1
|
Mahmud S, Ajadee A, Sarker A, Ahmmed R, Noor T, Pappu MAA, Islam MS, Mollah MNH. Exploring common genomic biomarkers to disclose common drugs for the treatment of colorectal cancer and hepatocellular carcinoma with type-2 diabetes through transcriptomics analysis. PLoS One 2025; 20:e0319028. [PMID: 40127075 PMCID: PMC11932495 DOI: 10.1371/journal.pone.0319028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/25/2025] [Indexed: 03/26/2025] Open
Abstract
Type 2 diabetes (T2D) is a crucial risk factor for both colorectal cancer (CRC) and hepatocellular carcinoma (HCC). However, so far, there was no study that has investigated common drugs against HCC and CRC during their co-occurrence with T2D patients. Consequently, patients often require multiple disease-specific multiple drugs, which can lead toxicities and adverse effects to the patients due to drug-drug interactions. This study aimed to identify common genomic biomarkers (cGBs) and associated pathogenetic mechanisms underlying CRC, HCC, and T2D to uncover potential common therapeutic compounds against these three diseases. Firstly, we identified 86 common differentially expressed genes (cDEGs) capable of separating each of CRC, HCC and T2D patients from control groups based on transcriptomic profiling. Of these cDEGs, 37 genes were upregulated and 49 were downregulated. Genetic association studies based on average of Log2 fold-change (aLog2FC) of cDEGs suggested a genetic association among CRC, HCC and T2D. Subsequently, six top-ranked cDEGs (MYC, MMP9, THBS1, IL6, CXCL1, and SPP1) were identified as common genomic biomarkers (cGBs) through protein-protein interaction (PPI) network analysis. Further analysis of these cGBs with GO-terms and KEGG pathways revealed shared pathogenetic mechanisms of three diseases, including specific biological processes, molecular functions, cellular components and signaling pathways. The gene co-regulatory network analysis identified two transcription factors (FOXC1 and GATA2) and three miRNAs (hsa-mir-195-5p, hsa-mir-124a-3p, and hsa-mir-34a-5p) as crucial transcriptional and post-transcriptional regulators of the cGBs. Finally, cGBs-guided seven candidate drugs (Digitoxin, Camptosar, AMG-900, Imatinib, Irinotecan, Midostaurin, and Linsitinib) as the common treatment against T2D, CRC and HCC were identified through molecular docking, cross-validation, and ADME/T (Absorption-Distribution-Metabolism-Excretion-Toxicity) analysis. Most of these findings received support by the literature review of diseases specific individual studies. Thus, this study offers valuable insights for researchers and clinicians to improve the diagnosis and treatment of CRC and/or HCC patients during the co-occurrence of T2D.
Collapse
Affiliation(s)
- Sabkat Mahmud
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Bangladesh
| | - Alvira Ajadee
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Bangladesh
| | - Arnob Sarker
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Bangladesh
- Department of Biochemistry & Molecular Biology, University of Rajshahi, Bangladesh
| | - Reaz Ahmmed
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Bangladesh
- Department of Biochemistry & Molecular Biology, University of Rajshahi, Bangladesh
| | - Tasfia Noor
- Department of Computer Science and Engineering, Rajshahi University of Engineering & Technology (RUET), Bangladesh
| | - Md. Al Amin Pappu
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Bangladesh
| | - Md. Saiful Islam
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Bangladesh
| | | |
Collapse
|
2
|
Chun JW, Lee DE, Han N, Heo S, Kim H, Lee MR, Park HM, Han SS, Park SJ, Kim TH, Lee WJ, Kim YH, Kong SY, Woo SM. Mutant KRAS and GATA6 Stratify Survival in Patients Treated with Chemotherapy for Pancreatic Adenocarcinoma: A Prospective Cohort Study. Cancers (Basel) 2025; 17:896. [PMID: 40075743 PMCID: PMC11899085 DOI: 10.3390/cancers17050896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Several pancreatic adenocarcinoma (PA) biomarkers beyond the traditional carbohydrate antigen (CA)19-9 have been identified but are lacking large-scale prospective validation. This prospective cohort study evaluated the prognostic impact of potential PA biomarkers. METHODS We enrolled 238 of 288 patients with histologically proven PA. We assessed candidate biomarkers, including CA19-9, germline BRCA1/2, and ATM mutations, as well as mutant KRAS circulating tumor DNA (ctDNA) in blood samples. Additionally, we evaluated the expression of SLC29A1 (hENT1), DCK, CES2, and GATA6. We examined the association of candidate biomarkers with progression-free survival (PFS) and overall survival (OS). RESULTS We analyzed biomarker efficacy in 200 (median age 65 years; 55% male) of the enrolled patients who received chemotherapy. A high mutant KRAS ctDNA concentration (hazard ratio [HR]: 1.508 and 95% confidence interval [CI]: 1.052-2.161 for PFS; HR: 1.796 and 95% CI: 1.203-2.681 for OS) and high CA19-9 level (HR: 1.647 and 95% CI: 1.177-2.306 for PFS; HR: 1.803 and 95% CI: 1.248-2.605 for OS) were associated with poor prognosis. High GATA6 RNA expression was linked to longer PFS (HR: 0.336 and 95% CI: 0.195-0.582) and OS (HR: 0.304 and 95% CI: 0.165-0.560). CONCLUSIONS Plasma mutant KRAS ctDNA concentrations and GATA6 expression could serve as significant prognostic biomarkers in patients with PA, potentially guiding therapeutic decisions and prognostication.
