Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.311
Filter
1.
Acta Chim Slov ; 71(2): 215-225, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38919104

ABSTRACT

1H-indole-2,3-dione 3-[4-(4-sulfamoylphenyl)thiosemicarbazones] (6a-j) were evaluated against Para-influenza-3, Reovirus-1, Sindbis, Coxsackie B4 and Punto Toro viruses. New 1-methyl-1H-indole-2,3-dione 3-[4-(4-sulfamoylphenyl)thiosemicarbazones] (7a-c) were synthesized to evaluate the contribution of methyl substitution at position 1- of the indole ring to antiviral activity. The test results showed that compounds 5-trifluoromethoxy- substituted 6c (EC50: 2-9 µM) and 5-bromo- substituted 6f (EC50: 2-3 µM) have non-toxic selective antiviral activity while not all standards are active against Reovirus-1. Molecular docking studies of 6c and 6f were carried out to determine the possible binding positions with Reovirus-1. Trifluoromethoxy and bromine substitutions at position 5- of the indole ring provided selective antiviral activity, while methyl substitution at position 1- of the indole ring significantly decreased the activity and increased toxicity against Reovirus-1.


Subject(s)
Antiviral Agents , Thiosemicarbazones , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/chemical synthesis , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemistry , Thiosemicarbazones/chemical synthesis , Molecular Docking Simulation , Animals , Indoles/pharmacology , Indoles/chemistry , Humans , Structure-Activity Relationship
2.
An Acad Bras Cienc ; 96(3): e20230811, 2024.
Article in English | MEDLINE | ID: mdl-38865509

ABSTRACT

Cancer is a complex and multifactorial disease characterized by uncontrolled cell growth and is one of the main causes of death in the world. This work aimed to evaluate a small series of 10 different indole-thiosemicarbazone compounds as potential antitumor agents. This is a pioneering study. For this, the antioxidant and cytotoxic capacity against normal and tumor cells was evaluated. The results showed that the compounds were able to promote moderate to low antioxidant activity for the ABTS radical scavenging assay. ADMET in silico assays showed that the compounds exhibited good oral bioavailability. As for toxicity, they were able to promote low cytotoxicity against normal cells, in addition to not being hemolytic. The compounds showed promising in vitro antitumor activity against the T47D, MCF-7, Jurkat and DU-145 strains, not being able to inhibit the growth of the Hepg2 strain. Through this in vitro study, it can be concluded that the compounds are potential candidates for antitumor agents.


Subject(s)
Antineoplastic Agents , Antioxidants , Indoles , Thiosemicarbazones , Humans , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacokinetics , Indoles/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antioxidants/pharmacology , Cell Line, Tumor , Computer Simulation , Drug Screening Assays, Antitumor , Cell Proliferation/drug effects
3.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38928235

ABSTRACT

The reaction mechanism of tthe formation of azomethine ylides from isatins and sarcosine is addressed in the literature in a general manner. This computational study aims to explore the mechanistic steps for this reaction in detail and to assess the reactivity of formed ylide in a 1,3-dipolar cycloaddition reaction with 7-oxabenzonorbornadiene. For this purpose, density functional theory (DFT) calculations at the M06-2X(SMD,EtOH)/6-31G(d,p) level were employed. The results indicate that CO2 elimination is the rate-determining step, the activation barrier for 1,3-dipolar cycloaddition is lower, and the formed ylide will readily react with dipolarophiles. The substitution of isatine with electron-withdrawal groups slightly decreases the activation barrier for ylide formation.


Subject(s)
Azo Compounds , Cycloaddition Reaction , Sarcosine , Thiosemicarbazones , Thiosemicarbazones/chemistry , Azo Compounds/chemistry , Sarcosine/chemistry , Sarcosine/analogs & derivatives , Isatin/chemistry , Models, Molecular , Density Functional Theory , Norbornanes/chemistry , Molecular Structure
4.
J Inorg Biochem ; 257: 112577, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38714060

