Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Chem Biodivers ; 19(9): e202200116, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35983935

RESUMO

A set of new heterocyclic analogs (Compounds I-IX), comprising of 6,7 dimethyl Quinoxalines were found to be active against the receptor GSK3ß (Compounds IV-V) (Chem. Biodiversity 2021, 18, e2100364). In an effort to modulate effective CDK5 inhibitors herein our hypothesis underpinned to fish out an appropriate derivative from the same quinoxaline series, as these two targets GSK3ß and CDK5 shared structural resemblance with each other. Aligned to the goal we have synthesized Compounds I-IX, characterized them using a combination of spectroscopic techniques and evaluated their activities against CDK5. Our analysis reflected that the adjacently located alkoxy/hydroxy functionality derivatives namely Compounds III and VI, to be the most potent (micromolar) amongst others in the series, backed by Density Functional Theory (DFT) calculations and molecular modelling studies. Also, the efficacy of the Compounds I-IX, were monitored in few other members of the CMGC family namely DYRK1A, CLK1and CK1δ that have been known to be directly involved in hyperphosphorylation of Tau. But unfortunately in none of the targets, our quinoxaline series were active. In a nut shell further optimisation of these intelligent nucleus, would not only lead to the discovery of novel pharmacophores, but also marked selectivity against a pool of kinases, thereby implementing a distinct roadmap towards the design of potential therapeutics against Alzheimer's.


Assuntos
Doença de Alzheimer , Animais , Doença de Alzheimer/tratamento farmacológico , Glicogênio Sintase Quinase 3 beta , Quinoxalinas/farmacologia
2.
Chem Biodivers ; 18(11): e2100364, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34486216

RESUMO

Untangling the most selective kinase inhibitors via pharmacological intervention remains one of the challenging affairs to date. In accordance to this drift, herein we describe the design and synthesis of a set of new heterocyclic analogs consisting of 6,7-dimethyl Quinoxaline, appended to a connector, employing Schiff base strategy (Compounds I-IX). The compounds were characterized by various spectroscopic techniques and the kinase inhibition assay were performed on few prime members of the CMGC family namely the GSK3ß, DYRK1A and CLK1 receptors, respectively, that have been known to be directly involved in hyperphosphorylation of Tau. Interestingly the biological evaluation results revealed that Compounds IV and V, with bromo/chloro functionalities in the aromatic core were advantaged of being highly selective towards the target GSK3ß over others. To strengthen our analysis, we adopted molecular modelling studies, where compounds IV/V were redocked in the same grid 4AFJ, as that of the reference ligand, 5-aryl-4-carboxamide-1,3-oxazole. Surprisingly, our investigation underpinned that for both the compounds IV/V, a primary H-bonding existed between the designed molecules (IV/V) and Val 135 residue in the receptor GSK3ß, in line with the reference ligand. We attribute this interaction to instigate potency in the compounds. Indeed the other non-covalent interaction, between the derivative's aromatic nucleus and Arg 141/Thr 138 in the receptor GSK3ß, might have been responsible for enhancing the selectivity in the targets. Overall, we feel that the present work depicts a logical demonstration towards fine tuning the efficacy of the inhibitors through systematic adjustment of electron density at appropriate positions in the aromatic ring be it the main quinoxaline or the other aromatic nucleus. Thus this pathway offers a convenient strategy for the development of efficient therapeutics for diversified neurodegenerative diseases like that of Alzheimer's.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinoxalinas/farmacologia , Doença de Alzheimer/metabolismo , Desenvolvimento de Medicamentos , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Modelos Moleculares , Estrutura Molecular , Fármacos Neuroprotetores/síntese química , Fármacos Neuroprotetores/química , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Quinoxalinas/síntese química , Quinoxalinas/química , Quinases Dyrk
3.
Molecules ; 26(14)2021 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-34299490