Collapse
Affiliation(s)
- Jung Won Chun
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Cancer Biomedical Science, National Cancer Center, Graduate School of Cancer Science and Policy, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Dong-eun Lee
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Nayoung Han
- Department of Pathology, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - SooBeen Heo
- Targeted Therapy Branch, Center for Rare Cancers, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Hyeji Kim
- Targeted Therapy Branch, Center for Rare Cancers, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Mi Rim Lee
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Hyeong Min Park
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Sung-Sik Han
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Cancer Biomedical Science, National Cancer Center, Graduate School of Cancer Science and Policy, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Sang-Jae Park
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Tae Hyun Kim
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Woo Jin Lee
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Yun-Hee Kim
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Cancer Biomedical Science, National Cancer Center, Graduate School of Cancer Science and Policy, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Sun-Young Kong
- Cancer Biomedical Science, National Cancer Center, Graduate School of Cancer Science and Policy, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Targeted Therapy Branch, Center for Rare Cancers, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Department of Laboratory Medicine, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| | - Sang Myung Woo
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
- Cancer Biomedical Science, National Cancer Center, Graduate School of Cancer Science and Policy, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Republic of Korea
| |
Collapse
|
3
|
Ajadee A, Mahmud S, Sarkar A, Noor T, Ahmmed R, Haque Mollah MN. Screening of common genomic biomarkers to explore common drugs for the treatment of pancreatic and kidney cancers with type-2 diabetes through bioinformatics analysis. Sci Rep 2025; 15:7363. [PMID: 40025145 PMCID: PMC11873208 DOI: 10.1038/s41598-025-91875-3] [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: 08/30/2024] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
Type 2 diabetes (T2D) is a crucial risk factor for both pancreatic cancer (PC) and kidney cancer (KC). However, effective common drugs for treating PC and/or KC patients who are also suffering from T2D are currently lacking, despite the probability of their co-occurrence. Taking disease-specific multiple drugs during the co-existence of multiple diseases may lead to adverse side effects or toxicity to the patients due to drug-drug interactions. This study aimed to identify T2D-, PC and KC-causing common genomic biomarkers (cGBs) highlighting their pathogenetic mechanisms to explore effective drugs as their common treatment. We analyzed transcriptomic profile datasets, applying weighted gene co-expression network analysis (WGCNA) and protein-protein interaction (PPI) network analysis approaches to identify T2D-, PC-, and KC-causing cGBs. We then disclosed common pathogenetic mechanisms through gene ontology (GO) terms, KEGG pathways, regulatory networks, and DNA methylation of these cGBs. Initially, we identified 78 common differentially expressed genes (cDEGs) that could distinguish T2D, PC, and KC samples from controls based on their transcriptomic profiles. From these, six top-ranked cDEGs (TOP2A, BIRC5, RRM2, ALB, MUC1, and E2F7) were selected as cGBs and considered targets for exploring common drug molecules for each of three diseases. Functional enrichment analyses, including GO terms, KEGG pathways, and regulatory network analyses involving transcription factors (TFs) and microRNAs, along with DNA methylation and immune infiltration studies, revealed critical common molecular mechanisms linked to PC, KC, and T2D. Finally, we identified six top-ranked drug molecules (NVP.BHG712, Irinotecan, Olaparib, Imatinib, RG-4733, and Linsitinib) as potential common treatments for PC, KC and T2D during their co-existence, supported by the literature reviews. Thus, this bioinformatics study provides valuable insights and resources for developing a genome-guided common treatment strategy for PC and/or KC patients who are also suffering from T2D.