ABSTRACT

A new pyrazole based thiosemicarbazone ligand, 5-methyl-3-formylpyrazole-N(4)-isopropylthiosemicarbazone, (HMPzNHPri) (compound I), and its cobalt(III) and nickel(II) complexes, [Co(MPzNHPri)2]Cl (compound II) and [Ni(HMPzNHPri)2]Br2 (compound III), respectively, have been synthesized and characterized through various physico-chemical and spectroscopic studies. Both the reported Co(III) and Ni(II) complexes are cationic in nature and behave as 1:1 and 1:2 electrolytes in MeOH, respectively. Electronic spectral features of the complexes have classified them as distorted octahedral ones. IR spectral data (4000-450 cm-1) have suggested a monoprotic tridentate (NNS) function of compound I coordinating to the Co(III) ion via the pyrazolyl (tertiary) ring nitrogen, azomethine nitrogen and thiolato sulphur atom; while for compound III, compound I has been found to act as neutral NNS tridentate one, coordinating to Ni(II) via the pyrazolyl iminic nitrogen, azomethine nitrogen and thioketo sulphur. Structural features of all the compounds are confirmed by the single crystal X-ray data. All the compounds reported here have been found to exhibit significant photocatalytic activity towards degradation of Methylene Blue (MB) under UV radiation. Anticancer activity of all the three compounds against cancer cell lines (HeLa and A549) and a normal cell line (HEK293) have been investigated. Compound II has been found to be more efficient against the human cervical cancer cell (HeLa) and the lung cancer cell (A549) than compounds I and III. The ligand and both the complexes display potential activities against both gram-positive (Bacillus subtilis MTCC 7193) and gram-negative bacteria (E. coli MTCC 1610).


Subject(s)
Antineoplastic Agents , Cobalt , Coordination Complexes , Nickel , Pyrazoles , Thiosemicarbazones , Thiosemicarbazones/chemistry , Nickel/chemistry , Cobalt/chemistry , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrazoles/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Humans , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Crystallography, X-Ray/methods , Ligands , Cell Line, Tumor , Catalysis , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Microbial Sensitivity Tests
5.
J Med Chem ; 67(11): 9091-9103, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38778566

ABSTRACT

Induction of cuproptosis and targeting of multiple signaling pathways show promising applications in tumor therapy. In this study, we synthesized two thiosemicarbazone-copper complexes ([CuII(L)Cl] 1 and [CuII2CuI(L)2Cl3] 2, where HL is the (E)-N-methyl-2-(phenyl(pyridin-2-yl)methylene ligand), to assess their antilung cancer activities. Both copper complexes showed better anticancer activity than cisplatin and exhibited hemolysis comparable to that of cisplatin. In vivo experiments showed that complex 2 retarded the A549 cell growth in a mouse xenograft model with low systemic toxicity. Primarily, complex 2 kills lung cancer cells in vitro and in vivo by triggering multiple pathways, including cuproptosis. Complex 2 is the first mixed-valent Cu(I/II) complex to induce cellular events consistent with cuproptosis in cancer cells, which may stimulate the development of mixed-valent copper complexes and provide effective cancer therapy.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Copper , Lung Neoplasms , Thiosemicarbazones , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemical synthesis , Thiosemicarbazones/therapeutic use , Humans , Copper/chemistry , Animals , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Coordination Complexes/therapeutic use , Mice , Mice, Nude , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Cell Line, Tumor , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , A549 Cells , Cell Proliferation/drug effects , Xenograft Model Antitumor Assays , Signal Transduction/drug effects , Structure-Activity Relationship , Hemolysis/drug effects , Drug Screening Assays, Antitumor , Mice, Inbred BALB C
6.
J Steroid Biochem Mol Biol ; 242: 106545, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38762058

ABSTRACT

Most breast and prostate cancers are caused by abnormal production or action of steroidal hormones. Hormonal drugs based on steroid scaffolds represent a significant class of chemotherapeutics that are routinely used in chemotherapy. In this study, the synthesis of new 17a-homo lactone and 17α-(pyridine-2-ylmethyl) androstane derivatives with hydrazide and semicarbazone motifs is presented. All compounds were screened for their effect on cell viability against a panel of five cancer cell lines and one healthy cell line. Two compounds showed significant cytotoxicity against cancer cells, with low toxicity against healthy cells. The relative binding affinities of compounds for the ligand-binding domains of estrogen receptor α, estrogen receptor ß, androgen receptor and glucocorticoid receptor were tested using a fluorescence screen in yeast. Potential for inhibition of aldo-keto reductase 1C3 and 1C4 activity was measured in vitro. Experimental results are analyzed in the context of molecular docking simulations. Our results could help guide design of steroid compounds with improved anticancer properties against androgen- and estrogen-dependent cancers.


