Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Chem Rec ; 23(3): e202200247, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36762719

ABSTRACT

During last two decades, there has been an enormous growth in the discovery of innovative active inorganic anticancer complexes (exerting remarkable cytotoxicity at sub micro-molar levels) derived from myriad ligand scaffolds, mainly acting on cancerous vs healthy cells by either halting or inhibiting their uncontrolled growth. The phenomenal success of cisplatin to treat numerous forms of solid malignancies has placed metal-based drugs to the forefront of treatment strategies against cancers. More than 10,000 platinum anticancer complexes have been developed during the past 40 years, but only five drugs have been approved for usage in humans while ten more complexes are currently undergoing clinical trials. Most of the compounds have failed either at R&D stages or in preclinical trails. This has led to extensive investigations by researchers of medicinal chemistry, including our group to design and prepare tailored 3d-metallo-drugs and organotin(IV) compounds from some naturally occurring bioactive compounds, such as amino-acids, peptides, chromone derivatives and NSAID's etc. that were used either alone or in cocktail combination, capable of specifically targeting DNA, lnc RNAs and proteins. Furthermore, 3d-metal ions such as copper, cobalt and zinc etc. incorporated in these ligand framework are biocompatible and induce a unique multi-modal mechanism of cytotoxic action involving angiogenesis, ROS-induced DNA damage, apoptosis by p53 mitochondrial genes and caspases etc. The results observed a positive correlation between the binding affinity of complexes with DNA (as quantified by intrinsic binding constant values) and their cytotoxic behavior. Complexes with high DNA binding propensity were typically lethal against a diverse panel of malignant cell types compared to normal cells.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Neoplasms , Humans , Ligands , Antineoplastic Agents/chemistry , Neoplasms/drug therapy , Copper/chemistry , DNA/chemistry , Coordination Complexes/chemistry
2.
Toxicol Appl Pharmacol ; 447: 116072, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35613639

ABSTRACT

Sorafenib is an FDA-approved chemotherapeutic drug used as standard therapy for advanced-stage cancers. However, Sorafenib-induced multiple adverse effects are a major limitation that directly impacts patients' physical and physiological well-being. Therefore, it is vital to identify agents that can lessen the associated adverse effects and enhance efficacy. Apigenin, a dietary plant flavone, is a bioactive-compound present in fruits and vegetables having anti-oxidant, anti-inflammatory, and anti-cancer properties. Our study aimed to investigate Sorafenib-induced toxic effects at genomic, cellular, and tissue level and the potential protective effects of Apigenin. To achieve our goal, we treated Swiss albino mice with Apigenin (50 mg/kg bw) alone or in combination with Sorafenib (40 mg/kg bw). Next, we performed DNA interaction, genotoxicity, oxidative damages, anti-oxidant activities, liver enzyme levels, and histopathological studies. We demonstrated that Apigenin and Sorafenib bind DNA via electrostatic interaction. Further, Sorafenib induces genetic, oxidative, and tissue damages characterized by an increase in chromosomal aberrations and micronucleus, reactive oxygen species (ROS) and reactive nitrogen species (RNS), oxidative and DNA damage, lipid peroxidation, and hepato-renal damages, and a decrease in antioxidant-enzymes. Interestingly, the Sorafenib-induced adverse effects were ameliorated by Apigenin. Our findings indicate that Apigenin has protective effects against Sorafenib-induced toxicity and could be combined with Sorafenib to lessen its adverse effects and enhance its efficacy. However, further pre-clinical and clinical studies are required to evaluate Apigenin's effectiveness with Sorafenib.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Antineoplastic Agents/toxicity , Antioxidants/pharmacology , Apigenin/pharmacology , Apoptosis , Humans , Mice , Neoplasms/drug therapy , Oxidative Stress , Sorafenib/toxicity
3.
Molecules ; 27(21)2022 Nov 05.
Article in English | MEDLINE | ID: mdl-36364418

ABSTRACT

Graphene oxide-based nanodrug delivery systems are considered one of the most promising platforms to deliver therapeutic drugs at the target site. In this study, Ru(II)(ƞ6-p-cymene) complexes containing the benzothiazole ligand were covalently anchored on graphene oxide using the ultrasonication method. The nanoconjugates GO-NCD-1 and GO-NCD-2 were characterized by FT-IR, UV-visible, 1H NMR, TGA, SEM, and TEM techniques, which confirmed the successful loading of both the complexes (NCD 1 and NCD 2) on the carrier with average particle diameter sizes of 17 ± 6.9 nm and 25 ± 6.5 nm. In vitro DNA binding studies of the nanoconjugates were carried out by employing various biophysical methods to investigate the binding interaction with the therapeutic target biomolecule and to quantify the intrinsic binding constant values useful to understand their binding affinity. Our results suggest (i) high Kb and Ksv values of the graphene-loaded conjugates (ii) effective cleavage of plasmid DNA at a lower concentration of 7.5 µM and 10 µM via an oxidative pathway, and (iii) fast release of NCD 2 at an acidic pH that could have a good impact on the controlled delivery of drug. It was found that 90% of the drug was released in an acidic pH (5.8 pH) environment in 48 h, therefore suggesting pH-responsive behavior of the drug delivery system. Molecular docking, DFT studies, and cytotoxicity activity against three cancer cell lines by SRB assay were also performed.