RESUMO

In this study, we designed, synthesized and evaluated, in vitro, novel chalcone analogs containing dialkylamino pharmacophores in the cervical cancer cell line, OV2008. The compound, DML6 was selective and significantly decreased the proliferation of OV2008 and HeLa cells in sub-micromolar concentrations, compared to prostate, lung, colon, breast or human embryonic kidney cell line (HEK293). DML6, at 5 µM, arrested the OV2008 cells in the G2 phase. Furthermore, DML6, at 5 µM, increased the levels of reactive oxygen species and induced a collapse in the mitochondrial membrane potential, compared to OV2008 cells incubated with a vehicle. DML6, at 5 µM, induced intrinsic apoptosis by significantly (1) increasing the levels of the pro-apoptotic proteins, Bak and Bax, and (2) decreasing the levels of l the anti-apoptotic protein, Bcl-2, compared to cell incubated with a vehicle. Furthermore, DML6, at 5 and 20 µM, induced the cleavage of caspase-9, followed by subsequent cleavage of the executioner caspases, caspase-3 and caspase-7, which produced OV2008 cell death. Overall, our data suggest that DML6 is an apoptosis-inducing compound that should undergo further evaluation as a potential treatment for cervical cancer.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Chalconas/farmacologia , Mitose/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Neoplasias do Colo do Útero/tratamento farmacológico , Células A549 , Animais , Células CHO , Linhagem Celular , Linhagem Celular Tumoral , Cricetulus , Feminino , Células HEK293 , Células HeLa , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Neoplasias do Colo do Útero/metabolismo
4.
Molecules ; 26(15)2021 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-34361570

RESUMO

A novel series of 4-anilinoquinazoline analogues, DW (1-10), were evaluated for anticancer efficacy in human breast cancer (BT-20) and human colorectal cancer (CRC) cell lines (HCT116, HT29, and SW620). The compound, DW-8, had the highest anticancer efficacy and selectivity in the colorectal cancer cell lines, HCT116, HT29, and SW620, with IC50 values of 8.50 ± 2.53 µM, 5.80 ± 0.92 µM, and 6.15 ± 0.37 µM, respectively, compared to the non-cancerous colon cell line, CRL1459, with an IC50 of 14.05 ± 0.37 µM. The selectivity index of DW-8 was >2-fold in colon cancer cells incubated with vehicle. We further determined the mechanisms of cell death induced by DW-8 in SW620 CRC cancer cells. DW-8 (10 and 30 µM) induced apoptosis by (1) producing cell cycle arrest at the G2 phase; (2) activating the intrinsic apoptotic pathway, as indicated by the activation of caspase-9 and the executioner caspases-3 and 7; (3) nuclear fragmentation and (4) increasing the levels of reactive oxygen species (ROS). Overall, our results suggest that DW-8 may represent a suitable lead for developing novel compounds to treat CRC.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias do Colo , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Células HCT116 , Células HT29 , Humanos
5.
Bioorg Chem ; 92: 103221, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31499261

RESUMO

A series of ten N-(3-(1H-tetrazole-5-yl)phenyl)acetamide derivatives (NM-07 to NM-16) designed from a lead molecule identified previously in our laboratory were synthesized and evaluated for protein tyrosine phosphatase 1B (PTP1B) inhibitory activity. Among the synthesized molecules, NM-14, a 5-Cl substituted benzothiazole analogue elicited significant PTP1B inhibition with an IC50 of 1.88 µM against reference standard suramin (IC50 ≥ 10 µM). Furthermore, this molecule also showed good in vivo antidiabetic activity which was comparable to that of standard antidiabetic drugs metformin and glimepiride. Overall, the results of the study clearly reveal that the reported tetrazole derivatives especially NM-14 are valuable prototypes for the development of novel non-carboxylic inhibitors of PTP1B with antidiabetic potential.


Assuntos
Acetamidas/farmacologia , Diabetes Mellitus Experimental/tratamento farmacológico , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Hipoglicemiantes/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/antagonistas & inibidores , Tetrazóis/farmacologia , Acetamidas/síntese química , Acetamidas/química , Animais , Glicemia/efeitos dos fármacos , Diabetes Mellitus Experimental/induzido quimicamente , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Hipoglicemiantes/síntese química , Hipoglicemiantes/química , Estrutura Molecular , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Ratos , Estreptozocina , Relação Estrutura-Atividade , Tetrazóis/síntese química , Tetrazóis/química
6.
Bioorg Med Chem Lett ; 28(13): 2244-2249, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29853331