Subject(s)
Antineoplastic Agents , Molecular Docking Simulation , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Hydrazines/pharmacology , Hydrazines/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Steroids/chemistry , Steroids/pharmacology , Semicarbazones/pharmacology , Semicarbazones/chemistry , Semicarbazones/chemical synthesis , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemistry , Drug Screening Assays, Antitumor
7.
J Med Chem ; 67(11): 9069-9090, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38771959

ABSTRACT

The development of copper(II) thiosemicarbazone complexes as potential anticancer agents, possessing dual functionality as inhibitors of R2 ribonucleotide reductase (RNR) and tubulin polymerization by binding at the colchicine site, presents a promising avenue for enhancing therapeutic effectiveness. Herein, we describe the syntheses and physicochemical characterization of four isomeric proligands H2L3-H2L6, with the methylmorpholine substituent at pertinent positions of the pyridine ring, along with their corresponding Cu(II) complexes 3-6. Evidently, the position of the morpholine moiety and the copper(II) complex formation have marked effects on the in vitro antiproliferative activity in human uterine sarcoma MES-SA cells and the multidrug-resistant derivative MES-SA/Dx5 cells. Activity correlated strongly with quenching of the tyrosyl radical (Y•) of mouse R2 RNR protein, inhibition of RNR activity in the cancer cells, and inhibition of tubulin polymerization. Insights into the mechanism of antiproliferative activity, supported by experimental results and molecular modeling calculations, are presented.


Subject(s)
Antineoplastic Agents , Copper , Morpholines , Ribonucleotide Reductases , Thiosemicarbazones , Tubulin , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemical synthesis , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Ribonucleotide Reductases/antagonists & inhibitors , Ribonucleotide Reductases/metabolism , Tubulin/metabolism , Animals , Morpholines/pharmacology , Morpholines/chemistry , Morpholines/chemical synthesis , Copper/chemistry , Mice , Cell Line, Tumor , Cell Proliferation/drug effects , Structure-Activity Relationship , Polymerization/drug effects , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Pyridines/pharmacology , Pyridines/chemistry , Pyridines/chemical synthesis , Tubulin Modulators/pharmacology , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Drug Screening Assays, Antitumor , Models, Molecular
8.
Bioorg Chem ; 148: 107486, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38788367

ABSTRACT

The study aims to synthesize a novel bis(thiosemicarbazone) derivative based on platinum (thioPt) and evaluate its anticancer properties against MFC-7 and MDA-MB-231 breast cancer cells. A new platinum complex was synthesised by reacting K2PtCl4 with 2,2'-(1,2-diphenylethane-1,2-diylidene)bis(hydrazine-1-carbothioamide) in ethanol in the presence of K2CO3. In the obtained complex, the platinum atom is coordinated by a conjugated system = N-NC-S-The structures of the new compound were characterised using NMR spectroscopy, HR MS, IR, and X-ray structural analysis. The obtained results of the cytotoxicity assay indicate that compound thioPt had potent anticancer activity (MCF-7: 61.03 ± 3.57 µM, MDA-MB-231: 60.05 ± 5.40 µM) with less toxicity against normal MCF-10A breast epithelial cells, even compared to the reference compound (cisplatin). In addition, subsequent experiments found that thioPt induces apoptosis through both an extrinsic (↑caspase 8 activity) and intrinsic (↓ΔΨm) pathway, which ultimately leads to an increase in active caspase 3/7 levels. The induction of autophagy and levels of proteins involved in this process (LC3A/B and Beclin-1) were examined in MCF-7 and MDA-MB-231 breast cancer cells exposed to tested compounds (thio, thioPt, cisPt) at a concentration of 50 µM for 24 h. Based on these results, it can be concluded that thio and thioPt do not significantly affect the autophagy process. This demonstrates their superiority over cisplatin, which can stimulate cancer cell survival through its effect on stimulation of autophagy.


