1
|
Wang L, Zhou S, Ruan Y, Wu X, Zhang X, Li Y, Ying D, Lu Y, Tian Y, Cheng G, Zhang J, Lv K, Zhou X. Hypoxia-Challenged Pancreatic Adenocarcinoma Cell-Derived Exosomal circR3HCC1L Drives Tumor Growth Via Upregulating PKM2 Through Sequestering miR-873-5p. Mol Biotechnol 2025; 67:762-777. [PMID: 38526683 DOI: 10.1007/s12033-024-01091-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/24/2024] [Indexed: 03/27/2024]
Abstract
Pancreatic adenocarcinoma (PAAD) is a fatal disease with poor survival. Increasing evidence show that hypoxia-induced exosomes are associated with cancer progression. Here, we aimed to investigate the function of hsa_circ_0007678 (circR3HCC1L) and hypoxic PAAD cell-derived exosomal circR3HCC1L in PAAD progression. Through the exoRBase 2.0 database, we screened for a circular RNA circR3HCC1L related to PAAD. Changes of circR3HCC1L in PAAD samples and cells were analyzed with real-time quantitative polymerase chain reaction (RT-qPCR). Cell proliferation, migration, invasion were analyzed by colony formation, cell counting, and transwell assays. Measurements of glucose uptake and lactate production were done using corresponding kits. Several protein levels were detected by western blotting. The regulation mechanism of circR3HCC1L was verified by dual-luciferase reporter, RNA immunoprecipitation, and RNA pull-down assays. Exosomes were separated by differential ultracentrifugation. Animal experiments were used to verify the function of hypoxia-derived exosomal circR3HCC1L. CircR3HCC1L was upregulated in PAAD samples and hypoxic PAAD cells. Knockdown of circR3HCC1L decreased hypoxia-driven PAAD cell proliferation, migration, invasion, and glycolysis. Hypoxic PAAD cell-derived exosomes had higher levels of circR3HCC1L, hypoxic PAAD cell-derived exosomal circR3HCC1L promoted normoxic cancer cell malignant transformation and glycolysis in vitro and xenograft tumor growth in mouse models in vivo. Mechanistically, circR3HCC1L regulated pyruvate kinase M2 (PKM2) expression via sponging miR-873-5p. Also, PKM2 overexpression or miR-873-5p silencing offset circR3HCC1L knockdown-mediated effects on hypoxia-challenged PAAD cell malignant transformation and glycolysis. Hypoxic PAAD cell-derived exosomal circR3HCC1L facilitated PAAD progression through the miR-873-5p/PKM2 axis, highlighting the contribution of hypoxic PAAD cell-derived exosomal circR3HCC1L in PAAD.
Collapse
Affiliation(s)
- Luoluo Wang
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Shuping Zhou
- Ningbo College of Health Sciences, No.51, Xuefu Road, Yinzhou District, Ningbo, 315040, Zhejiang, China.
| | - Yi Ruan
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Xiang Wu
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
- Medical School of Ningbo University, Ningbo, 315040, Zhejiang, China
| | - Xueming Zhang
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Yi Li
- College of Computer Science and Artificial Intelligence Wenzhou University, Wenzhou, 325000, Zhejiang, China
| | - Dongjian Ying
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Yeting Lu
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Yuan Tian
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Gong Cheng
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Jing Zhang
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Kaiji Lv
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Xinhua Zhou
- Department of Abdominal Minimally Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, No.1111, Jiangnan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China.