Collapse
Affiliation(s)
- Alvira Ajadee
- Bioinformatics Lab, Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Sabkat Mahmud
- Bioinformatics Lab, Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Arnob Sarkar
- Bioinformatics Lab, Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
- Department of Biochemistry & Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Tasfia Noor
- Department of Computer Science and Engineering, Rajshahi University of Engineering & Technology (RUET), Rajshahi, 6204, Bangladesh
| | - Reaz Ahmmed
- Bioinformatics Lab, Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
- Department of Biochemistry & Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md Nurul Haque Mollah
- Bioinformatics Lab, Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| |
Collapse
|
4
|
Chen Q, Hao Q, Yang Y, Li L, Li D, Zhao R, Wei W, Deng L, Su J, Liang Z, Tang S, Lu Y, Liang Y, Zhang Z, Zhou X, Xiao X, Li P, Huang Y, Zhao W. Carboxylesterase 4A Inhibits the Malignant Biological Behavior of Nasopharyngeal Carcinoma via the PI3K/AKT Pathway. Technol Cancer Res Treat 2025; 24:15330338251319144. [PMID: 39912257 PMCID: PMC11800256 DOI: 10.1177/15330338251319144] [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: 08/06/2024] [Revised: 12/10/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Carboxylesterase 4A (CES4A) belongs to the member of the carboxylesterase family, yet there has been limited research into its malignant biological behavior in malignant tumors. Here, we aim to investigate the expression, cellular biological functions, and the potential underlying mechanism of CES4A in nasopharyngeal carcinoma (NPC). METHOD A standardized mean difference (SMD) analysis was used to analyze the dysregulation of CES4A based on the gene expression omnibus (GEO) database. qRT-PCR and immunohistochemical staining (IHC) were used to identify the mRNA and protein levels of CES4A in NPC cell lines and tissues, respectively. CCK-8, colony formation, wound healing and transwell assays were utilized to estimate cellular growth and metastasis, respectively. Western blot was conducted to evaluate the activity of PI3K/AKT signaling pathway. RESULT Both mRNA and protein expression of CES4A was significantly diminished both in NPC cell lines and primary tumor tissues. Ectopic expression of CES4A restrains the proliferation, colony formation, migration and invasion of NPC. Additionally, KEGG analysis based on GEO data and high-throughput transcriptome sequencing of cell lines all strongly suggested that CES4A was involved in regulating phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway. It was observed that AKT and phosphorylated AKT were remarkably reduced in CES4A overexpressing NPC cells, indicating that PI3K/AKT signaling pathway is hindered by CES4A. CONCLUSION CES4A expression is silenced in NPC, functioning as a tumor suppressor by negatively modulating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Qiaoli Chen
- Department of Pathology, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China
- Key Laboratory of Oral Infectious Disease Prevention and Control, Health Commission of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Regional Key Laboratory of Early Prevention and Treatment of High Incidence Tumors, Nanning, China
| | - Quanxiang Hao
- Department of Pathology, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China
- Key Laboratory of Oral Infectious Disease Prevention and Control, Health Commission of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Regional Key Laboratory of Early Prevention and Treatment of High Incidence Tumors, Nanning, China
| | - Yanping Yang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Limei Li
- Department of Pathology, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China
- Key Laboratory of Oral Infectious Disease Prevention and Control, Health Commission of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Danping Li
- Department of Pathology, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China
- Key Laboratory of Oral Infectious Disease Prevention and Control, Health Commission of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ran Zhao
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Wanqi Wei
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lixian Deng
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Jiaming Su
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ziyuan Liang
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Shiyue Tang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yaomin Lu
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Yushan Liang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhe Zhang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaoying Zhou