Subject(s)
Antineoplastic Agents , Apoptosis , Breast Neoplasms , Cell Proliferation , Drug Screening Assays, Antitumor , Thiosemicarbazones , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemistry , Thiosemicarbazones/chemical synthesis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Apoptosis/drug effects , Structure-Activity Relationship , Molecular Structure , Cell Proliferation/drug effects , Female , Dose-Response Relationship, Drug , Cell Line, Tumor , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/chemical synthesis , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Platinum/chemistry , Platinum/pharmacology , Autophagy/drug effects
9.
An Acad Bras Cienc ; 96(2): e20231247, 2024.
Article in English | MEDLINE | ID: mdl-38808881

ABSTRACT

Thiosemicarbazones are promising classes of compounds with antitumor activity. For this study, six 2,4-dihydroxy-benzylidene-thiosemicarbazones compounds were synthesized. These compounds were submitted to different assays in silico, in vitro and in vivo to evaluate the toxicological, antioxidant and antitumor effects. The in silico results were evaluated by the SwissADME and pkCSM platforms and showed that all compounds had good oral bioavailability profiles. The in vitro and in vivo toxicity assays showed that the compounds showed low cytotoxicity against different normal cells and did not promote hemolytic effects. The single dose acute toxicity test (2000 mg/kg) showed that none of the compounds were toxic to mice. In in vitro antioxidant activity assays, the compounds showed moderate to low activity, with PB17 standing out for the ABTS radical capture assay. The in vivo antioxidant activity highlighted the compounds 1, 6 and 8 that promoted a significant increase in the concentration of liver antioxidant enzymes. Finally, all compounds showed promising antitumor activity against different cell lines, especially MCF-7 and DU145 lines, in addition, they inhibited the growth of sarcoma 180 at concentrations lower than 50 mg/kg. These results showed that the evaluated compounds can be considered as potential antitumor agents.


Subject(s)
Antineoplastic Agents , Antioxidants , Thiosemicarbazones , Animals , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemistry , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Mice , Humans , Male , Cell Line, Tumor , Computer Simulation , Drug Screening Assays, Antitumor , Female , Benzylidene Compounds/pharmacology , Benzylidene Compounds/chemistry
10.
Bioorg Chem ; 147: 107338, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38583253

ABSTRACT

Macrozones are novel conjugates of azithromycin and thiosemicarbazones, which exhibit very good in vitro antibacterial activities against susceptible and some resistant bacterial strains thus showing a potential for further development. A combination of spectrometric (fluorimetry, STD and WaterLOGSY NMR) and molecular docking studies provided insights into atomic details of interactions between selected macrozones and biological receptors such as E. coli ribosome and bovine serum albumin. Fluorimetric measurements revealed binding constants in the micro-molar range while NMR experiments provided data on binding epitopes. It has been demonstrated that both STD and WaterLOGSY gave comparable and consistent results unveiling atoms in intimate contacts with biological receptors. Docking studies pointed towards main interactions between macrozones and E. coli ribosome which included specific π - π stacking and hydrogen bonding interactions with thiosemicarbazone part extending down the ribosome exit tunnel. The results of the docking experiments were in fine correlation with those obtained by NMR and fluorimetry. Our investigation pointed towards a two-site binding mechanism of interactions between macrozones and E. coli ribosome which is the most probable reason for their activity against azithromycin-resistant strains. Much better activity of macrozone-nickel coordinated compound against E. coli ribosome compared to other macrozones has been attributed to the higher polarity which enabled better bacterial membrane penetration and binding of the two thiosemicarbazone units thus additionally contributing to the overall binding energy. The knowledge gained in this study should play an important role in anti-infective macrolide design in the future.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Fluorometry , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Molecular Docking Simulation , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Escherichia coli/drug effects , Binding Sites , Molecular Structure , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Structure-Activity Relationship , Ribosomes/metabolism , Ribosomes/drug effects , Dose-Response Relationship, Drug , Animals , Cattle , Azithromycin/pharmacology , Azithromycin/chemistry , Azithromycin/metabolism
11.
Chem Biodivers ; 21(5): e202400363, 2024 May.
Article in English | MEDLINE | ID: mdl-38470083