| |
Collapse
|
2
|
Samir S, El-Ashry M, Soliman W, Hassan M. Urinary biomarkers analysis as a diagnostic tool for early detection of pancreatic adenocarcinoma: Molecular quantification approach. Comput Biol Chem 2024; 112:108171. [PMID: 39159599 DOI: 10.1016/j.compbiolchem.2024.108171] [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: 01/29/2024] [Revised: 07/17/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND AND AIMS Pancreatic ductal adenocarcinoma (PDAC) is infrequent. Currently, non-invasive biomarkers for early detection of PDAC are not accessible. Here, we intended to identify a set of urine markers able to discriminate patients with early-stage PDAC from healthy individuals. PATIENTS AND METHODS Seventy-five urine samples from PDAC patients and 50 healthy controls were assayed using quantitative real-time PCR (qPCR). The chosen biomarkers were lymphatic vessel endothelial HA receptor (LYVE-1), regenerating islet-derived 1 alpha (REG1A), and trefoil factor family (TFF1). RESULTS LYVE-1, REG1A, and TFF1 expression in PDAC proved to be significantly elevated compared to healthy individuals (p < 0.05). Determination of these markers' expression might be useful for early tumor diagnosis with a sensitivity of 96 %, 100 %, and 73.33 % respectively, and a specificity of 100 %, 82 %, and 100 % respectively. CONCLUSION We have recognized three diagnostic biomarkers REG1A, TFF1, and LYVE1 that can detect patients with early-stage pancreatic cancer in non-invasive urine specimens with improved sensitivity and specificity. To the best of our knowledge, there have been no prior investigations examining the mRNA expression levels of them in urine within the Egyptian population.
Collapse
Affiliation(s)
- Safia Samir
- Biochemistry and Molecular Biology Department, Theodor Bilharz Research Institute, Giza 12411, Egypt.
| | - Mohamed El-Ashry
- Surgery Department, Theodor Bilharz Research Institute, Giza 12411, Egypt.
| | - Waleed Soliman
- Gastroenterology and Hepatology Department, Theodor Bilharz Research Institute, Giza 12411, Egypt.
| | - Marwa Hassan
- Immunology Department, Theodor Bilharz Research Institute, Giza 12411, Egypt.
| |
Collapse
|
3
|
Cortiana V, Abbas RH, Chorya H, Gambill J, Mahendru D, Park CH, Leyfman Y. Personalized Medicine in Pancreatic Cancer: The Promise of Biomarkers and Molecular Targeting with Dr. Michael J. Pishvaian. Cancers (Basel) 2024; 16:2329. [PMID: 39001391 PMCID: PMC11240738 DOI: 10.3390/cancers16132329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Pancreatic cancer, with its alarming rising incidence, is predicted to become the second deadliest type of solid tumor by 2040, highlighting the urgent need for improved diagnostic and treatment strategies. Despite medical advancements, the five-year survival rate for pancreatic cancer remains about 14%, dropping further when metastasized. This review explores the promise of biomarkers for early detection, personalized treatment, and disease monitoring. Molecular classification of pancreatic cancer into subtypes based on genetic mutations, gene expression, and protein markers guides treatment decisions, potentially improving outcomes. A plethora of clinical trials investigating different strategies are currently ongoing. Targeted therapies, among which those against CLAUDIN 18.2 and inhibitors of Claudin 18.1, have shown promise. Next-generation sequencing (NGS) has emerged as a powerful tool for the comprehensive genomic analysis of pancreatic tumors, revealing unique genetic alterations that drive cancer progression. This allows oncologists to tailor therapies to target specific molecular abnormalities. However, challenges remain, including limited awareness and uptake of biomarker-guided therapies. Continued research into the molecular mechanisms of pancreatic cancer is essential for developing more effective treatments and improving patient survival rates.
