Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Med Chem ; 67(4): 2529-2548, 2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38331432

RESUMO

Tuberculosis (TB) is the leading cause of global morbidity and mortality resulting from infectious disease, with over 10.6 million new cases and 1.4 million deaths in 2021. This global emergency is exacerbated by the emergence of multidrug-resistant MDR-TB and extensively drug-resistant XDR-TB; therefore, new drugs and new drug targets are urgently required. From a whole cell phenotypic screen, a series of azetidines derivatives termed BGAz, which elicit potent bactericidal activity with MIC99 values <10 µM against drug-sensitive Mycobacterium tuberculosis and MDR-TB, were identified. These compounds demonstrate no detectable drug resistance. The mode of action and target deconvolution studies suggest that these compounds inhibit mycobacterial growth by interfering with cell envelope biogenesis, specifically late-stage mycolic acid biosynthesis. Transcriptomic analysis demonstrates that the BGAz compounds tested display a mode of action distinct from the existing mycobacterial cell wall inhibitors. In addition, the compounds tested exhibit toxicological and PK/PD profiles that pave the way for their development as antitubercular chemotherapies.


Assuntos
Azetidinas , Tuberculose Extensivamente Resistente a Medicamentos , Mycobacterium tuberculosis , Tuberculose Resistente a Múltiplos Medicamentos , Humanos , Azetidinas/farmacologia , Azetidinas/uso terapêutico , Antituberculosos/farmacologia , Antituberculosos/uso terapêutico , Tuberculose Resistente a Múltiplos Medicamentos/tratamento farmacológico , Tuberculose Extensivamente Resistente a Medicamentos/tratamento farmacológico , Testes de Sensibilidade Microbiana
2.
Toxicol Lett ; 345: 34-45, 2021 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-33865918

RESUMO

HepG2 cells continue to be a valuable tool in early drug discovery and pharmaceutical development. In the current study we develop a 3D in vitro liver model, using HepG2/C3A cells that is predictive of human genotoxic exposure. HepG2/C3A cells cultured for 7-days in agarose-coated microplates formed spheroids which were uniform in shape and had well defined outer perimeters and no evidence of a hypoxic core. Quantitative real-time-PCR analysis showed statistically significant transcriptional upregulation of xenobiotic metabolising genes (CYP1A1, CYP1A2, UG1A1, UGT1A3, UGT1A6, EPHX, NAT2) and genes linked to liver function (ALB, CAR) in 3D cultures. In response to three model pro-genotoxicants: benzo[a]pyrene, amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) and 2-aminoanthracene (2-AA), we observed further transcriptional upregulation of xenobiotic metabolising genes (CYP1A1, CYP1A2, NAT1/2, SULT1A2, UGT1A1, UGT1A3) compared to untreated spheroids. Consistent with this, spheroids were more sensitive than 2D monolayers to compound induced single- and double- stranded DNA-damage as assessed by the comet assay and γH2AX phosphorylation respectively. In contrast, levels of DNA-damage induced by the direct acting mutagen 4-nitroquinoline N-oxide (4NQO) was the same in spheroids and monolayers. In support of the enhanced genotoxic response in spheroids we also observed transcriptional upregulation of genes relating to DNA-damage and cellular stress response (e.g. GADD45A and CDKN1A) in spheroids. In conclusion, HepG2/C3A 3D spheroids are a sensitive model for in vitro genotoxicity assessment with potential applications in early stage drug development.


Assuntos
4-Nitroquinolina-1-Óxido/toxicidade , Alternativas aos Testes com Animais , Antracenos/toxicidade , Benzo(a)pireno/toxicidade , Ensaio Cometa , Hepatócitos/efeitos dos fármacos , Imidazóis/toxicidade , Fígado/efeitos dos fármacos , 4-Nitroquinolina-1-Óxido/metabolismo , Ativação Metabólica , Antracenos/metabolismo , Benzo(a)pireno/metabolismo , Dano ao DNA , Regulação Enzimológica da Expressão Gênica , Células Hep G2 , Hepatócitos/enzimologia , Hepatócitos/patologia , Histonas/metabolismo , Humanos , Imidazóis/metabolismo , Fígado/enzimologia , Fígado/patologia , Fosforilação , Esferoides Celulares , Fatores de Tempo
3.
Medchemcomm ; 10(8): 1379-1390, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-32952998

RESUMO

Parthenolide is a natural product that exhibits anti-leukaemic activity, however, its clinical use is limited by its poor bioavailability. It may be extracted from feverfew and protocols for growing, extracting and derivatising it are reported. A novel parthenolide derivative with good bioavailability and pharmacological properties was identified through a screening cascade based on in vitro anti-leukaemic activity and calculated "drug-likeness" properties, in vitro and in vivo pharmacokinetics studies and hERG liability testing. In vitro studies showed the most promising derivative to have comparable anti-leukaemic activity to DMAPT, a previously described parthenolide derivative. The newly identified compound was shown to have pro-oxidant activity and in silico molecular docking studies indicate a prodrug mode of action. A synthesis scheme is presented for the production of amine 7 used in the generation of 5f.

4.
Arch Toxicol ; 87(8): 1569-79, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23609606

RESUMO

The carboxylic acid NSAID fenclozic acid exhibited an excellent preclinical safety profile and promising clinical efficacy, yet was withdrawn from clinical development in 1971 due to hepatotoxicity observed in clinical trials. A variety of modern in vitro approaches have been used to explore potential underlying mechanisms. Covalent binding studies were undertaken with [(14)C]-fenclozic acid to investigate the possible role of reactive metabolites. Time-dependent covalent binding to protein was observed in NADPH-supplemented liver microsomes, although no metabolites were detected in these incubations or in reactive metabolite trapping experiments. In human hepatocytes, covalent binding was observed at lower levels than in microsomes and a minor uncharacterizable metabolite was also observed. In addition, covalent binding was observed in incubations undertaken with dog and rat hepatocytes, where a taurine conjugate of the drug was detected. Although an acyl glucuronide metabolite was detected when liver microsomes from human, rat and dog were supplemented with UDPGA, there was no detectable UDPGA-dependent covalent binding. No effects were observed when fenclozic acid was assessed for P450-dependent and P450-independent cytotoxicity to THLE cell lines, time-dependent inhibition of five major human cytochrome P450 enzymes, inhibition of the biliary efflux transporters BSEP and MRP2 or mitochondrial toxicity to THLE or HepG2 cells. These data suggest that Phase 1 bioactivation plays a role in the hepatotoxicity of fenclozic acid and highlight the unique insight into mechanisms of human drug toxicity that can be provided by investigations of biotransformation and covalent binding to proteins.


Assuntos
Microssomos Hepáticos/efeitos dos fármacos , Tiazóis/farmacocinética , Tiazóis/toxicidade , Testes de Toxicidade/métodos , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Linhagem Celular Transformada , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/metabolismo , Cães , Células Hep G2/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Fígado/citologia , Fígado/efeitos dos fármacos , Masculino , Microssomos Hepáticos/metabolismo , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/antagonistas & inibidores , Ratos , Ratos Wistar , Tiazóis/metabolismo
5.
Xenobiotica ; 43(5): 421-31, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23153058

RESUMO

1. In vitro screens are sought as informative, alternatives to the use of animals in vivo and to improve upon the current use of fish liver 9000 g supernatants (S9) in environmental risk assessment. 2. The rates of ethoxyresorufin-O-deethylation (relative to S9 protein) measured under different conditions of culture of rainbow trout hepatocytes were significantly higher than those detected in S9, in the order of freshly isolated hepatocytes > 10-day spheroid cultures > primary hepatocytes in culture > S9. The percentage of conjugated metabolites was also similar between freshly isolated and spheroid cultured hepatocytes (9.9 and 13.5%). 3. The rate of oxidation was enhanced (1.7 fold) when S9 was supplemented with cofactors for phase II conjugation but this was only approximately one tenth of the rate in freshly isolated hepatocytes (7.1 pmol/min/mg S9 protein equivalent). 4. Hepatocytes also hydroxylated ibuprofen, producing two metabolites, in contrast to only one (identified as the 1-hydroxy derivative) using hepatic S9 fractions. 5. Since the bioaccumulation potential of chemicals is often based on un-supplemented S9 in incubations ≥ 1 h when activity declines, it is recommended that predictability would be greatly improved through the use of hepatocyte spheroids, due to their maintenance of activity and longevity.


Assuntos
Monitoramento Ambiental/métodos , Truta/metabolismo , Poluentes Químicos da Água/metabolismo , Xenobióticos/metabolismo , Animais , Modelos Animais , Modelos Químicos
6.
Toxicol Pathol ; 40(8): 1106-16, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22673116

RESUMO

The aims of this study were to assess the utility of the PXB mouse model of a chimeric human/mouse liver in studying human-specific effects of an important human hepatotoxic drug, the PPARγ agonist, troglitazone. When given orally by gavage for 7 days, at dose levels of 300 and 600 ppm, troglitazone induced specific changes in the human hepatocytes of the chimeric liver without an effect on the murine hepatic portions. The human hepatocytes, in the vehicle-treated PXB mouse, showed an accumulation of electron-dense lipid droplets that appeared as clear vacuoles under the light microscope in H&E-stained sections. Following dosing with troglitazone, there was a loss of the large lipid droplets in the human hepatocytes, a decrease in the amount of lipid as observed in frozen sections of liver stained by Oil-red-O, and a decrease in the expression of two bile acid transporters, BSEP and MRP2. None of these changes were observed in the murine remnants of the chimeric liver. No changes were observed in the expression of three CYPs, CYP 3A2, CYP 1A1, and CYP 2B1, in either the human or murine hepatocytes, even though the baseline expression of the enzymes differed significantly between the two hepatocyte species with the mouse hepatocytes consistently showing increased expression of the protein of all three enzymes. This study has shown that the human hepatocytes, in the PXB chimeric mouse liver, retain an essentially normal phenotype in the mouse liver and, the albeit limited CYP enzymes studied show a more human, rather than a murine, expression pattern. In line with this conclusion, the study has shown a differential response of the human versus the mouse hepatocytes, and the effects observed are highly suggestive of a differential handling of the compound by the two hepatocyte species although the exact reasons are not as yet clear. The PXB chimeric mouse system therefore holds the clear potential to explore human hepatic-specific features, such as metabolism, prior to dosing human subjects, and as such should have considerable utility in drug discovery and development.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Cromanos/toxicidade , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Tiazolidinedionas/toxicidade , Quimeras de Transplante , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Animais , Pré-Escolar , Sistema Enzimático do Citocromo P-450/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Feminino , Hepatócitos/metabolismo , Hepatócitos/transplante , Hepatócitos/ultraestrutura , Humanos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos SCID , Camundongos Transgênicos , Proteína 2 Associada à Farmacorresistência Múltipla , Tamanho do Órgão/efeitos dos fármacos , PPAR gama/agonistas , Especificidade da Espécie , Troglitazona
7.
Xenobiotica ; 42(11): 1128-37, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22642803

RESUMO

The pharmacokinetics and biotransformation of midazolam were investigated following single oral doses of 0.1, 1 and 10 mg/kg to chimeric mice with humanised livers (PXB mice) and to severe combined immunodeficient (SCID) mice used as controls. Pharmacokinetic analysis, on whole blood, revealed rapid absorption of the administered midazolam with a higher C(max) in PXB compared to SCID. The exposure to 1'-hydroxymidazolam was approximately 14-fold greater than to midazolam in the SCID mice and close to equivalent in the PXB mice. The metabolism of midazolam in SCID mice was faster than in the PXB mice such that pharmacokinetic data for midazolam in SCID mice could not be generated from the lowest dose in these animals. Both oxidative and conjugative metabolic pathways were identified in the PXB mice. All the major circulating metabolites observed in humans; 1'-hydroxymidazolam, 4'-hydroxymidazolam, 1',4'-dihydroxymidazolam and 1'-hydroxymidazolam glucuronide, were detected in the blood of PXB mice. However, 4'-hydroxymidazolam and the 1'-hydroxymidazolam glucuronide were not detected in blood samples obtained from SCID mice. The midazolam metabolite profile in the PXB mouse was similar to that previously reported for human suggesting that the PXB mouse model can provide a model system for predicting circulating human metabolites.


Assuntos
Hipnóticos e Sedativos/metabolismo , Midazolam/metabolismo , Animais , Biotransformação , Pré-Escolar , Quimera , Humanos , Hipnóticos e Sedativos/sangue , Hipnóticos e Sedativos/farmacocinética , Masculino , Camundongos , Camundongos SCID , Midazolam/análogos & derivados , Midazolam/sangue , Midazolam/farmacocinética
8.
Xenobiotica ; 42(2): 195-205, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21955249

RESUMO

The distribution, metabolism, excretion and hepatic effects of diclofenac were investigated following a single oral dose of 10 mg/kg to wild type and hepatic reductase null (HRN) mice. For the HRN strain the bulk of the [(14)C]-diclofenac-related material was excreted in the urine/aqueous cagewash within 12 h of administration (~82%) with only small amounts eliminated via the faeces (~2% in 24 h). Wild type mice excreted the radiolabel more slowly with ca. 52 and 15% of the dose recovered excreted in urine and faeces, respectively, by 24 h post dose. The metabolic profiles of the HRN mice were dominated by acyl conjugation to either taurine or glucuronic acid. Wild type mice produced relatively small amounts of the acyl glucuronide. Whole Body Autoradiography (WBA) of mice sacrificed at 24 h post dose indicated increased retention of radioactivity in the livers of HRN mice compared to wild type mice. Covalent binding studies showed no differences between the two strains. Metabolism of diclofenac in HRN mice involved mainly acyl glucuronide formation and taurine amide conjugation. This mouse model may find utility in understanding the impact of reactive metabolite formation via routes that involve the production of acyl-CoA or acyl glucuronides of acidic drugs.


Assuntos
Diclofenaco/farmacocinética , Fígado/metabolismo , NADPH-Ferri-Hemoproteína Redutase/genética , Animais , Cromatografia Líquida de Alta Pressão , Diclofenaco/química , Diclofenaco/farmacologia , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Espectrometria de Massas , Taxa de Depuração Metabólica , Desintoxicação Metabólica Fase II , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Xenobióticos/química , Xenobióticos/farmacocinética , Xenobióticos/farmacologia
9.
Xenobiotica ; 42(2): 179-94, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21955289

RESUMO

The metabolism of [(14)C]-diclofenac in mice was investigated following a single oral dose of 10 mg/kg. The majority of the drug-related material was excreted in the urine within 24 h of administration (49.7 %). Liquid chromatographic analyses of urine and faecal extracts revealed extensive metabolism to at least 37 components, with little unchanged diclofenac excreted. Metabolites were identified using a hybrid linear ion-trap mass spectrometer via exact mass determinations of molecular ions and subsequent multi-stage fragmentation. The major routes of metabolism identified included: 1) conjugation with taurine; and 2) hydroxylation (probably at the 4'-and 5-arene positions) followed by conjugation to taurine, glucuronic acid or glucose. Ether, rather than acyl glucuronidation, predominated. There was no evidence for p-benzoquinone-imine formation (i.e. no glutathione or mercapturic acid conjugates were detected). A myriad of novel minor drug-related metabolites were also detected, including ribose, glucose, sulfate and glucuronide ether-linked conjugates of hydroxylated diclofenac derivatives. Combinations of these hydroxylated derivatives with acyl conjugates (glucose, glucuronide and taurine) or N-linked sulfation or glucosidation were also observed. Acyl- or amide-linked-conjugates of benzoic acid metabolites and several indolinone derivatives with further hydroxylated and conjugated moieties were also evident. The mechanisms involved in the generation of benzoic acid and indolinone products indicate the formation reactive intermediates in vivo that may possibly contribute to hepatotoxicity.


Assuntos
Diclofenaco/farmacocinética , Animais , Cromatografia Líquida de Alta Pressão , Descarboxilação , Diclofenaco/química , Diclofenaco/urina , Glucose/química , Glucose/metabolismo , Ácido Glucurônico/química , Ácido Glucurônico/metabolismo , Hidroxilação , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Taurina/química , Taurina/metabolismo
10.
Xenobiotica ; 42(6): 503-17, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22201515

RESUMO

The pharmacokinetics, biotransformation and hepatic transporter effects of troglitazone were investigated following daily oral dosing, at 300 and 600 mg/kg, for 7 days to control (SCID) and chimeric (PXB) mice with humanized livers. Clinical chemistry revealed no consistent pattern of changes associated with troglitazone treatment in the PXB mouse. Human MRP2 but not mouse mrp2 was down-regulated following troglitazone treatment. Pharmacokinetic analysis revealed similar T(max) values for troglitazone in both mouse groups, a mono- and bi-phasic elimination phase in PXB and SCID mice, respectively, but a 3- to 5- and 2- to 5-fold higher C(max) and AUC, respectively, in PXB mice. Oxidative and conjugative metabolic pathways were identified, with the sulfate being the predominant metabolite in PXB compared to SCID mice (4- to 13-fold increase in liver and blood, respectively). The glucuronide conjugate was predominant in SCID mice. There was no evidence of glutathione conjugation. The primary oxidative pathways were mono- and di-oxidations which may also be attributed to quinone or hydroquinone derivatives. Several metabolites were observed in PXB mice only. As the troglitazone metabolic profiles in the PXB mouse were similar to reported human data the PXB mouse model can provide a useful first insight into circulating human metabolites of xenobiotics metabolized in the liver.


Assuntos
Cromanos/farmacologia , Cromanos/farmacocinética , Hepatócitos/transplante , Hipoglicemiantes , Fígado/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Tiazolidinedionas/farmacocinética , Transplante Heterólogo , Animais , Cromanos/metabolismo , Feminino , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos SCID , Proteína 2 Associada à Farmacorresistência Múltipla , Oxirredução , Sulfatos/metabolismo , Tiazolidinedionas/metabolismo , Quimeras de Transplante , Troglitazona
11.
Drug Metab Dispos ; 38(10): 1688-97, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20634336

RESUMO

The in vitro metabolism of cediranib (4-[(4-fluoro-2-methyl-1H-indol-5-yl)oxy]-6-methoxy-7-[3-(1-pyrrolidinyl)propoxy]quinazoline), a vascular endothelial growth factor (VEGF) tyrosine kinase inhibitor (TKI) of all three VEGF receptors in late-stage development for the treatment of colorectal cancer and recurrent glioblastoma was investigated in hepatic proteins from preclinical species and humans using radiolabeled material. In human hepatocyte cultures, oxidative and conjugative metabolic pathways were identified, with pyrrolidine N(+)-glucuronidation being the major route. The primary oxidative pathways were di-and trioxidations and pyrrolidine N-oxidation. All metabolites with the exception of the N(+)-glucuronide metabolite were observed in rat and cynomolgus monkey hepatocyte preparations. Additional metabolism studies in liver microsomes from these or other preclinical species (CD-1 mouse, Han Wistar rat, Dunkin Hartley guinea pig, Göttingen mini-pig, New Zealand White rabbit, beagle dog, and cynomolgus and rhesus monkey) indicated that the N(+)-glucuronide metabolite was not formed in these additional species. Incubations with recombinant flavin-containing monooxygenase (FMO) and UDP-glucuronosyltransferase (UGT) enzymes and inhibition studies using the nonselective cytochrome P450 (P450) chemical inhibitor 1-aminobenzotriazole in human hepatocytes indicated that FMO1 and FMO3 contributed to cediranib N-oxidation, whereas UGT1A4 had a major role in cediranib N(+)-glucuronidation. P450 enzymes had only a minor role in the metabolism of cediranib. In conclusion, species differences in the formation of the N(+)-glucuronide metabolite of cediranib were observed. All other metabolites of cediranib found in humans were also detected in rat and cynomolgus monkey. Non-P450 enzymes are predominantly involved in the metabolism of cediranib, and this suggests that clinical drug interactions involving other coadministered drugs are unlikely.


Assuntos
Fígado/enzimologia , Fígado/metabolismo , Quinazolinas/farmacocinética , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Adulto , Animais , Células Cultivadas , Sistema Enzimático do Citocromo P-450/metabolismo , Cães , Feminino , Glucuronosiltransferase/metabolismo , Cobaias , Hepatócitos/enzimologia , Hepatócitos/metabolismo , Humanos , Macaca fascicularis , Macaca mulatta , Masculino , Desentoxicação Metabólica Fase I , Desintoxicação Metabólica Fase II , Camundongos , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Pessoa de Meia-Idade , Oxigenases/metabolismo , Quinazolinas/metabolismo , Quinazolinas/farmacologia , Coelhos , Ratos , Ratos Wistar , Especificidade da Espécie , Suínos , Porco Miniatura
12.
J Pharm Biomed Anal ; 53(3): 526-36, 2010 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-20409669

