Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Molecules ; 28(1)2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36615533

ABSTRACT

Ag(I) coordination compounds have recently attracted much attention as antiproliferative and antibacterial agents against a wide range of cancer cell lines and pathogens. The bioactivity potential of these complexes depends on their structural characteristics and the nature of their ligands. Herein, we present a series of four Ag(I) coordination compounds bearing as ligands the CH3-substituted thiadiazole-based thioamide 5-methyl-1,3,4-thiadiazole-2-thiol (mtdztH) and phosphines, i.e., [AgCl(mtdztH)(PPh3)2] (1), [Ag(mtdzt)(PPh3)3] (2), [AgCl(mtdztH)(xantphos)] (3), and [AgmtdztH)(dppe)(NO3)]n (4), where xantphos = 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene and dppe = 1,2-bis(diphenylphosphino)ethane, and the assessment of their in vitro antibacterial and anti-cancer efficiency. Among them, diphosphine-containing compounds 3 and 4 were found to exhibit broad-spectrum antibacterial activity characteristics against both Gram-(+) and Gram-(-) bacterial strains, showing high in vitro bioactivity with IC50 values as low as 4.6 µΜ. In vitro cytotoxicity studies against human ovarian, pancreatic, lung, and prostate cancer cell lines revealed the strong cytotoxic potential of 2 and 4, with IC50 values in the range of 3.1-24.0 µΜ, while 3 and 4 maintained the normal fibroblast cells' viability at relatively higher levels. Assessment of these results, in combination with those obtained for analogous Ag(I) complexes bearing similar heterocyclic thioamides, suggest the pivotal role of the substituent groups of the thioamide heterocyclic ring in the antibacterial and anti-cancer efficacy of the respective Ag(I) complexes. Compounds 1-4 exhibited moderate in vitro antioxidant capacity for free radicals scavenging, as well as reasonably strong ability to interact with calf-thymus DNA, suggesting the likely implication of these properties in their bioactivity mechanisms. Complementary insights into the possible mechanism of their anti-cancer activity were provided by molecular docking calculations, exploring their ability to bind to the overexpressed fibroblast growth factor receptor 1 (FGFR1), affecting cancer cells' functionalities.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Neoplasms , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Proliferation , Coordination Complexes/chemistry , Molecular Docking Simulation , Silver/chemistry , Thioamides/pharmacology
2.
Molecules ; 27(21)2022 Nov 04.
Article in English | MEDLINE | ID: mdl-36364398

ABSTRACT

Since there is an urgent need for novel treatments to combat the current coronavirus disease 2019 (COVID-19) pandemic, in silico molecular docking studies were implemented as an attempt to explore the ability of selected bioactive constituents of extra virgin olive oil (EVOO) to act as potent SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) antiviral compounds, aiming to explore their ability to interact with SARS-CoV-2 Spike key therapeutic target protein. Our results suggest that EVOO constituents display substantial capacity for binding and interfering with Spike (S) protein, both wild-type and mutant, via the receptor-binding domain (RBD) of Spike, or other binding targets such as angiotensin-converting enzyme 2 (ACE2) or the RBD-ACE2 protein complex, inhibiting the interaction of the virus with host cells. This in silico study provides useful insights for the understanding of the mechanism of action of the studied compounds at a molecular level. From the present study, it could be suggested that the studied active phytochemicals could potentially inhibit the Spike protein, contributing thus to the understanding of the role that they can play in future drug designing and the development of anti-COVID-19 therapeutics.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Humans , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Olive Oil , Molecular Docking Simulation , Peptidyl-Dipeptidase A/metabolism , Binding Sites , Protein Binding
3.
Dalton Trans ; 51(24): 9412-9431, 2022 Jun 21.
Article in English | MEDLINE | ID: mdl-35674362

