1
|
García-Castro M, Fuentes-Rios D, López-Romero JM, Romero A, Moya-Utrera F, Díaz-Morilla A, Sarabia F. n-Tuples on Scaffold Diversity Inspired by Drug Hybridisation to Enhance Drugability: Application to Cytarabine. Mar Drugs 2023; 21:637. [PMID: 38132958 PMCID: PMC10744741 DOI: 10.3390/md21120637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
A mathematical concept, n-tuples are originally applied to medicinal chemistry, especially with the creation of scaffold diversity inspired by the hybridisation of different commercial drugs with cytarabine, a synthetic arabinonucleoside derived from two marine natural products, spongouridine and spongothymidine. The new methodology explores the virtual chemical-factorial combination of different commercial drugs (immunosuppressant, antibiotic, antiemetic, anti-inflammatory, and anticancer) with the anticancer drug cytarabine. Real chemical combinations were designed and synthesised for 8-duples, obtaining a small representative library of interesting organic molecules to be biologically tested as proof of concept. The synthesised library contains classical molecular properties regarding the Lipinski rules and/or beyond rules of five (bRo5) and is represented by the covalent combination of the anticancer drug cytarabine with ibuprofen, flurbiprofen, folic acid, sulfasalazine, ciprofloxacin, bortezomib, and methotrexate. The insertion of specific nomenclature could be implemented into artificial intelligence algorithms in order to enhance the efficiency of drug-hunting programs. The novel methodology has proven useful for the straightforward synthesis of most of the theoretically proposed duples and, in principle, could be extended to any other central drug.
Collapse
Affiliation(s)
- Miguel García-Castro
- Department of Organic Chemistry, Faculty of Sciences, University of Malaga, Campus de Teatinos s/n, 29071 Malaga, Spain
| | | | | | | | | | | | | |
Collapse
|
2
|
Poonacha KNT, Villa TG, Notario V. The Interplay among Radiation Therapy, Antibiotics and the Microbiota: Impact on Cancer Treatment Outcomes. Antibiotics (Basel) 2022; 11:331. [PMID: 35326794 PMCID: PMC8944497 DOI: 10.3390/antibiotics11030331] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 12/01/2022] Open
Abstract
Radiation therapy has been used for more than a century, either alone or in combination with other therapeutic modalities, to treat most types of cancer. On average, radiation therapy is included in the treatment plans for over 50% of all cancer patients, and it is estimated to contribute to about 40% of curative protocols, a success rate that may reach 90%, or higher, for certain tumor types, particularly on patients diagnosed at early disease stages. A growing body of research provides solid support for the existence of bidirectional interaction between radiation exposure and the human microbiota. Radiation treatment causes quantitative and qualitative changes in the gut microbiota composition, often leading to an increased abundance of potentially hazardous or pathogenic microbes and a concomitant decrease in commensal bacteria. In turn, the resulting dysbiotic microbiota becomes an important contributor to worsen the adverse events caused in patients by the inflammatory process triggered by the radiation treatment and a significant determinant of the radiation therapy anti-tumor effectiveness. Antibiotics, which are frequently included as prophylactic agents in cancer treatment protocols to prevent patient infections, may affect the radiation/microbiota interaction through mechanisms involving both their antimicrobial activity, as a mediator of microbiota imbalances, and their dual capacity to act as pro- or anti-tumorigenic effectors and, consequently, as critical determinants of radiation therapy outcomes. In this scenario, it becomes important to introduce the use of probiotics and/or other agents that may stabilize the healthy microbiota before patients are exposed to radiation. Ultimately, newly developed methodologies may facilitate performing personalized microbiota screenings on patients before radiation therapy as an accurate way to identify which antibiotics may be used, if needed, and to inform the overall treatment planning. This review examines currently available data on these issues from the perspective of improving radiation therapy outcomes.