Subject(s)
Antineoplastic Agents , Graphite , Noncommunicable Diseases , Humans , Graphite/chemistry , Nanoconjugates , Molecular Docking Simulation , Spectroscopy, Fourier Transform Infrared , Cell Line, Tumor , Antineoplastic Agents/chemistry , Drug Delivery Systems , DNA/chemistry , Hydrogen-Ion Concentration
4.
Bioorg Chem ; 104: 104327, 2020 11.
Article in English | MEDLINE | ID: mdl-33142405

ABSTRACT

Copper-based antitumor drug entities 1-3 derived from substituted (F-, Br-, -CH3) 3-formylchromone pharmacophore were synthesized and thoroughly characterized by spectroscopic and single X-ray crystallographic studies. These complexes show structural novelty due to presence of the X-bonds in chromone scaffold which could facilitate higher propensity for nucleic acids via sigma σ-hole interactions. Therefore, structure-activity relationship of 1-3 was studied by performing ct-DNA binding, pBR322 cleavage and cytotoxicity activity to validate their potential to act as chemotherapeutic drug entities. The binding studies of 1-3 with ct- DNA were carried out employing many biophysical techniques and the corroborative results of these experiments showed intercalation mode of binding and the order of binding was found to be 2 > 1 > 3. The structure of drug entities could facilitated strong halogen bonding interaction (in case of 1 &2) and stability of X bond was rationalized by sigma hole region of positive electrostatic potential on the surface of C-X covalent bond, as determined by gas phase B3LYP computational DFT studies. Interestingly, 2 exhibited most avid binding affinity due to presence of Br- electron withdrawing and polarizable group. Further, cleavage studies of 1-3 with pBR322 plasmid DNA were performed which demonstrated significant cleavage activity, the supercoiled form (Form I) of plasmid DNA was converted to nicked form (Form II) with the appearance of linearized form (Form III) in between two, implicating lethal double strand breaks of DNA. 2 showed predominantly higher cleavage activity following the similar trend as observed for binding studies. The cytotoxicity of the complexes 1-3 was evaluated by MTT assay against the human liver carcinoma (Huh-7) and prostate cancer (DU-145) cell lines; complex 2 exhibited specific and selective cytotoxicity for the DU-145 cancer cell line with LC50 value of 1.6 µM.


Subject(s)
Antineoplastic Agents/pharmacology , Chromones/pharmacology , Coordination Complexes/pharmacology , Copper/pharmacology , DNA/chemistry , Halogens/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chromones/chemistry , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Copper/chemistry , Crystallography, X-Ray , Density Functional Theory , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Halogens/chemistry , Humans , Models, Molecular , Molecular Structure , Particle Size , Structure-Activity Relationship , Surface Properties
5.
Bioorg Chem ; 94: 103427, 2020 01.
Article in English | MEDLINE | ID: mdl-31735357

ABSTRACT

New tailored copper(II)-based intercalating complexes [Cu(L1)2] (1) and [Cu(L2)2] (2) were synthesized from Schiff base scaffold HL1 and HL2(E)-4-(2-((2-hydroxy-3-methoxybenzylidene)amino)ethyl)benzenesulfonamide and (E)-4-(2-((2-hydroxybenzylidene)amino)ethyl)benzenesulfonamide, respectively. The structure elucidation of complexes 1 and 2 was carried out by employing various spectroscopic techniques viz., FT-IR, UV-vis, ESI-MS, EPR and single X-ray crystal diffraction studies. The complexes 1 and 2 were crystallized in monoclinic P21/n and triclinic P-1 space group, respectively possessing square planar geometry around Cu(II) coordinated with N,O-donor Schiff base ligands. An analysis of Hirshfeld surfaces of complexes 1 and 2 were performed to ascertain different intra and intermolecular non-covalent interactions (H-bonding, CH⋯ πetc.) responsible for the stabilization of crystal lattices. Calculations based on Density functional theory (B3LYP/DFT), have been carried out to obtain energies of Frontier molecular orbitals. Comparative in vitro binding profile of complexes 1 and 2 with ct-DNA was evaluated employing various biophysical techniques viz., UV-vis, fluorescence, circular dichroism and cyclic voltammetry which suggested non-covalent intercalative binding mode with more avid binding propensity of complex 1 compared to complex 2. The cleavage experiments of complex 1 was performed by gel electrophoretic assay which revealed efficient cleavage mediated via oxidative pathway. Furthermore, topoisomerase I enzymatic activity of complex 1 was carried out employing gel electrophoretic assay which demonstrated significant inhibitory effects at a low concentration of 25 µM. The cytotoxic potential of complex 1 was analyzed by SRB assay on a panel of selected human cancer cell lines which revealed selective activity for MCF-7 (breast cancer) cell line with GI50 = 16.21 µg/ml.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , DNA Topoisomerases, Type I/metabolism , DNA/drug effects , Density Functional Theory , Topoisomerase I Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Copper/chemistry , Copper/pharmacology , DNA Cleavage , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Structure , Schiff Bases/chemical synthesis , Schiff Bases/chemistry , Schiff Bases/pharmacology , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology , Topoisomerase I Inhibitors/chemical synthesis , Topoisomerase I Inhibitors/chemistry , Benzenesulfonamides
6.
Bioorg Chem ; 88: 102963, 2019 07.
Article in English | MEDLINE | ID: mdl-31071506