Collapse
Affiliation(s)
- Viviana Cortiana
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | | | | | | | - Diksha Mahendru
- Global Remote Research Scholars Program, St. Paul, MN 55101, USA
| | | | - Yan Leyfman
- Icahn School of Medicine at Mount Sinai South Nassau, Oceanside, NY 11572, USA
| |
Collapse
|
4
|
Elango A, Nesam VD, Sukumar P, Lawrence I, Radhakrishnan A. Postbiotic butyrate: role and its effects for being a potential drug and biomarker to pancreatic cancer. Arch Microbiol 2024; 206:156. [PMID: 38480544 DOI: 10.1007/s00203-024-03914-8] [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: 01/19/2024] [Revised: 02/17/2024] [Accepted: 02/26/2024] [Indexed: 04/16/2024]
Abstract
Postbiotics are produced by microbes and have recently gained importance in the field of oncology due to their beneficial effects to the host, effectiveness against cancer cells, and their ability to suppress inflammation. In particular, butyrate dominates over all other postbiotics both in quantity and anticancer properties. Pancreatic cancer (PC), being one of the most malignant and lethal cancers, reported a decreased 5-year survival rate in less than 10% of the patients. PC causes an increased mortality rate due to its inability to be detected at an early stage but still a promising strategy for its diagnosis has not been achieved yet. It is necessary to diagnose Pancreatic cancer before the metastatic progression stage. The available blood biomarkers lack accurate and proficient diagnostic results. Postbiotic butyrate is produced by gut microbiota such as Rhuminococcus and Faecalibacterium it is involved in cell signalling pathways, autophagy, and cell cycle regulation, and reduction in butyrate concentration is associated with the occurrence of pancreatic cancer. The postbiotic butyrate is a potential biomarker that could detect PC at an early stage, before the metastatic progression stage. Thus, this review focused on the gut microbiota butyrate's role in pancreatic cancer and the immuno-suppressive environment, its effects on histone deacetylase and other immune cells, microbes in major butyrate synthesis pathways, current biomarkers in use for Pancreatic Cancer.
Collapse
Affiliation(s)
- Abinaya Elango
- Department of Pharmacology, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Chengalpattu, Tamil Nadu, 603103, India
| | - Vineeta Debbie Nesam
- Department of Pharmacology, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Chengalpattu, Tamil Nadu, 603103, India
| | - Padmaja Sukumar
- Department of Pharmacology, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Chengalpattu, Tamil Nadu, 603103, India
| | - Infancia Lawrence
- Priyadharshani Research and Development, Kelambakkam, Chengalpattu, Tamil Nadu, 603103, India
| | - Arunkumar Radhakrishnan
- Department of Pharmacology, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Chengalpattu, Tamil Nadu, 603103, India.
| |
Collapse
|
5
|
Tajik F, Fattahi F, Rezagholizadeh F, Bouzari B, Babaheidarian P, Baghai Wadji M, Madjd Z. Nuclear overexpression of DNA damage-inducible transcript 4 (DDIT4) is associated with aggressive tumor behavior in patients with pancreatic tumors. Sci Rep 2023; 13:19403. [PMID: 37938616 PMCID: PMC10632485 DOI: 10.1038/s41598-023-46484-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023] Open
Abstract
DNA damage-inducible transcript 4 (DDIT4) is induced in various cellular stress conditions. Several studies showed that the dysregulation of DDIT4 is involved in different malignancies with paradoxical expressions and roles. Therefore, this study investigated the clinical significance, prognostic, and diagnostic value of DDIT4 in different types of pancreatic tumors (PT). The expression of DDIT4 and long non-coding RNA (TPTEP1) in mRNA level was examined in 27 fresh PT samples using Real-time quantitative PCR (RT-qPCR). Moreover, 200 formalin-fixed paraffin-embedded PT tissues, as well as 27 adjacent normal tissues, were collected to evaluate the clinical significance, prognostic, and diagnosis value of DDIT4 expression by immunohistochemistry (IHC) on tissue microarrays (TMA) slides. The results of RT-qPCR showed that the expression of DDIT4 in tumor samples was higher than in normal samples which was associated with high tumor grade (P = 0.015) and lymphovascular invasion (P = 0.048). Similar to this, IHC findings for nucleus, cytoplasm, and membrane localization showed higher expression of DDIT4 protein in PT samples rather than in nearby normal tissues. A statistically significant association was detected between a high level of nuclear expression of DDIT4 protein, and lymphovascular invasion (P = 0.025), as well as advanced TNM stage (P = 0.034) pancreatic ductal adenocarcinoma (PDAC) and in pancreatic neuroendocrine tumor (PNET), respectively. In contrast, a low level of membranous expression of DDIT4 protein showed a significant association with advanced histological grade (P = 0.011), margin involvement (P = 0.007), perineural invasion (P = 0.023), as well as lymphovascular invasion (P = 0.005) in PDAC. No significant association was found between survival outcomes and expression of DDIT4 in both types. It was found that DDIT4 has rational accuracy and high sensitivity as a diagnostic marker. Our results revealed a paradoxical role of DDIT4 expression protein based on the site of nuclear and membranous expression. The findings of this research indicated that there is a correlation between elevated nuclear expression of DDIT4 and the advancement and progression of disease in patients with PT. Conversely, high membranous expression of DDIT4 was associated with less aggressive tumor behavior in patients with PDAC. However, further studies into the prognostic value and biological function of DDIT4 are needed in future studies.