ABSTRACT

In recent years, there has been an increasing interest in the study of Ag(I) coordination compounds as potent antibacterial and anticancer agents. Herein, a series of Ag(I) complexes bearing phosphines and heterocyclic thioamide ligands with highly electronegative NH2- and CF3-group substituents, i.e. [AgCl(atdztH)(xantphos)] (1), [Ag(µ-atdztH)(DPEphos)]2(NO3)2 (2), [Ag(atdzt)(PPh3)3] (3), [Ag(µ-atdzt)(DPEphos)]2 (4), and [Ag(µ-mtft)(DPEphos)]2 (5), where atdztH = 5-amino-1,3,4-thiadiazole-2-thiol, mtftH = 4-methyl-5-(trifluoromethyl)-1,2,4-triazol-3-thiol, xantphos = 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene, and DPEphos = bis(2-diphenylphosphino-phenyl)ether, were synthesized, and their in vitro antibacterial and anticancer properties were evaluated. Complexes 1-4 bearing the NH2-substituted thioamide exhibited moderate-to-high activity against S. aureus, B. subtilis, B. cereus and E. coli bacterial strains. A high antiproliferative activity was also observed for 1-3 against SKOV-3, Hup-T3, DMS114 and PC3 cancer cell lines (IC50 = 4.0-11.7 µM), as well as some degree of selectivity against MRC-5 normal cells. Interestingly, 5 bearing the CF3-substituted thioamide is completely inactive in all bioactivity studies. Binding of 1-3 to drug-carrier proteins BSA and HSA is reasonably strong for their uptake and subsequent release to possible target sites. The three complexes show a significant in vitro antioxidant ability for scavenging free radicals, suggesting likely implication of this property in the mechanism of their bioactivity, but a low potential to destroy the double-strand structure of CT-DNA by intercalation. Complementary insights into possible bioactivity mechanisms were provided by molecular docking calculations, exploring the ability of complexes to bind to bacterial DNA gyrase, and to the overexpressed in the aforementioned cancer cells Fibroblast Growth Factor Receptor 1, affecting their functionalities.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Escherichia coli , Ligands , Molecular Docking Simulation , Silver/chemistry , Silver/pharmacology , Staphylococcus aureus , Thioamides/pharmacology
4.
J Inorg Biochem ; 231: 111805, 2022 06.
Article in English | MEDLINE | ID: mdl-35334392

ABSTRACT

In silico molecular docking studies, in vitro toxicity and in silico predictions on the biological activity profile, pharmacokinetic properties, drug-likeness, ADMET (absorption, distribution, metabolism, excretion, and toxicity) physicochemical pharmacokinetic data, and target proteins and toxicity predictions were performed on six copper(II) complexes with the non-steroidal anti-inflammatory drugs ibuprofen, loxoprofen, fenoprofen and clonixin as ligands, in order to investigate the ability of these complexes to interact with the key therapeutic target proteins of SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) 3C-like cysteine main protease (3CLpro/Mpro), viral papain-like protease (PLpro), RNA-dependent RNA polymerase (RdRp), and non-structural proteins (Nsps) Nsp16-Nsp10 2'-O-methyltransferase complex, and their capacity to act as antiviral agents, contributing thus to understanding the role they can play in the context of coronavirus 2019 (COVID-19) pandemic. Cytotoxic activity against five human cancer and normal cell lines were also evaluated.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Anti-Inflammatory Agents , Antiviral Agents/chemistry , Copper , Humans , Molecular Docking Simulation , SARS-CoV-2
5.
J Inorg Biochem ; 228: 111695, 2022 03.
Article in English | MEDLINE | ID: mdl-35007963

ABSTRACT

A series of heteroleptic Ag(I) complexes bearing 4,6-dimethyl-2-pyrimidinethiol (dmp2SH), i.e., [AgCl(dmp2SH)(PPh3)2] (1), [Ag(dmp2SH)(PPh3)2]NO3 (2), [Ag(dmp2SΗ)(xantphos)]NO3 (3), [Ag(µ-dmp2S)(PPh3)]2 (4), [Ag(dmp2S)(xantphos)] (5), [Ag(µ-dmp2S)(DPEphos)]2 (6) (xantphos = 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene and DPEPhos = bis[(2-diphenylphosphino)phenyl]ether) were synthesized. The complexes display systematic variation of particular structural characteristics which were proved to have a significant impact on their in vitro cytotoxicity and antimicrobial properties. A moderate-to-high potential for bacteria growth inhibition was observed for all complexes, with 2, 3 and 5 being particularly effective against Gram-(+) bacteria (IC50 = 1.6-4.5 µM). The three complexes exhibit high in vitro cytotoxicity against HeLa and MCF-7 cancer cells (IC50 = 0.32-3.00 µΜ), suggesting the importance of coordination unsaturation and cationic charge for effective bioactivity. A very low cytotoxicity against HDFa normal cells was observed, revealing a high degree of selectivity (selectivity index ~10) and, hence, biocompatibility. Fluorescence microscopy using 2 showed effective targeting on the membrane of the HeLa cancer cells, subsequently inducing cell death. Binding of the complexes to serum albumin proteins is reasonably strong for potential uptake and subsequent release to target sites. A moderate in vitro antioxidant capacity for free radicals scavenging was observed and a low potential to destroy the double-strand structure of calf-thymus DNA by intercalation, suggesting likely implication of these properties in the bioactivity mechanisms of these complexes. Further insight into possible mechanisms of bioactivity was obtained by molecular modeling calculations, by exploring their ability to act as potential inhibitors of DNA-gyrase, human estrogen receptor alpha, human cyclin-dependent kinase 6, and human papillomavirus E6 oncoprotein.