Collapse
Affiliation(s)
| | - Tomás G. Villa
- Department of Microbiology, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15705 La Coruña, Spain;
| | - Vicente Notario
- Department of Radiation Medicine, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
3
|
Ji Y, Yin Y, Zhang W. Integrated Bioinformatic Analysis Identifies Networks and Promising Biomarkers for Hepatitis B Virus-Related Hepatocellular Carcinoma. Int J Genomics 2020; 2020:2061024. [PMID: 32775402 PMCID: PMC7407030 DOI: 10.1155/2020/2061024] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/09/2020] [Accepted: 06/27/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic infection with hepatitis B virus (HBV) has long been recognized as a dominant hazard factor for hepatocellular carcinoma (HCC) and accounts for at least half of HCC instances globally. However, the underlying molecular mechanism of HBV-linked HCC has not been completely elucidated. Here, three microarray datasets, totally containing 170 tumoral samples and 181 adjacent normal tissues from the liver of patients suffering from HBV-related HCC assembled from the Gene Expression Omnibus (GEO) database, were subjected to integrated analysis of differentially expressed genes (DEGs). Subsequently, the analysis of function and pathway enrichment as well as the protein-protein interaction network (PPI) was performed. The ten hub genes screened out from the PPI network were further subjected to expression profile and survival analysis. Overall, 329 DEGs (67 upregulated and 262 downregulated) were identified. Ten DEGs with the highest degree of connectivity included cyclin-dependent kinase 1 (CDK1), cyclin B1 (CCNB1), cyclin B2 (CCNB2), PDZ-binding kinase (PBK), abnormal spindle microtubule assembly (ASPM), nuclear division cycle 80 (NDC80), aurora kinase A (AURKA), targeting protein for xenopus kinesin-like protein 2 (TPX2), kinesin family member 2C (KIF2C), and centromere protein F (CENPF). Kaplan-Meier analysis unveiled that overexpression levels of KIF2C and TPX2 were relevant to both the poor overall survival and relapse-free survival. In summary, the hub genes validated in the present study may provide promising targets for the diagnosis, prognosis, and therapy of HBV-associated HCC. Additionally, our work uncovers various crucial biological components (e.g., extracellular exosome) and signaling pathways that participate in the progression of HCC induced by HBV, serving comprehensive knowledge of the mechanisms regarding HBV-related HCC.
Collapse
Affiliation(s)
- Yun Ji
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
4
|
Jonnalagadda SK, Wielenberg K, Ronayne CT, Jonnalagadda S, Kiprof P, Jonnalagadda SC, Mereddy VR. Synthesis and biological evaluation of arylphosphonium-benzoxaborole conjugates as novel anticancer agents. Bioorg Med Chem Lett 2020; 30:127259. [DOI: 10.1016/j.bmcl.2020.127259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/07/2020] [Accepted: 05/09/2020] [Indexed: 01/09/2023]
|
5
|
Kaur H, Singh J, Narasimhan B. Synthesis and evaluation of novel naphthol diazenyl scaffold based Schiff bases as potential antimicrobial and cytotoxic agents against human colorectal carcinoma cell line (HT-29). BMC Chem 2019; 13:49. [PMID: 31384797 PMCID: PMC6661811 DOI: 10.1186/s13065-019-0558-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 03/15/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND In search of new antimicrobial and cytotoxic agents, a series of new naphthol diazenyl scaffold based Schiff bases (NS1-NS23) was efficiently synthesized by condensation of 2-hydroxy naphthaldehyde azo dyes with various substituted aromatic/heteroaromatic/aliphatic amines. METHODOLOGY The synthesized derivatives were characterized by various physicochemical and spectral techniques and assessed for in vitro antimicrobial and cytotoxic potential against human colorectal carcinoma cell line (HT-29). The active derivatives were further evaluated for their apoptotic potential by Annexin-V/propidium iodide double staining assay using flow cytometer and analyzed for cell-cycle arrest studies. RESULTS AND CONCLUSION The derivative NS-2 was found maximum active against E. coli, S. enterica and B. subtilis. The derivatives NS-12, NS-15, NS-21, and NS-23 showed maximum antifungal activity against A. fumigatus. The maximum cytotoxicity was observed from the derivatives NS-2, NS-8, NS-21, and NS-23 towards HT-29 cell line with IC50 between 4 and 19 μg/ml. More than 90% and 62% of the cells were found in the apoptotic phase on treatment with NS-2 and NS-21 respectively in comparison to the 68% for doxorubicin. Further, these derivatives arrested the cell growth in S and G2/M phase of the cell cycle.
