Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.906
Filter
1.
Int J Mol Sci ; 25(17)2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39273451

ABSTRACT

Interest and research focusing on the design of novel pharmaceutical agents is always growing [...].


Subject(s)
Drug Discovery , Heterocyclic Compounds , Drug Discovery/methods , Humans , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/therapeutic use
2.
Inorg Chem ; 63(37): 16949-16963, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39226133

ABSTRACT

A series of new gold(I) and silver(I) N-heterocyclic carbenes bearing a 1-thio-ß-d-glucose tetraacetate moiety was synthesized and chemically characterized. The compounds' stability and solubility in physiological conditions were investigated employing a multitechnique approach. Interaction studies with biologically relevant proteins, such as superoxide dismutase (SOD) and human serum albumin (HSA), were conducted via UV-vis absorption spectroscopy and high-resolution ESI mass spectrometry. The biological activity of the compounds was evaluated in the A2780 and A2780R (cisplatin-resistant) ovarian cancer cell lines and the HSkMC (human skeletal muscle) healthy cell line. Inhibition studies of the selenoenzyme thioredoxin reductase (TrxR) were also carried out. The results highlighted that the gold complexes are more stable in aqueous environment and capable of interaction with SOD and HSA. Moreover, these carbenes strongly inhibited the TrxR activity. In contrast, the silver ones underwent structural alterations in the aqueous medium and showed greater antiproliferative activity.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Drug Screening Assays, Antitumor , Gold , Heterocyclic Compounds , Methane , Silver , Thioredoxin-Disulfide Reductase , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Silver/chemistry , Silver/pharmacology , Gold/chemistry , Gold/pharmacology , Methane/analogs & derivatives , Methane/chemistry , Methane/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemical synthesis , Cell Proliferation/drug effects , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism , Hydrophobic and Hydrophilic Interactions , Molecular Structure , Structure-Activity Relationship , Cell Line, Tumor , Superoxide Dismutase/metabolism , Superoxide Dismutase/antagonists & inhibitors
3.
Bioorg Med Chem ; 112: 117901, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39232465

ABSTRACT

Multidrug-resistant (MDR) bacterial infections are becoming a life-threatening issue in public health; therefore, it is urgent to develop novel antibacterial agents for treating infections caused by MDR bacteria. The 20(S)-protopanaxadiol (PPD) derivative 9 was identified as a novel antibacterial hit compound in screening of our small synthetic natural product-like (NPL) library. A series of novel PPD derivatives with heterocyclic rings fused at the C-2 and C-3 positions of the A-ring were synthesized and their antibacterial activities against Staphylococcus aureus (S. aureus) Newman strain and MDR S. aureus strains (USA300, NRS-1, NRS-70, NRS-100, NRS-108, NRS-271, XJ017, and XJ036) were evaluated. Among these compounds, quinoxaline derivative 56 (SH617) exhibited the highest activity with MICs of 0.5-4 µg/mL against the S. aureus Newman strain and the eight MDR S. aureus strains. Its antibacterial activity was comparable to that of the positive control, vancomycin. In the zebrafish, 56 revealed no obvious toxicity even at a high administered dose. In vivo, following a lethal infection induced by USA300 strains in zebrafish, 56 exhibited significantly increased survival rates in a dose-dependent manner.


Subject(s)
Anti-Bacterial Agents , Microbial Sensitivity Tests , Sapogenins , Staphylococcus aureus , Zebrafish , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Sapogenins/pharmacology , Sapogenins/chemistry , Sapogenins/chemical synthesis , Staphylococcus aureus/drug effects , Animals , Structure-Activity Relationship , Molecular Structure , Dose-Response Relationship, Drug , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/chemical synthesis
4.
Int J Nanomedicine ; 19: 9213-9226, 2024.
Article in English | MEDLINE | ID: mdl-39263631