RESUMO

A series of lH-pyrazolo[3,4-b]quinolin-3-amine derivatives were synthesized and evaluated for anticancer efficacy in a panel of ten cancer cell lines, including breast (MDAMB-231 and MCF-7), colon (HCT-116, HCT-15, HT-29 and LOVO), prostate (DU-145 and PC3), brain (LN-229), ovarian (A2780), and human embryonic kidney (HEK293) cells, a non-cancerous cell line. Among the eight derivatives screened, compound QTZ05 had the most potent and selective antitumor efficacy in the four colon cancer cell lines, with IC50 values ranging from 2.3 to 10.2 µM. Furthermore, QTZ05 inhibited colony formation in HCT-116 cells in a concentration-dependent manner. Cell cycle analysis data indicated that QTZ05 caused an arrest in the sub G1 cell cycle in HCT-116 cells. QTZ05 induced apoptosis in HCT-116 cells in a concentration-dependent manner that was characterized by chromatin condensation and increase in the fluorescence of fluorochrome-conjugated Annexin V. The findings from our study suggest that QTZ05 may be a valuable prototype for the development of chemotherapeutics targeting apoptotic pathways in colorectal cancer cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Pirazóis/farmacologia , Quinolinas/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Células HEK293 , Humanos , Pirazóis/síntese química , Pirazóis/toxicidade , Quinolinas/síntese química , Quinolinas/toxicidade
7.
Bioorg Chem ; 80: 145-150, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29925050

RESUMO

Described herein is the synthesis and biological evaluation of a series of non-carboxylic inhibitors of Protein Tyrosine Phosphatase 1B designed using bioisosteric replacement strategy. Six N-(3-(1H-tetrazol-5-yl)phenyl)acetamide derivatives designed employing the aforementioned strategy were synthesized and screened for PTP1B inhibitory activity. Among the synthesized compounds, compound NM-03 exhibited the most potent inhibitory activity with IC50 value of 4.48 µM. Docking studies with NM-03 revealed the key interactions with desired amino acids in the binding site of PTP1B. Furthermore, compound NM-03 also elicited good in vivo activity. Taken together, the results of this study establish N-(3-(1H-tetrazole-5-yl)phenyl)-2-(benzo[d]oxazol-2-ylthio)acetamide (NM-03) as a valuable lead molecule with great potential for PTP1B inhibitor development targeting diabetes.


Assuntos
Acetamidas/química , Inibidores Enzimáticos/síntese química , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Acetamidas/metabolismo , Acetamidas/uso terapêutico , Animais , Sítios de Ligação , Glicemia/análise , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/tratamento farmacológico , Desenho de Fármacos , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/uso terapêutico , Teste de Tolerância a Glucose , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Estrutura Terciária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 1/antagonistas & inibidores , Ratos , Relação Estrutura-Atividade , Tetrazóis/química
8.
Bioorg Med Chem Lett ; 27(12): 2663-2667, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28487075

RESUMO

Described herein is the design, synthesis and biological evaluation of a series of N-(1H-pyrazol-3-yl)quinazolin-4-amines against a panel of eight disease relevant protein kinases. The kinase inhibition results indicated that two compounds inhibited casein kinase 1δ/ε (CK1δ/ε) with some selectivity over related kinases, namely CDK5/p25, GSK-3α/ß, and DYRK1A. Docking studies with 3c and 3d revealed the key interactions with desired amino acids in the ATP binding site of CK1δ. Furthermore, compound 3c also elicited selective cytotoxic activity against the pancreas ductal adenocarcinoma (PANC-1) cell line. Taken together, the results of this study establish N-(1H-pyrazol-3-yl)quinazolin-4-amines especially 3c and 3d as valuable lead molecules with great potential for CK1δ/ε inhibitor development targeting neurodegenerative disorders and cancer.


Assuntos
Caseína Quinase 1 épsilon/antagonistas & inibidores , Caseína Quinase Idelta/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Quinazolinas/farmacologia , Caseína Quinase 1 épsilon/metabolismo , Caseína Quinase Idelta/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Modelos Moleculares , Estrutura Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Pirazóis/síntese química , Pirazóis/química , Quinazolinas/síntese química , Quinazolinas/química , Relação Estrutura-Atividade
9.
Biochem Biophys Res Commun ; 460(4): 1002-7, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25847597