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Xue Xiao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ping Li
- Department of Pathology, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China
- Key Laboratory of Oral Infectious Disease Prevention and Control, Health Commission of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yi Huang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Weilin Zhao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
5
|
Cybulski M, Zaremba-Czogalla M, Trzaskowski B, Kubiszewski M, Tobiasz J, Jaromin A, Krzeczyński P, Gubernator J, Michalak O. The conjugates of 5'-deoxy-5-fluorocytidine and hydroxycinnamic acids - synthesis, anti-pancreatic cancer activity and molecular docking studies. RSC Adv 2024; 14:13129-13141. [PMID: 38655481 PMCID: PMC11036175 DOI: 10.1039/d4ra01683a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024] Open
Abstract
New amide conjugates 1-6 of hydroxycinnamic acids (HCA) and 5'-deoxy-5-fluorocytidine (5-dFCR), the prodrug of 5-fluorouracil (5-FU), were synthesized and tested in vitro against pancreatic cancer lines (PDAC). The compounds showed slightly higher efficacy against primary BxPC-3 cells (IC50 values of 14-45 μM) than against metastatic AsPC-1 (IC50 values of 37-133 μM), and similar to that of 5-FU for both PDAC lines. Compound 1, which has a para-(acetyloxy)coumaroyl substituent, was found to be the most potent (IC50 = 14 μM) with a selectivity index of approximately 7 to normal dermal fibroblasts (IC50 = 96 μM). The potential pharmacological profiles were discussed on the basis of the ADME data. Docking to the carboxylesterase CES2 showed that the synthesized compounds have the ability to bind via hydrogen bonding between a specific acetate group of the sugar moiety and Ser228, which belongs to the catalytic triad that causes hydrolysis. Docking to albumin, a major transport protein in the circulatory system, revealed a strong interaction of the conjugates at the binding site which is native to warfarin and responsible for its transport in the body.
Collapse
Affiliation(s)
- Marcin Cybulski
- Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute Rydygiera 8 01-793 Warsaw Poland +48 453 056 175 +48 453 056 177
| | - Magdalena Zaremba-Czogalla
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw Fryderyka Joliot-Curie 14a 50-383 Wroclaw Poland
| | - Bartosz Trzaskowski
- Chemical and Biological Systems Simulation Laboratory, Center of New Technologies, University of Warsaw Banacha 2c 02-097 Warsaw Poland
| | - Marek Kubiszewski
- Pharmaceutical Analysis Laboratory, Łukasiewicz Research Network-Industrial Chemistry Institute Rydygiera 8 01-793 Warsaw Poland
| | - Joanna Tobiasz
- Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute Rydygiera 8 01-793 Warsaw Poland +48 453 056 175 +48 453 056 177
| | - Anna Jaromin
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw Fryderyka Joliot-Curie 14a 50-383 Wroclaw Poland
| | - Piotr Krzeczyński
- Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute Rydygiera 8 01-793 Warsaw Poland +48 453 056 175 +48 453 056 177
| | - Jerzy Gubernator
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw Fryderyka Joliot-Curie 14a 50-383 Wroclaw Poland
| | - Olga Michalak
- Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute Rydygiera 8 01-793 Warsaw Poland +48 453 056 175 +48 453 056 177
| |
Collapse
|
6
|
Wang X, Gao J, Fan C, Gao Y, Yang X, Chen L. New Near-Infrared Fluorescence Imaging Platform with Large Stokes Shift for Carboxylesterase 2 Detection in Thyroid Cancer and Inflammatory Diseases Diagnosis. Anal Chem 2024; 96:3772-3779. [PMID: 38372636 DOI: 10.1021/acs.analchem.3c04399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Development of new near-infrared fluorophores is one of the eternal themes in the field of biosensing and biological imaging. In this work, we constructed a novel fluorophore platform MOR by replacing methylindole of hemicyanine fluorophore (CyR) with benzoxazole to acquire better fluorescence characteristics. Based on the platform, a near infrared (NIR) fluorescent probe MOR-CES2 was synthesized for the specific "off-on" response to carboxylesterase 2 (CES2). The probe exhibited excellent properties including near-infrared emission (735 nm), large Stokes shift (105 nm), high sensitivity (LOD, 0.3 ng/mL), and rapid response (15 min). The successful application of MOR-CES2 in biological imaging of CES2 in mice with thyroid cancer and inflammatory bowel disease demonstrated that the probe could identify cancer cells and tissues and sensitively respond to inflammation. The results proved the potency of MOR-CES2 as an efficient imaging tool to assist in the surgical resection of CES2-related tumors.
