Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Zhonghua Yu Fang Yi Xue Za Zhi ; 57(1): 35-42, 2023 Jan 06.
Artigo em Chinês | MEDLINE | ID: mdl-36655255

RESUMO

Objective: To investigate the epidemiological characteristics of human adenovirus (HADV) 2, 3 and 7 in hospitalized children with respiratory infection. Methods: A total of 25 686 children with respiratory infection hospitalized at Children's Hospital of Hebei Province from January 2018 to December 2020 were retrospectively included.Deep sputum or nasopharyngeal aspirates of those children were collected. Then thirteen common respiratory pathogens were detected by multiplex PCR. 510 HADV positive specimens were randomly selected via random number and classified for type 2, 3 and 7 using a multiplex real-time quantitative PCR. SPSS 21.0 software was used to perform all of the statistical analyses. Enumeration data were expressed by frequency and percentage. χ2 test was used for comparison between groups. Results: The HADV-positive rate was 7.99% (2 052/25 686). Children at age 3-<6 years had the highest HADV-positive rate (11.44%). The HADV-positive rate in 2019 was highest (10.64%). Among the 510 HADV-positive specimens, the proportion of type 3 was the highest (31.16%), followed by type 7 (21.37%) and type 2 (11.18%). The rate of type 2 in 2019 was significantly lower than that in 2018 and 2020 (χ2=8.954 and 16.354; P=0.003 and <0.01), while the rate of type 3 was significantly higher than that in 2018 and 2020 (χ2=5.248 and 4.811; P=0.022 and 0.028). The rate of type 2, type 3 and type 7 were lowest in winter, spring and autumn, respectively. The rate of type 2 increased significantly in autumn and the rate of type 3 and type 7 increased significantly in winter.The co-detection rate of HADV with other respiratory pathogens was 43.33%(221/510). Among, the co-detection rate of type 3 was highest (47.32%), and the co-detection rate of type 2, 3 and 7 was significantly higher than the alone detection rate (χ2=20.438, P<0.01; χ2=42.105, P<0.01; χ2=27.573, P<0.01).The proportion of severe pneumonia in children with type 7 positive (15.89%) was higher than that in children with non-type 7 positive (8.23%) (χ2=5.260, P=0.022). Conclusion: HADV is one of the important pathogens of children with respiratory infection in Children's Hospital of Hebei Province. The susceptible population of HADV is preschool children aged 3 to 6 years. HADV often co-detects with other respiratory pathogens. Type 3 and 7 is likely to be the dominant genotypes in this region, and type 7 may be one of the risk factors of severe pneumonia in children.


Assuntos
Infecções por Adenovirus Humanos , Adenovírus Humanos , Pneumonia , Infecções Respiratórias , Pré-Escolar , Criança , Humanos , Lactente , Adenovírus Humanos/genética , Criança Hospitalizada , Estudos Retrospectivos , Infecções por Adenovirus Humanos/epidemiologia , Infecções Respiratórias/epidemiologia , Hospitais
2.
Zhonghua Yu Fang Yi Xue Za Zhi ; 55(11): 1321-1327, 2021 Nov 06.
Artigo em Chinês | MEDLINE | ID: mdl-34749476

RESUMO

Objective: To investigate the epidemiological characteristics of human coronavirus (HCoV) in hospitalized children with respiratory tract infection in Hebei region, providing evidence for the diagnosis and prevention of children with respiratory tract infection. Methods: A retrospective study was conducted on 1 062 HCoV positive children hospitalized for respiratory tract infection in Children's Hospital of Hebei Province from January 2015 to December 2020, aged from 33 days to 14 years, with a median age of 2 years. 27 932 (60.9%) were males and 17 944(39.1%) were females. And the gender, ages, seasonal distribution, HCoV-positive rates, co-detection distribution and clinical diagnosis of HCoV positive cases were analyzed by SPSS 25.0. Enumeration data were expressed by frequency and percentage; categorical variable were compared by the Pearson χ2test. Results: The overall HCoV-positive rate was 2.31% (1 062/45 876), which was 2.37% (662/27 932) in male children and 2.23% (400/17 944) in female children. There was no statistically significant difference between genders (χ²=0.916, P=0.339). Children at age groups<1 years (2.44%) and 1-<3 years (2.63%) had higher HCoV-positive rates than those at age groups 3-<5 years (1.97%) and ≥5 years (1.38%) (χ²=27.332,P<0.01). The HCoV-positive rates from 2015 to 2018 were 2.13%, 2.45%, 2.28% and 2.23%. The HCoV-positive rate of 2019 (1.71%) was significantly lower than in 2016 (χ²=12.05, P<0.01), 2017 (χ²=7.34, P=0.01) and 2018 (χ²=6.78, P=0.01), but there was no significant difference compared with 2015 (χ²=2.84, P=0.09). The HCoV-positive rate of 2020 (3.37%) was significantly higher than in 2015 (χ²=13.636, P<0.01), 2016 (χ²=11.099, P<0.01), 2017 (χ²=15.482, P<0.01), 2018(χ²=18.601, P<0.01) and 2019(χ²=45.580, P<0.01). The positive rate was highest in spring (March to May) in 2015 and 2017 to 2018. February to April and July to September of 2016 were the peak periods of positive detection. No obvious seasonal change was observed in 2019 and the HCoV-positive rate of 2020 was extremely low from January to July, following significantly increased from August to December. 26.37% (280/1 062) of HCoV were co-detected with other respiratory pathogens and the most frequently identified mixed detection was RSV. Three or more pathogens were detected in 7.34% (78/1 062) of the HCoV-positive samples. Bronchopneumonia and bronchiolitis were more frequently observed in the single HCoV positive (61.89% and 16.75%) children compared to co-detected children(34.29% and 9.64%)(χ²=63.394 and 8.228, P<0.01). However, compared to those with HCoV mono-detection, co-detected children were more likely to have severe pneumonia (4.6% and 47.14%) (χ²=280.171, P<0.01). Conclusions: HCoV is one of the respiratory pathogens in children in Hebei region and more prevalent in spring. The susceptible population of HCoV is mainly children under the age of 3 years old. HCoV often co-detects with other respiratory pathogens, and the co-infection is one of the risk factors of severe pneumonia in children with respiratory infection.