ABSTRACT

Introduction: Targeting, imaging, and treating tumors represent major clinical challenges. Developing effective theranostic agents to address these issues is an urgent need. Methods: We introduce an "all-in-one" tumor-targeted theranostic platform using CuFeSe2-based composite nanoparticles (CuFeSe2@PA) for magnetic resonance (MR) and computed tomography (CT) dual model imaging-guided hyperthermia tumor ablation. Plerixafor (AMD3100) is bonded to the surface of CuFeSe2 as a targeting unit. Due to the robust interaction between AMD3100 and the overexpressed Chemokine CXC type receptor 4 (CXCR4) on the membrane of 4T1 cancer cells, CuFeSe2@PA specifically recognizes 4T1 cancer cells, enriching the tumor region. Results: CuFeSe2@PA serves as a contrast agent for T2-weighted MR imaging (relaxivity value of 1.61 mM-1 s-1) and CT imaging. Moreover, it effectively suppresses tumor growth through photothermal therapy (PTT) owing to its high photothermal conversion capability and stability, with minimized side effects demonstrated both in vitro and in vivo. Discussion: CuFeSe2@PA nanoparticles show potential as dual-mode imaging contrast agents for MR and CT and provide an effective means of tumor treatment through photothermal therapy. The surface modification with Plerixafor enhances the targeting ability of the nanoparticles, performing more significant efficacy and biocompatibility in the 4T1 cancer cell model. The study demonstrates that CuFeSe2@PA is a promising multifunctional theranostic platform with clinical application potential.


Subject(s)
Copper , Magnetic Resonance Imaging , Photothermal Therapy , Receptors, CXCR4 , Theranostic Nanomedicine , Tomography, X-Ray Computed , Animals , Receptors, CXCR4/metabolism , Theranostic Nanomedicine/methods , Photothermal Therapy/methods , Cell Line, Tumor , Magnetic Resonance Imaging/methods , Mice , Copper/chemistry , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Mice, Inbred BALB C , Female , Humans , Contrast Media/chemistry , Nanoparticles/chemistry , Cyclams/pharmacology , Cyclams/chemistry , Benzylamines/chemistry
5.
Bull Exp Biol Med ; 177(3): 307-312, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39123088

ABSTRACT

We studied the effects of some nitrogen-containing, heterocyclic, and cyclic compounds on the rate of oxidative deamination of polyamines and putrescine in tissues with a high proliferation rate. For this purpose, the specific activities of the main enzymes of polyamine oxidative degradation - spermine oxidase (SMO), polyamine oxidase (PAO), and diamine oxidase (DAO) were determined using a cell-free test system from regenerating rat liver. The compounds methyl 2-(5-formylfuran-2-yl)benzoate and 2,7-bis-[2-(diethylamino)ethoxy]-9H-fluoren-9-one (and in the form of dihydrochloride) showed mainly activating effect on oxidative degradation of putrescine, spermidine, and spermine, which indirectly indicates their antiproliferative effect. Nitrogen-free compounds inhibited this process, thus exhibiting potentially carcinogenic properties. Correlations were calculated for activity of DAO, PAO, and SMO with 5 topological indices: Wiener (W), Rouvray (R), Balaban (J) in the Trinaistich modification, detour (Ip), and electropy (Ie). The highest dependence was noted for DAO and the Balaban index (R=-0.55), for PAO and the detour index (R=0.78), and for SMO and the electropy index (R=0.53). The remaining dependencies showed insignificant correlation strength.


Subject(s)
Amine Oxidase (Copper-Containing) , Oxidation-Reduction , Oxidoreductases Acting on CH-NH Group Donors , Animals , Rats , Oxidation-Reduction/drug effects , Deamination , Amine Oxidase (Copper-Containing)/metabolism , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Polyamine Oxidase , Putrescine/metabolism , Putrescine/pharmacology , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemistry , Cell-Free System , Liver/metabolism , Liver/drug effects , Polyamines/metabolism , Spermine/metabolism , Spermine/pharmacology , Spermidine/metabolism , Male , Nitrogen/metabolism , Rats, Wistar
6.
Food Chem ; 460(Pt 3): 140576, 2024 Dec 01.
Article in English | MEDLINE | ID: mdl-39106755

ABSTRACT

The inhibition of amino acids on the formation of protein-bound HAs was assessed in both model systems and roast beef patties, and the synergism between these amino acids was also investigated. The amino acids can promote the formation of protein-bound HAs at low addition amount, and the total content of protein-bound HAs increased from 444.05 ± 4.98 ng/g of the control group to 517.36 ± 16.51 ng/g when 0.05 % cysteine was added. Amino acid combinations exhibited stable inhibitory effects, with the maximum inhibitory rate of 64 % in the treatment with histidine-proline combination (1:4). The synergistic inhibition may be caused by simultaneously scavenging intermediates and competing for the binding sites of muscle proteins, and the reaction with protein-bound HAs to form adduct can serve as supporting factors to co-mitigate the promotion in protein-bound HAs from increased protein solubility. These findings proposed the potential mitigation strategies against protein-bound HAs formation.


