the regulatory role of PITX2 in tumor cell ferroptosis within the context of pancreatic cancer. We conducted PITX2 knockdown experiments using lentiviral infection in two pancreatic cancer cell lines, namely PANC-1 and BxPC-3. We assessed protein expression through immunoblotting and mRNA expression through RT-PCR. To confirm PITX2 as a transcription factor for GPX4, we employed Chromatin Immunoprecipitation (ChIP) and Dual-luciferase assays. Furthermore, we used flow cytometry to measure reactive oxygen species (ROS), lipid peroxidation, and apoptosis and employed confocal microscopy to assess mitochondrial membrane potential. Additionally, electron microscopy was used to observe mitochondrial structural changes and evaluate PITX2's regulation of ferroptosis in pancreatic cancer cells. Our findings demonstrated that PITX2, functioning as a transcription factor for GPX4, promoted GPX4 expression, thereby exerting an inhibitory effect on ferroptosis in pancreatic cancer cells and consequently promoting tumor progression. Moreover, PITX2 enhanced the invasive and migratory capabilities of pancreatic cancer cells by activating the WNT signaling pathway. Knockdown of PITX2 increased ferroptosis and inhibited the proliferation of PANC-1 and BxPC-3 cells. Notably, the inhibitory effect on ferroptosis resulting from PITX2 overexpression in these cells could be countered using RSL3, an inhibitor of GPX4. Overall, our study established PITX2 as a transcriptional regulator of GPX4 that could promote tumor progression in pancreatic cancer by reducing ferroptosis. These findings suggest that PITX2 may serve as a potential therapeutic target for combating ferroptosis in pancreatic cancer.
Collapse
Affiliation(s)
- Zhiliang Wang
- Department of Hepatopancreatobiliary Surgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Di Wu
- Department of Hepatopancreatobiliary Surgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Yue Zhang
- Department of Hepatopancreatobiliary Surgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Weibo Chen
- Department of Hepatopancreatobiliary Surgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Yang Yang
- Department of Hepatopancreatobiliary Surgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Yue Yang
- Department of Hepatopancreatobiliary Surgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Guangchen Zu
- Department of Hepatopancreatobiliary Surgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Yong An
- Department of Hepatopancreatobiliary Surgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Department of Oncology, Shanghai Medical College, Shanghai Pancreatic Cancer Institute, Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Department of Oncology, Shanghai Medical College, Shanghai Pancreatic Cancer Institute, Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Department of Oncology, Shanghai Medical College, Shanghai Pancreatic Cancer Institute, Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xuemin Chen
- Department of Hepatopancreatobiliary Surgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China.
| |
Collapse
|
50
|
Zhang J, Sun Y, Ma J, Guo X. Deciphering the molecular mechanism of long non-coding RNA HIF1A-AS1 regulating pancreatic cancer cells. Ann Med Surg (Lond) 2024; 86:3367-3377. [PMID: 38846874 PMCID: PMC11152846 DOI: 10.1097/ms9.0000000000002097] [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: 01/25/2024] [Accepted: 04/11/2024] [Indexed: 06/09/2024] Open
Abstract
Background HIF1A-AS1, an antisense transcript of HIF1α gene, is a 652-bp LncRNA that is globally expressed in multiple tissues of animals. Recent evidence indicated that HIF1A-AS1 was involved in tumorigenesis of several types of cancer. However, the role of lncRNA in PC has not been reported, and the molecular mechanism remains elusive. Results In order to investigate the role of HIF1A-AS1 in PC, it was overexpressed in some PC cell lines (PANC-1, PATU8988 and SW1990), and a series of experiments including cell viability detection, flow cytometry, transwell migration, clone formation and wound healing were performed. Functionally, the results indicated that overexpression of HIF1A-AS1 could greatly inhibit proliferation and migration and promote apoptosis of PC cells. Moreover, the isobaric tags for relative and absolute quantification (iTRAQ) quantitative proteomics analysis was implemented to explore the underlying mechanism and the results indicated that OE of HIF1A-AS1 globally affected the expression levels of multiple proteins associated with metabolism of cancer. At last, the network analysis revealed that most of these differentially expressed proteins (DEPs) were integrated and severed essential roles in regulatory function. In view of this, we guessed HIF1A-AS1 overexpression induced the dysfunction of metabolism and disordered proteins' translation, which may account for its excellent tumour suppressor effect. Conclusions HIF1A-AS1 altered the cell function of PC cell lines via affecting the expression of numerous proteins. In summary, HIF1A-AS1 may exhibit a potential therapeutic effect on PC, and our study provided useful information in this filed.
Collapse
Affiliation(s)
- Jiaxin Zhang
- School of Physical Education, Xinxiang Medical University, Xinxiang, Henan
| | - Yifeng Sun
- Department of Occupational Health and Occupational Disease, School of Public Health, Zhengzhou University, Zhengzhou
| | - Jiahui Ma
- Zhengzhou Central Hospital Affiliated to Zhengzhou University, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiang Guo
- Zhengzhou Central Hospital Affiliated to Zhengzhou University, Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|