RESUMO

Lymphoma is the most common hematopoietic tumor in dogs and humans, with similar pathogenesis and therapeutic responses. Anticancer drugs like vincristine (VCR) and doxorubicin (DOX) are often used in treating lymphoma. However, the cure rate is generally poor due to chemoresistance. Here, we sought to determine whether stearidonic acid (SDA), a plant-based dietary fatty acid, sensitizes chemoresistant canine lymphoid-tumor cells. GL-1 B-cell lymphoid-tumor cells were found to be highly sensitive to the antitumor-activity of VCR and DOX, while OSW T-cell and 17-71 B-cell lymphoid-tumor cells were moderately and fully resistant, respectively. SDA, at its non-toxic concentrations, significantly promoted the antitumor action of VCR and DOX in both OSW and 17-71 cells. SDA-mediated chemosensitization was associated with SDA inhibition of P-glycoprotein (P-gp) function. This was confirmed in HEK293 cells stably expressing P-gp as well as by increased binding-affinity of SDA to P-gp in P-gp docking analysis. SDA at its chemosensitizing concentrations did not affect the viability of healthy dog peripheral blood mononuclear cells, suggesting that SDA is non-toxic to normal dog peripheral blood leucocytes at its chemosensitizing concentrations. Our study identifies a novel dietary fatty acid that may be used as a dietary supplement in combination with chemotherapy to promote the antitumor efficacy of the chemotherapy drugs in dogs and possibly in humans with chemoresistant lymphoma.


Assuntos
Antineoplásicos/uso terapêutico , Ácidos Graxos Ômega-3/farmacologia , Linfoma de Células B/tratamento farmacológico , Plantas/química , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Linhagem Celular Tumoral , Cães , Sinergismo Farmacológico , Ácidos Graxos Ômega-3/uso terapêutico
10.
Bioorg Med Chem ; 23(3): 602-11, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25537531

RESUMO

Naturally occurring condensed quinolines have anticancer properties. In efforts to find active analogues, we designed and synthesized eight polycyclic heterocycles with a pyrimido[1″,2″:1,5]pyrazolo[3,4-b]quinoline framework (IND series). The compounds were evaluated for activity against colon (HCT-116 and S1-MI-80), prostate (PC3 and DU-145), breast (MCF-7 and MDAMB-231), ovarian (ov2008 and A2780), and hepatocellular (HepG2) cancer cells and against non-cancerous Madin Darby canine kidney (MDCK), mouse embryonic fibroblast (NIH/3T3), and human embryonic kidney cells (HEK293). IND-2, a 4-chloro-2-methyl pyrimido[1″,2″:1,5]pyrazolo[3,4-b]quinoline, exhibited more than ten-fold selectivity and potent cytotoxic activity against colon cancer cells relative to the other cancer and non-cancer cells. With five additional colon cancer cell lines (HT-29, HCT-15, LS-180, LS-174, and LoVo), IND-2 had similar cytotoxicity and selectivity, and sub-micromolar concentrations caused changes in the morphology of HCT-116 and HCT-15 cells. IND-2 did not activate the transactivating function of the pregnane X receptor (PXR), indicating that it does not induce PXR-regulated ABCB1 or ABCG2 transporters. Indeed, IND-2 was not a substrate of ABCB1 or ABCG2, and it induced cytotoxicity in HEK293 cells overexpressing ABCB1 or ABCG2 to the same extent as in normal HEK293 cells. IND-2 was cytotoxic to resistant colon carcinoma S1-MI-80 cells, approximately three- and five-fold more than SN-38 and topotecan, respectively. In HCT-116 colon cancer cells, IND-2 produced concentration-dependent changes in mitochondrial membrane potential, leading to apoptosis, and sub-micromolar concentrations caused chromosomal DNA fragmentation. These findings suggest that, by increasing apoptosis, IND-2 has potential therapeutic efficacy for colorectal cancer.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Quinolinas/farmacologia , Animais , Antineoplásicos/farmacologia , Cães , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Células HCT116 , Células HEK293 , Células Hep G2 , Humanos , Células MCF-7 , Células Madin Darby de Rim Canino , Camundongos , Pirimidinas/farmacologia
11.
Bioorg Med Chem ; 22(6): 1909-15, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24530227

RESUMO

A series of novel 4-anilinoquinazoline derivatives (3a-3j) has been synthesized and evaluated as potential inhibitors for protein kinases implicated in Alzheimer's disease. Among all the synthesized compounds, compound 3e (N-(3,4-dimethoxyphenyl)-6,7-dimethoxyquinazolin-4-amine) exhibited the most potent inhibitory activity against CLK1 and GSK-3α/ß kinase with IC50 values of 1.5 µM and 3 µM, respectively. Docking studies were performed to elucidate the binding mode of the compounds to the active site of CLK1 and GSK-3ß. The results of our study suggest that compound 3e may serve as a valuable template for the design and development of dual inhibitors of CLK1 and GSK-3α/ß enzymes with potential therapeutic application in Alzheimer's disease.