ABSTRACT

Cobalt (II) phen-based drug candidates of the formulation Co(phen)2Cl2,1, Co(phen)2L, 2 where L = 1H-pyrazole-3,5-dicarboxylic acid were synthesized and thoroughly characterized by spectroscopic methods and single X-ray crystallography. DNA binding interaction of 1 and 2 was carried out employing biophysical techniques {UV-visible, fluorescence, thermal denaturation and cyclic voltammetry} to validate their potential to act as antitumor agents. The interpretations of these biophysical studies of 1 and 2 supported the non-covalent intercalative binding mode; furthermore, a higher binding trend of 2 was observed as compared to 1, phen and 1H-pyrazole-3,5-dicarboxylic acid alone. Cleavage studies of 1 and 2 with pBR322 were assessed by gel electrophoresis and it was observed that both drug candidates cleave DNA by hydrolytic pathway involving hydroxyl radical (OH). Cytotoxic activity of 1 and 2 against human cancer cell lines [MCF-7 (breast), HeLa (cervical), MIA-PA-CA 2 (pancreatic), A-498 (kidney), Hep-G2 (hepatoma)] was evaluated by SRB assay. The obtained results showed that drug candidate 1 showed significantly low GI50 value (<10 µg/ml) against MCF-7 and HeLa cell lines. However, candidate 2 revealed excellent cytotoxicity (<10 µg/ml) against all the tested cancer cell lines. The in vivo genotoxicity of 2 was evaluated by micronucleus (MN) test and chromosomal aberration (CA) in bone marrow cells of the Wistar rats to check cobalt(II)-induced systemic toxicity. The results showed that no significant chromosomal aberrations and micronucleus formation was observed at 5 mg/kg and 10 mg/kg in presence of drug candidate 2 implicating that it could be administered safely at a low dosage. However, an elevated percentage of chromosomal aberration and micronucleated polychromatic erythrocytes (MNPCE) was observed only at higher doses (20 mg/kg and 40 mg/kg) of drug candidate 2.


Subject(s)
Antineoplastic Agents/pharmacology , Chromosome Aberrations/drug effects , Cobalt/pharmacology , Organometallic Compounds/pharmacology , Phenanthrenes/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cobalt/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Male , Molecular Docking Simulation , Molecular Structure , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Particle Size , Phenanthrenes/chemistry , Rats , Rats, Wistar , Structure-Activity Relationship , Surface Properties , Tumor Cells, Cultured
7.
Dalton Trans ; 53(24): 10126-10141, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38817206

ABSTRACT

Bilastine (BLA), 2-(4-(2-(4-(1-(2-ethoxyethyl)-1H-benzo[d]imidazole-2-yl)-piperidin-1-yl)-ethyl)-phenyl)-2-methylpropanoic acid, is an active antihistamine drug. With the idea of repurposing drugs from the existing pool of 'active' pharmaceutical ingredients, the therapeutic potency of bilastine as an anticancer agent was investigated via the tailored synthesis of a metal-based anticancer drug formulation of the type [BLA(phen)2M(II)]+·X-, where M = Co, Cu, and Zn and X- = NO3 and ClO4. The synthesized metal-based chemotherapeutics derived from the bilastine drug that acts as a ligand were thoroughly characterized using spectroscopic techniques, namely, UV-vis, FT-IR, and EPR (in the case of 1 and 2); 1H-NMR and 13C-NMR (in the case of 3); ESI-MS and single-crystal X-ray diffraction studies. Comprehensive biological studies (DNA binding, cleavage, and cytotoxic activity) using various biophysical and gel electrophoretic methods were carried out to validate their potential as anticancer agents. The cytotoxic activity of 'therapeutically promising' copper(II)-based drug candidate 2 was evaluated against MCF-7, MBA-MD-231, HeLa, HepG2, and Mia-PaCa-2 cancer cells via an SRB assay, and the results demonstrated 2 as a potent anticancer agent at low nanomolar concentrations against all tested cancer cells, preferably with a much superior anticancer efficacy against human pancreatic cancer cells.