RESUMO

Cediranib (4-[(4-fluoro-2-methyl-1H-indol-5-yl)oxy]-6-methoxy-7-[3-(1-pyrrolidinyl)propoxy]quinazoline; RECENTIN), a vascular endothelial growth factor (VEGF) tyrosine kinase inhibitor (TKI) of all three VEGF receptors, is currently in Phase III clinical trials for the first-line treatment of colorectal cancer and the treatment of recurrent glioblastoma. During its clinical development a unique human metabolite, an N(+)-glucuronide, was identified as a major circulating metabolite and one of the major metabolites excreted into faeces. Given the possibility of four sites for the conjugation of the glucuronic acid moiety, determination of the location of the conjugation site on cediranib was warranted. A small quantity of the N(+)-glucuronide metabolite of cediranib was initially generated using recombinant human uridine glucuronosyltransferase 1A4 (UGT1A4) enzymes. The metabolite generated was characterised by HPLC-UV and mass spectrometric (HPLC-MS(n)) detection and confirmed by (1)H NMR spectroscopy. However, the exact site of conjugation could not be determined without generating more of the metabolite. Hence a subsequent biosynthetic scale-up experiment was devised to generate a sufficiently large quantity for full structural characterisation by (1)H NMR spectroscopy. The identity of the N(+)-glucuronide metabolite generated in the UGT1A4 scale-up experiment was confirmed by HPLC-MS(n) and displayed the same retention time, molecular mass and mass fragmentation data as the metabolite generated in previous human liver microsomal and hepatocyte incubations. (1)H NMR spectroscopy clearly showed the characteristic anomeric doublet at approximately 4.7 ppm, which, following irradiation during selective Rotating frame Overhauser Effect Spectroscopy (ROESY) experiments, enabled the site of glucuronidation to be confirmed on the pyrrolidine nitrogen. With the exception of the N(+)-glucuronide metabolite, all other human metabolites of cediranib were observed following incubation with hepatocytes from rat and cynomolgus monkey, the species used for toxicology testing of the drug [6]. As the N(+)-glucuronide was not detected in the preclinical species, it is suggested that its formation is more likely in human and higher primates (great apes), a finding widely supported in the literature.


Assuntos
Glucuronídeos/análise , Quinazolinas/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Cães , Feminino , Cobaias , Hepatócitos/metabolismo , Humanos , Macaca fascicularis , Macaca mulatta , Espectroscopia de Ressonância Magnética , Masculino , Camundongos , Coelhos , Ratos , Ratos Wistar , Suínos , Porco Miniatura
13.
Anal Biochem ; 351(2): 266-72, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16473319

RESUMO

Idiosyncratic adverse drug reactions (ADRs) are one of the most common causes of pharmaceutical withdrawals and labeling changes. Most ADRs are caused by drugs that form reactive species that can bind covalently to macromolecules such as proteins. The current methodology for the measurement of covalent binding relies on the use of radiolabeled material that requires an investment in time and resources not typically expended until later in the discovery process. Efforts are also made to identify reactive intermediates by the use of chemical trapping agents, such as reduced glutathione and cyanide, to form stable adducts that are characterized by liquid chromatography-tandem mass spectrometry and/or nuclear magnetic resonance spectroscopy. Here, we describe a high-throughput assay for the measurement of reactive intermediate formation. The method involves incubation of cold compound with liver microsomes in the presence of [14C]potassium cyanide. Hard electrophilic species would react with the trapping agent, resulting in the formation of a radiolabeled conjugate. Unreacted trapping agent is removed using solid-phase extraction, and the amount of radiolabeled conjugate present is determined by liquid scintillation counting. This newly developed screen has proved to be specific, sensitive, robust, and a powerful tool for assessing bioactivation potential.


Assuntos
Biotransformação , Microssomos Hepáticos/metabolismo , Preparações Farmacêuticas/metabolismo , Autoanálise , Radioisótopos de Carbono , Avaliação Pré-Clínica de Medicamentos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Humanos , Nicotina/metabolismo , Cianeto de Potássio , Contagem de Cintilação/métodos , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...