Subject(s)
Amines , Amino Acids , Animals , Cattle , Amino Acids/chemistry , Amino Acids/metabolism , Amines/chemistry , Amines/metabolism , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Myofibrils/chemistry , Myofibrils/metabolism , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/chemistry
7.
Bioorg Chem ; 151: 107686, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39111120

ABSTRACT

A series of novel quinazoline-derived EGFR/HER-2 dual-target inhibitors were designed and synthesized by heterocyclic-containing tail approach. The inhibitory activities against four human epidermal growth factor receptor (HER) isozymes (EGFR, HER-2, HER-3 and HER-4) of all new compounds so designed were investigated in vitro. Compound 12k was found to be the most effective and rather selective dual-target inhibitor of EGFR and HER-2 with inhibitory constant (IC50) values of 6.15 and 9.78 nM, respectively, which was more potent than the clinical used agent Lapatinib (IC50 = 8.41 and 9.41 nM). Meanwhile, almost all compounds showed excellent antiproliferative activities against four cancer cell models (A549, NCI-H1975, SK-BR-3 and MCF-7) and low damage to healthy cells. Among them, compound 12k also exhibited the most prominent antitumor activity. Moreover, the hit compound 12k could bind to EGFR and HER-2 stably in molecular docking and dynamics studies. The following wound healing assay revealed that compound 12k could inhibit the migration of SK-BR-3 cells. Further studies found that compound 12k could arrest cell cycle in the G0/G1 phase and induce SK-BR-3 cells apoptosis. Notably, compound 12k could effectively inhibit breast cancer growth with little toxicity in the SK-BR-3 cell xenograft model. Taken together, in vitro and in vivo results disclosed that compound 12k had high drug potential as a dual-target inhibitor of EGFR/HER-2 to inhibit breast cancer growth.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , ErbB Receptors , Protein Kinase Inhibitors , Quinazolines , Receptor, ErbB-2 , Humans , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Quinazolines/pharmacology , Quinazolines/chemistry , Quinazolines/chemical synthesis , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Cell Proliferation/drug effects , Structure-Activity Relationship , Molecular Structure , Animals , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Mice , Cell Line, Tumor , Molecular Docking Simulation , Apoptosis/drug effects , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/chemical synthesis , Female
8.
Eur J Med Chem ; 277: 116746, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39146831

ABSTRACT

Metal complexes showing dual activity against cancer and bacterial infections are currently the focus of significant interest for their potential in treating life-threatening diseases. Aiming to investigate the impact of ligand substituents on these bioactivity properties of Group 11 d10 metal complexes, we herein present a series of mononuclear Cu(I) and Ag(I) complexes featuring the bis-NH2-substituted heterocyclic thioamide dap2SH (=4,6-diaminopyrimidine-2-thione), namely [AgCl(dap2SH)(PPh3)2] (1), [CuBr(dap2SH)(PPh3)2] (2), [CuBr(dap2SH)(xantphos)] (3), [Ag(dap2S)(xantphos)] (4), and [Cu(dap2S)(xantphos)] (5) (xantphos = 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene). Complexes were characterized by means of different physicochemical methods (i.e., single crystal X-ray diffraction as well as FTIR, NMR, UV-Vis and fluorescence spectroscopy), and studied in-vitro for their antibacterial and anticancer activity against a variety of bacterial strains and cancer cell lines. Complexes 1-3 effectively inhibited both Gram (+) and Gram (-) bacterial growth, while cellular uptake studies for the most potent complex 1 against E. coli bacteria revealed the accumulation of Ag(I) ions in the periplasm of the bacteria. A high anti-proliferative effect was observed for 1 and 5 against A549, MCF7 and PC3 cancer cell lines, with 1 being capable of inducing apoptosis in A549 cells, as suggested by flow cytometry analysis. DNA interaction studies revealed the capacity of 1 to intercalate between base-pairs of CT DNA. All complexes had a moderate-to-high capacity to scavenge free radicals preventing oxidative stress. Molecular docking calculations, in combination with the experimentally obtained data, provided insights for potential mechanisms of the bioactivity of the complexes.