Subject(s)
Antineoplastic Agents , Cobalt , Coordination Complexes , Copper , Drug Repositioning , Zinc , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Copper/chemistry , Copper/pharmacology , Zinc/chemistry , Zinc/pharmacology , Crystallography, X-Ray , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Cobalt/chemistry , Cobalt/pharmacology , Models, Molecular , Cell Line, Tumor , Cell Proliferation/drug effects , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Benzimidazoles/chemical synthesis , Drug Screening Assays, Antitumor , Molecular Structure , Piperidines/chemistry , Piperidines/pharmacology , Drug Resistance, Neoplasm/drug effects , Cell Survival/drug effects , Histamine Antagonists/chemistry , Histamine Antagonists/pharmacology , Histamine Antagonists/chemical synthesis
8.
Int J Biol Macromol ; 254(Pt 1): 127521, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37898256

ABSTRACT

New Quercetin-phenylalanine metal-based therapeutic agents of the formulation [Qu(Phe)M(II).(H2O)2].NO3 where M(II) = Co(II) and Ni(II) and [Qu(Phe)Cu(II).(H2O)2] were synthesized and their structure was predicted by IR, UV-vis, EPR and ESI-MS spectroscopic techniques. The bio-molecular interaction studies of the Quercetin-phenylalanine complexes, 1-3 with ct-DNA and BSA were performed using a battery of complimentary biophysical techniques. The corroborative results of these experiments revealed strong binding propensity via electrostatic interactions probably through minor grove binding towards ct-DNA, therapeutic target. The binding affinity of Quercetin-phenylalanine complexes 1-3 was quantified by determining binding constants values, Kb, Ksv, and the magnitude of binding propensity followed the order 3 > 1 > 2, implicating the preferential binding of Cu(II) complex 3 with ct-DNA. The cleavage studies were performed with complexes using gel electrophoretic mobility assay. The complexes 1-3 demonstrated efficient cleaving ability by the hydrolytic cleavage pathway involving hydroxyl (OH) radicals. BSA binding profile of Quercetin-phenylalanine metal therapeutics 1-3 was studied in order to understand the drug carrier potential of these compounds and found that complex 3 was capable of binding preferentially with BSA as compared to other complexes.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Quercetin/pharmacology , Quercetin/chemistry , Phenylalanine , DNA/chemistry , Metals , Coordination Complexes/chemistry , DNA Cleavage , Copper/chemistry , Antineoplastic Agents/chemistry , Serum Albumin, Bovine/chemistry
9.
Drug Discov Today ; 29(7): 104055, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38852835

ABSTRACT

Metal-based drugs hold promise as potent anticancer agents owing to their unique interactions with cellular targets. This review discusses recent advances in our understanding of the intricate molecular interactions of metal-based anticancer compounds with specific therapeutic targets in cancer cells. Advanced computational and experimental methodologies delineate the binding mechanisms, structural dynamics and functional outcomes of these interactions. In addition, the review sheds light on the precise modes of action of these drugs, their efficacy and the potential avenues for further optimization in cancer-treatment strategies and the development of targeted and effective metal-based therapies for combating various forms of cancer.


Subject(s)
Antineoplastic Agents , DNA , Neoplasms , RNA , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Metals/chemistry , Animals
10.
Biophys Chem ; 308: 107214, 2024 May.
Article in English | MEDLINE | ID: mdl-38428228

ABSTRACT

In the recent past, there has been an ever-increasing interest in the search for metal-based therapeutic drug candidates for protein misfolding disorders (PMDs) particularly neurodegenerative disorders such as Alzheimer's, Parkinson's, Prion's diseases, and amyotrophic lateral sclerosis. Also, different amyloidogenic variants of human lysozyme (HL) are involved in hereditary systemic amyloidosis. Metallo-therapeutic agents are extensively studied as antitumor agents, however, they are relatively unexplored for the treatment of non-neuropathic amyloidoses. In this work, inhibition potential of a novel ionic cobalt(II) therapeutic agent (CoTA) of the formulation [Co(phen)(H2O)4]+[glycinate]- is evaluated against HL fibrillation. Various biophysical techniques viz., dye-binding assays, dynamic light scattering (DLS), differential scanning calorimetry (DSC), electron microscopy, and molecular docking experiments validate the proposed mechanism of inhibition of HL fibrillation by CoTA. The experimental corroborative results of these studies reveal that CoTA can suppress and slow down HL fibrillation at physiological temperature and pH. DLS and 1-anilino-8-naphthalenesulfonate (ANS) assay show that reduced fibrillation in the presence of CoTA is marked by a significant decrease in the size and hydrophobicity of the aggregates. Fluorescence quenching and molecular docking results demonstrate that CoTA binds moderately to the aggregation-prone region of HL (Kb = 6.6 × 104 M-1), thereby, inhibiting HL fibrillation. In addition, far-UV CD and DSC show that binding of CoTA to HL does not cause any change in the stability of HL. More importantly, CoTA attenuates membrane damaging effects of HL aggregates against RBCs. This study identifies inorganic metal complexes as a therapeutic intervention for systemic amyloidosis.


Subject(s)
Amyloid , Amyloidosis , Humans , Amyloid/chemistry , Muramidase/chemistry , Molecular Docking Simulation , Amyloidosis/drug therapy , Amyloidosis/metabolism , Dynamic Light Scattering , Protein Aggregates
11.
Spectrochim Acta A Mol Biomol Spectrosc ; 303: 123152, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37467591

ABSTRACT

An oxo-bridged Sn (IV) Cluster, (TOC) was synthesized and fully characterized by FT-IR, UV-vis, 1H NMR, 119Sn NMR, Mass spectrometry and single crystal X-ray diffraction studies. The single-crystal X-ray analysis revealed that the crystal crystallizes in the monoclinic crystal system possessing the P 21/c space group and exhibited a distorted trigonal bipyramidal geometry. The TOC exhibited a unique turn-off fluorescence response for the selective detection of dopamine (DA) over other analytes. The stoichiometry between the TOC and DA was calculated using Job's plot. The value of the detection limit was found to be 1.33 µM. The Hirshfeld surface analysis was carried out on the crystal structure to investigate the H-H, Cl-H, Cl-Cl, Sn-Cl and Cl-C interaction studies in the molecule. Density Functional Theory (DFT) studies further supported the sensing mechanism, which closely agreed with the experimental results. Furthermore, the TOC chemosensor was used to detect DA in human blood plasma, and molecular docking studies validated the interaction between the chemosensor and protein. Confocal fluorescence imaging studies were carried out and validated TOC sensing ability for DA in human blood plasma.


Subject(s)
Dopamine , Tin , Humans , Tin/chemistry , Molecular Docking Simulation , Spectroscopy, Fourier Transform Infrared , Coloring Agents
12.
Dalton Trans ; 52(16): 5141-5154, 2023 Apr 25.
Article in English | MEDLINE | ID: mdl-36961247

ABSTRACT

Bio-compatible water-soluble conjugates of Co(II), Cu(II) and Zn(II) (1-3), [Co(Boc-L-valine)2(imidazole)2], [Cu(Boc-L-valine)2(imidazole)2], and [Zn(Boc-L-valine)2(imidazole)2], were synthesized and comprehensively characterized by various spectroscopic techniques (UV-visible, FT-IR, ESI-MS, EPR, 1H NMR, 13C NMR) and single crystal X-ray diffraction which showed that the complexes 1-3 crystallized in an orthorhombic crystal system, in a slightly distorted octahedral geometry having the space group P21212. Density functional theory calculations were performed to correlate the energy of frontier molecular orbitals with the stability and reactivity of the complexes. In vitro DNA binding interaction studies of complexes were performed by employing various biophysical techniques and their corroborative results revealed (i) the electrostatic mode of binding in the groove region of DNA, (ii) pBR322 plasmid cleavage at a low concentration of 5-12.5 µM via an oxidative pathway in complexes 1 and 2 and the hydrolytic mechanism in the case of 3, (iii) changes in the 1H NMR chemical shift values of the NH2 group of GMP after interaction with complex 3, (iv) alteration in the EPR parameters of complex 2 after complexation with DNA, (v) SOD mimetic activity of complex 2 with the IC50 value of 2.08 µM and (vi) a good and selective cytotoxicity profile against chemo-resistant MCF-7 and MDA-MB-231 cancer cell lines by complex 1. Molecular docking studies complemented the spectroscopic results and confirmed the electrostatic interaction of complexes in the groove region of DNA.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Coordination Complexes/chemistry , Molecular Docking Simulation , Spectroscopy, Fourier Transform Infrared , Water , Copper/chemistry , DNA/chemistry , Zinc/chemistry , Superoxide Dismutase/metabolism , Imidazoles , DNA Cleavage , Antineoplastic Agents/chemistry
13.
Pharmacol Ther ; 241: 108335, 2023 01.
Article in English | MEDLINE | ID: mdl-36567056

ABSTRACT

Metal-based complexes have occupied a pioneering niche in the treatment of many chronic diseases, including various types of cancers. Despite the phenomenal success of cisplatin for the treatment of many solid malignancies, a limited number of metallo-drugs are in clinical use against cancer chemotherapy till date. While many other prominent platinum and non­platinum- based metallo-drugs (e.g. NAMI-A, KP1019, carboplatin, oxaliplatin, titanocene dichloride, casiopeinas® etc) have entered clinical trials, many have failed at later stages of R&D due to deleterious toxic effects, intrinsic resistance and poor pharmacokinetic response and low therapeutic efficacy. Nonetheless, research in the area of medicinal inorganic chemistry has been increasing exponentially over the years, employing novel target based drug design strategies aimed at improving pharmacological outcomes and at the same time mitigating the side-effects of these drug entities. Over the last few decades, natural products became one of the key structural motifs in the anticancer drug development. Many eminent researchers in the area of medicinal chemistry are devoted to develop new 3d-transition metal-based anticancer drugs/repurpose the existing bioactive compounds derived from myriad pharmacophores such as coumarins, flavonoids, chromones, alkaloids etc. Metal complexes of natural alkaloids and their analogs such as luotonin A, jatrorrhizine, berberine, oxoaporphine, 8-oxychinoline etc. have gained prominence in the anticancer drug development process as the naturally occurring alkaloids can be anti-proliferative, induce apoptosis and exhibit inhibition of angiogenesis with better healing effect. While some of them are inhibitors of ERK signal-regulated kinases, others show activity based on cyclooxygenases-2 (COX-2) and telomerase inhibition. However, the targets of these alkaloid complexes are still unclear, though it is well-established that they demonstrate anticancer potency by interfering with multiple pathways of tumorigenesis and tumor progression both in vitro and in vivo. Over the last decade, many significant advances have been made towards the development of natural alkaloid-based metallo-drug therapeutics for intervention in cancer chemotherapy that have been summarized below and reviewed in this article.


Subject(s)
Alkaloids , Antineoplastic Agents , Coordination Complexes , Neoplasms , Humans , Antineoplastic Agents/adverse effects , Neoplasms/drug therapy , Neoplasms/pathology , Coordination Complexes/therapeutic use , Drug Delivery Systems , Alkaloids/pharmacology , Alkaloids/therapeutic use
14.
Int J Biol Macromol ; 240: 124429, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37062375

ABSTRACT

RNA-targeted drugs are considered as safe treatment option for the cure of many chronic diseases preventing off-targeted delivery and acute toxic manifestations. FDA has approved many such RNA therapies in different phases of clinical trials, validating their use for the treatment of various chronic diseases. We report herein, new water-soluble (µ-oxo) bridged polymeric Cu(II) complexes of taurine (2-aminoethane sulfonic acid) complexes 1 and 2. The therapeutic potency of 1 and 2 was ascertained by studying biophysical interactions with tRNA/ct-DNA. The experimental results demonstrated that the complexes interacted avidly to nucleic acids through intercalation mode depicting a specific preference for tRNA in comparison to ct-DNA and, moreover 2 showed higher binding propensity than 1. The electrophoretic behaviour of the complexes with plasmid pBR322 DNA and tRNA were examined by gel mobility assay that revealed a concentration-dependent activity with complex 2 performing more efficient cleavage as compared to complex 1. Cytotoxicity results on cancer cell strains displayed higher cytotoxicity than complex 1 against treated cancer cells. The synthesized copper(II) taurine complexes have met the basic criteria of anticancer drug design as they are structurally well-characterized, exhibiting good solubility in water, lipophilic in nature with superior intercalating propensity towards tRNA and cytotoxic in nature.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Neoplasms , Saccharomyces cerevisiae/metabolism , Antineoplastic Agents/chemistry , Copper/chemistry , RNA, Transfer , RNA , DNA/chemistry , Coordination Complexes/chemistry
15.
Spectrochim Acta A Mol Biomol Spectrosc ; 293: 122438, 2023 May 15.
Article in English | MEDLINE | ID: mdl-36758364

ABSTRACT

A new mixed-ligand Cu(II) complex formulated as [Cu(dipic)(amp)(H2O)].H2O (dipic: pyridine-2,6-dicarboxylic acid, amp: 2-amino-4-methylpyridine), was synthesized and structurally characterized by FTIR spectroscopy, CHN analysis, and the single-crystal X-ray crystallographic method. The complex crystallizes in an orthorhombic space group Pna21, and the coordination environment around the metal center was found to be a pentacoordinate CuN2O2OW distorted square-pyramidal geometry. In order to systematically explore a detailed in vitro and in silico study of the DNA binding of the title complex, various biophysical (UV-Vis absorption spectroscopy, fluorescence, competitive binding with ethidium bromide) and theoretical (DFT, molecular docking simulation, and QM/MM) methods were applied which revealed that the complex could intercalate with the insertion of the amp ligand between the DNA base pairs. The experimental thermodynamic parameters of the interaction revealed the spontaneity of the process and the domination of the hydrophobic interactions in the association and stabilization of the DNA-Cu(II) complex adduct, which was in line with the docking and QM/MM data. In vitro cytotoxic potential of the complex against the human breast adenocarcinoma (MCF-7) cells was examined using MTT assay, which indicated that cancerous cells showed inhibition in presence of the complex.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Humans , Intercalating Agents/chemistry , Molecular Docking Simulation , Ligands , Coordination Complexes/chemistry , Copper/chemistry , DNA/chemistry , Antineoplastic Agents/pharmacology
16.
Chirality ; 24(12): 977-86, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23001645