Assuntos
Compostos de Anilina/farmacologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinazolinas/farmacologia , Compostos de Anilina/síntese química , Compostos de Anilina/química , Relação Dose-Resposta a Droga , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Modelos Moleculares , Estrutura Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Quinazolinas/síntese química , Quinazolinas/química , Relação Estrutura-Atividade
12.
J Liposome Res ; 24(2): 150-62, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24328725

RESUMO

CONTEXT: Strategy of dual therapy has been proposed to minimize the amount of each drug and to achieve the synergistic effect for cancer therapies. OBJECTIVE: The aim of this study was to develop an effective drug delivery system for the simultaneous topical delivery of two anti-tumor agents, cisplatin and imiquimod. MATERIAL AND METHODS: The preformulation studies were carried out in terms of tests for identification, solubility profile, determination of partition coefficient and simultaneous estimation of both drugs by UV-Visible spectrophotometer and High Performance Liquid Chromatography (HPLC). Drug-drug and drug-excipients interactions were examined by thin layer chromatography, infrared spectroscopy, differential scanning calorimetry (DSC) and X-ray diffraction (XRD). Provesicular drug delivery system (protransfersome gel formulation) have been prepared and characterized by in vitro and in vivo parameters. RESULTS: The mean size, poly dispersity index (PDI) and zeta potential of transfersomal vesicles formed by protransfersome hydration were 429.5 nm, 0.631 and -68.1 Mv, respectively. The prepared formulation showed toxicity on cutaneous squamous cell carcinoma cell line (A-431) at 200 µg (cisplatin) and 1 mg (imiquimod) concentration of drug in combination against control. The cisplatin- and imiquimod-loaded provesicular dual-drug delivery system achieved an optimal antitumor effect, increase in lifespan, antiviral, and toxicity reduction, revealing the advantage of site specific drug delivery and the modified combination therapy. DISCUSSION: Cisplatin delivery through protransfersome gel in combination with imiquimod may potentiate the activity against solid tumors of epidermal origin. CONCLUSION: Data revealed that combination therapy considerably enhances antitumor efficacy of the drug for skin-cited malignancies.


Assuntos
Aminoquinolinas/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Carcinoma de Células Escamosas/tratamento farmacológico , Cisplatino/administração & dosagem , Neoplasias Cutâneas/tratamento farmacológico , Administração Tópica , Animais , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Humanos , Imiquimode , Camundongos , Microscopia de Fluorescência , Ratos Wistar , Neoplasias Cutâneas/patologia
13.
J Labelled Comp Radiopharm ; 57(6): 425-33, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24889134

RESUMO

The present investigation deals with the development of topical (top.) formulation for co-delivery of cisplatin and imiquimod to enhance the antitumor efficacy of the drug for skin-cited malignancies even in immune compromised patient. Cisplatin (CDDP) and imiquimod-loaded protransfersome gel (CDDP-Imi-Pts gel) formulation was characterized for entrapment efficiency, pH, and viscosity. Further, fluorescence-labeled (6-carboxyfluorescin) and radiolabeled ((99m) technetium) drug-loaded formulations were compared with respect to biodistribution and pharmacokinetic studies. Gamma scintigraphy of mice following radiolabeled formulation administrations was performed to accomplish the localization of drugs in various organs. The percentage entrapment efficiency of cisplatin and imiquimod in the protransfersome gel formulations were found to be 36.22 ± 6.41 and 63.11 ± 3.73. The skin/blood localization ratio of 1.096, 120.13, 0.174, and 349.88 was found for intraperitoneal radiolabeled drug solution ((99m-)Tc-CDDP-Imi-Sol), top. radiolabeled drug-loaded protransfersome gel formulation ((99m-)Tc-CDDP-Imi-Pts gel), intraperitoneal 6-carboxyfluorescin labeled drug solution (6-Cf-CDDP-Imi-Sol), top. 6-carboxyfluorescin labeled drug-loaded protransfersome gel formulation (6-Cf-CDDP-Imi-Pts gel), respectively after 0.5h of administration. CDDP-Imi-Pts gel has a potential for site specific delivery and reduces the systemic toxicity of anti cancer drugs. These findings suggest that the cisplatin-imiquimod co-delivery offers an attractive, novel approach for treatment of skin-cited malignancies.