Collapse
Affiliation(s)
- Fatemeh Tajik
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Surgery, University of California, Irvine, CA, USA
| | - Fahimeh Fattahi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Fereshteh Rezagholizadeh
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behnaz Bouzari
- Department of Pathology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Pegah Babaheidarian
- Department of Pathology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Baghai Wadji
- Department of Surgery, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Department of Pathology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Kisling SG, Atri P, Shah A, Cox JL, Sharma S, Smith LM, Ghersi D, Batra SK. A Novel HOXA10-Associated 5-Gene-Based Prognostic Signature for Stratification of Short-term Survivors of Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2023; 29:3759-3770. [PMID: 37432996 PMCID: PMC10529249 DOI: 10.1158/1078-0432.ccr-23-0825] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/02/2023] [Accepted: 07/06/2023] [Indexed: 07/13/2023]
Abstract
PURPOSE Despite the significant association of molecular subtypes with poor prognosis in patients with pancreatic ductal adenocarcinoma (PDAC), few efforts have been made to identify the underlying pathway(s) responsible for this prognosis. Identifying a clinically relevant prognosis-based gene signature may be the key to improving patient outcomes. EXPERIMENTAL DESIGN We analyzed the transcriptomic profiles of treatment-naïve surgically resected short-term survivor (STS) and long-term survivor (LTS) tumors (GSE62452) for expression and survival, followed by validation in several datasets. These results were corroborated by IHC analysis of PDAC-resected STS and LTS tumors. The mechanism of this differential survival was investigated using CIBERSORT and pathway analyses. RESULTS We identified a short-surviving prognostic subtype of PDAC with a high degree of significance (P = 0.018). One hundred thirty genes in this novel subtype were found to be regulated by a master regulator, homeobox gene HOXA10, and a 5-gene signature derived from these genes, including BANF1, EIF4G1, MRPS10, PDIA4, and TYMS, exhibited differential expression in STSs and a strong association with poor survival. This signature was further associated with the proportion of T cells and macrophages found in STSs and LTSs, demonstrating a potential role in PDAC immunosuppression. Pathway analyses corroborated these findings, revealing that this HOXA10-driven prognostic signature is associated with immune suppression and enhanced tumorigenesis. CONCLUSIONS Overall, these findings reveal the presence of a HOXA10-associated prognostic subtype that can be used to differentiate between STS and LTS patients of PDAC and inform on the molecular interactions that play a role in this poor prognosis.
Collapse
Affiliation(s)
- Sophia G. Kisling
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA
| | - Pranita Atri
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA
| | - Ashu Shah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA
| | - Jesse L. Cox
- Department of Pathology and Microbiology, University of Nebraska Medical Center, NE, 68198, USA
| | - Sunandini Sharma
- Department of Pathology and Microbiology, University of Nebraska Medical Center, NE, 68198, USA
| | - Lynette M. Smith
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, NE, 68198, USA
| | - Dario Ghersi
- School of Interdisciplinary Informatics, College of Information Science & Technology, University of Nebraska Omaha, NE, 68182, USA
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, NE, 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, NE, 68198, USA
| |
Collapse
|
7
|
Zhao Q, Wang Y, Huo T, Li F, Zhou L, Feng Y, Wei Z. Exploration of Risk Factors for Pancreatic Cancer and Development of a Clinical High-Risk Group Rating Scale. J Clin Med 2023; 12:358. [PMID: 36615158 PMCID: PMC9821400 DOI: 10.3390/jcm12010358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
(1) Background: There are few studies on people at high risk for clinical pancreatic cancer (PC). We aimed to explore the risk factors of PC and establish a scale for identifying high-risk populations of clinical PC. (2) Methods: We conducted a matched case-control study, retrospectively collecting demographic data and common clinical indicators from all subjects. Logistic regression was used to explore the risk factors of PC. Based on these factors, we created a high-risk population rating scale, which showed a higher diagnostic value. (3) Results: 385 cases and 428 controls were finally enrolled in our study. Multivariate analysis showed that body mass index (BMI) below 18.5 kg/m2 (OR 5.944, 95%CI: 1.759~20.084), smoking (OR 2.745, 95%CI: 1.555~4.844), new-onset diabetes (OR 5.239, 95%CI: 2.091~13.125), low high-density lipoprotein cholesterol (HDL-C) levels (OR 1.790, 95%CI: 1.044~3.069), and carbohydrate antigen 19-9 (CA19-9) levels no less than 35 U/mL (OR 160.328, 95%CI: 83.392~308.243) were associated with an increased risk of PC, whereas high total cholesterol (TC) levels were related to a lower risk of PC (OR 0.392, 95%CI: 0.211~0.730). The high-risk population scale, whose area under the receiver operating curve reached 0.948 (p < 0.001), showed a greater clinical diagnostic value. (4) Conclusions: Smoking history, new-onset diabetes, BMI, TC, HDL-C, and CA19-9 levels were associated with the risk of PC. The high-risk population rating scale might be used for early clinical PC screening.
Collapse
Affiliation(s)
- Qian Zhao
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Yan Wang
- Hepatobiliary and Pancreatic Surgery and Liver Transplantation Center, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Tianyu Huo
- Hepatobiliary and Pancreatic Surgery and Liver Transplantation Center, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Furong Li
- Department of Pathology & Pathophysiology, School of Basic Medicine Shanxi Medical University, Taiyuan 030001, China
| | - Lu Zhou
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Yongliang Feng
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Zhigang Wei
- Hepatobiliary and Pancreatic Surgery and Liver Transplantation Center, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
8
|
Zhang CY, Liu S, Yang M. Clinical diagnosis and management of pancreatic cancer: Markers, molecular mechanisms, and treatment options. World J Gastroenterol 2022; 28:6827-6845. [PMID: 36632312 PMCID: PMC9827589 DOI: 10.3748/wjg.v28.i48.6827] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/04/2022] [Accepted: 11/28/2022] [Indexed: 12/26/2022] Open
Abstract
Pancreatic cancer (PC) is the third-leading cause of cancer deaths. The overall 5-year survival rate of PC is 9%, and this rate for metastatic PC is below 3%. However, the PC-induced death cases will increase about 2-fold by 2060. Many factors such as genetic and environmental factors and metabolic diseases can drive PC development and progression. The most common type of PC in the clinic is pancreatic ductal adenocarcinoma, comprising approximately 90% of PC cases. Multiple pathogenic processes including but not limited to inflammation, fibrosis, angiogenesis, epithelial-mesenchymal transition, and proliferation of cancer stem cells are involved in the initiation and progression of PC. Early diagnosis is essential for curable therapy, for which a combined panel of serum markers is very helpful. Although some mono or combined therapies have been approved by the United States Food and Drug Administration for PC treatment, current therapies have not shown promising outcomes. Fortunately, the development of novel immunotherapies, such as oncolytic viruses-mediated treatments and chimeric antigen receptor-T cells, combined with therapies such as neoadjuvant therapy plus surgery, and advanced delivery systems of immunotherapy will improve therapeutic outcomes and combat drug resistance in PC patients. Herein, the pathogenesis, molecular signaling pathways, diagnostic markers, prognosis, and potential treatments in completed, ongoing, and recruiting clinical trials for PC were reviewed.