Assuntos
Coinfecção , Infecções por Coronavirus , Coronavirus , Infecções Respiratórias , Criança , Criança Hospitalizada , Pré-Escolar , Infecções por Coronavirus/epidemiologia , Feminino , Humanos , Lactente , Masculino , Infecções Respiratórias/epidemiologia , Estudos Retrospectivos , Estações do Ano
3.
Xenobiotica ; 33(2): 169-80, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12623759

RESUMO

1. The main purpose was to develop a high-performance liquid chromatography (HPLC)-based method to assay serotonin glucuronidation activity using liver microsomal fractions. Application of this method was then demonstrated by determining serotonin UDP-glucuronosyltransferase (UGT) enzyme kinetics using human liver microsomes and recombinant human UGT1A6. Interspecies differences were also evaluated using liver microsomes from 10 different mammalian species. 2. Incubation of liver microsomes with serotonin, UDP-glucuronic acid and magnesium resulted in the formation of a single product peak using HPLC with fluorescence and ultraviolet absorbance detection. This peak was confirmed as serotonin glucuronide based on sensitivity to beta-glucuronidase and by obtaining the expected mass of 352 with positive-ion mass spectrometry. 3. Following a preparative HPLC isolation, the structure of this metabolite was established as serotonin-5-O-glucuronide by (1)H-NMR spectroscopy. 4. Enzyme kinetic studies showed apparent K(m) and V(max) of 8.8 +/- 0.3 mM and 43.4 +/- 0.4 nmoles min(-1) mg(-1) protein, respectively, for human liver microsomes, and 5.9 +/- 0.2 mM and 15.8 +/- 0.2 nmoles min(-1) mg(-1), respectively, for recombinant UGT1A6. 5. The order of serotonin-UGT activities in animal liver microsomes was rat > mouse > human > cow > pig > horse > dog > rabbit > monkey > ferret. Cat livers showed no serotonin-UGT activity. Heterozygous and homozygous mutant Gunn rat livers had 40 and 13%, respectively, of the activity of the normal Wistar rat, indicating a significant contribution by a rat UGT1A isoform to serotonin glucuronidation. 6. This assay provides a novel sensitive and specific technique for the measurement of serotonin-UGT activity in vitro.


Assuntos
Microssomos Hepáticos/metabolismo , Serotonina/metabolismo , Adulto , Idoso , Animais , Bovinos , Cromatografia Líquida de Alta Pressão , Cães , Feminino , Furões , Glucuronidase/metabolismo , Glucuronídeos/metabolismo , Glucuronosiltransferase/metabolismo , Cavalos , Humanos , Técnicas In Vitro , Cinética , Espectroscopia de Ressonância Magnética , Masculino , Espectrometria de Massas , Camundongos , Pessoa de Meia-Idade , Coelhos , Ratos , Proteínas Recombinantes/metabolismo , Especificidade da Espécie , Suínos
4.
J Pharmacol Exp Ther ; 299(3): 998-1006, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11714888

RESUMO

Interindividual variability in acetaminophen (APAP) glucuronidation may contribute to differences in susceptibility to APAP intoxication in humans. The purpose of this study was to identify the relevant UDP-glucuronosyltransferase (UGT) isoforms mediating APAP-UGT activity in human liver microsomes (HLMs). APAP-UGT activities and enzyme kinetics were determined using HLMs from 56 donors and nine recombinant human UGTs. Activities mediated by UGT1A1, UGT1A4, UGT1A9, and UGT2B7, and relative UGT1A6 protein content were quantified using 20 livers. More than 15-fold variation in liver microsomal APAP-UGT activities was observed with a distribution skewed toward lower activities. Although most UGTs could glucuronidate APAP, UGT1A1, UGT1A6, and UGT1A9 were most active. UGT1A6 was a relatively high-affinity (K(m) = 2.2 mM), low-capacity enzyme; UGT1A1 was intermediate in affinity (K(m) = 9.4 mM) and capacity; and UGT1A9 was a low-affinity (K(m) = 21 mM), high-capacity enzyme. K(m) values were similar to UGT1A1 in high- and intermediate-activity HLMs (6-10 mM) and UGT1A9 in low-activity HLMs (10-55 mM). APAP-UGT activities correlated best with propofol-UGT (r = 0.85; UGT1A9) and bilirubin-UGT (r = 0.66; UGT1A1) activities, but poorly with UGT1A6 protein (r = 0.30). A kinetic model was constructed from these data that identified UGT1A9 as the predominant APAP-UGT (>55% total activity) in HLMs over a clinically relevant APAP concentration range (50 microM-5 mM). UGT1A1 was also predicted to contribute substantially at toxic concentrations (>1 mM; >28% activity), whereas UGT1A6 was most active at relatively low concentrations (<50 microM; >29% activity).