Subject(s)
Anti-Infective Agents/pharmacology , Coordination Complexes/chemistry , Silver/chemistry , Thioamides/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/chemistry , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Bacteria/drug effects , Cyclin-Dependent Kinase 6/metabolism , DNA/metabolism , DNA Gyrase/metabolism , HeLa Cells , Humans , Ligands , MCF-7 Cells , Microbial Sensitivity Tests/methods , Models, Molecular , Molecular Docking Simulation/methods , Phosphines/chemistry , Silver/pharmacology , Thioamides/pharmacology , Xanthenes/chemistry
6.
J Inorg Biochem ; 226: 111659, 2022 01.
Article in English | MEDLINE | ID: mdl-34801971

ABSTRACT

The synthesis of five neutral zinc(II) complexes of 3,5-dibromo-salicyladehyde (3,5-diBr-saloH) in the presence of nitrogen-donor co-ligands 2,2'-bipyridine (bipy), 1,10-phenanthroline (phen), 2,9-dimethyl-1,10-phenanthroline (neoc), or 2,2'-bipyridylamine (bipyam) was undertaken and complexes [Zn(3,5-diBr-salo)2(H2O)2] (1), [Zn(3,5-diBr-salo)2(bipy)] (2), [Zn(3,5-diBr-salo)2(phen)].3,5-diBr-saloΗ (3), [Zn(3,5-diBr-salo)2(neoc)] (4) and [Zn(3,5-diBr-salo)2(bipyam)] (5) were characterized by various techniques. The crystal structures of complexes 3 and 5 were determined by X-ray crystallography, revealing the co-existence of two different coordination modes of 3,5-diBr-salo- ligands. The new complexes show selective in vitro antibacterial activity against two Gram-positive and two Gram-negative bacterial strains. The complexes may scavenge 1,1-diphenyl-picrylhydrazyl and 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) radicals and reduce H2O2. The complexes may intercalate in-between the calf-thymus DNA-bases and have exhibited low-to-moderate ability to cleave supercoiled circular pBR322 plasmid DNA. The complexes may bind tightly and reversibly to bovine and human serum albumins. In order to explain the in vitro activity of the compounds, molecular docking studies were adopted on the crystal structure of calf-thymus DNA, human and bovine serum albumin, Escherichia coli and Staphylococcus aureus DNA-gyrase, 5-lipoxygenase, and 5-lipoxygenase activating protein. The employed in silico studies aimed to explore the ability of the compounds to bind to these target biomacromolecules, establishing a possible mechanism of action and were in accordance with the in vitro studies.


Subject(s)
Aldehydes/chemistry , Coordination Complexes , Enzyme Inhibitors , Escherichia coli Proteins , Escherichia coli/enzymology , Staphylococcus aureus/enzymology , Zinc/chemistry , Animals , Cattle , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , Humans
7.
J Inorg Biochem ; 224: 111563, 2021 11.
Article in English | MEDLINE | ID: mdl-34399232

ABSTRACT

Six novel copper(II) complexes with the non-steroidal anti-inflammatory drugs ibuprofen, loxoprofen, fenoprofen and clonixin as ligands were synthesized and characterized by diverse techniques including single-crystal X-ray crystallography. The in vitro scavenging activity of the complexes against 1,1-diphenyl-picrylhydrazyl and 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) free radicals and the ability to reduce H2O2 were studied in the context of the antioxidant activity studies. The complexes may interact with calf-thymus DNA via intercalation as revealed by the techniques employed. The affinity of the complexes for bovine and human serum albumins was evaluated by fluorescence emission spectroscopy and the corresponding binding constants were determined. Molecular docking simulations on the crystal structure of calf-thymus DNA, human and bovine serum albumins were also employed in order to study in silico the ability of the studied compounds to bind to these target biomacromolecules, in terms of impairment of DNA and transportation through serum albumins, to explain the observed in vitro activity and to establish a possible mechanism of action.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Coordination Complexes/chemistry , Copper/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/chemistry , Clonixin/chemistry , Coordination Complexes/pharmacology , Copper/pharmacology , Crystallography, X-Ray/methods , DNA/chemistry , Fenoprofen/chemistry , Free Radical Scavengers/chemistry , Humans , Hydrogen Peroxide/chemistry , Ibuprofen/chemistry , Intercalating Agents/chemistry , Molecular Docking Simulation/methods , Phenylpropionates/chemistry , Serum Albumin, Bovine/chemistry , Serum Albumin, Human/chemistry
8.
Biomedicines ; 9(8)2021 Aug 17.
Article in English | MEDLINE | ID: mdl-34440232

ABSTRACT

We evaluated three newly synthesized B-lactam hybrid homo-aza-steroidal alkylators (ASA-A, ASA-B and ASA-C) for their PARP1/2 inhibition activity and their DNA damaging effect against human ovarian carcinoma cells. These agents are conjugated with an alkylating component (POPA), which also served as a reference molecule (positive control), and were tested against four human ovarian cell lines in vitro (UWB1.289 + BRCA1, UWB1.289, SKOV-3 and OVCAR-3). The studied compounds were thereafter compared to 3-AB, a known PARP inhibitor, as well as to Olaparib, a standard third-generation PARP inhibitor, on a PARP assay investigating their inhibitory potential. Finally, a PARP1 and PARP2 mRNA expression analysis by qRT-PCR was produced in order to measure the absolute and the relative gene expression (in mRNA transcripts) between treated and untreated cells. All the investigated hybrid steroid alkylators and POPA decreased in vitro cell growth differentially, according to the sensitivity and different gene characteristics of each cell line, while ASA-A and ASA-B presented the most significant anticancer activity. Both these compounds induced PARP1/2 enzyme inhibition, DNA damage (alkylation) and upregulation of PARP mRNA expression, for all tested cell lines. However, ASA-C underperformed on average in the above tasks, while the compound ASA-B induced synthetic lethality effects on the ovarian cancer cells. Nevertheless, the overall outcome, leading to a drug-like potential, provides strong evidence toward further evaluation.

9.
J Inorg Biochem ; 221: 111402, 2021 08.
Article in English | MEDLINE | ID: mdl-33975249

ABSTRACT

An oxovanadium(IV) - curcumin based complex, viz. [VO(cur)(2,2´-bipy)(H2O)] where cur is curcumin and bipy is bipyridine, previously synthesized, has been studied for interaction with albumin and DNA. Fluorescence emission spectroscopy was used to evaluate the interaction of the complex with bovine serum albumin (BSA) and the BSA-binding constant (Kb) was calculated to be 2.56 x 105 M-1, whereas a single great-affinity binding site was revealed. Moreover, the hemocompatibility test demonstrated that the complex presented low hemolytic fraction (mostly below 1%), in all concentrations tested (0-250 µΜ of complex, 5% DMSO) assuring a safe application in interaction with blood. The binding of the complex to DNA was also investigated using absorption, fluorescence, and viscometry methods indicating a binding through a minor groove mode. From competitive studies with ethidium bromide the apparent binding constant value to DNA was estimated to be 4.82 x 106 M-1. Stern-Volmer quenching phenomenon gave a ΚSV constant [1.92 (± 0.05) x 104 M-1] and kq constant [8.33 (± 0.2) x 1011 M-1s-1]. Molecular docking simulations on the crystal structure of BSA, calf thymus DNA, and DNA gyrase, as well as pharmacophore analysis for BSA target, were also employed to study in silico the ability of [VO(cur)(2,2´-bipy)(H2O)] to bind to these target bio-macromolecules and explain the observed in vitro activity.


Subject(s)
Coordination Complexes/metabolism , Curcumin/metabolism , DNA Gyrase/metabolism , DNA/metabolism , Serum Albumin, Bovine/metabolism , Animals , Binding Sites , Cattle , Coordination Complexes/chemistry , Coordination Complexes/toxicity , Curcumin/analogs & derivatives , Curcumin/toxicity , DNA/chemistry , DNA Gyrase/chemistry , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Hemolysis/drug effects , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Serum Albumin, Bovine/chemistry , Vanadium/chemistry , Vanadium/toxicity , Viscosity/drug effects
10.
J Inorg Biochem ; 212: 111213, 2020 11.
Article in English | MEDLINE | ID: mdl-32889129

ABSTRACT

The interaction of cobalt chloride with the non-steroidal anti-inflammatory drug indomethacin (Hindo) led to the formation of the polymeric complex [Co(indo-O)2(H2O)2(µ-Cl)]n·n(MeOH·H2O) bearing one chlorido bridge between the cobalt atoms. The presence of the nitrogen-donor co-ligands 2,2'-bipyridine (bipy), 2,2'-bipyridylamine (bipyam), 1,10-phenanthroline (phen) or 1H-imidazole (Himi) resulted in the isolation of complexes [Co2(µ-indo-O,O')2(indo-O)2(bipy)2(µ-H2O)]·3.3MeOH, [Co(indo-O,O')2(bipyam)]·0.9MeOH·0.2H2O, [Co(indo-O,O')2(phen)] (4) and [Co(indo-O)2(Himi)2] (5), respectively, where the indomethacin ligands were coordinated in diverse manners. The study of the affinity of the complexes for calf-thymus DNA revealed their intercalation between the DNA-bases. The binding of the complexes to albumins was also examined and the corresponding binding constants and binding subdomain were determined. The free radical scavenging activity of the compounds was evaluated towards 1,1-diphenyl-picrylhydrazyl and 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid). Molecular modeling calculations may usually provide a molecular basis for the understanding of both the impairment of DNA by its binding with the studied complexes and the ability of these compounds to transportation through serum albumin proteins. This study can provide information for the elucidation of the mechanism of action of the compounds in a molecular level.


Subject(s)
Cobalt/chemistry , Coordination Complexes/chemistry , Indomethacin/chemistry , Computer Simulation , In Vitro Techniques
11.
J Inorg Biochem ; 210: 111167, 2020 09.
Article in English | MEDLINE | ID: mdl-32653633

ABSTRACT

Herein we report on the synthesis and molecular structures of six silver(I) mixed-ligand complexes containing a heterocyclic thioamide [4-phenyl-imidazole-2-thione (phimtH) or 2,2,5,5-tetramethyl-imidazolidine-4-thione (tmimdtH)] and a tertiary arylphosphane [triphenylphosphine (PPh3), tri-o-tolylphosphane (totp)] or diphosphane [(1,2-bis(diphenylphosphano)ethane (dppe), bis(2-diphenylphosphano-phenyl)ether (DPEphos) or 4,5-bis(diphenylphosphano)-9,9-dimethylxanthene) (xantphos)]. The interaction of the compounds with calf-thymus DNA (CT DNA), as monitored directly via UV-vis spectroscopy and DNA-viscosity measurements and indirectly via its competition with ethidium bromide for DNA-intercalation sites, is suggested to take place via an intercalative mode. The new complexes show selective significant in vitro antibacterial activity against four bacterial strains. The antiproliferative effects and cytostatic efficacies of the complexes against four human cancer cell lines were evaluated. The best cytostatic and cytotoxic activity was appeared for the complexes bearing the phimtH moiety. In order to explain the described in vitro activity of the complexes, and to approach a possible mechanism of action, molecular docking studies were adopted on the crystal structure of CT DNA, DNA-gyrase, human estrogen receptor alpha and a cell-cycle specific target protein, human cyclin-dependent kinase 6.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Intercalating Agents/pharmacology , Organophosphorus Compounds/pharmacology , Thioamides/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Bacteria/drug effects , Cattle , Coordination Complexes/chemical synthesis , Coordination Complexes/metabolism , Cyclin-Dependent Kinase 6/metabolism , DNA/metabolism , DNA Gyrase/metabolism , Escherichia coli Proteins/metabolism , Estrogen Receptor alpha/metabolism , Humans , Intercalating Agents/chemical synthesis , Intercalating Agents/metabolism , Ligands , Microbial Sensitivity Tests , Molecular Docking Simulation , Organophosphorus Compounds/chemical synthesis , Organophosphorus Compounds/metabolism , Protein Binding , Silver/chemistry , Thioamides/chemical synthesis , Thioamides/metabolism
12.
Future Med Chem ; 12(1): 19-35, 2020 01.
Article in English | MEDLINE | ID: mdl-31729254

ABSTRACT

Aim: Steroidal prodrugs of nitrogen mustards such as estramustine and prednimustine have proven effective anticancer agents in clinical use since the 1970s. In this work, we aimed to develop steroidal prodrugs of the novel nitrogen mustard POPAM-NH2. POPAM-NH2 is a melphalan analogue that was coupled with three different steroidal lactams. Methodology: The new conjugates were preclinically tested for anticancer activity against nine human and one rodent cancer experimental models, in vitro and in vivo. Results & conclusion: All the steroidal alkylators showed high antitumor activity, in vitro and in vivo, in the experimental systems tested. Moreover, these hybrid compounds showed by far superior anticancer activity compared with the alkylating agents, melphalan and POPAM-NH2.


Subject(s)
Aniline Mustard/analogs & derivatives , Antineoplastic Agents/pharmacology , Drug Discovery , Lactams/pharmacology , Propionates/pharmacology , Steroids/pharmacology , Aniline Mustard/administration & dosage , Aniline Mustard/chemistry , Aniline Mustard/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , HT29 Cells , Humans , Injections, Intraperitoneal , Lactams/administration & dosage , Lactams/chemistry , Male , Mice , Mice, SCID , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Propionates/administration & dosage , Propionates/chemistry , Steroids/administration & dosage , Steroids/chemistry , Structure-Activity Relationship
13.
Amino Acids ; 50(2): 279-291, 2018 02.
Article in English | MEDLINE | ID: mdl-29185031

ABSTRACT

Quercetin is a flavonoid presenting cytotoxicity against different cancer cell lines. We hypothesized that its core could serve as a scaffold for generating more potent compounds. A quercetin-alanine bioconjugate was synthesized, its cellular internalization was monitored through confocal microscopy and its cytotoxic activity was explored against ten different cell lines. The bioconjugate consistently illustrated enhanced cytotoxic activity with respect to the parent compound. A threefold enhancement in its cytotoxicity was revealed for HeLa, A549, MCF-7 and LNCaP cells. In silico studies suggested that quercetin-alanine possesses enhanced binding affinity to human estrogen receptor alpha corroborating to its activity to MCF-7, overexpressing this receptor. Spectrofluorimetric, calorimetric and in silico studies revealed that quercetin-alanine binds primarily to Sudlow site I of serum albumin mainly through hydrogen bonding. Through this array of experiments we discovered that the specific compound bears a more refined pharmaceutical profile in contrast to quercetin in terms of cytotoxicity, while at the same time preserves its affinity to serum albumin. Natural products could thus offer a potent scaffold to develop bioconjugates with amplified therapeutic window.


Subject(s)
Antineoplastic Agents/pharmacology , Quercetin/analogs & derivatives , Quercetin/pharmacology , Alanine/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Biological Products/chemistry , Biological Products/metabolism , Biological Products/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Flavonoids/chemistry , Flavonoids/metabolism , Flavonoids/pharmacology , Humans , Inhibitory Concentration 50 , Mice , Molecular Docking Simulation , Protein Binding/drug effects , Quercetin/chemistry , Quercetin/metabolism , Serum Albumin/metabolism , Structure-Activity Relationship
14.
Org Biomol Chem ; 15(37): 7956-7976, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28902204

ABSTRACT

Anti-apoptotic proteins, like the Bcl-2 family proteins, present an important therapeutic cancer drug target. Their activity is orchestrated through neutralization upon interaction of pro-apoptotic protein counterparts that leads to immortality of cancer cells. Therefore, generating compounds targeting these proteins is of immense therapeutic importance. Herein, Induced Fit Docking (IFD) and Molecular Dynamics (MD) simulations were performed to rationally design quercetin analogues that bind in the BH3 site of the Bcl-xL protein. IFD calculations determined their binding cavity while Molecular Mechanics Poisson Boltzmann Surface Area (MM-PBSA) and Molecular Mechanics Generalised Born Surface Area (MM-GBSA) calculations provided an insight into the binding enthalpies of the analogues. The quercetin analogues were synthesized and their binding to Bcl-xL was verified with fluorescence spectroscopy. The binding affinity and the thermodynamic parameters between Bcl-xL and quercetin-glutamic acid were estimated through Isothermal Titration Calorimetry. 2D 1H-15N HSQC NMR chemical shift perturbation mapping was used to chart the binding site of the quercetin analogues in the Bcl-xL that overlapped with the predicted poses generated by both IFD and MD calculations. Furthermore, evaluation of the four conjugates against the prostate DU-145 and PC-3 cancer cell lines, revealed quercetin-glutamic acid and quercetin-alanine as the most potent conjugates bearing the higher cytostatic activity. This pinpoints that the chemical space of natural products can be tailored to exploit new hits for difficult tractable targets such as protein-protein interactions.


Subject(s)
Amino Acids/pharmacology , Antineoplastic Agents/pharmacology , Cytostatic Agents/pharmacology , Drug Design , Quercetin/pharmacology , bcl-X Protein/antagonists & inhibitors , Amino Acids/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cytostatic Agents/chemical synthesis , Cytostatic Agents/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Structure , Quercetin/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
15.
J BUON ; 21(4): 764-779, 2016.
Article in English | MEDLINE | ID: mdl-27685895

ABSTRACT

Conventional drug design embraces the "one gene, one drug, one disease" philosophy. Nowadays, new generation of anti- cancer drugs, able to inhibit more than one pathway, is believed to play a major role in contemporary anticancer drug research. In this way, polypharmacology, focusing on multi-target drugs, has emerged as a new paradigm in drug discovery. A number of recent successful drugs have in part or in whole emerged from a structure-based research approach. Many advances including crystallography and informatics are behind these successes. Increasing insight into the genetics and molecular biology of cancer has resulted in the identification of an increasing number of potential molecular targets, for anticancer drug discovery and development. These targets can be approached through exploitation of emerging structural biology, "rational" drug design, screening of chemical libraries, or a combination of these methods. The result is the rapid discovery of new anticancer drugs. In this article we discuss the application of molecular modeling, molecular docking and virtual high-throughput screening to multi-targeted anticancer drug discovery. Efforts have been made to employ in silico methods for facilitating the search and design of selective multi-target agents. These computer aided molecular design methods have shown promising potential in facilitating drug discovery directed at selective multiple targets and is expected to contribute to intelligent lead anticancer drugs.


Subject(s)
Antineoplastic Agents/chemistry , Drug Discovery/methods , Molecular Docking Simulation/methods , Small Molecule Libraries/chemistry , Antineoplastic Agents/therapeutic use , Drug Design , High-Throughput Screening Assays , Humans , Models, Molecular , Neoplasms/drug therapy , Small Molecule Libraries/therapeutic use
16.
Steroids ; 115: 1-8, 2016 11.
Article in English | MEDLINE | ID: mdl-27473822

ABSTRACT

Alkylating agents are still nowadays one of the most important classes of cytotoxic drugs, which display a wide range of therapeutic use for the treatment of various cancers. We have synthesized and tested four hybrid homo-azasteroidal alkylating esters for antileukemic activity against five sensitive to alkylating agents human leukemia cell lines in vitro and against P388 murine leukemia in vivo. Comparatively, melphalan and 3-(4-(bis(2-chloroethyl)amino)phenoxy)propanoic acid (POPAM) were also examined. All the homo-aza-steroidal alkylators showed relatively lower acute toxicity, very promising and antileukemic activity both in vitro and in vivo.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Lactams/chemical synthesis , Lactams/therapeutic use , Leukemia P388/drug therapy , Steroids/chemical synthesis , Steroids/therapeutic use , Aniline Mustard/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Humans , Jurkat Cells , K562 Cells , Lactams/chemistry , Lactams/pharmacology , Male , Mechlorethamine/chemistry , Mice, Inbred BALB C , Steroids/chemistry , Steroids/pharmacology , Structure-Activity Relationship
17.
J BUON ; 21(6): 1337-1358, 2016.
Article in English | MEDLINE | ID: mdl-28039691

ABSTRACT

Conventional drug design embraces the "one gene, one drug, one disease" philosophy. Nowadays, new generation of anticancer drugs, able to inhibit more than one pathway, is believed to play a major role in contemporary anticancer drug research. In this way, polypharmacology, focusing on multi-target drugs, has emerged as a new paradigm in drug discovery. A number of recent successful drugs have in part or in whole emerged from a structure-based research approach. Many advances including crystallography and informatics are behind these successes. In this part II we will review the role and methodology of ligand-, structure- and fragment-based computer-aided drug design computer aided drug desing (CADD), virtual high throughput screening (vHTS), de novo drug design, fragment-based design and structure-based molecular docking, homology modeling, combinatorial chemistry and library design, pharmacophore model chemistry and informatics in modern drug discovery.


Subject(s)
Antineoplastic Agents/pharmacology , Combinatorial Chemistry Techniques , Computer-Aided Design , Drug Design , High-Throughput Screening Assays , Molecular Targeted Therapy , Neoplasms/drug therapy , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Binding Sites , Humans , Ligands , Molecular Docking Simulation , Molecular Structure , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , Protein Conformation , Signal Transduction/drug effects , Structure-Activity Relationship
18.
Eur J Med Chem ; 96: 47-57, 2015.
Article in English | MEDLINE | ID: mdl-25874330