Collapse
Affiliation(s)
- Chun-Ye Zhang
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, United States
| | - Shuai Liu
- The First Affiliated Hospital, Zhejiang University, Hangzhou 310006, Zhejiang Province, China
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65211, United States
| |
Collapse
|
9
|
Identification of MYEOV-Associated Gene Network as a Potential Therapeutic Target in Pancreatic Cancer. Cancers (Basel) 2022; 14:cancers14215439. [DOI: 10.3390/cancers14215439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
The molecular mechanism that promotes pancreatic cancer remains unclear, so it is important to find the molecular network of important genes related to pancreatic cancer. To find the key molecule of pancreatic cancer, differential gene expression analyses were analyzed by the Deseq2 package, edgeR package, and limma-voom package, respectively. Pancreatic cancer survival-related genes were analyzed by COX survival analysis. Finally, we integrated the results to obtain the significantly differentially expressed gene, MYEOV (myeloma overexpressed gene), most strongly related to survival in pancreatic cancer. Experimental verification by qRT-PCR confirmed that transcription levels of MYEOV mRNA markedly increased in pancreatic cancer cells relative to normal human pancreatic ductal epithelial cells (HPDE). Through the comprehensive analysis of multiple databases, we constructed a molecular network centered on MYEOV and found specific links between molecules in this network and tumor-associated immune cells. It was noted that MYEOV could serve as a ceRNA by producing molecular sponging effects on hsa-miR-103a-3p and hsa-miR-107, thus affecting the role of GPRC5A, SERPINB5, EGFR, KRAS, EIF4G2, and PDCD4 on pancreatic cancer progression. Besides, we also identified that infiltrated immune cells are potential mediators for the molecules in the MYEOV-related network to promote pancreatic cancer progression. It is the first report to focus on the possibility that MYEOV may act as a competing endogenous RNA (ceRNA) to form an interactive network with some pancreatic cancer-related genes such as KRAS and serve as a key therapeutic target of pancreatic cancer treatment.
Collapse
|
10
|
Zhang CY, Liu S, Yang M. Crosstalk between gut microbiota and COVID-19 impacts pancreatic cancer progression. World J Gastrointest Oncol 2022; 14:1456-1468. [PMID: 36160747 PMCID: PMC9412935 DOI: 10.4251/wjgo.v14.i8.1456] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/26/2022] [Accepted: 07/05/2022] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most common causes of cancer-associated death worldwide, with a low rate of 5-year survival. Currently, the pathogenesis of PC is complicated, with no efficient therapy. Coronavirus disease 2019 (COVID-19) disease caused by severe acute respiratory syndrome coronavirus 2 further exacerbates the challenge of patients with PC. The alteration of gut microbiota caused by COVID-19 infection may impact PC progression in patients via immune regulation. The expression of inflammatory immune mediators such as interleukin (IL)-6, IL-8, and IL-10 has been found to increase in both PC and COVID-19 patients, which is associated with the disease severity and prognostic outcome. Gut microbiome serves as a critical connector between viral infection and PC. It can regulate host systemic immune response and impact the efficacy of immunotherapy. Here, we first demonstrated the features of inflammatory cytokines in both diseases and their impact on disease outcomes. Then, we demonstrated the importance of immunotherapeutic strategies. This includes the immune modulation that targets a single or dual receptors using a single agent or their combinations for the treatment of PC in patients who get infected with COVID-19. Additionally, we explored the possibility of managing the disease by regulating gut microbiome. Overall, modulation of the lung-gut-pancreases axis can boost anti-cancer immunotherapy and reduce adverse prognostic outcomes.
Collapse
Affiliation(s)
- Chun-Ye Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, United States
| | - Shuai Liu
- The First Affiliated Hospital, Zhejiang University, Hangzhou 310006, Zhejiang Province, China
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65211, United States
| |
Collapse
|