Assuntos
Acetaminofen/análogos & derivados , Acetaminofen/análise , Acetaminofen/metabolismo , Glucuronosiltransferase/isolamento & purificação , Isoenzimas/isolamento & purificação , Microssomos Hepáticos/metabolismo , Glucuronosiltransferase/metabolismo , Humanos , Técnicas In Vitro , Isoenzimas/metabolismo , Cinética , Microssomos Hepáticos/enzimologia , Fatores Sexuais , Estatística como Assunto
5.
J Clin Pharmacol ; 41(1): 85-91, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11225565

RESUMO

The capacity of three clinically available nonnucleoside reverse transcriptase inhibitors (NNRTIs) to inhibit the activity of human cytochromes P450 (CYPs) was studied in vitro using human liver microsomes. Delavirdine, nevirapine, and efavirenz produced negligible inhibition of phenacetin O-deethylation (CYP1A2) or dextromethorphan O-demethylation (CYP2D6). Nevirapine did not inhibit hydroxylation of tolbutamide (CYP2C9) or S-mephenytoin (CYP2C19), but these CYP isoforms were importantly inhibited by delavirdine and efavirenz. This indicates the likelihood of significantly impaired clearance of CYP2C substrate drugs (such as phenytoin, tolbutamide, and warfarin) upon initial exposure to these two NNRTIs. Delavirdine and efavirenz (but not nevirapine) also were strong inhibitors of CYP3A, consistent with clinical hazards of initial cotreatment with either of these drugs and substrates of CYP3A. The in vitro microsomal model provides relevant predictive data on probable drug interactions with NNRTIs when the mechanism is inhibition of CYP-mediated drug biotransformation. However, the model does not incorporate interactions attributable to enzyme induction.


Assuntos
Fármacos Anti-HIV/farmacologia , Inibidores das Enzimas do Citocromo P-450 , Inibidores da Transcriptase Reversa/farmacologia , Alcinos , Benzoxazinas , Ciclopropanos , Sistema Enzimático do Citocromo P-450/metabolismo , Delavirdina/farmacologia , Humanos , Hidrólise , Concentração Inibidora 50 , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Nevirapina/farmacologia , Oxazinas/farmacologia , Triazolam/metabolismo
6.
Anesthesiology ; 94(1): 110-9, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11135730

RESUMO

BACKGROUND: Oxidation of propofol to 4-hydroxypropofol represents a significant pathway in the metabolism of this anesthetic agent in humans. The aim of this study was to identify the principal cytochrome P-450 (CYP) isoforms mediating this biotransformation. METHODS: Propofol hydroxylation activities and enzyme kinetics were determined using human liver microsomes and cDNA-expressed CYPs. CYP-specific marker activities and CYP2B6 protein content were also quantified in hepatic microsomes for correlational analyses. Finally, inhibitory antibodies were used to ascertain the relative contribution of CYPs to propofol hydroxylation by hepatic microsomes. RESULTS: Propofol hydroxylation by hepatic microsomes showed more than 19-fold variability and was most closely correlated to CYP2B6 protein content (r = 0.904), and the CYP2B6 marker activities, S-mephenytoin N-demethylation (r = 0.919) and bupropion hydroxylation (r = 0.854). High- and intermediate-activity livers demonstrated high-affinity enzyme kinetics (K(m) < 8 microm), whereas low-activity livers displayed low-affinity kinetics (K(m) > 80 microm). All of the CYPs evaluated were capable of hydroxylating propofol; however, CYP2B6 and CYP2C9 were most active. Kinetic analysis indicated that CYP2B6 is a high-affinity (K(m) = 10 +/- 2 microm; mean +/- SE of the estimate), high-capacity enzyme, whereas CYP2C9 is a low-affinity (K(m) = 41 +/- 8 microm), high-capacity enzyme. Furthermore, immunoinhibition showed a greater contribution of CYP2B6 (56 +/- 22% inhibition; mean +/- SD) compared with CYP2C isoforms (16 +/- 7% inhibition) to hepatic microsomal activity. CONCLUSIONS: Cytochrome P-450 2B6, and to a lesser extent CYP2C9, contribute to the oxidative metabolism of propofol. However, CYP2B6 is the principal determinant of interindividual variability in the hydroxylation of this drug by human liver microsomes.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/fisiologia , Microssomos Hepáticos/metabolismo , Oxirredutases N-Desmetilantes/fisiologia , Propofol/metabolismo , Esteroide 16-alfa-Hidroxilase , Biotransformação , Citocromo P-450 CYP2B6 , Citocromo P-450 CYP2C9 , Sistema Enzimático do Citocromo P-450/metabolismo , Sistema Enzimático do Citocromo P-450/farmacologia , Humanos , Hidroxilação , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Oxirredutases N-Desmetilantes/metabolismo , Propofol/farmacocinética , Esteroide Hidroxilases/metabolismo , Esteroide Hidroxilases/farmacologia
7.
Drug Metab Dispos ; 28(10): 1176-83, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10997936

RESUMO

The in vitro biotransformation of bupropion to hydroxybupropion was studied in human liver microsomes and microsomes containing heterologously expressed human cytochromes P450 (CYP). The mean (+/-S.E.) K(m) in four human liver microsomes was 89 (+/-14) microM. In microsomes containing cDNA-expressed CYPs, hydroxybupropion formation was mediated only by CYP2B6 at 50 microM bupropion (K(m) 85 microM). A CYP2B6 inhibitory antibody produced more than 95% inhibition of bupropion hydroxylation in four human livers. Bupropion hydroxylation activity at 250 microM was highly correlated with S-mephenytoin N-demethylation activity (yielding nirvanol), another CYP2B6-mediated reaction, in a panel of 32 human livers (r = 0.94). The CYP2B6 content of 12 human livers highly correlated with bupropion hydroxylation activity (r = 0.96). Thus bupropion hydroxylation is mediated almost exclusively by CYP2B6 and can serve as an index reaction reflecting activity of this isoform. IC(50) values for inhibition of a CYP2D6 index reaction (dextromethorphan O-demethylation) by bupropion and hydroxybupropion were 58 and 74 microM, respectively. This suggests a low inhibitory potency versus CYP2D6, the clinical importance of which is not established. Since bupropion is frequently coadministered with other antidepressants, IC(50) values (microM) for inhibition of bupropion hydroxylation were determined as follows: paroxetine (1.6), fluvoxamine (6.1), sertraline (3.2), desmethylsertraline (19.9), fluoxetine (59.5), norfluoxetine (4.2), and nefazodone (25.4). Bupropion hydroxylation was only weakly inhibited by venlafaxine, O-desmethylvenlafaxine, citalopram, and desmethylcitalopram. The inhibition of bupropion hydroxylation in vitro by a number of newer antidepressants suggests the potential for clinical drug interactions.


Assuntos
Antidepressivos de Segunda Geração/metabolismo , Hidrocarboneto de Aril Hidroxilases , Bupropiona/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Fluoxetina/análogos & derivados , Oxirredutases N-Desmetilantes/metabolismo , Sertralina/análogos & derivados , Anticorpos/farmacologia , Antidepressivos de Segunda Geração/farmacocinética , Biotransformação , Bupropiona/farmacocinética , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP2B6 , Sistema Enzimático do Citocromo P-450/imunologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Fluoxetina/farmacologia , Fluvoxamina/farmacologia , Humanos , Hidroxilação/efeitos dos fármacos , Isoenzimas/metabolismo , Cinética , Microssomos Hepáticos/metabolismo , Oxirredutases N-Desmetilantes/imunologia , Paroxetina/farmacologia , Piperazinas , Sertralina/farmacologia , Triazóis/farmacologia
8.
Eur J Clin Pharmacol ; 56(3): 259-61, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10952482

RESUMO

OBJECTIVE: Biotransformation of triazolam to its alpha-hydroxy and 4-hydroxy metabolites by human liver microsomes in vitro was used as an index of human cytochrome P450 3A (CYP3A) activity. RESULTS: The reaction was strongly inhibited by co-incubation with the viral protease inhibitors ritonavir (IC50 = 0.14 microM) and amprenavir (IC50 = 2.5 2.9 microM), and by the azole derivative ketoconazole (IC50 = 0.07 microM). Pre-incubation of microsomes with ritonavir or amprenavir increased inhibitory potency (IC50 reduced to 0.07 microM and 1.4 microM, respectively). This was not the case with ketoconazole. CONCLUSIONS: Thus, ritonavir and amprenavir are highly potent mechanism-based inhibitors of human CYP3A isoforms.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Inibidores das Enzimas do Citocromo P-450 , Inibidores Enzimáticos/farmacologia , Cetoconazol/farmacologia , Oxirredutases N-Desmetilantes/antagonistas & inibidores , Ritonavir/farmacologia , Sulfonamidas/farmacologia , Carbamatos , Citocromo P-450 CYP3A , Furanos , Humanos , Hidroxilação , Triazolam/metabolismo
9.
Biol Psychiatry ; 46(6): 839-49, 1999 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10494454

RESUMO

BACKGROUND: Biotransformation of citalopram (CT), a newly available selective serotonin reuptake inhibitor antidepressant, to its principal metabolite, desmethycitalopram (DCT), and the capacity of CT and DCT to inhibit human cytochromes P450, were studied in vitro. METHODS: Formation of DCT from CT was evaluated using human liver microsomes and microsomes from cDNA-transfected human lymphoblastoid cells. Cytochrome inhibition by CT and DCT in liver microsomes was studied using isoform-specific index reactions. RESULTS: Formation of DCT from CT in liver microsomes had a mean apparent K(m) of 174 mumol/L. Coincubation with 1 mumol/L ketoconazole reduced reaction velocity to 46 to 58% of control values, while omeprazole, 10 mumol/L, reduced velocity to 80% of control. Quinidine produced minimal inhibition. DCT was formed from CT by heterologously expressed human P450-2D6, -2C19, -3A4. After accounting for the relative abundance of individual cytochromes, 3A4 and 2C19 were estimated to make major contributions to net reaction velocity, with a possible contribution of 2D6 at therapeutic CT concentrations. CT and DCT themselves produced negligible inhibition of 2C9, 2E1, and 3A, and only weak inhibition of 1A2, 2C19, and 2D6. CONCLUSIONS: Formation of DCT from CT is mediated mainly by P450-3A4 and 2C19, with an additional contribution of 2D6. CT at therapeutic doses in humans may produce a small degree of inhibition of P450-1A2, -2C19, and -2D6, but negligible inhibition of P450-2C9, -2E1, and -3A.


Assuntos
Citalopram/análogos & derivados , Citalopram/farmacocinética , Citocromos/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacocinética , Biotransformação/fisiologia , Linhagem Celular Transformada/efeitos dos fármacos , Células Cultivadas , Cromatografia Líquida de Alta Pressão/métodos , DNA Complementar/efeitos dos fármacos , Humanos , Técnicas In Vitro , Microssomos Hepáticos/metabolismo , Transfecção/efeitos dos fármacos
10.
Br J Clin Pharmacol ; 48(1): 89-97, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10383565

RESUMO

AIMS: To determine the human cytochromes mediating biotransformation of the imidazopyridine hypnotic, zolpidem, and the clinical correlates of the findings. METHODS: Kinetic properties of zolpidem biotransformation to its three hydroxylated metabolites were studied in vitro using human liver microsomes and heterologously expressed individual human cytochromes. RESULTS: The metabolic product termed M-3 accounted for more than 80% of net intrinsic clearance by liver microsomes in vitro. Microsomes containing human cytochromes CYP1A2, 2C9, 2C19, 2D6, and 3 A4 expressed by cDNA-transfected human lymphoblastoid cells mediated zolpidem metabolism in vitro. The kinetic profile for zolpidem metabolite formation by each individual cytochrome was combined with estimated relative abundances based on immunological quantification, yielding projected contributions to net intrinsic clearance of: 61% for 3 A4, 22% for 2C9, 14% for 1A2, and less than 3% for 2D6 and 2C19. These values were consistent with inhibitory effects of ketoconazole and sulfaphenazole on zolpidem biotransformation by liver microsomes. Ketoconazole had a 50% inhibitory concentration (IC50 ) of 0.61 microm vs formation of the M-3 metabolite of zolpidem in vitro; in a clinical study, ketoconazole coadministration reduced zolpidem oral clearance by approximately 40%, somewhat less than anticipated based on the IC50 value and total plasma ketoconazole levels, but much more than predicted based on unbound plasma ketoconazole levels. CONCLUSIONS: The incomplete dependence of zolpidem clearance on CYP3A activity has clinical implications for susceptibility to metabolic inhibition.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Hipnóticos e Sedativos/metabolismo , Microssomos Hepáticos/metabolismo , Piridinas/metabolismo , Biotransformação , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/genética , Inibidores Enzimáticos/farmacologia , Humanos , Técnicas In Vitro , Cetoconazol/farmacologia , Taxa de Depuração Metabólica , Transfecção , Células Tumorais Cultivadas , Zolpidem
11.
J Pharm Sci ; 87(10): 1184-9, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9758674

RESUMO

Pharmacokinetic drug interactions with viral protease inhibitors are of potential clinical importance. An in vitro model was applied to the quantitative identification of possible interactions of protease inhibitors with substrates of cytochrome P450-2D6. Biotransformation of desipramine (DMI) to hydroxydesipramine (OH-DMI), an index reaction used to profile activity of human cytochrome P450-2D6, was studied in vitro using human liver microsomes. Quinidine and four viral protease inhibitors currently used to treat human immunodeficiency virus infection were tested as chemical inhibitors in this system. Formation of OH-DMI from DMI was consistent with Michaelis-Menten kinetics, having a mean Km value of 11.7 microM (range: 9.9-15.3 microM). Quinidine, a highly potent and relatively selective inhibitor of P450-2D6, strongly inhibited OH-DMI formation with an apparent competitive mechanism, having a mean inhibition constant of 0.16 microM (range: 0.13-0.18 microM). All four protease inhibitors impaired OH-DMI formation; the pattern was consistent with a mixed competitive-noncompetitive mechanism. Mean inhibition constants (small numbers indicating greater inhibiting potency) were as follows: ritonavir, 4.8 microM; indinavir, 15.6 microM; saquinavir, 24.0 microM; nelfinavir, 51.9 microM. In a clinical pharmacokinetic study, coadministration of ritonavir with DMI inhibited DMI clearance by an average of 59%. The in vitro findings, together with observed plasma ritonavir concentrations, provided a reasonable quantitative forecast of this interaction, whereas estimated unbound plasma or intrahepatic ritonavir concentrations yielded poor quantitative forecasts. Thus the in vitro model correctly identifies ritonavir as a potent and clinically important inhibitor of human P450-2D6. Other protease inhibitors may also inhibit 2D6 activity in humans, but with lower potency than ritonavir.


Assuntos
Antidepressivos Tricíclicos/antagonistas & inibidores , Inibidores do Citocromo P-450 CYP2D6 , Desipramina/antagonistas & inibidores , Inibidores da Protease de HIV/farmacologia , Quinidina/farmacologia , Ritonavir/farmacologia , Antidepressivos Tricíclicos/farmacocinética , Área Sob a Curva , Biotransformação , Citocromo P-450 CYP2D6/metabolismo , Desipramina/farmacocinética , Humanos , Hidroxilação , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo
12.
Clin Pharmacol Ther ; 64(3): 278-85, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9757151

RESUMO

BACKGROUND: Macrolide antimicrobial agents may impair hepatic clearance of drugs metabolized by cytochrome P4503A isoforms. Potential interactions of triazolam, a substrate metabolized almost entirely by cytochrome P4503A in humans, with 3 commonly prescribed macrolides were identified using an in vitro metabolic model. The actual interactions, and their pharmacodynamic consequences, were verified in a controlled clinical study. METHODS: In an in vitro model using human liver microsomes, 250 mumol/L triazolam was incubated with ascending concentrations (0 to 250 mumol/L of troleandomycin, azithromycin, erythromycin, and clarithromycin. In a randomized, double-blind, 5-trial clinical pharmacokinetic-pharmacodynamic study, 12 volunteers received 0.125 mg triazolam orally, together with placebo, azithromycin, erythromycin, or clarithromycin. In a fifth trial they received placebo plus placebo. RESULTS: Mean 50% inhibitory concentrations versus 4-hydroxytriazolam formation in vitro were as follows: 3.3 mumol/L troleandomycin, 27.3 mumol/L erythromycin, 25.2 mumol/L clarithromycin, and greater than 250 mumol/L azithromycin. Apparent oral clearance of triazolam when given with placebo or azithromycin was nearly identical (413 and 416 mL/min), as were peak plasma concentrations (1.25 and 1.32 ng/mL) and elimination half-life (2.7 and 2.6 hours). Apparent oral clearance was significantly reduced (P < .05) during erythromycin and clarithromycin trials (146 and 95 mL/min). Peak plasma concentration was correspondingly increased, and elimination half-life was prolonged. The effects of triazolam on dynamic measures were nearly identical when triazolam was given with placebo or azithromycin, but benzodiazepine agonist effects were enhanced during erythromycin and clarithromycin trials. CONCLUSION: The in vitro model identifies macrolides that may impair triazolam clearance. Anticipated interactions, and their pharmacodynamic consequences in volunteer subjects, were verified in vivo.


Assuntos
Antibacterianos/farmacologia , Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Hipnóticos e Sedativos/farmacocinética , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Oxirredutases N-Desmetilantes/efeitos dos fármacos , Triazolam/farmacocinética , Administração Oral , Azitromicina/farmacologia , Claritromicina/farmacologia , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/metabolismo , Método Duplo-Cego , Eritromicina/farmacologia , Humanos , Hipnóticos e Sedativos/metabolismo , Técnicas In Vitro , Microssomos Hepáticos/enzimologia , Oxirredutases N-Desmetilantes/metabolismo , Fatores de Tempo , Triazolam/metabolismo , Troleandomicina/farmacologia
13.
J Clin Psychopharmacol ; 18(4): 338-41, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9690701

RESUMO

The activity of D-fenfluramine, L-fenfluramine, and phentermine as inhibitors of five human cytochromes P450 was evaluated using human liver microsomes in vitro. All three compounds produced negligible inhibition of P450-1A2, -2C9, -2E1, and -3A. Phentermine also did not inhibit P450-2D6. However, D- and L-fenfluramine significantly inhibited P450-2D6 activity as measured by dextromethorphan O-demethylation, with mean 50% inhibitory concentrations (15.1 microM) within one order of magnitude of that for fluoxetine (2.7 microM). Findings from the in vitro assay are consistent with clinical studies showing significant inhibition of desipramine clearance by coadministration of fenfluramine.


Assuntos
Depressores do Apetite/farmacologia , Inibidores do Citocromo P-450 CYP2D6 , Fenfluramina/farmacologia , Microssomos Hepáticos/efeitos dos fármacos , Fentermina/farmacologia , Técnicas de Cultura , Humanos , Microssomos Hepáticos/enzimologia
14.
J Pharm Pharmacol ; 50(3): 265-74, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9600717

RESUMO

The kinetics of the N-demethylation of adinazolam to N-desmethyladinazolam (NDMAD), and of NDMAD to didesmethyladinazolam (DDMAD), were studied with human liver microsomes using substrate concentrations in the range 10-1000 microM. The specific cytochrome P450 (CYP) isoforms mediating the biotransformations were identified using microsomes containing specific recombinant CYP isozymes expressed in human lymphoblastoid cells, and by the use of CYP isoform-selective chemical inhibitors. Adinazolam was demethylated by human liver microsomes to NDMAD, and NDMAD was demethylated to DDMAD; the substrate concentrations, Km, at which the reaction velocities were 50% of the maximum were 92 and 259 microM, respectively. Another metabolite of yet undetermined identity (U) was also formed from NDMAD (Km 498 microM). Adinazolam was demethylated by cDNA-expressed CYP 2C19 (Km 39 microM) and CYP 3A4 (Km 83 microM); no detectable activity was observed for CYPs 1A2, 2C9, 2D6 and 2E1. Ketoconazole, a relatively specific CYP 3A4 inhibitor, inhibited the reaction; the concentration resulting in 50% of maximum inhibition, IC50, was 0.15 microM and the inhibition constant, Ki, was < 0.04 microM in five of six livers tested. Troleandomycin, a specific inhibitor of CYP 3A4, inhibited adinazolam N-demethylation with an IC50 of 1.96 microM. The CYP 2C19-inhibitor omeprazole resulted in only partial inhibition (IC50 21 microM) and sulphaphenazole, alpha-naphthoflavone, quinidine and diethyldithiocarbamate did not inhibit the reaction. NDMAD was demethylated by cDNA-expressed CYP 3A4 (Km 220 microM, Hill number A 1.21), CYP 2C19 (Km 187 microM, Hill number A 1.29) and CYP 2C9 (Km 1068 microM). Formation of U was catalysed by CYP 3A4 alone. Ketoconazole strongly inhibited NDMAD demethylation (IC50 0.14 microM) and formation of U (IC50 < 0.1 microM) whereas omeprazole and sulphaphenazole had no effect on reaction rates. These results show that CYP 3A4 is the primary hepatic CYP isoform mediating the N-demethylation of adinazolam and NDMAD. Co-administration of adinazolam with CYP 3A4 inhibitors such as ketoconazole or erythromycin might lead to reduced efficacy, since adinazolam by itself has relatively weak benzodiazepine agonist activity, with much of the pharmacological activity of adinazolam being attributable to its active metabolite NDMAD.