Assuntos
Aminoquinolinas/administração & dosagem , Aminoquinolinas/química , Cisplatino/administração & dosagem , Cisplatino/química , Sistemas de Liberação de Medicamentos/métodos , Fluoresceínas/química , Tecnécio/química , Administração Tópica , Aminoquinolinas/metabolismo , Aminoquinolinas/farmacocinética , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Química Farmacêutica , Cisplatino/metabolismo , Cisplatino/farmacocinética , Corantes Fluorescentes/química , Imiquimode , Marcação por Isótopo , Camundongos , Permeabilidade , Cintilografia , Pele/diagnóstico por imagem , Pele/metabolismo , Distribuição Tecidual
14.
Anticancer Agents Med Chem ; 23(4): 383-403, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35708082

RESUMO

Cyclin Dependent Kinase 9 (CDK9), which controls transcriptional elongation, is a promising pharmacological target for a variety of cancerous cells, specifically those characterized by transcriptional dysregulation. CDK9 promotes the pause or release of RNA polymerase II, a rate-limiting stage in normal transcriptional regulation that is often disturbed in cancers. New indications suggest that selective CDK9 antagonism may be beneficial in the treatment of some cancers. CDK9 modulators (inhibitors and degraders) have gained a lot of attention recently, and many molecules are currently in clinical trials. In this review, the CDK9 antagonists under clinical and preclinical trials have been discussed, as well as the structure-activity relationship has been studied, which will help scientists generate more target- specific drug molecules in the future with less toxicity.


Assuntos
Quinase 9 Dependente de Ciclina , Neoplasias , Humanos , Quinase 9 Dependente de Ciclina/genética , Quinase 9 Dependente de Ciclina/metabolismo , Regulação da Expressão Gênica , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Neoplasias/tratamento farmacológico
15.
J Enzyme Inhib Med Chem ; 27(2): 201-7, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21635210

RESUMO

A series of 1,2,4-trisubstituted 5-imidazolinone derivatives were synthesized by Erlenmeyer condensation of benzoylglycine (hippuric acid) with different aldehydes in the presence of sodium acetate and acetic anhydride. The derivatives of the compounds were prepared by condensation of some known sulpha drugs with 5-oxazolone derivatives. The anticonvulsant activity of the compounds was determined by the protection of pentylenetetrazole-induced convulsions that was ranged from 10 to 60%. The compounds with p-OCH3, p-OH and o-Cl substitutions in the phenyl ring on 4(th) position of the imidazolinone ring exhibited good anticonvulsant activity. In silico metabolic and toxicity studies showed that all the compounds in the series are not likely to exhibit toxicity except the compounds IIIa, IIIb, VIa and VIb, that is predicted to show 29% mutagenicity and 53% irritation in comparison to the other compounds. The predicted lethal effect and hERG toxicity of the compounds showed that IIa, IVa, Va and Vb might be toxic at higher concentrations. The results successfully establish the synthesized imidazolinone derivatives as novel compounds with anticonvulsant properties, low predicted cardiotoxicity and lethal effects thus can be promising leads for further development as novel anticonvulsants.


Assuntos
Anticonvulsivantes/síntese química , Anticonvulsivantes/farmacologia , Anticonvulsivantes/toxicidade , Imidazolinas/síntese química , Imidazolinas/farmacologia , Imidazolinas/toxicidade , Convulsões/prevenção & controle , Animais , Convulsivantes/toxicidade , Camundongos , Estrutura Molecular , Pentilenotetrazol/toxicidade , Convulsões/induzido quimicamente , Software , Relação Estrutura-Atividade
16.
Hell J Nucl Med ; 15(1): 9-15, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22413106