ABSTRACT

Recent science evidenced the interlinkage of oxidative stress and cancer. Due to the inherent complexity of cancer and its accompanying effect of oxidative stress, novel molecules, containing combinatorial functionalities should be targeted. Herein, we synthesized gemcitabine-5'-O-lipoate derived from a regioselective coupling of the chemotherapeutic drug gemcitabine (GEM), a first-line agent for cancer therapy and α-lipoic acid (LA), a potent antioxidant. gemcitabine-5'-O-lipoate was obtained in 4 chemical steps. To avoid the tedious and laborious chemical steps we also utilized biocatalysis using immobilized Candida antarctica lipase B (CALB), and the optimum conditions for the regioselective and one-pot synthesis of gemcitabine-5'-O-lipoate were established by exploiting different solvents (organic solvents and ionic liquids) and enzyme immobilization (acrylic resin and carbon nanotubes). Cytotoxic activity of co-administrating GEM and LA was proven to be synergistic against non-small cell lung cancer cells whereas antagonistic for bladder cancer cells. In contrast, the gemcitabine-5'-O-lipoate hybrid was found to be superior to the parent compounds against both non-small cell lung cancer and bladder cancer cells as also was found to preserve the redox activity of the parent compound LA. The regioselective biotransformation mediated synthesis of the anticancer-antioxidant hybrid illustrates the capacity of biocatalysis to act as an asset in molecular hybridization techniques.


Subject(s)
Antineoplastic Agents/pharmacology , Deoxycytidine/analogs & derivatives , Fungal Proteins/metabolism , Lipase/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Biocatalysis , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/chemistry , Deoxycytidine/metabolism , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Humans , Molecular Structure , Oxidation-Reduction , Stereoisomerism , Structure-Activity Relationship , Gemcitabine
19.
Anticancer Res ; 31(3): 831-42, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21498703

ABSTRACT

BACKGROUND: In order to reduce toxicity and to enhance anticancer activity of nitrogen mustards, three hybrid steroidal esters were synthesized and tested in vitro against human pancreatic cancer cells expressing uridine phosphorylase (UPase). The inhibition potency against a target protein implicated in the chemotherapy of solid tumors, such as UPase, is of fundamental importance in the design and synthesis of new anticancer drugs. MATERIALS AND METHODS: MTT colorimetric assay and molecular docking were employed for the in vitro and in silico drug evaluation, respectively. RESULTS: A difference in cell sensitivity was found, which followed the known different UPase expression in the cell lines. Molecular docking studies on UPase protein, revealed the tested compounds to be bound to the binding cavity of the protein, with different affinity. Between the two D-modified compounds, the D-homo-aza (lactam)-hybrid compound (C2) was found to interact with the protein in a more efficient way. CONCLUSION: The molecular docking data were in accordance with the in vitro results, where the lactam steroid alkylator showed significantly higher cytostatic and cytotoxic activity than the non-D-modified compounds, which also correlated with the level of UPase expression in the pancreatic cancer cells.


Subject(s)
Alkylating Agents/pharmacology , Antineoplastic Agents/pharmacology , Computational Biology , Molecular Targeted Therapy , Steroids/pharmacology , Uridine Phosphorylase/antagonists & inhibitors , Alkylating Agents/chemistry , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Fluorouracil/chemistry , Fluorouracil/metabolism , Humans , Ligands , Models, Molecular , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Steroids/chemistry , Thermodynamics , Thiouracil/analogs & derivatives , Thiouracil/chemistry , Thiouracil/metabolism , Uridine Phosphorylase/chemistry , Uridine Phosphorylase/metabolism
20.
Anticancer Res ; 30(10): 4201-4, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21036741

ABSTRACT

BACKGROUND: Modified steroidal derivatives (PK11-PK14) of p-bis(2-chloroethyl)aminophenyl propenate (PK15) were used to study their antitumour activity on Lewis lung carcinoma (LLC) and their effect on sister chromatid exchanges (SCEs) and human lymphocyte proliferation kinetics. MATERIALS AND METHODS: LLC was tested in this study. C57BL mice were used for in vivo chemotherapy evaluation and the antitumour activity was assessed. Lymphocyte cultures were used to study the genotoxic effect in vitro. RESULTS: PK15 and PK11 were the most effective against LLC, causing significant inhibition of tumour growth. PK11 and PK15 induced significant increase in SCE rates. A correlation was observed between the cytogenetic effect and the antitumour effectiveness. CONCLUSION: The order of the antitumour effectiveness of PK11-PK15 resembled the order of the cytogenetic damage induced by the same compounds in vitro.


Subject(s)
Carcinoma, Lewis Lung/drug therapy , Steroids/pharmacology , Animals , Cells, Cultured , Female , Humans , Lymphocytes/drug effects , Mice , Mice, Inbred C57BL , Phenylpropionates/pharmacology , Phenylpropionates/toxicity , Steroids/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...