Assuntos
Ansiolíticos/metabolismo , Hidrocarboneto de Aril Hidroxilases , Benzodiazepinas/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Microssomos Hepáticos/enzimologia , Oxigenases de Função Mista/metabolismo , Esteroide 16-alfa-Hidroxilase , Biotransformação , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP2C19 , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP2E1/metabolismo , Citocromo P-450 CYP3A , Inibidores das Enzimas do Citocromo P-450 , Inibidores Enzimáticos/farmacologia , Humanos , Técnicas In Vitro , Cetoconazol/farmacologia , Cinética , Macrolídeos , Oxigenases de Função Mista/antagonistas & inibidores , Omeprazol/farmacologia , Esteroide Hidroxilases/metabolismo
15.
J Clin Pharmacol ; 38(2): 106-11, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9549640

RESUMO

Four protease inhibitor antiviral agents (ritonavir, indinavir, nelfinavir, saquinavir) were evaluated as in vitro inhibitors of the activity of six human cytochromes using an in vitro model based on human liver microsomes. Ritonavir was a highly potent inhibitor of P450-3A activity (triazolam hydroxylation), having inhibitory potency slightly less than ketoconazole. Indinavir was also a potent 3A inhibitor, while nelfinavir and saquinavir were less potent. Ritonavir had high inhibition potency against cytochrome P450-2C9 (tolbutamide hydroxylation), -2C19 (S-mephenytoin hydroxylation), and -2D6 (dextromethorphan O-demethylation and desipramine hydroxylation), while the other protease inhibitors had one or more orders of magnitude lower inhibitory activity against these reactions. None of the protease inhibitors had important inhibitory potency against P450-1A2 (phenacetin O-deethylation) or -2E1 (chlorzoxazone hydroxylation). Thus, among available protease inhibitors, ritonavir carries the highest risk of incurring drug interactions due to inhibition of cytochrome P450 activity.


Assuntos
Antivirais/farmacologia , Inibidores das Enzimas do Citocromo P-450 , Inibidores da Protease de HIV/farmacologia , Microssomos Hepáticos/efeitos dos fármacos , Ritonavir/farmacologia , Antivirais/efeitos adversos , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Inibidores da Protease de HIV/efeitos adversos , Humanos , Microssomos Hepáticos/enzimologia , Fatores de Risco , Ritonavir/efeitos adversos
16.
Psychopharmacology (Berl) ; 132(4): 402-7, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9298519

RESUMO

Biotransformation of the selective serotonin reuptake inhibitor antidepressant, fluoxetine, to its principal metabolite, norfluoxetine, was evaluated in human liver microsomes and in microsomes from transfected cell lines expressing pure human cytochromes. In human liver microsomes, formation of norfluoxetine from R,S-fluoxetine was consistent with Michaelis-Menten kinetics (mean K(m) = 33 microM), with evidence of substrate inhibition at high substrate concentrations in a number of cases. The reaction was minimally inhibited by coincubation with chemical probes inhibitory for P450-2D6 (quinidine), -1A2 (furafylline, alpha-naphthoflavone), and -2E1 (diethyldithiocarbamate). Substantial inhibition was produced by coincubation with sulfaphenazole (Ki = 2.8 microM), an inhibitory probe for P450-2C9, and by ketoconazole (Ki = 2.5 microM) and fluvoxamine (Ki = 5.2 microM). However, ketoconazole, relatively specific for P450-3A isoforms only at low concentrations, reduced norfluoxetine formation by only 20% at 1 microM, and triacetyloleandomycin (> or = 5 microM) reduced the velocity by only 20-25%. Microsomes from cDNA-transfected human lymphoblastoid cells containing human P450-2C9 produced substantial quantities of norfluoxetine when incubated with 100 microM fluoxetine. Smaller amounts of product were produced by P450-2C19 and -2D6, but no product was produced by P450-1A2, -2E1, or 3A4. Cytochrome P450-2C9 appears to be the principal human cytochrome mediating fluoxetine N-demethylation. P450-2C19 and -3A may make a further small contribution, but P450-2D6 is unlikely to make an important contribution.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Citocromo P-450 CYP2E1/metabolismo , Fluoxetina/metabolismo , Microssomos Hepáticos/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/metabolismo , Esteroide 16-alfa-Hidroxilase , Sistema Enzimático do Citocromo P-450/metabolismo , Fluoxetina/análogos & derivados , Humanos , Esteroide Hidroxilases/metabolismo
18.
Psychopharmacology (Berl) ; 128(4): 398-407, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8986010

RESUMO

Biotransformation of phenacetin via O-deethylation to acetaminophen, an index reaction reflecting activity of Cytochrome P450-1A2, was studied in microsomal preparations from a series of human livers. Acetaminophen formation was consistent with a double Michaelis-Menten system, with low-Km (mean Km1 = 68 microM) and high-Km (mean Km2 = 7691 microM) components. The low-K(m) enzyme accounted for an average of 96% of estimated intrinsic clearance, and was predicted to contribute more than 50% of net reaction velocity at phenacetin concentrations less than 2000 microM. Among index inhibitor probes, alpha-naphthoflavone was a highly potent inhibitor of the low-Km enzyme (Ki1 = 0.013 microM); furafylline also was a moderately active inhibitor (Ki1 = 4.4 microM), but its inhibiting potency was increased by preincubation with microsomes. Ketoconazole was a relatively weak inhibitor (Ki1 = 32 microM); quinidine and cimetidine showed minimal inhibiting activity. Among six selective serotonin reuptake inhibitor (SSRI) antidepressants, fluvoxamine was a potent inhibitor of 1A2 (mean Ki1 = 0.24 microM). The other SSRIs were more than tenfold less potent. Mean Ki1 values were: fluoxetine, 4.4 microM; norfluoxetine, 15.9 microM; sertraline, 8.8 microM; desmethylsertraline, 9.5 microM; paroxetine, 5.5 microM. The antidepressant nefazodone and four of its metabolites (meta-chloro-phenylpiperazine, two hydroxylated derivatives, and a triazoledione) were very weak inhibitors of P450-1A2. Venlafaxine and its O- and N-desmethyl metabolites showed minimal inhibitory activity.