Collapse
Affiliation(s)
- Xiaochun Wang
- Liaoning Key Laboratory of Development and Utilization for Natural Products Active Molecules, School of Chemistry and Life Science, Anshan Normal University, Anshan 114007, China
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jian Gao
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Chuanfeng Fan
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yingkai Gao
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xintong Yang
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Lingxin Chen
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Shandong Key Laboratory of Coastal Environmental Processes, Research Center for Coastal Environmental Engineering and Technology, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
- College of Chemistry and Chemical Engineering, Shaoxing University, Shaoxing 312000, China
| |
Collapse
|
7
|
Kailass K, Casalena D, Jenane L, McEdwards G, Auld DS, Sadovski O, Kaye EG, Hudson E, Nettleton D, Currie MA, Beharry AA. Tight-Binding Small-Molecule Carboxylesterase 2 Inhibitors Reduce Intracellular Irinotecan Activation. J Med Chem 2024; 67:2019-2030. [PMID: 38265364 DOI: 10.1021/acs.jmedchem.3c01850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
As the primary enzyme responsible for the activatable conversion of Irinotecan (CPT-11) to SN-38, carboxylesterase 2 (CES2) is a significant predictive biomarker toward CPT-11-based treatments for pancreatic ductal adenocarcinoma (PDAC). High SN-38 levels from high CES2 activity lead to harmful effects, including life-threatening diarrhea. While alternate strategies have been explored, CES2 inhibition presents an effective strategy to directly alter the pharmacokinetics of CPT-11 conversion, ultimately controlling the amount of SN-38 produced. To address this, we conducted a high-throughput screening to discover 18 small-molecule CES2 inhibitors. The inhibitors are validated by dose-response and counter-screening and 16 of these inhibitors demonstrate selectivity for CES2. These 16 inhibitors inhibit CES2 in cells, indicating cell permeability, and they show inhibition of CPT-11 conversion with the purified enzyme. The top five inhibitors prohibited cell death mediated by CPT-11 when preincubated in PDAC cells. Three of these inhibitors displayed a tight-binding mechanism of action with a strong binding affinity.
Collapse
Affiliation(s)
- Karishma Kailass
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada L5L 1C6
| | - Dominick Casalena
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Lina Jenane
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada L5L 1C6
| | - Gregor McEdwards
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada, L5L 1C6
| | - Douglas S Auld
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Oleg Sadovski
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada L5L 1C6
| | - Esther G Kaye
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada L5L 1C6
| | - Elyse Hudson
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada L5L 1C6
| | - David Nettleton
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Mark A Currie
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada, L5L 1C6
| | - Andrew A Beharry
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada L5L 1C6
| |
Collapse
|
8
|
de Jesus VHF, Riechelmann RP. Current Treatment of Potentially Resectable Pancreatic Ductal Adenocarcinoma: A Medical Oncologist's Perspective. Cancer Control 2023; 30:10732748231173212. [PMID: 37115533 PMCID: PMC10155028 DOI: 10.1177/10732748231173212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/01/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Pancreatic cancer has traditionally been associated with a dismal prognosis, even in early stages of the disease. In recent years, the introduction of newer generation chemotherapy regimens in the adjuvant setting has improved the survival of patients treated with upfront resection. However, there are multiple theoretical advantages to deliver early systemic therapy in patients with localized pancreatic cancer. So far, the evidence supports the use of neoadjuvant therapy for patients with borderline resectable pancreatic cancer. The benefit of this treatment sequence for patients with resectable disease remains elusive. In this review, we summarize the data on adjuvant therapy for pancreatic cancer and describe which evidence backs the use of neoadjuvant therapy. Additionally, we address important issues faced in clinical practice when treating patients with localized pancreatic cancer.