Collapse
Affiliation(s)
- Harmeet Kaur
- Faculty of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001 India
| | - Jasbir Singh
- College of Pharmacy, Postgraduate Institute of Medical Sciences, Rohtak, 124001 India
| | | |
Collapse
|
6
|
Kaur H, Singh J, Narasimhan B. Antimicrobial, antioxidant and cytotoxic evaluation of diazenyl chalcones along with insights to mechanism of interaction by molecular docking studies. BMC Chem 2019; 13:87. [PMID: 31384834 PMCID: PMC6661766 DOI: 10.1186/s13065-019-0596-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 06/25/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In continuation of our work, new diazenyl chalcones scaffolds (C-18 to C-27) were efficiently synthesized from substituted acetophenone azo dyes (A-E) by base catalyzed Claisen-Schmidt condensation with different substituted aromatic/heteroaromatic aldehydes. METHODOLOGY The synthesized chalcones were assessed for their in vitro antimicrobial potential towards several pathogenic microbial strains by tube dilution method and further evaluated for antioxidant potential by DPPH assay. These derivatives were also assessed for the cytotoxicity towards the human lung cancer cell line (A549) and normal cell line (HEK) by MTT assay. The most active antimicrobial compounds were docked using Schrodinger v18.1 software with the various potential bacterial receptors to explore the mechanism of interaction. RESULTS The derivative C-22 exhibited high antibacterial activity with very low MIC (1.95-3.90 µg ml-1) and MBC (3.90-7.81 µg ml-1) values. The derivatives C-23, C-24 and C-27 have demonstrated good antioxidant potential (IC50 = 7-18 µg ml-1) correlated to the ascorbic acid (IC50 = 4.45 µg ml-1). The derivative C-25 had shown comparable cytotoxicity to camptothecin against A549 cell line. The docking studies predicted the bacterial dihydrofolate reductase (PDB ID: 3SRW) and bacterial DNA gyrase (PDB ID: 4ZVI) as the possible targets for most of the active antimicrobial compounds. These derivatives affirmed their safety by presenting less cytotoxicity towards HEK cells. Further the ADME prediction by qikprop module of the Schrodinger proved that these compounds exhibited drug-like attributes. CONCLUSION Hence, these compounds have shown their potential as lead for future expansion of novel antimicrobial and cytotoxic drugs.
Collapse
Affiliation(s)
- Harmeet Kaur
- Faculty of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001 India
| | - Jasbir Singh
- College of Pharmacy, Postgraduate Institute of Medical Sciences, Rohtak, 124001 India
| | | |
Collapse
|
7
|
Natarajan A, Thangarajan R, Kesavan S. Repurposing Drugs by In Silico Methods to Target BCR Kinase Domain in Chronic Myeloid Leukemia. Asian Pac J Cancer Prev 2019; 20:3399-3406. [PMID: 31759365 PMCID: PMC7063026 DOI: 10.31557/apjcp.2019.20.11.3399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Targeted therapy in the form of highly selective tyrosine kinase inhibitors (TKIs) has transformed the treatment of chronic myeloid leukemia (CML). However, mutations in the kinase domain contribute to drug resistance against TKIs which compromises the treatment response. Our aim is to explore regions outside the BCR-ABL oncoprotein to identify potential therapeutic targets to curb drug resistance by targeting growth factor receptor-bound protein-2 (Grb-2) which binds to BCR-ABL at the phosphorylated tyrosine (Y177) thereby activating the Ras and PI3K/AKT signaling pathway. METHODS We have used in silico methods to repurpose drugs for identifying their potential to inhibit the binding of Grb-2 with Y177 by occupying the active binding site of the BCR domain. RESULTS Differentially expressed genes from GEO dataset were found to be associated with hematopoietic cell lineage, NK cell-mediated cytotoxicity, NF-κB and chemokine signaling, cytokine-cytokine receptor interaction, histidine metabolism and transcriptional misregulation in cancer. The fold recognition method of SPARKS-X tool was used to model the BCR domain (Z-score = 8.21). Connectivity Map generated a drug list based on the gene expression profile, which were docked with BCR. Schrodinger XP glide docking identified Diphosphopyridine nucleotide, Hesperidin, Butirosin, Ovoflavin, and Nor-dihydroguaiaretic acid to show strong interaction in close proximity to the active binding pocket containing Y177 of the target protein and was further validated using iGEMDOCK and Parallelized Open Babel and AutoDock suite Pipeline (POAP). CONCLUSION Our study not only extends our current knowledge about repurposing drugs for newer indications but also provides a route towards combinatorial therapy with standard drugs used for CML treatment. However, the efficacy of these repurposed drugs needs to be further investigated using in vitro and in vivo studies.<br />.