RESUMO

The main aim of this study was to develop and evaluate nanoparticulate system of paclitaxel loaded polymeric biodegradable Poly (ε-caprolactone) nanoparticles for targeting to neuroendocrine pancreatic tumors in mice. Nanoparticles were prepared by simple emulsion technique having surface modification with pluronic F-68. All formulations were evaluated for particle shape and size, zeta potential, encapsulation efficiency and in vitro drug release. Radiolabelling of nanoparticles with (99m)Tc was done for scintigraphy and biodistribution studies. Cytotoxicity studies were performed on BON-1 cell line using MTT cell proliferation assay. The in vivo tumor inhibition study was performed after i.v. administration of paclitaxel nanoparticles. Our results showed that optimized nanoformulation was found to have size range from 100 ± 0.03 nm to 250 ± 0.06 nm with smooth spherical shape. Negative zeta potential value confirmed the surface modification and stability of nanoformulations. The amount of drug released after 24h from the formulation was found to be 73.3% ± 2.7%. More pronounced cytotoxicity was found with nanoparticulate formulation as compared with paclitaxel. The PCL-Ptx nanoparticles reduced tumor volume significantly in comparison with paclitaxel. Higher concentration of Ptx-NPs were found in tumor which was also revealed by high quality scintigraphic image of BON-1 tumor bearing mouse model. In conclusion, polymeric nanoparticulate formulation of paclitaxel was very much efficient for chemotherapy of human pancreatic neuroendocrine tumor in mice.


Assuntos
Nanocápsulas/administração & dosagem , Tumores Neuroendócrinos/tratamento farmacológico , Tumores Neuroendócrinos/metabolismo , Paclitaxel/administração & dosagem , Paclitaxel/farmacocinética , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Implantes Absorvíveis , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Preparações de Ação Retardada/administração & dosagem , Taxa de Depuração Metabólica , Camundongos , Camundongos Nus , Nanocápsulas/química , Poliésteres/química , Polietilenoglicóis/química , Distribuição Tecidual , Resultado do Tratamento
17.
Med Chem ; 18(10): 1044-1059, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35240964

RESUMO

BACKGROUND: Pyrazole is a bioactive heterocyclic congener with numerous biological and pharmacological functionalities. Due to their multiple prospective applications, developing innovative and novel pyrazoles and analogs, revealing revolutionary methods for synthesizing this nucleus, investigating diverse potencies of that heterocycle, and exploring possible pyrazole applications are becoming increasingly relevant. OBJECTIVES: Pyrazole scaffolds have been proven successful as antimicrobial, anticancer, and antimalarial therapeutics against multiple targets like DNA gyrase, topoisomerase IV, Hsp90, and several kinase enzymes. For this variability in the biotic zone, their moiety has gained the attention of many scientists interested in researching chemical and pharmacological profiles. RESULTS: The review covers pyrazole scaffolds with a variety of biological functions and attempts to connect the structure-activity relationship. Multiple pyrazole analogs have been produced as lead compounds, and their activities have been evaluated. CONCLUSION: The combination of pyrazole with other pharmacophores in a molecule might lead to novel potent therapeutic medicines, which could aid in the development of potent lead compounds.


Assuntos
Anti-Infecciosos , Antimaláricos , Antineoplásicos , Antibacterianos , Pirazóis , Relação Estrutura-Atividade
18.
Curr Cancer Drug Targets ; 22(3): 221-233, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35232350

RESUMO

Cancer is a leading cause of death worldwide. The Janus kinase (JAK) signal transducer and activator of transcription (STAT) signalling pathway are activated abnormally, which promotes carcinogenesis. Several cytokines are important cancer drivers. These proteins bind to receptors and use the Janus kinase (JAK) and STAT pathways to communicate their responses. Cancer risks are linked to genetic differences in the JAK-STAT system. JAK inhibitors have been shown to reduce STAT initiation, tissue propagation, and cell existence in preclinical investigations involving solid tumour cell line models. JAK inhibitors, notably ruxolitinib, JAK1 or 2 blockers, make cell lines and mouse models more susceptible to radiotherapy, biological response modifier therapy, and oncolytic viral treatment. Numerous JAK antagonists have been or are now being evaluated in cancerous patients as monotherapy or by combining with other drugs in clinical studies. In preclinical investigations, certain JAK inhibitors showed promising anticancer effects; however, clinical trials explicitly evaluating their effectiveness against the JAK/STAT system in solid tumours have yet to be completed. JAK inhibition is a promising strategy to target the JAK/STAT system in solid tumours, and it deserves to be tested further in clinical studies. The function of directing Janus kinases (JAKs), an upstream accelerator of STATs, as a technique for lowering STAT activity in various malignant circumstances is summarized in this article, which will help scientists to generate more specific drug molecules in the future.