ABSTRACT

Reactions between sodium tetrachloropalladate and 2- (or 4-) substituted 4-phenyl-3-thiosemicarbazone ligands (HLR), with various electron-donating and electron-withdrawing substituents (R = OCH3, NO2, and Cl), afford square-planar complexes of the general formula [Pd(LR)2]. Ground-state geometry optimization and the vibrational analysis of cis- and trans-isomers of the complexes were carried out to get an insight into the stereochemistry of the complexes. Natural bond orbital analysis was used to analyze how the nature of the substituent affects the natural charge of the metal center, the type of hybridization, and the strength of the M-N and M-S bonds. Using spectrophotometry, the stability of the complexes, and their DNA binding abilities were assessed. The Pd(II) complexes showed moderate cytotoxicity against MCF-7 and Caco-2 cell lines, two of the assessed malignant cell lines, resulting in all known cell death types, including early apoptotic bodies and late apoptotic vacuoles as well as evident necrotic bodies.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Palladium , Thiosemicarbazones , Humans , Palladium/chemistry , Palladium/pharmacology , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Ligands , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Drug Screening Assays, Antitumor , Caco-2 Cells , Cell Proliferation/drug effects , MCF-7 Cells , Molecular Structure , Apoptosis/drug effects , Cell Death/drug effects , Structure-Activity Relationship , DNA/chemistry , DNA/metabolism , DNA/drug effects
12.
Int J Biol Macromol ; 266(Pt 2): 131068, 2024 May.
Article in English | MEDLINE | ID: mdl-38531526

ABSTRACT

An extensive range of new biologically active morpholine based thiosemicarbazones derivatives 3a-r were synthesized, characterized by spectral techniques and evaluated as inhibitors of ENPP isozymes. Most of the novel thiosemicarbazones exhibit potent inhibition towards NPP1 and NPP3 isozymes. Compound 3 h was potent inhibitor of NPP1 with IC50 value of 0.55 ±â€¯0.02. However, the most powerful inhibitor of NPP3 was 3e with an IC50 value of 0.24 ±â€¯0.02. Furthermore, Lineweaver-Burk plot for compound 3 h against NPP1 and for compound 3e against NPP3 was devised through enzymes kinetics studies. Molecular docking and in silico studies was also done for analysis of interaction pattern of all newly synthesized compounds. The results were further validated by molecular dynamic (MD) simulation where the stability of conformational transformation of the best protein-ligand complex (3e) were justified on the basis of RMSD and RMSF analysis.


Subject(s)
Molecular Docking Simulation , Molecular Dynamics Simulation , Morpholines , Phosphoric Diester Hydrolases , Pyrophosphatases , Thiosemicarbazones , Morpholines/chemistry , Morpholines/pharmacology , Morpholines/chemical synthesis , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/metabolism , Pyrophosphatases/antagonists & inhibitors , Pyrophosphatases/chemistry , Pyrophosphatases/metabolism , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemical synthesis , Humans , Kinetics , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/chemical synthesis , Computer Simulation , Structure-Activity Relationship , Ligands
13.
Spectrochim Acta A Mol Biomol Spectrosc ; 313: 124117, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38461559

ABSTRACT

Cancer's global impact necessitates innovative and less toxic treatments. Thiosemicarbazones (TSCs), adaptable metal chelators, offer such potential. In this study, we have synthesized N (4)-substituted heterocyclic TSCs from syringaldehyde (TSL1, TSL2), and also report the unexpected copper-mediated cyclization of the TSCs to form thiadiazoles (TSL3, TSL4), expanding research avenues. This work includes extensive characterization and studies such as DNA/protein binding, molecular docking, and theoretical analyses to demonstrate the potential of the as-prepared TSCs and thiadiazoles against different cancer cells. The DFT results depict that the thiadiazoles exhibit greater structural stability and reduced reactivity compared to the corresponding TSCs. The docking results suggest superior EGFR inhibition for TSL3 with a binding constant value of - 6.99 Kcal/mol. According to molecular dynamics studies, the TSL3-EGFR complex exhibits a lower average RMSD (1.39 nm) as compared to the TSL1-EGFR complex (3.29 nm) suggesting that both the thiadiazole and thiosemicarbazone examined here can be good inhibitors of EGFR protein, also that TSL3 can inhibit EGFR better than TSL1. ADME analysis indicates drug-likeness and oral availability of the thiadiazole-based drugs. The DNA binding experiment through absorption and emission spectroscopy discovered that TSL3 is more active towards DNA which is quantitatively calculated with a Kb value of 4.74 × 106 M-1, Kq value of 4.04 × 104 M-1and Kapp value of 5 × 106 M-1. Furthermore, the BSA binding studies carried out with fluorescence spectroscopy showed that TSL3 shows better binding capacity (1.64 × 105 M-1) with BSA protein. All the compounds show significant cytotoxicity against A459-lung, MCF-7-breast, and HepG2-liver cancer cell lines; TSL3 exhibits the best cytotoxicity, albeit less effective than cisplatin. Thiadiazoles demonstrate greater cytotoxicity than the TSCs. Overall, the promise of TSCs and thiadiazoles in cancer research is highlighted by this study. Furthermore, it unveils unexpected copper-mediated cyclization of the TSCs to thiadiazoles.