Collapse
Affiliation(s)
- Aparna Natarajan
- Department of Molecular Oncology, Cancer Institute (WIA), Adyar, Chennai, India
| | | | - Sabitha Kesavan
- Department of Molecular Oncology, Cancer Institute (WIA), Adyar, Chennai, India
| |
Collapse
|
8
|
Meng C, Bai C, Brown TD, Hood LE, Tian Q. Human Gut Microbiota and Gastrointestinal Cancer. GENOMICS PROTEOMICS & BIOINFORMATICS 2018. [PMID: 29474889 DOI: 10.1016/j.gpb.2017.06.002.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Human gut microbiota play an essential role in both healthy and diseased states of humans. In the past decade, the interactions between microorganisms and tumors have attracted much attention in the efforts to understand various features of the complex microbial communities, as well as the possible mechanisms through which the microbiota are involved in cancer prevention, carcinogenesis, and anti-cancer therapy. A large number of studies have indicated that microbial dysbiosis contributes to cancer susceptibility via multiple pathways. Further studies have suggested that the microbiota and their associated metabolites are not only closely related to carcinogenesis by inducing inflammation and immune dysregulation, which lead to genetic instability, but also interfere with the pharmacodynamics of anticancer agents. In this article, we mainly reviewed the influence of gut microbiota on cancers in the gastrointestinal (GI) tract (including esophageal, gastric, colorectal, liver, and pancreatic cancers) and the regulation of microbiota by diet, prebiotics, probiotics, synbiotics, antibiotics, or the Traditional Chinese Medicine. We also proposed some new strategies in the prevention and treatment of GI cancers that could be explored in the future. We hope that this review could provide a comprehensive overview of the studies on the interactions between the gut microbiota and GI cancers, which are likely to yield translational opportunities to reduce cancer morbidity and mortality by improving prevention, diagnosis, and treatment.
Collapse
Affiliation(s)
- Changting Meng
- Institute for Systems Biology, Seattle, WA 98109, USA; Department of Oncology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Chunmei Bai
- Department of Oncology, Peking Union Medical College Hospital, Beijing 100730, China
| | | | - Leroy E Hood
- Institute for Systems Biology, Seattle, WA 98109, USA; Swedish Cancer Institute, Seattle, WA 98104, USA
| | - Qiang Tian
- Institute for Systems Biology, Seattle, WA 98109, USA; P4 Medicine Institute, Seattle, WA 98109, USA.
| |
Collapse
|
9
|
Meng C, Bai C, Brown TD, Hood LE, Tian Q. Human Gut Microbiota and Gastrointestinal Cancer. GENOMICS, PROTEOMICS & BIOINFORMATICS 2018; 16:33-49. [PMID: 29474889 PMCID: PMC6000254 DOI: 10.1016/j.gpb.2017.06.002] [Citation(s) in RCA: 241] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 06/08/2017] [Accepted: 07/04/2017] [Indexed: 02/06/2023]
Abstract
Human gut microbiota play an essential role in both healthy and diseased states of humans. In the past decade, the interactions between microorganisms and tumors have attracted much attention in the efforts to understand various features of the complex microbial communities, as well as the possible mechanisms through which the microbiota are involved in cancer prevention, carcinogenesis, and anti-cancer therapy. A large number of studies have indicated that microbial dysbiosis contributes to cancer susceptibility via multiple pathways. Further studies have suggested that the microbiota and their associated metabolites are not only closely related to carcinogenesis by inducing inflammation and immune dysregulation, which lead to genetic instability, but also interfere with the pharmacodynamics of anticancer agents. In this article, we mainly reviewed the influence of gut microbiota on cancers in the gastrointestinal (GI) tract (including esophageal, gastric, colorectal, liver, and pancreatic cancers) and the regulation of microbiota by diet, prebiotics, probiotics, synbiotics, antibiotics, or the Traditional Chinese Medicine. We also proposed some new strategies in the prevention and treatment of GI cancers that could be explored in the future. We hope that this review could provide a comprehensive overview of the studies on the interactions between the gut microbiota and GI cancers, which are likely to yield translational opportunities to reduce cancer morbidity and mortality by improving prevention, diagnosis, and treatment.
Collapse
Affiliation(s)
- Changting Meng
- Institute for Systems Biology, Seattle, WA 98109, USA; Department of Oncology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Chunmei Bai
- Department of Oncology, Peking Union Medical College Hospital, Beijing 100730, China
| | | | - Leroy E Hood
- Institute for Systems Biology, Seattle, WA 98109, USA; Swedish Cancer Institute, Seattle, WA 98104, USA
| | - Qiang Tian
- Institute for Systems Biology, Seattle, WA 98109, USA; P4 Medicine Institute, Seattle, WA 98109, USA.
| |
Collapse
|