1
|
da Silva EL, Mesquita FP, Pinto LC, Gomes BPS, de Oliveira EHC, Burbano RMR, Moraes MEAD, de Souza PFN, Montenegro RC. Transcriptome analysis displays new molecular insights into the mechanisms of action of Mebendazole in gastric cancer cells. Comput Biol Med 2025; 184:109415. [PMID: 39566281 DOI: 10.1016/j.compbiomed.2024.109415] [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: 06/22/2024] [Revised: 10/15/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
Gastric cancer (GC) is a common cancer worldwide. Therefore, searching for effective treatments is essential, and drug repositioning can be a promising strategy to find new potential drugs for GC therapy. For the first time, we sought to identify molecular alterations and validate new mechanisms related to Mebendazole (MBZ) treatment in GC cells through transcriptome analysis using microarray technology. Data revealed 1066 differentially expressed genes (DEGs), of which 345 (2.41 %) genes were upregulated, 721 (5.04 %) genes were downregulated, and 13,231 (92.54 %) genes remained unaltered after MBZ exposure. The overexpressed genes identified were CCL2, IL1A, and CDKN1A. In contrast, the H3C7, H3C11, and H1-5 were the top 3 underexpressed genes. Gene set enrichment analysis (GSEA) identified 8 pathways significantly overexpressed in the treated group (p < 0.05 and FDR<0.25). The validation of the expression of top desregulated genes by RT-qPCR confirmed the transcriptome results, where MBZ increased the CCL2, IL1A, and CDKN1A and reduced the H3C7, H3C11, and H1-5 transcript levels. Expression analysis in samples from TCGA databases correlated that the lower ILI1A and higher H3C11 and H1-5 gene expression are associated with decreased overall survival rates in patients with GC, indicating that MBZ treatment can improve the prognosis of patients. Thus, the data demonstrated that the drug MBZ alters the transcriptome of the AGP-01 lineage, mainly modulating the expression of histone proteins and inflammatory cytokines, indicating a possible epigenetic and immunological effect on tumor cells, these findings highlight new mechanisms of action related to MBZ treatment. Additional studies are still needed to better clarify the epigenetic and immune mechanism of MBZ in the therapy of GC.
Collapse
Affiliation(s)
- Emerson Lucena da Silva
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, Fortaleza, Brazil
| | - Felipe Pantoja Mesquita
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, Fortaleza, Brazil
| | - Laine Celestino Pinto
- Laboratory of Experimental Neuropathology, Biological Science Institute, Federal University of Pará, Mundurucus Street, Belém, Brazil
| | - Bruna Puty Silva Gomes
- Laboratory of Cytogenomics and Environmental Mutagenesis, Environment Section (SAMAM), Evandro Chagas Institute (IEC), Ananindeua, Brazil
| | | | - Rommel Mario Rodríguez Burbano
- Molecular Biology Laboratory, Ophir Loyola Hospital, Av. Governador Magalhães Barata, Belém, Brazil; Laboratory of Human Cytogenetics, Institute of Biological Sciences, Federal University of Pará, Augusto Correa Avenue, Belém, Brazil
| | - Maria Elisabete Amaral de Moraes
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, Fortaleza, Brazil
| | - Pedro Filho Noronha de Souza
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, Fortaleza, Brazil; Visiting Researcher at the Cearense Foundation to Support Scientific and Technological Development, Brazil; National Institute of Science and Technology in Human Pathogenic Fungi, Ribeirão Preto, Brazil.
| | - Raquel Carvalho Montenegro
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, Fortaleza, Brazil; Red Latinoamericana de Implementación y Validación de guias clinicas Farmacogenomicas (RELIVAF), Brazil.
| |
Collapse
|
2
|
Sabale P, Sayyad N, Sabale V, Begum T, Murali Prakash J, Gobalakriahnan P, Hemalatha K, Parupathi P, Kumar Reddy KT, Kolli D, Ali Alshehri M, Obaidur Rab S, Bin Emran T. Molecular Docking, Synthesis and Biological Evaluation of New Benzimidazole‐Pyridine Derivatives as Potential Aromatase Inhibitor for the Treatment of Cancer. ChemistrySelect 2024; 9. [DOI: 10.1002/slct.202401797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/05/2024] [Indexed: 01/12/2025]
Abstract
AbstractThis study describes the synthesis of N5‐(4‐(1H‐benzo[d]imidazol‐2‐yl)phenyl)‐N2‐phenylpyridine‐2,5‐diamine derivatives from Orthophenylenediamine (1) and 4‐aminobenzoic acid (2) and all the synthesized chemical moieties screened against a panel of cancer cell lines resulted in the identification compound 6 a with good anti‐cancer potential and a GI50 of 2.95 μM, 3.35 μM, 2.27 μM, 8.46 nM and 1.56 μM against MDAMB‐231, MCF‐7, A‐549, NCI‐H23 and A‐498 respectively. As the second greatest cause of death globally, cancer continues to pose a serious threat to public health. An essential enzyme called aromatase catalyses the last, rate‐limiting step in the production of oestrogens. As a well‐researched endocrine therapeutic strategy, aromatase inhibitors (AIs) efficiently block the production of oestrogen, which is necessary for aromatase activity.