Subject(s)
Antineoplastic Agents , Thiadiazoles , Thiosemicarbazones , Molecular Docking Simulation , Density Functional Theory , Copper/pharmacology , Copper/chemistry , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemistry , Cyclization , Thiadiazoles/pharmacology , Thiadiazoles/chemistry , Spectrometry, Fluorescence , DNA/chemistry , ErbB Receptors/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry
14.
Anticancer Agents Med Chem ; 24(9): 649-667, 2024.
Article in English | MEDLINE | ID: mdl-38367264

ABSTRACT

INTRODUCTION: Carbonic anhydrases (CAs) are widespread metalloenzymes with the core function of catalyzing the interconversion of CO2 and HCO3 -. Targeting these enzymes using selective inhibitors has emerged as a promising approach for the development of novel therapeutic agents against multiple diseases. METHODS: A series of novel thiosemicarbazone-containing derivatives were synthesized, characterized, and tested for their inhibitory activity against pharmaceutically important human CA I (hCA I), II (hCA II), IX (hCA IX), and XII (hCA XII) using the single tail approach. RESULTS: The compounds generally inhibited the isoenzymes at low nanomolar concentrations, with compound 6b having Ki values of 7.16, 0.31, 92.5, and 375 nM against hCA I, II, IX and XII, respectively. Compound 6e exhibited Ki values of 27.6, 0.34, 872, and 94.5 nM against hCA I, II, IX and XII, respectively. CONCLUSION: To rationalize the inhibition data, molecular docking studies were conducted, providing insight into the binding mechanisms, molecular interactions, and selectivity of the compounds towards the isoenzymes.


Subject(s)
Benzenesulfonamides , Carbonic Anhydrase Inhibitors , Carbonic Anhydrases , Sulfonamides , Thiosemicarbazones , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/chemistry , Humans , Sulfonamides/chemistry , Sulfonamides/pharmacology , Sulfonamides/chemical synthesis , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemical synthesis , Carbonic Anhydrases/metabolism , Structure-Activity Relationship , Molecular Structure , Molecular Docking Simulation , Dose-Response Relationship, Drug
15.
Molecules ; 29(2)2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38276575

ABSTRACT

The aim of this work is to explore a new library of coordination compounds for medicinal applications. Gallium is known for its various applications in this field. Presently, indium is not particularly important in medicine, but it shares a lot of chemical traits with its above-mentioned lighter companion, gallium, and is also used in radio imaging. These metals are combined with thiosemicarbazones, ligating compounds increasingly known for their biological and pharmaceutical applications. In particular, the few ligands chosen to interact with these hard metal ions share the ideal affinity for a high charge density. Therefore, in this work we describe the synthesis and the characterization of the resulting coordination compounds. The yields of the reactions vary from a minimum of 21% to a maximum of 82%, using a fast and easy procedure. Nuclear Magnetic Resonance (NMR) and Infra Red (IR) spectroscopy, mass spectrometry, elemental analysis, and X-ray Diffraction (XRD) confirm the formation of stable compounds in all cases and a ligand-to-metal 2:1 stoichiometry with both cations. In addition, we further investigated their chemical and biological characteristics, via UV-visible titrations, stability tests, and cytotoxicity and antibiotic assays. The results confirm a strong stability in all explored conditions, which suggests that these compounds are more suitable for radio imaging applications rather than for antitumoral or antimicrobic ones.


Subject(s)
Coordination Complexes , Gallium , Thiosemicarbazones , Gallium/pharmacology , Gallium/chemistry , Indium/chemistry , Thiosemicarbazones/chemistry , Ligands , Magnetic Resonance Spectroscopy , Coordination Complexes/chemistry
16.
J Inorg Biochem ; 251: 112438, 2024 02.
Article in English | MEDLINE | ID: mdl-38029536

ABSTRACT

Cancer continues to pose a global threat, underscoring the urgent need for more effective and safer treatment options. Gold-based compounds have recently emerged as promising candidates due to their diverse range of biological activities. In this study, three gold(III) complexes derived from thiosemicarbazone ligands have been synthesized, fully characterized, including their X-ray crystal structures. We conducted initial mode-of-action studies on DNA and BSA, followed by a comprehensive investigation into the cytotoxic effects of these novel gold(III) complexes on lung cancer cells (A549, H2052, and H28). The results demonstrated a concentration-dependent cytotoxic response, with H28 cells exhibiting the highest sensitivity to the treatment. Furthermore, the analysis of the cell cycle revealed that these compounds induce cell cycle arrest and promote apoptosis as a response to treatment. We also observed distinct morphological changes and increased oxidative stress, contributing significantly to cell death. Notably, these complexes exhibited the ability to suppress interleukin-6 production in mesothelioma cell lines, and this highlights their anti-inflammatory potential. To gain an initial understanding of cytotoxicity on healthy cells, hemolysis tests were conducted against human blood cells, with no evidence of hemolysis. Furthermore, a toxicity assessment through the in vivo Galleria mellonella model underscored the absence of detectable toxicity. These findings prove that these complexes are promising novel therapeutic agents for lung cancer.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Lung Neoplasms , Thiosemicarbazones , Humans , Gold/chemistry , Lung Neoplasms/drug therapy , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemistry , Hemolysis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Ligands , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Cell Line, Tumor
17.
Biometals ; 37(1): 247-265, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37938497

ABSTRACT

Malaria, a relentless and ancient adversary, continues to cast its shadow over vast swathes of the globe, afflicting millions of people and have a heavy toll on human health and well-being. Despite substantial progress in the fight against this parasitic disease in recent decades, malaria still persists as a substantial global health concern, especially in some specific region which have limited resources and vulnerable populations. Thus, to ascertain an combating agent for malaria and its associated dysfunction, 4-(4-ethylphenyl)-3-thiosemicarbazide and benzaldehydes based two new thiosemicarbazone ligands (1-2) and their cobalt(II), nickel(II), copper(II), zinc(II) metal complexes (3-10) were synthesized in the present research work. The synthesized compounds were comprehensive characterized through spectral and physical investigations, demonstrating octahedral stereochemistry of the complexes. Further, the antimalarial and antioxidant potential of the compounds (1-10) were analyzed by micro assay and DPPH assay protocols, respectively, to examine the therapeutic aspect of the compounds. The performed biological evaluations revealed that the complexes are more efficient in controlling infectious ailment in comparison of ligands. The complexes (5), (6), (10) shows significant efficiency for malarial and oxidant dysfunctions whereas Zn(II) complex (6) exhibit highest potency with 1.02 ± 0.07 and 2.28 ± 0.05 µM IC50 value. Furthermore, to support the highest antimalarial potency of the (3-6) complexes and their associated ligand (1), the computational studies like molecular docking, DFT, MESP and ADMET analysis were executed which were supported the biological efficacy of the complex (6) by providing numerous parameters like binding interaction electronegativity, electrophilicity, HOMO value and electron density.


Subject(s)
Antimalarials , Coordination Complexes , Malaria , Thiosemicarbazones , Humans , Antimalarials/pharmacology , Antimalarials/chemistry , Molecular Docking Simulation , Antioxidants/pharmacology , Antioxidants/chemistry , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemistry , Ligands , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Zinc/chemistry , Copper/chemistry , Chelating Agents
18.
J Biol Inorg Chem ; 29(1): 3-31, 2024 02.
Article in English | MEDLINE | ID: mdl-38148423

ABSTRACT

Thiosemicarbazones are biologically active substances whose structural formula is formed by an azomethine, an hydrazine, and a thioamide fragments, to generate a R2C=N-NR-C(=S)-NR2 backbone. These compounds often act as ligands to generate highly stable metal-organic complexes. In certain experimental conditions, however, thiosemicarbazones undergo reactions leading to the cleavage of the chain. Sometimes, the breakage involves desulfurization processes. The present work summarizes the different chemical factors that influence the desulfurization reactions of thiosemicarbazones, such as pH, the presence of oxidant reactants or the establishment of redox processes as those electrochemically induced, the effects of the solvent, the temperature, and the electromagnetic radiation. Many of these reactions require coordination of thiosemicarbazones to metal ions, even those present in the intracellular environment. The nature of the products generated in these reactions, their detection in vivo and in vitro, together with the relevance for the biological activity of these compounds, mainly as antineoplastic agents, is discussed.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Thiosemicarbazones , Metals , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Oxidation-Reduction , Thiosemicarbazones/chemistry , Ions , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry
19.
Int J Mol Sci ; 24(24)2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38139350

ABSTRACT

The number of people affected by cancer and antibiotic-resistant bacterial infections has increased, such that both diseases are already seen as current and future leading causes of death globally. To address this issue, based on a combined in silico and in vitro approach, we explored the anticancer potential of known antibacterials with a thiazolidinedione-thiosemicarbazone (TZD-TSC) core structure. A cytotoxicity assessment showed encouraging results for compounds 2-4, with IC50 values against T98G and HepG2 cells in the low micromolar range. TZD-TSC 3 proved to be most toxic to cancer cell lines, with IC50 values of 2.97 ± 0.39 µM against human hepatoma HepG2 cells and IC50 values of 28.34 ± 2.21 µM against human glioblastoma T98G cells. Additionally, compound 3 induced apoptosis and showed no specific hemolytic activity. Furthermore, treatment using 3 on cancer cell lines alters these cells' morphology and further suppresses migratory activity. Molecular docking, in turn, suggests that 3 would have the capacity to simultaneously target HDACs and PPARγ, by the activation of PPARγ and the inhibition of both HDAC4 and HDAC8. Thus, the promising preliminary results obtained with TZD-TSC 3 represent an encouraging starting point for the rational design of novel chemotherapeutics with dual antibacterial and anticancer activities.


Subject(s)
Antineoplastic Agents , Thiazolidinediones , Thiosemicarbazones , Humans , Structure-Activity Relationship , Cell Line, Tumor , Molecular Docking Simulation , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemistry , PPAR gamma , Drug Screening Assays, Antitumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Thiazolidinediones/pharmacology , Anti-Bacterial Agents/pharmacology , Molecular Structure , Cell Proliferation , Histone Deacetylases/metabolism , Repressor Proteins/metabolism
20.
PLoS One ; 18(11): e0295012, 2023.
Article in English | MEDLINE | ID: mdl-38032914

ABSTRACT

A series of 38 thiosemicarbazone derivatives based on camphene and limonene were evaluated for their antiproliferative activity. Among them, 19 were synthesized and characterized using proton and carbon-13 nuclear magnetic resonance (1H and 13C NMR). For initial compound selection, human melanoma cells (SK-MEL-37) were exposed to a single concentration of a compound (100 µM) for 24, 48, and 72 hours, and cell detachment was visually observed. Cell viability was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method. Nineteen compounds (4, 6, 8, 11, 13, 14, 15, 16, 17, 18, 20, 22, 25, 26, 31, 3', 4', 6', and 9') yielded cell viability below 20%. Subsequently, IC50 values for these compounds were determined, ranging from 11.56 to 55.38 µM, after 72 hours of treatment. Compound 17 (o-hydroxybenzaldehyde (-)-camphene-based thiosemicarbazone) demonstrated the lowest IC50 value, followed by compound 4 (benzaldehyde (-) camphene-based thiosemicarbazone) at 12.84 µM. Regarding compound 4, we observed the induction of a characteristic ladder pattern of DNA fragmentation through gel electrophoresis. Furthermore, fluorescence, flow cytometry and scanning microscopy assays revealed morphological changes consistent with apoptosis induction. Additionally, the measurement of caspase 6 and 8 activity in cellular extracts after treatment for 2, 4, 6, and 24 hours suggested the potential involvement of the extrinsic apoptosis pathway in the mechanism of action of compound 4. Further investigations, including molecular docking studies, are required to fully explore the potential of compound 4 and the other selected compounds, highlighting their promising role in future melanoma therapy research.


Subject(s)
Antineoplastic Agents , Melanoma , Thiosemicarbazones , Humans , Limonene/pharmacology , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemistry , Molecular Docking Simulation , Cell Proliferation , Melanoma/drug therapy , Melanoma/pathology , Apoptosis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Structure-Activity Relationship , Drug Screening Assays, Antitumor
SELECTION OF CITATIONS
SEARCH DETAIL
...