Collapse
|
9
|
Elkhanoufi S, Stefania R, Alberti D, Baroni S, Aime S, Geninatti Crich S. Highly Sensitive "Off/On" EPR Probes to Monitor Enzymatic Activity. Chemistry 2022; 28:e202104563. [PMID: 35175676 PMCID: PMC9314618 DOI: 10.1002/chem.202104563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Indexed: 11/16/2022]
Abstract
The assessment of unregulated level of enzyme activity is a crucial parameter for early diagnoses in a wide range of pathologies. In this study, we propose the use of electron paramagnetic resonance (EPR) as an easy method to probe carboxylesterase (CE) enzymatic activity in vitro. For this application, were synthesized two amphiphilic, nitroxide containing esters, namely Tempo-C12 (T-C12) and Tempo-2-C12 (T-2-C12). They exhibit low solubility in water and form stable micelles in which the radicals are EPR almost silent, but the hydrolysis of the ester bond yields narrows and intense EPR signals. The intensity of the EPR signals is proportional to the enzymatic activity. CEs1, CEs2 and esterase from porcine liver (PLE) were investigated. The obtained results show that T-C12 and T-2-C12-containing systems display a much higher selectivity toward the CEs2, with a Limit of Detection of the same order of those ones obtained with optical methods.
Collapse
Affiliation(s)
- Sabrina Elkhanoufi
- University of TorinoDepartment of Molecular Biotechnology and Health Sciencesvia Nizza 5210126TorinoItaly
| | - Rachele Stefania
- University of TorinoDepartment of Molecular Biotechnology and Health Sciencesvia Nizza 5210126TorinoItaly
| | - Diego Alberti
- University of TorinoDepartment of Molecular Biotechnology and Health Sciencesvia Nizza 5210126TorinoItaly
| | - Simona Baroni
- University of TorinoDepartment of Molecular Biotechnology and Health Sciencesvia Nizza 5210126TorinoItaly
| | - Silvio Aime
- University of TorinoDepartment of Molecular Biotechnology and Health Sciencesvia Nizza 5210126TorinoItaly
| | - Simonetta Geninatti Crich
- University of TorinoDepartment of Molecular Biotechnology and Health Sciencesvia Nizza 5210126TorinoItaly
| |
Collapse
|
10
|
Chen Y, Capello M, Rios Perez MV, Vykoukal JV, Roife D, Kang Y, Prakash LR, Katayama H, Irajizad E, Fleury A, Ferri-Borgogno S, Baluya DL, Dennison JB, Do KA, Fiehn O, Maitra A, Wang H, Chiao PJ, Katz MHG, Fleming JB, Hanash SM, Fahrmann JF. CES2 sustains HNF4α expression to promote pancreatic adenocarcinoma progression through an epoxide hydrolase-dependent regulatory loop. Mol Metab 2022; 56:101426. [PMID: 34971802 PMCID: PMC8841288 DOI: 10.1016/j.molmet.2021.101426] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/16/2021] [Accepted: 12/23/2021] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE Intra-tumoral expression of the serine hydrolase carboxylesterase 2 (CES2) contributes to the activation of the pro-drug irinotecan in pancreatic ductal adenocarcinoma (PDAC). Given other potential roles of CES2, we assessed its regulation, downstream effects, and contribution to tumor development in PDAC. METHODS Association between the mRNA expression of CES2 in pancreatic tumors and overall survival was assessed using The Cancer Genome Atlas. Cell viability, clonogenic, and anchorage-independent growth assays as well as an orthotopic mouse model of PDAC were used to evaluate the biological relevance of CES2 in pancreatic cancer. CES2-driven metabolic changes were determined by untargeted and targeted metabolomic analyses. RESULTS Elevated tumoral CES2 mRNA expression was a statistically significant predictor of poor overall survival in PDAC patients. Knockdown of CES2 in PDAC cells reduced cell viability, clonogenic capacity, and anchorage-independent growth in vitro and attenuated tumor growth in an orthotopic mouse model of PDAC. Mechanistically, CES2 was found to promote the catabolism of phospholipids resulting in HNF4α activation through a soluble epoxide hydrolase (sEH)-dependent pathway. Targeting of CES2 via siRNA or small molecule inhibitors attenuated HNF4α protein expression and reduced gene expression of classical/progenitor markers and increased basal-like markers. Targeting of the CES2-sEH-HNF4α axis using small molecule inhibitors of CES2 or sEH reduced cell viability. CONCLUSIONS We establish a novel regulatory loop between CES2 and HNF4α to sustain the progenitor subtype and promote PDAC progression and highlight the potential utility of CES2 or sEH inhibitors for the treatment of PDAC as part of non-irinotecan-containing regimens.