Collapse
Affiliation(s)
- Prafulla Sabale
- Department of Pharmaceutical Sciences Nagpur Rashtrasant Tukdoji Maharaj Nagpur, University Nagpur, MS 440033 India
| | - Nusrat Sayyad
- Department of Pharmaceutical Sciences Nagpur Rashtrasant Tukdoji Maharaj Nagpur, University Nagpur, MS 440033 India
| | - Vidya Sabale
- Dadasaheb Balpande College of Pharmacy Besa, Nagpur, MS 440037 India
| | - Touseef Begum
- Department of Pharmaceutical Sciences Ibn Sina National College for Medical Studies Jeddah 31906 Kingdom of Saudi Arabia
| | - Jatla Murali Prakash
- Department of Chemistry Koneru Lakshmaiah Education Foundation Green fields Vaddeswaram, Guntur, Andhra Pradesh 522502 India
| | - P. Gobalakriahnan
- Department of Pharmaceutical Chemistry Vagdevi College of Pharmacy and Research Centre Nellore, Andhra Pradesh India
| | - K. Hemalatha
- Department of Pharmaceutical Chemistry Saveetha College of Pharmacy Saveetha Institute of Medical and Technical Sciences, Saveetha University, Thandalam Chennai, Tamil Nadu 602105 India
| | - Prashanth Parupathi
- Division of Pharmaceutical Sciences Arnold & Marie Schwartz College of Pharmacy and Health Sciences Long Island University Brooklyn NY-11201 USA
| | - Konatham Teja Kumar Reddy
- Department of Pharmacy Practice St. Peter's Institute of Pharmaceutical Sciences Hanamkonda, Telangana 506331 India
| | - Deepti Kolli
- Department of Chemistry Koneru Lakshmaiah Education Foundation Green fields Vaddeswaram, Guntur, Andhra Pradesh 522502 India
| | - Mohammed Ali Alshehri
- Department of Biology Faculty of Science University of Tabuk Tabuk 71491 Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences College of Applied Medical Science King Khalid University Abha Saudi Arabia
- Central Labs King Khalid University AlQura'a, Abha P.O. Box 960 Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy Faculty of Allied Health Sciences Daffodil International University Dhaka 1207 Bangladesh
| |
Collapse
|
3
|
Dong W, Lu J, Li Y, Zeng J, Du X, Yu A, Zhao X, Chi F, Xi Z, Cao S. SIRT1: a novel regulator in colorectal cancer. Biomed Pharmacother 2024; 178:117176. [PMID: 39059350 DOI: 10.1016/j.biopha.2024.117176] [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] [Revised: 07/08/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024] Open
Abstract
The class-III histone deacetylase SIRT1 is the most extensively investigated sirtuin deacetylase. It is resistant to the broad deacetylase inhibitor trichostatin A and depends on oxidized nicotinamide adenine nucleotide (NAD+). SIRT1 plays a crucial role in the tumorigenesis of numerous types of cancers, including colorectal cancer (CRC). Accumulating evidence indicates that SIRT1 is a therapeutic target for CRC; however, the function and underlying mechanism of SIRT1 in CRC still need to be elucidated. Herein, we provide a detailed and updated review to illustrate that SIRT1 regulates many processes that go awry in CRC cells, such as apoptosis, autophagy, proliferation, migration, invasion, metastasis, oxidative stress, resistance to chemo-radio therapy, immune evasion, and metabolic reprogramming. Moreover, we closely link our review to the clinical practice of CRC treatment, summarizing the mechanisms and prospects of SIRT1 inhibitors in CRC therapy. SIRT1 inhibitors as monotherapy in CRC or in combination with chemotherapy, radiotherapy, and immune therapies are comprehensively discussed. From epigenetic regulation to its potential therapeutic effect, we hope to offer novel insights and a comprehensive understanding of SIRT1's role in CRC.
Collapse
Affiliation(s)
- Weiwei Dong
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China
| | - Jinjing Lu
- Department of Health Management, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China
| | - You Li
- Nursing Department, Liaoning Jinqiu Hospital, Shenyang, Liaoning Province 110016, China
| | - Juan Zeng
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China
| | - Xiaoyun Du
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China
| | - Ao Yu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China
| | - Xuechan Zhao
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China
| | - Feng Chi
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China.
| | - Zhuo Xi
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China.
| | - Shuo Cao
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China.
| |
Collapse
|
4
|
Baldasso-Zanon A, Silva AO, Franco N, Picon RV, Lenz G, Lopez PLDC, Filippi-Chiela EC. The rational modulation of autophagy sensitizes colorectal cancer cells to 5-fluouracil and oxaliplatin. J Cell Biochem 2024; 125:e30517. [PMID: 38224178 DOI: 10.1002/jcb.30517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/25/2023] [Accepted: 12/18/2023] [Indexed: 01/16/2024]
Abstract
Colorectal cancer (CRC) is the third most common and deadliest cancer globally. Regimens using 5-fluorouracil (5FU) and Oxaliplatin (OXA) are the first-line treatment for CRC, but tumor recurrence is frequent. It is plausible to hypothesize that differential cellular responses are triggered after treatments depending on the genetic background of CRC cells and that the rational modulation of cell tolerance mechanisms like autophagy may reduce the regrowth of CRC cells. This study proposes investigating the cellular mechanisms triggered by CRC cells exposed to 5FU and OXA using a preclinical experimental design mimicking one cycle of the clinical regimen (i.e., 48 h of treatment repeated every 2 weeks). To test this, we treated CRC human cell lines HCT116 and HT29 with the 5FU and OXA, combined or not, for 48 h, followed by analysis for two additional weeks. Compared to single-drug treatments, the co-treatment reduced tumor cell regrowth, clonogenicity and stemness, phenotypes associated with tumor aggressiveness and poor prognosis in clinics. This effect was exerted by the induction of apoptosis and senescence only in the co-treatment. However, a week after treatment, cells that tolerated the treatment had high levels of autophagy features and restored the proliferative phenotype, resembling tumor recurrence. The pharmacologic suppression of early autophagy during its peak of occurrence, but not concomitant with chemotherapeutics, strongly reduced cell regrowth. Overall, our experimental model provides new insights into the cellular mechanisms that underlie the response and tolerance of CRC cells to 5FU and OXA, suggesting optimized, time-specific autophagy inhibition as a new avenue for improving the efficacy of current treatments.
Collapse
Affiliation(s)
- Andréa Baldasso-Zanon
- Programa de Pós-Graduação Ciências em Gastroenterologia e Hepatologia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Centro de Pesquisas Experimental, Laboratório de Biologia Celular e Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Andrew Oliveira Silva
- Centro de Pesquisas Experimental, Laboratório de Biologia Celular e Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Unidade Centro RS, Faculdade Estácio do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Nayara Franco
- Programa de Pós-Graduação Ciências em Gastroenterologia e Hepatologia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Centro de Pesquisas Experimental, Laboratório de Biologia Celular e Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Rafael V Picon
- Programa de Pós-Graduação Ciências em Gastroenterologia e Hepatologia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Departamento de Medicina Interna, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Guido Lenz
- Departamento de Biofísica, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Patrícia Luciana da Costa Lopez
- Programa de Pós-Graduação Ciências em Gastroenterologia e Hepatologia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Centro de Pesquisas Experimental, Laboratório de Biologia Celular e Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Eduardo C Filippi-Chiela
- Programa de Pós-Graduação Ciências em Gastroenterologia e Hepatologia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Centro de Pesquisas Experimental, Laboratório de Biologia Celular e Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Departamento de Ciências Morfológicas, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
5
|
Youssif BGM, Morcoss MM, Bräse S, Abdel-Aziz M, Abdel-Rahman HM, Abou El-Ella DA, Abdelhafez ESMN. Benzimidazole-Based Derivatives as Apoptotic Antiproliferative Agents: Design, Synthesis, Docking, and Mechanistic Studies. Molecules 2024; 29:446. [PMID: 38257358 PMCID: PMC10819888 DOI: 10.3390/molecules29020446] [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: 11/06/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
A new class of benzimidazole-based derivatives (4a-j, 5, and 6) with potential dual inhibition of EGFR and BRAFV600E has been developed. The newly synthesized compounds were submitted for testing for antiproliferative activity against the NCI-60 cell line. All newly synthesized compounds 4a-j, 5, and 6 were selected for testing against a panel of sixty cancer cell lines at a single concentration of 10 µM. Some compounds tested demonstrated remarkable antiproliferative activity against the cell lines tested. Compounds 4c, 4e, and 4g were chosen for five-dose testing against 60 human tumor cell lines. Compound 4c demonstrated strong selectivity against the leukemia subpanel, with a selectivity ratio of 5.96 at the GI50 level. The most effective in vitro anti-cancer assay derivatives (4c, 4d, 4e, 4g, and 4h) were tested for EGFR and BRAFV600E inhibition as potential targets for antiproliferative action. The results revealed that compounds 4c and 4e have significant antiproliferative activity as dual EGFR/BRAFV600E inhibitors. Compounds 4c and 4e induced apoptosis by increasing caspase-3, caspase-8, and Bax levels while decreasing the anti-apoptotic Bcl2 protein. Moreover, molecular docking studies confirmed the potential of compounds 4c and 4e to act as dual EGFR/BRAFV600E inhibitors.
Collapse
Affiliation(s)
- Bahaa G. M. Youssif
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Martha M. Morcoss
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Nahda University, Beni-Suef 62513, Egypt;
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, IBCS-FMS, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Mohamed Abdel-Aziz
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt; (M.A.-A.); (E.S.M.N.A.)
| | - Hamdy M. Abdel-Rahman
- Department of Medicinal Chemistry, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt;
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Badr University in Assiut (BUA), Assiut 71536, Egypt
| | - Dalal A. Abou El-Ella
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy Ain Shams University, Cairo 11566, Egypt;
| | - El Shimaa M. N. Abdelhafez
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt; (M.A.-A.); (E.S.M.N.A.)
| |
Collapse
|
6
|
Lee YT, Tan YJ, Oon CE. BZD9L1 Differentially Regulates Sirtuins in Liver-Derived Cells by Inducing Reactive Oxygen Species. Biomedicines 2023; 11:3059. [PMID: 38002059 PMCID: PMC10669747 DOI: 10.3390/biomedicines11113059] [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: 06/26/2023] [Revised: 11/04/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Growing evidence has highlighted that mitochondrial dysfunction contributes to drug-induced toxicities and leads to drug attrition and post-market withdrawals. The acetylation or deacetylation of mitochondrial proteins can affect mitochondrial functions as the cells adapt to various cellular stresses and other metabolic challenges. SIRTs act as critical deacetylases in modulating mitochondrial function in response to drug toxicity, oxidative stress, reactive oxygen species (ROS), and energy metabolism. We previously showed that a recently characterised SIRT inhibitor (BZD9L1) is non-toxic in rodents in a short-term toxicity evaluation. However, the impact of BZD9L1 on mitochondrial function is unknown. This work aims to determine the effects of BZD9L1 on mitochondrial function in human normal liver and kidney-derived cell lines using the Agilent Seahorse Cell Mito Stress Test to complement our short-term toxicity evaluations in vivo. The Mito Stress assay revealed that BZD9L1 could potentially trigger oxidative stress by inducing ROS, which promotes proton leak and reduces coupling efficiency in liver-derived THLE cells. However, the same was not observed in human kidney-derived HEK293 cells. Interestingly, BZD9L1 had no impact on SIRT3 protein expression in both cell lines but affected SOD2 and its acetylated form at 72 h in THLE cells, indicating that BZD9L1 exerted its effect through SIRT3 activity rather than protein expression. In contrast, BZD9L1 reduced SIRT1 protein expression and impacted the p53 protein differently in both cell lines. Although BZD9L1 did not affect the spare respiratory capacity in vitro, these findings call for further validation of mitochondrial function through assessment of other mitochondrial parameters to evaluate the safety of BZD9L1.
Collapse
Affiliation(s)
| | | | - Chern Ein Oon
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor 11800, Penang, Malaysia; (Y.T.L.); (Y.J.T.)
| |
Collapse
|
7
|
Akkoc S, Sahin D, Muhammed MT, Yıldız M, Ilhan IO. Synthesis, characterization, antiproliferative activity, docking, and molecular dynamics simulation of new 1,3-dihydro-2 H-benzimidazol-2-one derivatives. J Biomol Struct Dyn 2023; 42:11495-11507. [PMID: 37787572 DOI: 10.1080/07391102.2023.2262601] [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: 05/10/2023] [Accepted: 09/17/2023] [Indexed: 10/04/2023]
Abstract
Cancer is a global public health problem that affects millions each year. Novel anticancer drug candidates are in need to treat various cancers and to overcome the resistance that exists against drugs in use. Benzimidazole derivatives have been reported as anticancer agents. These lead us to synthesize similar benzimidazole derivatives and investigate their anticancer activity. In this study, six new 1,3-dihydro-2H-benzimidazol-2-one-based molecules (2a-f) were synthesized. The structures of these molecules were verified by spectroscopic methods. The antiproliferative activities of molecules 2a-f were screened against a panel of human cancer cell lines, including the liver, colon, lung, and breast. The molecules were also tested towards normal human lung cell line to determine their selectivity. The results demonstrated that compound 2d had the highest cytotoxic effect compared to compounds 2a-c, 2e, and 2f against DLD-1 and MDA-MB-231 cell lines. The binding potential of the relatively active compound, 2d, with three targets was investigated through molecular docking. The stability of target-compound complexes procured from the docking was explored through molecular dynamics (MD) simulation. The docking and MD simulation studies revealed that compound 2d had the highest potential to bind to GALR3 among the targets. Furthermore, the computational pharmacokinetic study demonstrated that the synthesized compounds had drug-like properties.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Senem Akkoc
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Suleyman Demirel University, Isparta, Türkiye
- Faculty of Engineering and Natural Sciences, Bahçeşehir University, Istanbul, Türkiye
| | - Dicle Sahin
- Department of Pharmaceutical Research and Development, Institute of Health Sciences, Suleyman Demirel University, Isparta, Türkiye
| | - Muhammed Tilahun Muhammed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Suleyman Demirel University, Isparta, Türkiye
| | - Mustafa Yıldız
- Department of Nuclear Medicine, Faculty of Medicine, Suleyman Demirel University, Isparta, Türkiye
| | - Ilhan Ozer Ilhan
- Department of Chemistry, Faculty of Science, Erciyes University, Kayseri, Türkiye
| |
Collapse
|
8
|
Oon CE, Subramaniam AV, Ooi LY, Yehya AHS, Lee YT, Kaur G, Sasidharan S, Qiu B, Wang X. BZD9L1 benzimidazole analogue hampers colorectal tumor progression by impeding angiogenesis. World J Gastrointest Oncol 2023; 15:810-827. [PMID: 37275453 PMCID: PMC10237024 DOI: 10.4251/wjgo.v15.i5.810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/17/2023] [Accepted: 04/21/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND The development of new vasculatures (angiogenesis) is indispensable in supplying oxygen and nutrients to fuel tumor growth. Epigenetic dysregulation in the tumor vasculature is critical to colorectal cancer (CRC) progression. Sirtuin (SIRT) enzymes are highly expressed in blood vessels. BZD9L1 benzimidazole analogue is a SIRT 1 and 2 inhibitor with reported anticancer activities in CRC. However, its role has yet to be explored in CRC tumor angiogenesis.
AIM To investigate the anti-angiogenic potential of BZD9L1 on endothelial cells (EC) in vitro, ex vivo and in HCT116 CRC xenograft in vivo models.
METHODS EA.hy926 EC were treated with half inhibitory concentration (IC50) (2.5 μM), IC50 (5.0 μM), and double IC50 (10.0 μM) of BZD9L1 and assessed for cell proliferation, adhesion and SIRT 1 and 2 protein expression. Next, 2.5 μM and 5.0 μM of BZD9L1 were employed in downstream in vitro assays, including cell cycle, cell death and sprouting in EC. The effect of BZD9L1 on cell adhesion molecules and SIRT 1 and 2 were assessed via real-time quantitative polymerase chain reaction (qPCR). The growth factors secreted by EC post-treatment were evaluated using the Quantibody Human Angiogenesis Array. Indirect co-culture with HCT116 CRC cells was performed to investigate the impact of growth factors modulated by BZD9L1-treated EC on CRC. The effect of BZD9L1 on sprouting impediment and vessel regression was determined using mouse choroids. HCT116 cells were also injected subcutaneously into nude mice and analyzed for the outcome of BZD9L1 on tumor necrosis, Ki67 protein expression indicative of proliferation, cluster of differentiation 31 (CD31) and CD34 EC markers, and SIRT 1 and 2 genes via hematoxylin and eosin, immunohistochemistry and qPCR, respectively.
RESULTS BZD9L1 impeded EC proliferation, adhesion, and spheroid sprouting through the downregulation of intercellular adhesion molecule 1, vascular endothelial cadherin, integrin-alpha V, SIRT1 and SIRT2 genes. The compound also arrested the cells at G1 phase and induced apoptosis in the EC. In mouse choroids, BZD9L1 inhibited sprouting and regressed sprouting vessels compared to the negative control. Compared to the negative control, the compound also reduced the protein levels of angiogenin, basic fibroblast growth factor, platelet-derived growth factor and placental growth factor, which then inhibited HCT116 CRC spheroid invasion in co-culture. In addition, a significant reduction in CRC tumor growth was noted alongside the downregulation of human SIRT1 (hSIRT1), hSIRT2, CD31, and CD34 EC markers and murine SIRT2 gene, while the murine SIRT1 gene remained unaffected, compared to vehicle control. Histology analyses revealed that BZD9L1 at low (50 mg/kg) and high (250 mg/kg) doses reduced Ki-67 protein expression, while BZD9L1 at the high dose diminished tumor necrosis compared to vehicle control.
CONCLUSION These results highlighted the anti-angiogenic potential of BZD9L1 to reduce CRC tumor progression. Furthermore, together with previous anticancer findings, this study provides valuable insights into the potential of BZD9L1 to co-target CRC tumor vasculatures and cancer cells via SIRT1 and/or SIRT2 down-regulation to improve the therapeutic outcome.
Collapse
Affiliation(s)
- Chern Ein Oon
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Ayappa V Subramaniam
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Lik Yang Ooi
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Ashwaq Hamid Salem Yehya
- Cancer Research, Eman Biodiscoveries, Kedah 08000, Malaysia
- Vatche and Tamar Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Yeuan Ting Lee
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Gurjeet Kaur
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Sreenivasan Sasidharan
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Beiying Qiu
- Academic Clinical Program, Duke-NUS Medical School, National University of Singapore, Singapore 169857, Singapore
- Singapore National Eye Centre, Singapore Eye Research Institute, Singapore 168751, Singapore
| | - Xiaomeng Wang
- Academic Clinical Program, Duke-NUS Medical School, National University of Singapore, Singapore 169857, Singapore
- Singapore National Eye Centre, Singapore Eye Research Institute, Singapore 169857, Singapore
| |
Collapse
|
9
|
Lee YT, Tan YJ, Oon CE. Benzimidazole and its derivatives as cancer therapeutics: The potential role from traditional to precision medicine. Acta Pharm Sin B 2023; 13:478-497. [PMID: 36873180 PMCID: PMC9978992 DOI: 10.1016/j.apsb.2022.09.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/11/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
Cancer is the second leading cause of mortality globally which remains a continuing threat to human health today. Drug insensitivity and resistance are critical hurdles in cancer treatment; therefore, the development of new entities targeting malignant cells is considered a high priority. Targeted therapy is the cornerstone of precision medicine. The synthesis of benzimidazole has garnered the attention of medicinal chemists and biologists due to its remarkable medicinal and pharmacological properties. Benzimidazole has a heterocyclic pharmacophore, which is an essential scaffold in drug and pharmaceutical development. Multiple studies have demonstrated the bioactivities of benzimidazole and its derivatives as potential anticancer therapeutics, either through targeting specific molecules or non-gene-specific strategies. This review provides an update on the mechanism of actions of various benzimidazole derivatives and the structure‒activity relationship from conventional anticancer to precision healthcare and from bench to clinics.
Collapse
Affiliation(s)
- Yeuan Ting Lee
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia
| | - Yi Jer Tan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia
| |
Collapse
|
10
|
Zhang W, Zhang X, Zhao D, Hu M, Ge X, Xia L. An Individualized EMT-Related Gene Signature to Predict Recurrence-Free Survival in Stage II/III Colorectal Cancer Patients. Dig Dis Sci 2022; 67:5116-5126. [PMID: 35094253 DOI: 10.1007/s10620-021-07338-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 11/15/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND Approximately 30% of stage II and 50-60% of stage III colorectal cancer (CRC) patients who have undergone surgery will develop recurrence within 5 years. Thus, more reliable prognostic biomarkers are urgently needed to identify the high-risk subset of patients who will benefit from postoperative adjuvant therapy. METHODS We retrospectively analyzed 911 stage II/III CRC patients in multiple cohorts. Using a series of bioinformatic and statistical approaches, an individualized prognostic signature was established in the training cohort and validated in four other independent cohorts. An integrated decision tree was generated to improve risk stratification, and a nomogram was built to quantify risk assessment for individual patients. RESULTS Epithelial-mesenchymal transition (EMT) was identified as a dominant risk factor for recurrence-free survival (RFS) in stage II/III CRC patients. The EMT-related gene signature could discriminate high-risk subsets in a training cohort and four independent validation cohorts (with 473, 89, 130, 74 and 145 patients, respectively). Survival analyses demonstrated that the EMT-related gene signature served as an independent risk factor for RFS in different subgroups. The decision tree could optimize the risk stratification, and the nomogram could predict the 5-year RFS probability accurately. CONCLUSION The proposed EMT-related prognostic signature is a useful biomarker to predict RFS and identify the high-risk subset in stage II/III CRC patients.
Collapse
Affiliation(s)
- Wei Zhang
- Division of Gastrointestinal Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Xiaoyu Zhang
- Division of Gastrointestinal Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Di Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mengwen Hu
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Xiaoqin Ge
- Department of Endocrinology, The Second Affiliated Hospital of Nantong University and First People's Hospital of Nantong City, Jiangsu, China
| | - Li Xia
- Department of Surgery, The Fourth People's Hospital of Huai'an, Huai'an, China.
| |
Collapse
|
11
|
Lin MH, Islam A, Liu YH, Weng CW, Zhan JH, Liang RH, Tikhomirov AS, Shchekotikhin AE, Chueh PJ. Antibiotic heliomycin and its water-soluble 4-aminomethylated derivative provoke cell death in T24 bladder cancer cells by targeting sirtuin 1 (SIRT1). Am J Cancer Res 2022; 12:1042-1055. [PMID: 35411221 PMCID: PMC8984893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 02/19/2022] [Indexed: 06/14/2023] Open
Abstract
Bladder cancer is one of the most frequent cancers among males, and a poor survival rate reflects problems with aggressiveness and chemo-resistance. Accumulating evidence indicates that SIRT1 is involved in bladder cancer tumorigenesis and is positively associated with chemo-resistance and poor prognosis. We recently synthesized water-soluble chemical derivatives of heliomycin, an antibiotic from Streptomyces resistomycificus, and demonstrated that they possess anticancer properties. In this present study, we used the cellular thermal shift assay (CETSA) in T24 bladder cancer cells to show that heliomycin (designated compound (H1)) and its 4-(tert-butylamino)methyl derivative (HD2) directly engaged with SIRT1 in the native cellular environment, whereas another derivative (HD3) did not. Upon binding, heliomycin downregulated SIRT1 protein expression without altering its transcript level, and subsequently induced autophagy. Interestingly, the derivative (HD2) triggered apoptosis. The interaction between SIRT1 protein and heliomycin or its derivatives was also speculated by a molecular docking simulation, suggesting heliomycin (H1) and derivative (HD2) acting with the different binding modes to SIRT1. Given the increased water-solubility, hydrogen bonds were found on Ala262 and Ile347 residues in the docked complex of derivative (HD2) to produce more steady interaction and initiate signaling pathways that were not observed in the case of heliomycin. Meanwhile, it is evident that derivative (HD3) did not engage with SIRT1 by CETSA or molecular docking studies, nor did it downregulate SIRT1 expression. Taken together, these findings clearly show that SIRT1 is targeted and downregulated by heliomycin and its water-soluble 4-aminomethylated derivative (HD2) possibly through autophagic and/or proteasomal degradation, leading to cell death and growth suppression of T24 bladder cancer cells.
Collapse
Affiliation(s)
- Ming Hung Lin
- Division of Urology, Department of Surgery, An Nan Hospital, China Medical UniversityTainan 70965, Taiwan
- Division of Urology, Department of Surgery, Tri-service General HospitalTaipei 11490, Taiwan
| | - Atikul Islam
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung 40227, Taiwan
| | - Yen-Hui Liu
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung 40227, Taiwan
| | - Chia-Wei Weng
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung 40227, Taiwan
- Institute of Medicine, Chung Shan Medical UniversityTaichung 40201, Taiwan
| | - Jun-Han Zhan
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung 40227, Taiwan
| | - Ru-Hao Liang
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung 40227, Taiwan
| | | | | | - Pin Ju Chueh
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung 40227, Taiwan
- Department of Medical Research, China Medical University HospitalTaichung 40402, Taiwan
- Graduate Institute of Basic Medicine, China Medical UniversityTaichung 40402, Taiwan
| |
Collapse
|
12
|
Fakhri S, Zachariah Moradi S, DeLiberto LK, Bishayee A. Cellular senescence signaling in cancer: A novel therapeutic target to combat human malignancies. Biochem Pharmacol 2022; 199:114989. [DOI: 10.1016/j.bcp.2022.114989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 12/26/2022]
|
13
|
Chen WK, Oon CE, Kaur G, Sainson RC, Li JL. Downregulation of Manic fringe impedes angiogenesis and cell migration of renal carcinoma. Microvasc Res 2022; 142:104341. [DOI: 10.1016/j.mvr.2022.104341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/14/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022]
|
14
|
Correlation Analysis of Protein Expression of 10 HDAC/Sirtuin Isoenzymes with Sensitivities of 23 Anticancer Drugs in 17 Cancer Cell Lines and Potentiation of Drug Activity by Co-Treatment with HDAC Inhibitors. Cancers (Basel) 2021; 14:cancers14010187. [PMID: 35008351 PMCID: PMC8750037 DOI: 10.3390/cancers14010187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Protein expression profiles of 10 HDAC/Sirtuin isoenzymes in two panels of human cancer cell lines were compared with each other and with the potencies of various anticancer drugs by Pearson and Spearman correlation analysis to identify patterns of enzyme expression and anticancer activity. Furthermore, the NCI COMPARE database was used to identify possible correlations between the mRNA expression in a 60 cancer cell panel and the potency of the same anticancer drugs. While several interesting correlations were found within both data sets, none of these correlations were identical in the two sets of data, suggesting that protein and mRNA expression profiles are not comparable. Combination treatments with several HDAC inhibitors with a number of the anticancer drugs revealed interesting synergistic effects that were in keeping with some of the correlations predicted by our protein expression analysis. Abstract Inhibiting the activity of histone deacetylase (HDAC) is an ongoing strategy in anticancer therapy. However, to our knowledge, the relationships between the expression of HDAC proteins and the antitumor drug sensitivity of cancer cells have not been studied until now. In the current work, we investigated the relative expression profiles of 10 HDAC isoenzymes comprising the classes I–III (HDAC1/2/4/6; Sirt1/2/3/5/6/7) in a panel of 17 cancer cell lines, including the breast, cervix, oesophageal, lung, oral squamous, pancreas, as well as urinary bladder carcinoma cells. Correlations between the data of mRNA expression for these enzymes obtained from the National Cancer Institute (NCI) 60 cancer cell line program were also examined. Next, we performed univariate analysis between the expression patterns of HDAC/Sirt isoenzymes with the sensitivity of a 16 cell panel of cancer cell lines towards several antitumor drugs. In a univariate correlation analysis, we found a strong relation between Sirt2 expression and cytotoxicity caused by busulfan, etoposide, and hydroxyurea. Moreover, it was identified that Sirt5 correlates with the effects exerted by oxaliplatin or topotecan, as well as between HDAC4 expression and these two drugs. Correlations between the data of mRNA expression for enzymes with the potencies of the same anticancer agents obtained from the NCI 60 cancer cell line program were also found, but none were the same as those we found with our protein expression data. Additionally, we report here the effects upon combination of the approved HDAC inhibitor vorinostat and one other known inhibitor trichostatin A as well as newer hetero-stilbene and diazeno based sirtuin inhibitors on the potency of cisplatin, lomustine, and topotecan. For these three anticancer drugs, we found a significantly enhanced cytotoxicity when co-incubated with HDAC inhibitors, demonstrating a potentially beneficial influence of HDAC inhibition on anticancer drug treatment.
Collapse
|
15
|
Hong JY, Lin H. Sirtuin Modulators in Cellular and Animal Models of Human Diseases. Front Pharmacol 2021; 12:735044. [PMID: 34650436 PMCID: PMC8505532 DOI: 10.3389/fphar.2021.735044] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/24/2021] [Indexed: 12/22/2022] Open
Abstract
Sirtuins use NAD+ to remove various acyl groups from protein lysine residues. Through working on different substrate proteins, they display many biological functions, including regulation of cell proliferation, genome stability, metabolism, and cell migration. There are seven sirtuins in humans, SIRT1-7, each with unique enzymatic activities, regulatory mechanisms, subcellular localizations, and substrate scopes. They have been indicated in many human diseases, including cancer, neurodegeneration, microbial infection, metabolic and autoimmune diseases. Consequently, interests in development of sirtuin modulators have increased in the past decade. In this brief review, we specifically summarize genetic and pharmacological modulations of sirtuins in cancer, neurological, and cardiovascular diseases. We further anticipate this review will be helpful for scrutinizing the significance of sirtuins in the studied diseases.
Collapse
Affiliation(s)
- Jun Young Hong
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, United States
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, United States.,Department of Chemistry and Chemical Biology, Howard Hughes Medical Institute, Cornell University, Ithaca, NY, United States
| |
Collapse
|
16
|
Tan YJ, Lee YT, Mancera RL, Oon CE. BZD9L1 sirtuin inhibitor: Identification of key molecular targets and their biological functions in HCT 116 colorectal cancer cells. Life Sci 2021; 284:119747. [PMID: 34171380 DOI: 10.1016/j.lfs.2021.119747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 05/22/2021] [Accepted: 06/11/2021] [Indexed: 02/07/2023]
Abstract
BZD9L1 was previously described as a SIRT1/2 inhibitor with anti-cancer activities in colorectal cancer (CRC), either as a standalone chemotherapy or in combination with 5-fluorouracil. BZD9L1 was reported to induce apoptosis in CRC cells; however, the network of intracellular pathways and crosstalk between molecular players mediated by BZD9L1 is not fully understood. This study aimed to uncover the mechanisms involved in BZD9L1-mediated cytotoxicity based on previous and new findings for the prediction and identification of related pathways and key molecular players. BZD9L1-regulated candidate targets (RCTs) were identified using a range of molecular, cell-based and biochemical techniques on the HCT 116 cell line. BZD9L1 regulated major cancer pathways including Notch, p53, cell cycle, NFκB, Myc/MAX, and MAPK/ERK signalling pathways. BZD9L1 also induced reactive oxygen species (ROS), regulated apoptosis-related proteins, and altered cell polarity and adhesion profiles. In silico analyses revealed that most RCTs were interconnected, and were involved in the modulation of catalytic activity, metabolism and transcription regulation, response to cytokines, and apoptosis signalling pathways. These RCTs were implicated in p53-dependent apoptosis pathway. This study provides the first assessment of possible associations of molecular players underlying the cytotoxic activity of BZD9L1, and establishes the links between RCTs and apoptosis through the p53 pathway.
Collapse
Affiliation(s)
- Yi Jer Tan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia; Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI) and Curtin Institute for Computation, Curtin University, GPO Box U1987, Perth, WA 6845, Australia
| | - Yeuan Ting Lee
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Ricardo L Mancera
- Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI) and Curtin Institute for Computation, Curtin University, GPO Box U1987, Perth, WA 6845, Australia.
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia.
| |
Collapse
|
17
|
Cristóbal I, Rubio J, Santos A, Torrejón B, Caramés C, Imedio L, Mariblanca S, Luque M, Sanz-Alvarez M, Zazo S, Madoz-Gúrpide J, Rojo F, García-Foncillas J. MicroRNA-199b Downregulation Confers Resistance to 5-Fluorouracil Treatment and Predicts Poor Outcome and Response to Neoadjuvant Chemoradiotherapy in Locally Advanced Rectal Cancer Patients. Cancers (Basel) 2020; 12:cancers12061655. [PMID: 32580513 PMCID: PMC7352382 DOI: 10.3390/cancers12061655] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/18/2020] [Accepted: 06/18/2020] [Indexed: 12/17/2022] Open
Abstract
Neoadjuvant 5-fluorouracil (5-FU)-based chemoradiotherapy followed by mesorectal excision is the current standard treatment in locally advanced rectal cancer (LARC) and the lack of complete response represents a major problem that compromises long-term patient survival. However, there is a lack of robust established markers predictive of response to this preoperative treatment available in the clinical routine. The tumor suppressor microRNA (miR)-199b directly targets the PP2A inhibitor SET, which has been involved in 5-FU resistance, and its downregulation has been found to correlate with poor outcome in metastatic colorectal cancer. Here, we studied the functional effects of miR-199b on 5-FU sensitivity after its ectopic modulation, and its expression was quantified by real-time-PCR in a cohort of 110 LARC patients to evaluate its potential clinical significance. Interestingly, our findings demonstrate that miR-199b enhances the sensitivity of colorectal cancer cells to 5-FU in a SET-dependent manner, and that both miR-199b overexpression and SET inhibition are able to overcome resistance to this drug using an acquired 5-FU-resistant model. MiR-199b was found downregulated in 26.4% of cases and was associated with positive lymph node levels after chemoradiotherapy (CRT, p = 0.007) and high pathological stage (p = 0.029). Moreover, miR-199b downregulation determined shorter overall (p = 0.003) and event-free survival (p = 0.005), and was an independent predictor of poor response to preoperative CRT (p = 0.004). In conclusion, our findings highlight the clinical impact of miR-199b downregulation predicting poor outcome and pathological response in LARC, and suggest the miR-199b/SET signaling axis as a novel molecular target to prevent the development of resistance to 5-FU treatment.
Collapse
Affiliation(s)
- Ion Cristóbal
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS- Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain; (J.R.); (A.S.); (B.T.); (C.C.); (L.I.); (S.M.)
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain
- Correspondence: (I.C.); (J.G.-F.); Tel.: +34-915504800 (I.C. & J.G.-F.)
| | - Jaime Rubio
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS- Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain; (J.R.); (A.S.); (B.T.); (C.C.); (L.I.); (S.M.)
- Medical Oncology Department, University Hospital “Fundación Jiménez Díaz”, UAM, E-28040 Madrid, Spain
| | - Andrea Santos
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS- Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain; (J.R.); (A.S.); (B.T.); (C.C.); (L.I.); (S.M.)
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain
| | - Blanca Torrejón
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS- Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain; (J.R.); (A.S.); (B.T.); (C.C.); (L.I.); (S.M.)
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain
| | - Cristina Caramés
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS- Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain; (J.R.); (A.S.); (B.T.); (C.C.); (L.I.); (S.M.)
- Medical Oncology Department, University Hospital “Fundación Jiménez Díaz”, UAM, E-28040 Madrid, Spain
| | - Laura Imedio
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS- Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain; (J.R.); (A.S.); (B.T.); (C.C.); (L.I.); (S.M.)
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain
| | - Sofía Mariblanca
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS- Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain; (J.R.); (A.S.); (B.T.); (C.C.); (L.I.); (S.M.)
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain
| | - Melani Luque
- Pathology Department, IIS-Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain; (M.L.); (M.S.-A.); (S.Z.); (J.M.-G.); (F.R.)
| | - Marta Sanz-Alvarez
- Pathology Department, IIS-Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain; (M.L.); (M.S.-A.); (S.Z.); (J.M.-G.); (F.R.)
| | - Sandra Zazo
- Pathology Department, IIS-Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain; (M.L.); (M.S.-A.); (S.Z.); (J.M.-G.); (F.R.)
| | - Juan Madoz-Gúrpide
- Pathology Department, IIS-Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain; (M.L.); (M.S.-A.); (S.Z.); (J.M.-G.); (F.R.)
| | - Federico Rojo
- Pathology Department, IIS-Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain; (M.L.); (M.S.-A.); (S.Z.); (J.M.-G.); (F.R.)
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, E-28040 Madrid, Spain
- Medical Oncology Department, University Hospital “Fundación Jiménez Díaz”, UAM, E-28040 Madrid, Spain
- Correspondence: (I.C.); (J.G.-F.); Tel.: +34-915504800 (I.C. & J.G.-F.)
| |
Collapse
|