Assuntos
Inibidores de Janus Quinases , Neoplasias , Animais , Humanos , Janus Quinase 2/metabolismo , Janus Quinase 2/farmacologia , Inibidores de Janus Quinases/farmacologia , Inibidores de Janus Quinases/uso terapêutico , Janus Quinases , Camundongos , Neoplasias/tratamento farmacológico , Fatores de Transcrição STAT , Transdução de Sinais
19.
Anticancer Agents Med Chem ; 22(8): 1478-1495, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34382529

RESUMO

Cancer is considered one of the gruelling challenges and poses a grave health hazard across the globe. According to the International Agency for Research on Cancer (IARC), new cancer cases increased to 18.1 million in 2018, with 9.6 million deaths, bringing the global cancer rate to 23.6 million by 2030. In 1942, the discovery of nitrogen mustard as an alkylating agent was a tremendous breakthrough in cancer chemotherapy. It acts by binding to the DNA, and creating cross linkages between the two strands, leading to halt of DNA replication and eventual cell death. Nitrogen lone pairs of 'nitrogen mustard' produce an intermediate 'aziridinium ion' at the molecular level, which is very reactive towards DNA of tumour cells, resulting in multiple side effects with therapeutic consequences. Owing to its high reactivity and peripheral cytotoxicity, several improvements have been made with structural modifications for the past 75 years to enhance its efficacy and improve the direct transport of drugs to the tumour cells. Alkylating agents were among the first non-hormonal substances proven to be active against malignant cells and also the most valuable cytotoxic therapies available for the treatment of leukaemia and lymphoma patients. This review focus on the versatile use of alkylating agents and the Structure Activity Relationship (SAR) of each class of these compounds. This could provide an understanding for design and synthesis of new alkylating agents having enhanced target specificity and adequate bioavailability.


Assuntos
Antineoplásicos , Leucemia , Neoplasias , Alquilantes/química , Alquilantes/farmacologia , Alquilantes/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , DNA/química , Humanos , Leucemia/tratamento farmacológico , Mecloretamina/farmacologia , Mecloretamina/uso terapêutico , Neoplasias/tratamento farmacológico
20.
Life (Basel) ; 12(11)2022 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-36431014

RESUMO

In men, prostate cancer (PC) is the most frequently diagnosed cancer, causing an estimated 375,000 deaths globally. Currently, existing therapies for the treatment of PC, notably metastatic cases, have limited efficacy due to drug resistance and problematic adverse effects. Therefore, it is imperative to discover and develop novel drugs for treating PC that are efficacious and do not produce intolerable adverse or toxic effects. Condensed quinolines are naturally occurring anticancer compounds. In this study, we determined the in vitro efficacy of IND-2 (4-chloro-2-methylpyrimido[1″,2″:1,5]pyrazolo[3,4-b]quinolone) in the PC lines, PC-3 and DU-145. IND-2 significantly inhibited the proliferation of PC-3 and DU-145, with IC50 values of 3 µM and 3.5 µM, respectively. The incubation of PC-3 cells with 5 and 10 µM of IND-2 caused the loss of the mitochondrial membrane potential in PC-3 cells. Furthermore, IND-2, at 5 µM, increased the expression of cleaved caspase-3, cleaved caspase-7 and cleaved poly (ADP-ribose) polymerase (PARP). The incubation of PC-3 cells with 5 µM of IND-2 significantly decreased the expression of the apoptotic protein, B-cell lymphoma 2 (Bcl-2). Furthermore, 5 and 10 µM of IND-2 produced morphological changes in PC-3 cells characteristic of apoptosis. Interestingly, IND-2 (2.5, 5 and 10 µM) also induced mitotic catastrophe in PC-3 cells, characterized by the accumulation of multinuclei. The incubation of DU-145 cells with 1.25 and 5 µM of IND-2 significantly increased the levels of reactive oxygen species (ROS). Finally, IND-2, at 10 µM, inhibited the catalytic activity of topoisomerase IIα. Overall, our findings suggest that IND-2 could be a potential lead compound for the development of more efficacious compounds for the treatment of PC.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA