Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Med Educ ; 35(5): 424-5, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11328509
2.
Acad Med ; 75(4): 337-45, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10893115

RESUMO

Based on his work in Shanghai, China, the author believes that U.S. academic health centers (AHCs) should take a leadership role in global health initiatives. While acknowledging that most AHCs already have focused projects involving research or education with foreign institutions, he proposes a greater coordination of these projects into programs that, in some areas, could also be linked to clinical delivery systems where care may be provided. These AHC "platforms" overseas would be structured as a partnership between an AHC in the United States and one in the foreign country where the platform is located, to promote their missions of education, research, and service. For example, U.S. AHCs benefit, and are often dependent upon, international trainees who seek further clinical or research training in the United States. However, the identification of suitable candidates and career guidance, so the students' career choices could benefit their home countries, are often lacking. Thus, the United States is often viewed as facilitating a "brain drain" of future leaders in academic medicine from developing areas of the world. The author proposes a way to lessen this problem by shifting more on-site training to settings in the students' home countries, which could occur if AHCs were willing to develop overseas platforms. U.S. students would also benefit from access to medical training in foreign lands for both the cultural perspectives they offer and the unique diseases and medical situations encountered. He also suggests that shared platforms would lead to greater research opportunities for institutions in the United States and abroad. He argues for increased efforts at coordinating these activities with the rising demand for Western clinical services by multinational companies and U.S. expatriate communities overseas. The potential pitfalls of such initiatives as well as the need for permanent relationships are discussed. In conclusion, he believes that AHCs have an opportunity to establish global health initiatives through education, research, and patient care that will both enhance their own institutions and benefit the international community.


Assuntos
Centros Médicos Acadêmicos , Atenção à Saúde/métodos , Liderança , China , Atenção à Saúde/estatística & dados numéricos , Educação Médica/estatística & dados numéricos , Ciência de Laboratório Médico , Pesquisa , Fatores de Tempo , Estados Unidos
5.
Drug Metab Dispos ; 23(12): 1315-24, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8689938

RESUMO

The small intestinal metabolism of tacrolimus, which is used as an immunosuppressant in transplantation medicine, was investigated in this study. Tacrolimus was metabolized in vitro by isolated human, pig, and rat small intestinal microsomes. The metabolites generated were identified by HPLC/MS. Tacrolimus and its metabolites were quantified using HPLC or HPLC/MS. The cytochrome P450 (CYP) enzymes responsible for tacrolimus metabolism in small intestine were identified using specific CYP antibodies and inhibitors. For characterization of the interindividual variability, microsomes were isolated from small intestinal samples of patients who had undergone resection for various reasons. In an in vitro model using pig small intestinal microsomes, 32 drugs were analyzed for their interactions with tacrolimus metabolism. After incubation with human, rat, and pig small intestinal microsomes, the metabolites 13-O-demethyl and 13,15-O-demethyl tacrolimus were identified. The metabolism of tacrolimus by human small intestine was inhibited by anti-CYP3A, troleandomycin, and erythromycin, indicating that, as in the liver, CYP3A enzymes are the major enzymes for tacrolimus metabolism in the human small intestine. Metabolism of tacrolimus by small intestinal microsomes isolated from 14 different patients varied between 24 and 110 pmol/13-O-demethyl tacrolimus/min/mg microsomal protein, with a mean +/- SD of 54.2 +/- 29.2 pmol/min/mg. Of 32 drugs tested, 15 were found to inhibit small intestinal tacrolimus metabolism: bromocryptine, corticosterone, cyclosporine, dexamethasone, ergotamine, erythromycin, ethinyl estradiol, josamycin, ketoconazole, nifedipine, omeprazole, progesterone, rapamycin, troleandomycin, and verapamil. All of these drugs inhibited tacrolimus metabolism by human liver microsomes as well. It is concluded that tacrolimus is metabolized by cytochrome CYP3A enzymes in the small intestine. The rate of the CYP3A enzymatic activities varies about 5 times from patient to patient, and drugs that interfere with the in vitro metabolism of tacrolimus in the liver also inhibit its small intestinal metabolism.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Imunossupressores/metabolismo , Intestino Delgado/metabolismo , Tacrolimo/metabolismo , Animais , Biotransformação , Cromatografia Líquida de Alta Pressão , Inibidores das Enzimas do Citocromo P-450 , Interações Medicamentosas , Humanos , Immunoblotting , Técnicas In Vitro , Intestino Delgado/enzimologia , Masculino , Espectrometria de Massas , Microssomos/enzimologia , Microssomos/metabolismo , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Suínos
6.
Drug Metab Dispos ; 22(6): 947-55, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7895614

RESUMO

The CYP3A subfamily of cytochromes P450 metabolize many medications and environmental contaminants. CYP3A4 and, in 25% of patients, CYP3A5 seem to be the major CYP3A genes expressed in adult liver. Hepatic levels of CYP3A4 can be estimated by the erythromycin breath test and vary at least 10-fold among patients. CYP3A4 has also been shown to be present in small bowel where it is responsible for significant "first-pass" metabolism of orally administered substrates. However, it is not known whether there is significant interindividual variability in the intestinal expression of CYP3A4, or whether the liver and intestinal catalytic activities of CYP3A4 correlate within an individual. It is also not known whether CYP3A5 is expressed in the small intestine. To address these questions, we administered the erythromycin breath test to 20 patients and obtained biopsies from their small bowel. There was a 6-fold variation in CYP3A catalytic activity (midazolam hydroxylation), an 11-fold variation in CYP3A4 protein content, and an 8-fold variation in CYP3A4 mRNA content in intestinal biopsies. There was an excellent correlation between intestinal CYP3A4 protein level and catalytic activity (r = 0.86; p = 0.0001); however, neither parameter significantly correlated with hepatic CYP3A4 activity as measured by the erythromycin breath test result (r = 0.27; p = 0.24 and r = 0.33; p = 0.15, respectively). We also found that CYP3A5 protein was readily detectable in biopsies from 14 (70%) of the patients, indicating that CYP3A5 is commonly expressed in human small intestine.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Sistema Enzimático do Citocromo P-450/análise , Eritromicina/metabolismo , Intestino Delgado/enzimologia , Oxigenases de Função Mista/análise , Adulto , Idoso , Testes Respiratórios , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/genética , Feminino , Humanos , Masculino , Midazolam/metabolismo , Pessoa de Meia-Idade , Oxigenases de Função Mista/genética , RNA Mensageiro/análise
7.
Pharmacogenetics ; 4(5): 247-59, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7894497

RESUMO

CYP3A4, a major Phase I xenobiotic metabolizing enzyme present in liver, is also present in human small bowel epithelium where it appears to catalyse significant 'first pass' metabolism of some drugs. To determine whether CYP3A4 or the related enzymes CYP3A3, CYP3A5, and CYP3A7 are present in other regions of the digestive tract, we used CYP3A-specific antibodies to examine histological sections and epithelial microsomes obtained from a human organ donor. CYP3A-related proteins were detected in epithelia throughout the digestive tract and in gastric parietal cells, in pericentral hepatocytes, and in ductular cells of the pancreas. Immunoblot analysis suggested that the major CYP3A protein present in liver, jejunum, colon, and pancreas was CYP3A4 or CYP3A3, whereas CYP3A5 was the major protein present in stomach. Both CYP3A4 and CYP3A5 mRNA were detectable in all regions of the digestive tract using the polymerase chain reaction (PCR); however, only CYP3A4 could be detected by Northern blot analysis. CYP3A7 mRNA was consistently detected only in the liver by PCR and CYP3A3 mRNA was not detected in any of the tissues. We conclude that CYP3A4 and CYP3A5 are present throughout the human digestive tract and that differences in the expression of these enzymes may account for inter-organ differences in the metabolism of CYP3A substrates.


Assuntos
Sistema Enzimático do Citocromo P-450/genética , Regulação Enzimológica da Expressão Gênica , Mucosa Intestinal/enzimologia , Intestino Delgado/enzimologia , Oxigenases de Função Mista/genética , Adulto , Sequência de Bases , Western Blotting , Citocromo P-450 CYP2E1 , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/metabolismo , DNA Complementar , Feminino , Humanos , Imuno-Histoquímica , Oxigenases de Função Mista/metabolismo , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
8.
Gastroenterology ; 106(2): 433-9, 1994 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8299909

RESUMO

BACKGROUND/AIMS: Hepatic CYP3A enzymes have been implicated in the bioactivation of aflatoxin B1 (AFB1) to DNA binding metabolites. CYP3A enzymes are also abundant in the small bowel, and we therefore examined the ability of this tissue to form intracellular AFB1 adducts. METHODS: Immunohistochemistry using a antibody to the stable AFB1-DNA adduct was performed on small bowel sections obtained from rats orally gavaged with AFB1 and on human small bowel biopsy specimens maintained in explant culture. 3H-AFB1 was instilled into a loop of small bowel of untreated rats and rats pretreated with the CYP3A inducer dexamethasone during vivisection. DNA was isolated from the loop 2 hours later and assayed for specific activity. RESULTS: In both rats and humans, AFB1-adducts were detected exclusively in mature enterocytes in a pattern similar to the distribution of CYP3A enzymes. Induction of enterocyte CYP3A in rats resulted in an increase in enterocyte immunoreactive AFB1 adducts and in a 1.8-fold increase in 3H-AFB1-nucleic acid adducts (P = 0.01). CONCLUSIONS: Intracellular AFB1 adducts are formed in the small intestine, and this reflects, at least in part, the catalytic activity of CYP3A enzymes. Because these AFB1 adducts should ultimately pass in stool, enterocyte CYP3A may represent a regulatable barrier to dietary aflatoxins.


Assuntos
Aflatoxina B1/metabolismo , Adutos de DNA , DNA/metabolismo , Intestino Delgado/metabolismo , Animais , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/fisiologia , Feminino , Humanos , Imuno-Histoquímica , Intestino Delgado/citologia , Isoenzimas/fisiologia , Ratos , Ratos Sprague-Dawley
9.
Biochem Pharmacol ; 46(5): 905-18, 1993 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-8373442

RESUMO

Enzymes within the CYP3A subfamily are major Phase I drug-metabolizing enzymes present in hepatocytes and small bowel enterocytes. These enzymes are highly inducible in the liver by many structurally diverse compounds, including a number of commonly used medications. Studies indicate that CYP3A enzymes present in small bowel enterocytes are also inducible. However, the regulation of CYP3A enzymes in this tissue has not been well characterized, in part because in vivo studies are difficult, especially in humans. Our goals was to develop an in vitro model to study the regulation of CYP3A in enterocytes. To this end, we defined culture conditions under which adult rat jejunal explants maintained viable appearing villi for 21 hr. When dexamethasone, the prototypical inducer of CYP3A1 in rat hepatocytes, was added to the culture medium, there was a time-dependent induction of CYP3A1 mRNA and CYP3A protein in explant enterocytes which was essentially indistinguishable from the time course of induction of CYP3A1 mRNA and protein in enterocytes in vivo. This effect of dexamethasone appeared to be specific since dexamethasone had no consistent effect on the explant concentration of another enterocyte specific mRNA, intestinal fatty acid binding protein. Using this explant culture model, we found that CYP3A1 mRNA was also inducible by clotrimazole but we were unable to detect induction by rifampicin or troleandomycin. Our observations suggest that jejunal explants may provide an appropriate model for the study of the regulation of CYP3A and other drug-metabolizing enzymes.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/genética , Isoenzimas/genética , Jejuno/enzimologia , Animais , Clotrimazol/farmacologia , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/biossíntese , Dexametasona/farmacologia , Indução Enzimática/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica , Jejuno/citologia , Jejuno/ultraestrutura , Modelos Biológicos , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
11.
J Clin Invest ; 90(5): 1871-8, 1992 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-1430211

RESUMO

Enzymes within the P450IIIA (CYP3A) subfamily appear to account for significant "first pass" metabolism of some drugs in the intestine. To identify which of the known P450IIIA genes are expressed in intestine, enterocyte RNA was hybridized on Northern blots with synthetic oligonucleotides complementary to hypervariable regions of hepatic P450IIIA4, P450IIIA5, and P450IIIA7 cDNAs. Hybridization was detected only with the P450IIIA4-specific oligonucleotide. The identity of the hybridizing mRNA was confirmed to be P450IIIA4 by direct sequencing of a DNA fragment amplified from enterocyte cDNA by the polymerase chain reaction. To determine if enterocyte P450IIIA4 is inducible, biopsies of small bowel mucosa were obtained from five volunteers before and after they received 7d of treatment with rifampin, a known inducer of P450IIIA4 in liver. Rifampin treatment resulted in a five- or eightfold mean increase (P < 0.05) in the biopsy concentration of P450IIIA4 mRNA when normalized for content of sucrase isomaltase or intestinal fatty acid binding protein mRNAs, respectively. Rifampin also induced P450IIIA immunoreactive protein in enterocytes in each of the subjects, as judged by immunohistochemistry, and resulted in a 10-fold increase in P450IIIA4-specific catalytic activity (erythromycin N-demethylation) in the one patient studied. Our identification of inducible P450IIIA4 in enterocytes may in part account for drug interactions characteristic of P450IIIA4 substrates and suggests a strategy for controlling entry into the body of a major class of xenobiotics.


Assuntos
Sistema Enzimático do Citocromo P-450/biossíntese , Intestino Delgado/enzimologia , Rifampina/farmacologia , Adulto , Sequência de Bases , Ciclosporina/farmacologia , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/genética , Indução Enzimática , Feminino , Humanos , Masculino , Dados de Sequência Molecular , RNA Mensageiro/análise
12.
Clin Pharmacol Ther ; 52(3): 265-73, 1992 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1526083

RESUMO

The production of 14CO2 in the breath from an intravenous dose of [14C-N-methyl]-erythromycin (the erythromycin breath test [ERMBT]) and the measurement of the ratio of 6-beta-cortisol to free cortisol (6-beta-F/FF) in the urine have each been proposed as means of measuring hepatic P450IIIA catalytic activity in patients. We found that there was a significant correlation between the results of each test (r = 0.59, p less than 0.001) in 47 patients who were without liver disease and who were not taking medications believed to influence P450IIIA catalytic activity. In the 24 of these patients who were subsequently treated with the P450IIIA substrate cyclosporine, the ERMBT result was highly correlated with the mean trough cyclosporine blood level observed; however, there was no correlation between urinary 6-beta-F/FF and the cyclosporine blood levels. In a separate study of a patient during the anhepatic phase of liver transplantation surgery, the ERMBT result decreased by greater than 85%, whereas urinary 6-beta-F/FF decreased by just 50%. We conclude that the ERMBT and urinary 6-beta-F/FF do not always provide similar information about P450IIIA catalytic activity in patients, possibly because of extrahepatic production of 6-beta-F. Of the two tests, the ERMBT appears to provide the most relevant information for cyclosporine administration.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Testes Respiratórios/métodos , Sistema Enzimático do Citocromo P-450/metabolismo , Hidrocortisona/urina , Fígado/enzimologia , Oxirredutases N-Desmetilantes/metabolismo , Dióxido de Carbono/metabolismo , Ciclosporina/uso terapêutico , Citocromo P-450 CYP3A , Humanos , Psoríase/tratamento farmacológico , Análise de Regressão , Rifampina/uso terapêutico , Troleandomicina/uso terapêutico
14.
Gastroenterology ; 102(4 Pt 1): 1186-98, 1992 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-1551527

RESUMO

The P450IIIA (CYP3A) cytochromes are a major family of enzymes that play an important role in the metabolism of many medications, including cyclosporine A, as well as some dietary xenobiotics, including aflatoxin B1. The purpose of the studies was to detect, localize, and characterize P450IIIA enzymes present throughout the digestive tract. To this end, P450IIIA-specific antibodies were used to examine gut epithelial microsomes and histological tissue sections obtained from the digestive tract of both male and female rats. P450IIIA-related proteins were detected in epithelia throughout the gut; however, the specific proteins expressed appeared to differ among digestive organs and between male and female rats. RNA obtained from the gut epithelia was also analyzed using P450IIIA-specific synthetic oligonucleotides as probes on Northern blots and as primers for the polymerase chain reaction. P450IIIA1, which is a dexamethasone inducible enzyme in liver, was also found to be induced by dexamethasone treatment in epithelia from stomach and jejunum, but not from colon or esophagus. It was concluded that P450IIIA enzymes are present in mature epithelia throughout the gastrointestinal tract. However, expression of the P450IIIA enzymes is influenced by anatomic location and gender.


Assuntos
Sistema Enzimático do Citocromo P-450/análise , Sistema Digestório/enzimologia , Animais , Sequência de Bases , Sistema Enzimático do Citocromo P-450/genética , DNA/análise , Dexametasona/farmacologia , Epitélio/enzimologia , Feminino , Immunoblotting , Imuno-Histoquímica , Masculino , Dados de Sequência Molecular , RNA Mensageiro/análise , Ratos , Ratos Endogâmicos , Fatores Sexuais
15.
Transplantation ; 53(3): 596-602, 1992 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1549853

RESUMO

Cyclosporine is converted to its major metabolites (M-17, M-1, and M-21) in human liver by enzymes belonging to the P450IIIA subfamily. These enzymes are also present in rat and human enterocytes; however, the possibility that CsA is metabolized in enterocytes has not been previously investigated. We therefore directly compared metabolism of 3H-CsA in microsomes prepared from liver and jejunal enterocytes. M-17, M-1, and M-21 were the major CsA metabolites produced by enterocyte microsomes. This metabolism appeared to be catalyzed by P450IIIA, because pretreatment of rats with the P450IIIA inducer dexamethasone significantly increased the rate of CsA metabolism in enterocyte microsomes and preincubation of enterocyte microsomes with anti-P450IIIA IgG inhibited the production of CsA metabolites by greater than 95%. To determine if enterocyte P450IIIA metabolizes CsA in vivo, rats were pretreated with the P450IIIA inducer dexamethasone, the P450IIIA inhibitor erythromycin, or vehicle alone. At laparotomy, 2 mg/kg of 3H-CsA was injected into a sealed loop of jejunum, and after collection of the mesenteric venous blood draining this segment for 45 min, the production of M-17 and M-1 was measured. In the control group, a mean of 3.9% of the recovered radioactivity was found as M-1 and M-17. In the rats pretreated with dexamethasone, a mean of 8.4% of the radioactivity was found as M-1 and M-17 (P less than 0.05 relative to control) and this decreased to 2.3% in the group pretreated with erythromycin (P = 0.08 relative to control). We conclude that P450IIIA in jejunal enterocytes readily metabolizes CsA. Furthermore, the metabolism of CsA by enterocytes in vivo is substantial and likely contributes to "first pass metabolism" of orally administered CsA. Our observations provide novel hypotheses to explain some important drug interactions and interpatient differences in CsA dosing requirements.


Assuntos
Ciclosporina/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Jejuno/citologia , Administração Oral , Animais , Disponibilidade Biológica , Ciclosporina/administração & dosagem , Ciclosporina/farmacocinética , Dexametasona/farmacologia , Feminino , Jejuno/enzimologia , Jejuno/metabolismo , Masculino , Microssomos/enzimologia , Microssomos Hepáticos/enzimologia , Ratos , Ratos Endogâmicos
16.
Lancet ; 338(8781): 1488-90, 1991 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-1683920

RESUMO

Cyclosporin is thought to be exclusively metabolised in the liver. We instilled cyclosporin into the small bowel of 2 patients during the anhepatic phase of liver transplantation; cyclosporin metabolites were readily detected in portal venous blood. Our findings indicate that the small intestine is a major site of cyclosporin breakdown: such intestinal metabolism might help to explain the poor oral bioavailability and drug interactions of cyclosporin.


Assuntos
Ciclosporina/metabolismo , Intestino Delgado/metabolismo , Disponibilidade Biológica , Ciclosporina/sangue , Ciclosporina/farmacocinética , Humanos , Absorção Intestinal/efeitos dos fármacos , Transplante de Fígado , Veia Porta
18.
Hepatology ; 12(6): 1371-8, 1990 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-2258153

RESUMO

Progressive liver fibrosis in rats develops when they are fed a diet deficient in choline. This diet also results in a pronounced and selective decrease in the liver microsomal content of a phase I drug-metabolizing enzyme belonging to the cytochrome P-450III gene family. Because P-450III cytochromes characteristically catalyze the N-demethylation of erythromycin, we believed that the production of breath CO2 from erythromycin would be dramatically reduced in choline-deficient rats. However, when 12 choline-deficient rats were compared with 9 control rats, the reduction in CO2 production from erythromycin (mean decrease 71%) was essentially identical to that from aminopyrine (mean decrease 69%), a substrate believed to be metabolized normally by the hepatocyte in fibrotic liver disease. Furthermore, we found that the relative erythromycin and aminopyrine demethylase activities were comparable when measured in vitro in liver microsomes prepared from the choline-deficient rats. To determine the molecular basis for the erythromycin demethylase activity in the choline-deficient rats, the liver microsomes were subjected to immunoblot analysis using a variety of polyclonal and monoclonal antibodies capable of distinguishing individual P-450III-related proteins. Our studies confirm that a major erythromycin demethylase belonging to the P-450III family, termed P-450p, was greatly reduced in the choline-deficient rat liver. However, the specific concentration of a second P-450p-related protein was essentially normal and that of a third P-450p-related protein was actually increased in the choline-deficient rat liver.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Deficiência de Colina/complicações , Sistema Enzimático do Citocromo P-450/metabolismo , Eritromicina , Cirrose Hepática Experimental/enzimologia , Fígado/enzimologia , Aminopirina , Animais , Northern Blotting , Testes Respiratórios , Sistema Enzimático do Citocromo P-450/genética , Feminino , Immunoblotting , Cirrose Hepática Experimental/etiologia , Testes de Função Hepática , Masculino , Microssomos Hepáticos/enzimologia , Hibridização de Ácido Nucleico , Oxirredutases N-Desmetilantes/metabolismo , RNA Mensageiro/análise , Ratos , Ratos Endogâmicos
19.
Clin Pharmacol Ther ; 48(2): 120-9, 1990 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-2116259

RESUMO

The daily dose of cyclosporine required to attain a desired blood level can vary greatly among patients. Because elimination of cyclosporine depends on its metabolism in the liver by an enzyme (cytochrome P-450IIIA) that also demethylates erythromycin, we reasoned that the ability of patients to demethylate a test dose of erythromycin might be useful in estimating their appropriate daily doses of cyclosporine. Accordingly, the [14C-N-methyl] erythromycin breath test was administered to 32 patients before they received 3.0, 5.0, or 7.5 mg/kg/day cyclosporine to treat psoriasis. We found that a simple mathematical equation incorporating just the 14CO2 production, the age of the patient, and the daily dose of cyclosporine accounted for almost 80% (R2 = 0.78) of the interpatient variability in cyclosporine blood levels we observed. Our data indicate that P-450IIIA activity largely accounts for the relationship between dose of cyclosporine and blood levels for an individual patient. We conclude that the erythromycin breath test may be a convenient guide for cyclosporine dosing.


Assuntos
Testes Respiratórios , Ciclosporinas/sangue , Eritromicina , Adulto , Idoso , Dióxido de Carbono/metabolismo , Ciclosporinas/metabolismo , Ciclosporinas/uso terapêutico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Teóricos , Valor Preditivo dos Testes , Psoríase/sangue , Psoríase/tratamento farmacológico , Análise de Regressão
20.
Lancet ; 335(8680): 11-5, 1990 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-1967328

RESUMO

A 40-year-old male liver allograft recipient had neurological dysfunction and renal failure while his cyclosporin blood levels were in the therapeutic range; these features recurred on rechallenge. The hypothesis that this toxic effect might have resulted from abnormal metabolism of cyclosporin by liver cytochrome P-450 IIIA was investigated with the [14C]erythromycin breath test, which is a measure of this enzyme's activity. P-450 IIIA activity was decreased compared with that in controls, including other liver transplant recipients. Pretreatment with rifampicin, an inducer of P-450 IIIA, increased enzyme activity. After treatment with rifampicin the patient could be rechallenged with cyclosporin at a dose almost twice that which had previously been toxic. The patient died during a second transplantation and the microsomal content of P-450 IIIA was found to be low in the first transplant.


Assuntos
Ciclosporinas/efeitos adversos , Sistema Enzimático do Citocromo P-450/deficiência , Transplante de Fígado , Administração Oral , Adulto , Testes Respiratórios/métodos , Radioisótopos de Carbono , Cromatografia Líquida de Alta Pressão , Ciclosporinas/administração & dosagem , Ciclosporinas/sangue , Ciclosporinas/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Eritromicina , Estudos de Avaliação como Assunto , Humanos , Immunoblotting , Isoenzimas/deficiência , Masculino , Microssomos Hepáticos/enzimologia , Rifampina/administração & dosagem , Rifampina/farmacologia , Estimulação Química , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...