Assuntos
Antidepressivos de Segunda Geração/farmacologia , Cicloexanóis/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Inibidores Enzimáticos/farmacologia , Microssomos Hepáticos/efeitos dos fármacos , Fenacetina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Triazóis/farmacologia , Biotransformação/efeitos dos fármacos , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Inibidores das Enzimas do Citocromo P-450 , Humanos , Microssomos Hepáticos/metabolismo , Piperazinas , Cloridrato de Venlafaxina
19.
J Clin Pharmacol ; 36(9): 783-91, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8889898

RESUMO

Biotransformation of the imidazobenzodiazepine midazolam to its alpha-hydroxy and 4-hydroxy metabolites was studied in vitro using human liver microsomal preparations. Formation of alpha-hydroxy-midazolam was a high-affinity (Km = 3.3 mumol/L) Michaelis-Menten process coupled with substrate inhibition at high concentrations of midazolam. Formation of 4-hydroxy-midazolam had much lower apparent affinity (57 mumol/L), with minimal evidence of substrate inhibition. Based on comparison of Vmax/Km ratios for the two pathways, alpha-hydroxy-midazolam formation was estimated to account for 95% of net intrinsic clearance. Three azole antifungal agents were inhibitors of midazolam metabolism in vitro, with inhibition being largely consistent with a competitive mechanism. Mean competitive inhibition constants (Ki) versus alpha-hydroxy-midazolam formation were 0.0037 mumol/L for ketoconazole, 0.27 mumol/L for itraconazole, and 1.27 mumol/L for fluconazole. An in vitro-in vivo scaling model predicted inhibition of oral midazolam clearance due to coadministration of ketoconazole or itraconazole; the predicted inhibition was consistent with observed interactions in clinical pharmacokinetic studies. The selective serotonin reuptake inhibitor (SSRI) antidepressant fluoxetine and its principal metabolite, norfluoxetine, also were inhibitors of both pathways of midazolam biotransformation, with norfluoxetine being a much more potent inhibitor than was fluoxetine itself. This finding is consistent with results of other in vitro studies and of clinical studies, indicating that fluoxetine, largely via its metabolite norfluoxetine, may impair clearance of P450-3A substrates.


Assuntos
Anestésicos Intravenosos/metabolismo , Ansiolíticos/metabolismo , Antifúngicos/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Fluoxetina/análogos & derivados , Fluoxetina/farmacologia , Microssomos Hepáticos/efeitos dos fármacos , Midazolam/metabolismo , Oxigenases de Função Mista/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Citocromo P-450 CYP3A , Inibidores das Enzimas do Citocromo P-450 , Interações Medicamentosas , Humanos , Hidroxilação , Cinética , Microssomos Hepáticos/enzimologia , Oxigenases de Função Mista/antagonistas & inibidores
20.
J Clin Psychopharmacol ; 16(2): 104-12, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8690825

RESUMO

Biotransformation of the H-1 antagonist terfenadine to its desalkyl and hydroxy metabolites was studied in vitro using microsomal preparations of human liver. These metabolic reactions are presumed to be mediated by Cytochrome P450-3A isoforms. The azole antifungal agent ketoconazole was a highly potent inhibitor of both reactions, having mean inhibition constants (Ki) of 0.037 and 0.34 microM for desalkyl- and hydroxy-terfenadine formation, respectively. Itraconazole also was a potent inhibitor, with Ki values of 0.28 and 2.05 microM, respectively. Fluconazole, on the other hand, was a weak inhibitor. Six selective serotonin reuptake inhibitor antidepressants tested in this system were at least 20 times less potent inhibitors of terfenadine metabolism than was ketoconazole. An in vitro-in vivo scaling model used in vitro Ki values, typical clinically relevant plasma concentrations of inhibitors, and presumed liver:plasma partition ratios to predict the degree of terfenadine clearance impairment during coadministration of terfenadine with these inhibitors in humans. The model predicted a large and potentially hazardous impairment of terfenadine clearance by ketoconazole and, to a slightly lesser extent, by itraconazole. However, fluconazole and the six selective serotonin reuptake inhibitors (SSRIs) at usual clinical doses were not predicted to impair terfenadine clearance to a degree that would be of clinical importance. Caution is nonetheless warranted with the coadministration of SSRIs and terfenadine when high doses of SSRIs (particularly fluoxetine) are administered. Also, some individuals may be unusually susceptible to metabolic inhibition for a variety of reasons.


Assuntos
Antifúngicos/farmacologia , Hidrocarboneto de Aril Hidroxilases , Inibidores das Enzimas do Citocromo P-450 , Antagonistas dos Receptores Histamínicos H1/farmacocinética , Cetoconazol/farmacologia , Microssomos Hepáticos/efeitos dos fármacos , Oxirredutases N-Desmetilantes/antagonistas & inibidores , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Terfenadina/farmacocinética , Antifúngicos/toxicidade , Biotransformação/efeitos dos fármacos , Técnicas de Cultura , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/fisiologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Antagonistas dos Receptores Histamínicos H1/toxicidade , Humanos , Cetoconazol/toxicidade , Taxa de Depuração Metabólica/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Oxirredutases N-Desmetilantes/fisiologia , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Terfenadina/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...