Subject(s)
Anti-Bacterial Agents , Antineoplastic Agents , Coordination Complexes , Copper , Drug Screening Assays, Antitumor , Escherichia coli , Microbial Sensitivity Tests , Silver , Thioamides , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Escherichia coli/drug effects , Humans , Copper/chemistry , Copper/pharmacology , Thioamides/chemistry , Thioamides/pharmacology , Thioamides/chemical synthesis , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Silver/chemistry , Silver/pharmacology , Molecular Structure , Structure-Activity Relationship , Amines/chemistry , Amines/pharmacology , Amines/chemical synthesis , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Cell Line, Tumor , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemical synthesis
9.
Bioorg Chem ; 152: 107696, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39167870

ABSTRACT

The JAK-STAT signalling pathway is primarily involved in cytokine signalling and induces various factors namely, erythropoietin, thrombopoietin, interferons, interleukins, and granulocyte colony-stimulating factors. These factors tremendously influenced understanding human health and illness, specifically cancer. Inhibiting the JAK/STAT pathway offers enormous therapeutic promises against cancer. Many JAK inhibitors are now being studied due to their efficacy in various cancer treatments. Further, the Nitrogen-heterocyclic (N-heterocyclic) scaffold has always shown to be a powerful tool for designing and discovering synthetic compounds with diverse pharmacological characteristics. The review focuses on several FDA-approved JAK inhibitors and their systematic categorization. The medicinal chemistry perspective is highlighted and classified review on the basis of N-heterocyclic molecules. Several examples of designing strategies of N-heterocyclic rings including pyrrolo-azepine, purine, 1H-pyrazolo[3,4-d]pyrimidine, 1H-pyrrolo[2,3-b]pyridine, pyrazole, thieno[3,2-d] pyrimidine, and, pyrimidine-based derivatives and their structure-activity relationships (SAR) are discussed. Among the various N-heterocyclic-based JAK inhibitors pyrimidine-containing compound 1 exhibited excellent inhibition activity against JAK2WT and mutated-JAK2V617F with IC50 of 2.01 and 18.84 nM respectively. Amino pyrimidine-containing compound 6 and thiopheno[3,2-d]pyrimidine-containing compound 13 expressed admirable JAK3 inhibition activity with IC50 of 1.7 nM and 1.38 nM respectively. Our review will support the medicinal chemists in refining and directing the development of novel N-heterocyclic-based JAK inhibitors.


Subject(s)
Antineoplastic Agents , Heterocyclic Compounds , Janus Kinase Inhibitors , Animals , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemical synthesis , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/chemical synthesis , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Molecular Structure , Neoplasms/drug therapy , Nitrogen/chemistry , Structure-Activity Relationship , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology
10.
Eur J Med Chem ; 277: 116757, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39142149

ABSTRACT

N-heterocyclic carbenes (NHCs) represent suitable ligands for rapid and efficient drug design, because they offer the advantage of being easily chemically modified and can bind several substituents, including transition metals as, for instance, gold derivatives. Gold-NHC complexes possess various biological activities and were demonstrated good candidates as anticancer drugs. Besides, carbazole derivatives are characterized by various pharmacological properties, such as anticancer, antibacterial, anti-inflammatory, and anti-psychotropic. Amongst the latter, N-thioalkyl carbazoles were proved to inhibit cancer cells damaging the nuclear DNA, through the inhibition of human topoisomerases. Herein, we report the design, synthesis and biological evaluation of nine new hybrid molecules in which NHC-Au(I) complexes and N-alkylthiolated carbazoles are linked together, in order to obtain novel biological multitarget agents. We demonstrated that the lead hybrid complexes possess anticancer, anti-inflammatory and antioxidant properties, with a high potential as useful tools for treating distinct aspects of several diseases, amongst them cancer.


Subject(s)
Antineoplastic Agents , Carbazoles , Drug Design , Heterocyclic Compounds , Methane , Carbazoles/chemistry , Carbazoles/pharmacology , Carbazoles/chemical synthesis , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemical synthesis , Structure-Activity Relationship , Methane/analogs & derivatives , Methane/chemistry , Methane/pharmacology , Molecular Structure , Gold/chemistry , Gold/pharmacology , Drug Screening Assays, Antitumor , Cell Line, Tumor , Cell Proliferation/drug effects , Antioxidants/pharmacology , Antioxidants/chemistry , Antioxidants/chemical synthesis , Animals , Dose-Response Relationship, Drug , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Mice , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/chemical synthesis
11.
Int J Mol Sci ; 25(15)2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39125838

ABSTRACT

Climate change forces agriculture to face the rapidly growing virulence of biotrophic fungal pathogens, which in turn drives researchers to seek new ways of combatting or limiting the spread of diseases caused by the same. While the use of agrochemicals may be the most efficient strategy in this context, it is important to ensure that such chemicals are safe for the natural environment. Heterocyclic compounds have enormous biological potential. A series of heterocyclic scaffolds (1,3,4-thiadiazole, 1,3-thiazole, 1,2,4-triazole, benzothiazine, benzothiadiazine, and quinazoline) containing 2,4-dihydroxylaryl substituents were investigated for their ability to inhibit the growth and development of biotrophic fungal pathogens associated with several important cereal diseases. Of the 33 analysed compounds, 3 were identified as having high inhibitory potential against Blumeria and Puccinia fungi. The conducted research indicated that the analysed compounds can be used to reduce the incidence of fungal diseases in cereals; however, further thorough research is required to investigate their effects on plant-pathogen systems, including molecular studies to determine the exact mechanism of their activity.


Subject(s)
Edible Grain , Heterocyclic Compounds , Plant Diseases , Plant Diseases/microbiology , Plant Diseases/prevention & control , Edible Grain/microbiology , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemistry , Fungi/drug effects , Fungicides, Industrial/pharmacology , Fungicides, Industrial/chemistry , Thiadiazoles/pharmacology , Thiadiazoles/chemistry
12.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39126093

ABSTRACT

The history of effective anti-cancer medications begins with the discovery of cisplatin's anti-cancer properties. Second-generation analogue, carboplatin, with a similar range of effectiveness, made progress in improving these drugs with fewer side effects and better solubility. Renewed interest in platinum-based drugs has been increasing in the past several years. These developments highlight a revitalized enthusiasm and ongoing exploration in platinum chemotherapy based on the series of dinuclear platinum(II) complexes, [{Pt(L)Cl}2(µ-bridging ligand)]2+, which have been synthesized and evaluated for their biological activities. These complexes are designed to target various cancerous conditions, exhibiting promising antitumor, antiproliferative, and apoptosis-inducing activities. The current work aims to shed light on the potential of these complexes as next-generation platinum-based therapies, highlighting their enhanced efficacy and reduced side effects, which could revolutionize the approach to chemotherapy.


Subject(s)
Antineoplastic Agents , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Ligands , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/chemical synthesis , Apoptosis/drug effects , Platinum/chemistry , Platinum/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemical synthesis , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Neoplasms/drug therapy , Neoplasms/pathology
13.
J Med Chem ; 67(16): 14414-14431, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39119630

ABSTRACT

In this study, we synthesized novel Pd(II)-indenyl complexes using various N-heterocyclic carbene (NHC) ligands, including chelating NHC-picolyl, NHC-thioether, and diNHC ligands, and two monodentate NHCs. Transmetalation reactions between a Pd(II)-indenyl precursor and silver-NHC complexes were generally employed, except for chelating diNHC derivatives, which required direct reaction with bisimidazolium salts and potassium carbonate. Characterization included NMR, HRMS analysis, and single-crystal X-ray diffraction. In vitro on five ovarian cancer cell lines showed notable cytotoxicity, with IC50 values in the micro- and submicromolar range. Some compounds exhibited intriguing selectivity for cancer cells due to higher tumor cell uptake. Mechanistic studies revealed that monodentate NHCs induced mitochondrial damage while chelating ligands caused DNA damage. One chelating NHC-picolyl ligand showed promising cytotoxicity and selectivity in high-grade serous ovarian cancer models, supporting its consideration for preclinical study.


Subject(s)
Antineoplastic Agents , Heterocyclic Compounds , Methane , Ovarian Neoplasms , Palladium , Humans , Female , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Palladium/chemistry , Methane/analogs & derivatives , Methane/chemistry , Methane/pharmacology , Ligands , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Indenes/chemistry , Indenes/pharmacology , Indenes/chemical synthesis , Drug Screening Assays, Antitumor , Structure-Activity Relationship
14.
J Med Chem ; 67(17): 15494-15508, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39196554

ABSTRACT

From previous studies, it is evident that metal-organic gold(I) complexes have antiproliferative activities. The aim of this study is not only to find new anticancer agents but also to overcome existing cytostatic resistance in cancer cells. The synthesis and medicinal evaluation of two cationic 1,3-disubstituted gold(I) bis-tetrazolylidene complexes 1 and 2 are reported. To determine apoptosis-inducing properties of the complexes, DNA fragmentation was measured using propidium iodide staining followed by flow cytometry. Gold(I) complex 1 targets explicitly malignant cells, effectively inhibiting their growth and selectively inducing apoptosis without signs of necrosis. Even in cells resistant to common treatments such as doxorubicin, it overcomes multidrug resistance and sensitizes existing drug-resistant cells to common cytostatic drugs. It is assumed that gold(I) complex 1 involves the mitochondrial pathway in apoptosis and targets members of the BCL-2 family, enhancing its potential as a therapeutic agent in cancer treatment.


Subject(s)
Antineoplastic Agents , Apoptosis , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Gold , Proto-Oncogene Proteins c-bcl-2 , Humans , Apoptosis/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Drug Resistance, Neoplasm/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Drug Resistance, Multiple/drug effects , Gold/chemistry , Gold/pharmacology , Cell Line, Tumor , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/chemical synthesis , Leukemia/drug therapy , Leukemia/pathology , Leukemia/metabolism , Methane/analogs & derivatives , Methane/pharmacology , Methane/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Structure-Activity Relationship , Cell Proliferation/drug effects
15.
J Med Chem ; 67(16): 13737-13764, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39169825

ABSTRACT

Since the largest and most fatal Ebola virus epidemic during 2014-2016, there have been several consecutive filoviral outbreaks in recent years, including those in 2021, 2022, and 2023. Ongoing outbreak prevalence and limited FDA-approved filoviral therapeutics emphasize the need for novel small molecule treatments. Here, we showcase the structure-activity relationship development of N-substituted pyrrole-based heterocycles and their potent, submicromolar entry inhibition against diverse filoviruses in a target-based pseudovirus assay. Inhibitor antiviral activity was validated using replication-competent Ebola, Sudan, and Marburg viruses. Mutational analysis was used to map the targeted region within the Ebola virus glycoprotein. Antiviral counter-screen and phospholipidosis assays were performed to demonstrate the reduced off-target activity of these filoviral entry inhibitors. Favorable antiviral potency, selectivity, and drug-like properties of the N-substituted pyrrole-based heterocycles support their potential as broad-spectrum antifiloviral treatments.


Subject(s)
Antiviral Agents , Ebolavirus , Pyrroles , Virus Internalization , Pyrroles/pharmacology , Pyrroles/chemistry , Pyrroles/chemical synthesis , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/chemical synthesis , Humans , Structure-Activity Relationship , Ebolavirus/drug effects , Virus Internalization/drug effects , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/chemical synthesis , Filoviridae/drug effects , Marburgvirus/drug effects
16.
Biochem Biophys Res Commun ; 731: 150400, 2024 Oct 30.
Article in English | MEDLINE | ID: mdl-39024975

ABSTRACT

Neuromuscular signal transmission is affected in various diseases including myasthenia gravis, congenital myasthenic syndromes, and sarcopenia. We used an ATF2-luciferase system to monitor the phosphorylation of MuSK in HEK293 cells introduced with MUSK and LRP4 cDNAs to find novel chemical compounds that enhanced agrin-mediated acetylcholine receptor (AChR) clustering. Four compounds with similar chemical structures carrying benzene rings and heterocyclic rings increased the luciferase activities 8- to 30-folds, and two of them showed continuously graded dose dependence. The effects were higher than that of disulfiram, a clinically available aldehyde dehydrogenase inhibitor, which we identified to be the most competent preapproved drug to enhance ATF2-luciferase activity in the same assay system. In C2C12 myotubes, all the compounds increased the area, intensity, length, and number of AChR clusters. Three of the four compounds increased the phosphorylation of MuSK, but not of Dok7, JNK. ERK, or p38. Monitoring cell toxicity using the neurite elongation of NSC34 neuronal cells as a surrogate marker showed that all the compounds had no effects on the neurite elongation up to 1 µM. Extensive docking simulation and binding structure prediction of the four compounds with all available human proteins using AutoDock Vina and DiffDock showed that the four compounds were unlikely to directly bind to MuSK or Dok7, and the exact target remained unknown. The identified compounds are expected to serve as a seed to develop a novel therapeutic agent to treat defective NMJ signal transmission.


Subject(s)
Muscle Fibers, Skeletal , Receptors, Nicotinic , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Animals , Mice , Cell Line , Humans , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Genes, Reporter , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Multigene Family , Signal Transduction/drug effects , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neurites , Bungarotoxins/pharmacology , Benzene/pharmacology , Heterocyclic Compounds/pharmacology , Molecular Docking Simulation
17.
Eur J Med Chem ; 276: 116649, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-38972078

ABSTRACT

Guided by the X-ray cocrystal structure of the lead compound 4a, we developed a series of thieno[3,2-d]pyrimidine and heterocyclic fused pyrimidines demonstrating potent antiproliferative activity against four tumor cell lines. Two analogs, 13 and 25d, exhibited IC50 values around 1 nM and overcame P-glycoprotein (P-gp)-mediated multidrug resistance (MDR). At low concentrations, 13 and 25d inhibited both the colony formation of SKOV3 cells in vitro and tubulin polymerization. Furthermore, mechanistic studies showed that 13 and 25d induced G2/M phase arrest and apoptosis in SKOV3 cells, as well as dose-dependent inhibition of tumor cell migration and invasion at low concentrations. Most notably, the X-ray cocrystal structures of compounds 4a, 25a, and the optimal molecule 13 in complex with tubulin were elucidated. This study identifies thieno[3,2-d]pyrimidine and heterocyclic fused pyrimidines as representatives of colchicine-binding site inhibitors (CBSIs) with potent antiproliferative activity.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Drug Screening Assays, Antitumor , Pyrimidines , Tubulin Modulators , Tubulin , Humans , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Tubulin/metabolism , Structure-Activity Relationship , Tubulin Modulators/pharmacology , Tubulin Modulators/chemistry , Tubulin Modulators/chemical synthesis , Molecular Structure , Apoptosis/drug effects , Dose-Response Relationship, Drug , Cell Line, Tumor , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemical synthesis , Drug Discovery , Models, Molecular
18.
Sci Rep ; 14(1): 15522, 2024 07 05.
Article in English | MEDLINE | ID: mdl-38969677

ABSTRACT

A series of benzoquinoline-employing heterocycles was synthesized by treating 3-chlorobenzo[f]quinoline-2-carbaldehyde with N-phenyl-3-methylpyrazolone, 4-aminoacetophenone, 1,2-diaminoethane, and 2-cyanoethanohydrazide. Also, pyridine, chromene, α,ß-unsaturated nitrile, thiosemicarbazone, and 1,2-bis-aryl hydrazine derivatives were prepared from the cyanoethanohydrazone obtained. The DFT calculations and experiment outcomes were consistent. In vitro screening of their antiproliferative efficacy was examined against HCT116 and MCF7 cancer cell lines. The pyrazolone 2 and cyanoethanohydrazone 5 derivatives exhibited the most potency, which was demonstrated by their molecular docking towards the CDK-5 enzyme. The binding energies of compounds 2 and 5 were - 6.6320 kcal/mol (with RMSD of 0.9477 Å) and - 6.5696 kcal/mol (with RMSD of 1.4889 Å), respectively, which were near to that of co-crystallized ligand (EFP). This implies a notably strong binding affinity towards the CDK-5 enzyme. Thus, pyrazolone derivative 2 would be considered a promising candidate for further optimization to develop new chemotherapeutic agents. In addition, the ADME (absorption, distribution, metabolism, and excretion) analyses displayed its desirable drug-likeness and oral bioavailability properties.


Subject(s)
Antineoplastic Agents , Molecular Docking Simulation , Quinolines , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Quinolines/chemistry , Quinolines/chemical synthesis , Quinolines/pharmacology , MCF-7 Cells , Cell Proliferation/drug effects , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/pharmacology , Computer Simulation , HCT116 Cells , Cell Line, Tumor , Structure-Activity Relationship
19.
Bioorg Med Chem ; 110: 117834, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39029436

ABSTRACT

Currently, no effective treatment exists for premature ovarian failure (POF). To obtain compounds with protective effects against POF, we aimed to design and synthesize a series of spiroheterocyclic protective agents with a focus on minimizing toxicity while enhancing their protective effect against cisplatin-induced POF. This was achieved through systematic modifications of Michael receptors and linkers within the molecular structure of 1,5-diphenylpenta-1,4-dien-3-one analogs. To assess the cytotoxicity and activity of these compounds, we constructed quantitative conformational relationship models using an artificial intelligence random forest algorithm, resulting in R2 values exceeding 0.87. Among these compounds, j2 exhibited optimal protective activity. It significantly increased the survival of cisplatin-injured ovarian granulosa KGN cells, improved post-injury cell morphology, reduced apoptosis, and enhanced cellular estradiol (E2) levels. Subsequent investigations revealed that j2 may exert its protective effect via a novel mechanism involving the activation of the SIRT1/AKT signal pathway. Furthermore, in cisplatin-injured POF in rats, j2 was effective in increasing body, ovarian, and uterine weights, elevating the number of follicles at all levels in the ovary, improving ovarian and uterine structures, and increasing serum E2 levels in rats with cisplatin-injured POF. In conclusion, this study introduces a promising compound j2 and a novel target SIRT1 with substantial protective activity against cisplatin-induced POF.


Subject(s)
Cisplatin , Primary Ovarian Insufficiency , Sirtuin 1 , Female , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/pathology , Primary Ovarian Insufficiency/metabolism , Sirtuin 1/metabolism , Sirtuin 1/antagonists & inhibitors , Cisplatin/pharmacology , Animals , Rats , Humans , Structure-Activity Relationship , Up-Regulation/drug effects , Rats, Sprague-Dawley , Molecular Structure , Protective Agents/pharmacology , Protective Agents/chemistry , Protective Agents/chemical synthesis , Drug Discovery , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Dose-Response Relationship, Drug , Apoptosis/drug effects , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemical synthesis
20.
J Med Chem ; 67(14): 12118-12142, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38996194

ABSTRACT

Disrupting microtubule dynamics has emerged as a promising strategy for cancer treatment. However, drug resistance remains a challenge hindering the development of microtubule-targeting agents. In this work, a novel class of diaryl substituted fused heterocycles were designed, synthesized, and evaluated, which were demonstrated as effective dual katanin and tubulin regulators with antitumor activity. Following three rounds of stepwise optimization, compound 21b, featuring a 3H-imidazo[4,5-b]pyridine core, displayed excellent targeting capabilities on katanin and tubulin, along with notable antiproliferative and antimetastatic effects. Mechanistic studies revealed that 21b disrupts the microtubule network in tumor cells, leading to G2/M cell cycle arrest and apoptosis induction. Importantly, 21b exhibited significant inhibition of tumor growth in MDA-MB-231 and A549/T xenograft tumor models without evident toxicity and side effects. In conclusion, compound 21b presents a novel mechanism for disrupting microtubule dynamics, warranting further investigation as a dual-targeted antitumor agent with potential antimultidrug resistance properties.


Subject(s)
Antineoplastic Agents , Drug Resistance, Neoplasm , Heterocyclic Compounds , Katanin , Tubulin Modulators , Tubulin , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Tubulin/metabolism , Drug Resistance, Neoplasm/drug effects , Animals , Katanin/metabolism , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemical synthesis , Cell Line, Tumor , Tubulin Modulators/pharmacology , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Mice , Apoptosis/drug effects , Cell Proliferation/drug effects , Structure-Activity Relationship , Mice, Nude , Drug Discovery , Microtubules/drug effects , Microtubules/metabolism , Xenograft Model Antitumor Assays , Drug Screening Assays, Antitumor , Mice, Inbred BALB C , Female
SELECTION OF CITATIONS
SEARCH DETAIL