Collapse
Affiliation(s)
- Yihui Chen
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michela Capello
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mayrim V Rios Perez
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jody V Vykoukal
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Roife
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ya'an Kang
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laura R Prakash
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hiroyuki Katayama
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ehsan Irajizad
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alia Fleury
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sammy Ferri-Borgogno
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dodge L Baluya
- Departments of Center for Radiation Oncology Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer B Dennison
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kim-Anh Do
- Departments of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Oliver Fiehn
- UC Davis Genome Center - Metabolomics, University of California, Davis, 95616, CA, USA
| | - Anirban Maitra
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, USA; Departments of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Huamin Wang
- Departments of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul J Chiao
- Departments of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew H G Katz
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason B Fleming
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Samir M Hanash
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Johannes F Fahrmann
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
11
|
Determination of carboxylesterase 2 by fluorescence probe to guide pancreatic adenocarcinoma profiling. Chem Phys Lett 2021. [DOI: 10.1016/j.cplett.2021.139143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
12
|
Gómez-España MA, Montes AF, Garcia-Carbonero R, Mercadé TM, Maurel J, Martín AM, Pazo-Cid R, Vera R, Carrato A, Feliu J. SEOM clinical guidelines for pancreatic and biliary tract cancer (2020). Clin Transl Oncol 2021; 23:988-1000. [PMID: 33660222 PMCID: PMC8058005 DOI: 10.1007/s12094-021-02573-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2021] [Indexed: 11/10/2022]
Abstract
Pancreatic cancer (PC) and biliary tract cancer (BTC) are both aggressive and highly fatal malignancies. Nowadays we have a profound knowledge about the molecular landscape of these neoplasms and this has allowed new therapeutic options. Surgery is the only potentially curative therapy in both cancers, but disease recurrence is frequent. In PC, adjuvant treatment with mFOLFIRINOX has improved overall survival (OS) and in BTC adjuvant treatment with capecitabine seems to improve OS and relapse-free survival. Concomitant radio-chemotherapy could also be considered following R1 surgery in both neoplasms. Neoadjuvant treatment represents the best option for achieving an R0 resection in borderline PC. Upfront systemic chemotherapy is the treatment of choice in unresectable locally advanced PC and BTC; then locoregional therapy could be considered after an initial period of at least 3-4 months of systemic chemotherapy. In metastatic PC, FOLFIRINOX or Gemcitabine plus nab-paclitaxel have improved OS compared with gemcitabine alone. In metastatic BTC, cisplatin plus gemcitabine constitute the standard treatment. Progress in the knowledge of molecular biology has enabled the identification of new targets for therapy with encouraging results that could in the future improve the survival and quality of life of patients with PC and BTC.
Collapse
Affiliation(s)
- Mª A. Gómez-España
- Medical Oncology Department, Hospital Universitario Reina Sofía, IMIBIC, CIBERONC, Córdoba, Spain
| | - A. F. Montes
- Medical Oncology Department, Complexo Hospitalario Universitario de Ourense (CHUO), Orense, Spain
| | - R. Garcia-Carbonero
- Medical Oncology Department, Hospital Universitario, UCM, CNIO, CIBERONC, 12 de Octubre, IIS imas12, Madrid, Spain
| | - T. M. Mercadé
- Medical Oncology Department, Hospital Universitari Vall D´Hebron, Barcelona, Spain
| | - J. Maurel
- Medical Oncology Department, Hospital Clinic Barcelona, Barcelona, Spain
| | - A. M. Martín
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - R. Pazo-Cid
- Medical Oncology Department, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - R. Vera
- Medical Oncology Department, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - A. Carrato
- Medical Oncology Department, Hospital Universitario Ramón Y Cajal, Alcala University, IRYCIS, CIBERONC, Madrid, Spain
| | - J. Feliu
- Medical Oncology Department, Hospital Universitario La Paz, CIBERONC, IDIPAZ, Madrid, Spain
| |
Collapse
|