ABSTRACT

Novel chiral Schiff base ligands (R)/(S)-2-amino-3-(((1-hydroxypropan-2-yl)imino)methyl)-4H-chromen-4-one (L(1) and L(2)) derived from 2-amino-3-formylchromone and (R/S)-2-amino-1-propanol and their Cu(II)/Zn(II) complexes (R1, S1, R2, and S2) were synthesized. The complexes were characterized by elemental analysis, infrared (IR), hydrogen ((1) H) and carbon ((13)C) nuclear magnetic resonance (NMR), electrospray ionization-mass spectra (ESI-MS), and molar conductance measurements. The DNA binding studies of the complexes with calf thymus were carried out by employing different biophysical methods and molecular docking studies that revealed that complexes R1 and S1 prefers the guanine-cytosine-rich region, whereas R2 and prefers the adenine-thymine residues in the major groove of DNA. The relative trend in K(b) values followed the order R1>S1>R2>S2. This observation together with the findings of circular dichroic and fluorescence studies revealed maximal potential of (R)-enantiomeric form of complexes to bind DNA. Furthermore, the absorption studies with mononucleotides were also monitored to examine the base-specific interactions of the complexes that revealed a higher propensity of Cu(II) complexes for guanosine-5'-monophosphate disodium salt, whereas Zn(II) complexes preferentially bind to thymidine-5'-monophosphate disodium salt. The cleavage activity of R1 and R2 with pBR322 plasmid DNA was examined by gel electrophoresis that revealed that they are good DNA cleavage agents; nevertheless, R1 proved to show better DNA cleavage ability. Topoisomerase II inhibitory activity of complex R1 revealed that the complex inhibits topoisomerase II catalytic activity at a very low concentration (25 µM). Furthermore, in vitro antitumor activity of complexes R1 and S1 were screened against human carcinoma cell lines of different histological origin.


Subject(s)
Chromones/chemistry , Copper/chemistry , DNA/metabolism , Organometallic Compounds/chemistry , Organometallic Compounds/metabolism , Zinc/chemistry , Absorption , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Binding Sites , Cell Line, Tumor , DNA/chemistry , DNA Topoisomerases, Type II/metabolism , Drug Design , Goiter, Substernal , Guanosine Monophosphate/metabolism , Humans , Models, Molecular , Nucleic Acid Conformation , Organometallic Compounds/chemical synthesis , Organometallic Compounds/pharmacology , Stereoisomerism , Substrate Specificity , Thymidine Monophosphate/metabolism , Viscosity
17.
Dalton Trans ; 51(31): 11713-11729, 2022 Aug 09.
Article in English | MEDLINE | ID: mdl-35852297

ABSTRACT

To validate the effect of metal ions in analogous ligand scaffolds on DNA binding and cytotoxic response, we have synthesized a series of water-soluble ionic N-phthaloylglycinate conjugated bis(diaminocyclohexane)M2+ complexes where M = Ni(II), Cu(II) and Zn(II) (1-3). The structural characterization of the complexes (1-3) was achieved by spectroscopic {FT-IR, EPR, UV-vis absorption data, 1H NMR, ESI-MS and elemental analysis} and single crystal X-ray diffraction studies, which revealed different topologies for the late 3d-transition metals. The Ni(II) and Zn(II) complexes exhibited an octahedral geometry with coordinated labile water molecules in the P1̄ space group while the Cu(II) complex revealed a square planar geometry with the P21/c space lattice. In vitro DNA-complexation studies were performed employing various complementary biophysical methods to quantify the intrinsic binding constant Kb and Ksv values and to envisage the binding modes and binding affinity of (1-3) at the therapeutic targets. The corroborative results of these experiments revealed a substantial geometric and electronic effect of (1-3) on DNA binding and the following inferences were observed, (i) high Kb and Ksv values, (ii) remarkable cleavage efficiency via an oxidative pathway, (iii) condensation behavior and (iv) good cytotoxic response to HepG2 and PTEN-caP8 cancer cell lines, with copper(II) complex 2 outperforming the other two complexes as a most promising anticancer drug candidate. Copper(II) complexes have been proven in the literature to be good anticancer drug entities, displaying inhibition of uncontrolled-cell growth by multiple pathways viz., anti-angiogenesis, inducing apoptosis and reactive oxygen species mediated cell death phenomena. Nickel(II) and zinc(II) ionic complexes 1 and 3 have also demonstrated good chemotherapeutic potential in vitro and the bioactive 1,2-diaminocyclohexane fragment in these complexes plays an instrumental role in anticancer activity.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Copper/chemistry , Copper/pharmacology , Cyclohexylamines , DNA/chemistry , DNA Cleavage , Ions , Ligands , Spectroscopy, Fourier Transform Infrared , Water , Zinc/chemistry
18.
J Biomol Struct Dyn ; 40(16): 7381-7393, 2022 10.
Article in English | MEDLINE | ID: mdl-33685362

ABSTRACT

In the present work, we report the synthesis, characterization of two cobalt complexes (1 and 2) and their HSA binding studies by multispectroscopic methods. Hirshfeld surfaces analysis and fingerprint plot analysis were carried out to identify intermolecular interactions viz., N-H···O, O-H···O and C-H···O linkages in crystal framework of the complexes. Density functional theory (DFT) studies were carried out to ascertain the electronic structure and molecular geometry of the complexes 1 and 2, and determine the localization of HOMO and LUMO in the complexes. A comparative in vitro interaction study of complex 1 and 2 with human serum albumin protein was carried out by employing UV-vis, fluorescence, circular dichroism, FTIR and molecular docking techniques. Interestingly, the HSA binding affinity of complex 2 was found to be more than complex 1 which was evidenced from the higher binding constant values owing to its strong hydrophobic topology. Further, a significant conformational change in microenvironment of HSA was noticed upon binding with complexes 1 and 2, nevertheless more perturbations were noticed in presence of complex 1. Molecular docking studies were carried out to validate the spectroscopic results and ascertain the preferential binding mode of complexes at the specific target site of HSA.Communicated by Ramaswamy H. Sarma.


Subject(s)
Cobalt , Binding Sites , Circular Dichroism , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Docking Simulation , Protein Binding , Spectrometry, Fluorescence/methods , Thermodynamics
19.
J Biomol Struct Dyn ; 40(10): 4558-4569, 2022 07.
Article in English | MEDLINE | ID: mdl-33331234

ABSTRACT

In an attempt to identify suitable nano-carriers for drug delivery, natural drug umbelliferone was chosen to synthesize new modulated nanoconjugate of umbelliferone cobalt oxide with cobalt (II) nitrate in one pot assembly in the presence of tannic acid. The synthesized nanoconjugate drug (NCD) was then loaded on graphene oxide (GO) as drug carrier by simple ultrasonication method and thoroughly characterized by various spectroscopic techniques (FT-IR, SEM, TEM, XRD, EPR and thermogravimetric analysis) which revealed the successful loading of the nanoconjugate drug on GO. The UV-visible, fluorescence and electrochemical studies suggested that strong π-π stacking interactions exist between nanoconjugate drug and GO. The binding studies of NCD-GO with ct-DNA were performed by various optical and biophysical methods viz., UV-visible, fluorescence, circular dichroism (CD) and cyclic voltammetry (CV) which indicated electrostatic mode of binding towards the ct-DNA. Furthermore, condensate of nanoconjugate drug-loaded GO (NCD-GO) with ct-DNA was prepared and analyzed by scanning electron microscopy (SEM) which revealed that the interaction of NCD-GO with ct-DNA had occurred. Cleavage activity of NCD-GO with pBR322 was evaluated by gel electrophoresis and it was found that NCD-GO cleave DNA through hydrolytic pathway involving hydroxyl radical (OH). The cytotoxicity of NCD-GO was evaluated against human liver carcinoma (Huh-7), prostate cancer (Du-145) cell lines along with normal cell line (PNT 2). The results obtained showed selective cytotoxic activity of NCD-GO against Du-145 cell lines. The intracellular uptake was visualized by confocal microscopy which revealed the significant cellular uptake and internalization of nanoparticles by cells. Moreover, the adsorption of cobalt oxide umbelliferone on GO was studied by density functional theory. The process of adsorption was found exothermic in nature and the optimized geometry structure is quite stable. Communicated by Ramaswamy H. Sarma.


Subject(s)
Antineoplastic Agents , Drug Carriers , Graphite , Noncommunicable Diseases , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cobalt/chemistry , DNA/chemistry , Graphite/chemistry , Humans , Male , Nanoconjugates , Oxides , Spectroscopy, Fourier Transform Infrared , Umbelliferones/pharmacology
20.
Chirality ; 23(3): 250-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20928895

ABSTRACT

A new chiral series of potential antitumor metal-based complexes 1-3(a and b) of L- and D-tryptophan have been synthesized and thoroughly characterized. Both enantiomers of 1-3 bind DNA noncovalently via phosphate interaction with slight preference of metal center for covalent coordination to nucleobases. The K(b) values of L-enantiomer, however, possess higher propensity for DNA binding in comparison with the D-enantiomeric analogs. The relative trend in K(b) values is as follows: 2(a) > 2(b) > 3(a) > 1(a) > 3(b) > 1(b). These observations together with the findings of circular dichoric and fluorescence studies reveal maximal potential of L-enantiomeric form of copper complex to bind DNA, thereby exerting its therapeutic effect. The complex 2a exhibits a remarkable DNA cleavage activity with pBR322DNA in the presence of different activators such as H(2) O(2) , ascorbic acid, 3-mercaptopropionic acid, and glutathione, suggesting the involvement of active oxygen species for the DNA scission. In vitro anticancer activity of complexes 1-3(a) were screened against 14 different human carcinoma cell lines of different histological origin, and the results reveal that 2a shows significant antitumor activity in comparison with both 1a and 3a and is particularly selective for MIAPACA2 (pancreatic cancer cell line).


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Copper/chemistry , Cyclohexylamines/chemistry , DNA Cleavage/drug effects , DNA/chemistry , Organomercury Compounds/chemistry , Tryptophan/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Circular Dichroism/methods , Coordination Complexes/analysis , DNA/metabolism , Electron Spin Resonance Spectroscopy/methods , Guanosine Monophosphate/chemistry , Humans , Magnetic Resonance Spectroscopy/methods , Molecular Structure , Reactive Oxygen Species/chemistry , Spectrophotometry, Infrared/methods , Stereoisomerism , Thymidine Monophosphate/chemistry , Tryptophan/analysis , X-Ray Diffraction/methods
SELECTION OF CITATIONS
SEARCH DETAIL