Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Pharmacogenomics J ; 16(4): 303-4, 2016 08.
Article in English | MEDLINE | ID: mdl-27168097

ABSTRACT

Clinical applications of prenatal genetic screening currently focus on detection of aneuploidy and other genetic diseases in the developing fetus. Growing evidence suggests that the fetal genome may also be informative about fetal exposures through contributions to placental transport as well as placental and fetal metabolism. Possible clinical applications of prenatal pharmacogenomic screening include prospective optimization of medication selection and dosage, as well as retrospective assessment of whether a fetus was previously exposed to significant risk. Newly available noninvasive methods of prenatal genetic screening mean that relevant fetal genotypes could be made available to obstetricians for use in management of a current pregnancy. This promising area for research merits more attention than it has thus far received.The Pharmacogenomics Journal advance online publication, 10 May 2016; doi:10.1038/tpj.2016.33.


Subject(s)
Biomedical Research/methods , Cytochrome P-450 Enzyme System/genetics , Membrane Transport Proteins/genetics , Pharmacogenetics/methods , Pharmacogenomic Testing , Pharmacogenomic Variants , Prenatal Diagnosis/methods , Animals , Biotransformation , Cytochrome P-450 Enzyme System/metabolism , Female , Fetus/metabolism , Genotype , Humans , Membrane Transport Proteins/metabolism , Phenotype , Placenta/metabolism , Pregnancy
2.
Clin Pharmacol Ther ; 100(1): 53-62, 2016 07.
Article in English | MEDLINE | ID: mdl-27082931

ABSTRACT

The pharmacokinetics of many drugs are altered by pregnancy. Drug distribution and protein binding are changed by pregnancy. While some drug metabolizing enzymes have an apparent increase in activity, others have an apparent decrease in activity. Not only is drug metabolism affected by pregnancy, but renal filtration is also increased. In addition, pregnancy alters the apparent activities of multiple drug transporters resulting in changes in the net renal secretion of drugs.


Subject(s)
Pharmaceutical Preparations/metabolism , Pharmacokinetics , Pregnancy/metabolism , Female , Humans , Kidney/metabolism , Membrane Transport Proteins/metabolism , Pharmaceutical Preparations/administration & dosage , Protein Binding , Tissue Distribution
3.
Clin Pharmacol Ther ; 92(6): 737-45, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23073208

ABSTRACT

We evaluated the hypothesis that cytochrome P450 3A5 (CYP3A5) expression can affect intrarenal tacrolimus accumulation. Tacrolimus was administered orally to 24 healthy volunteers who were selected on the basis of their CYP3A5 genotype. As compared with CYP3A5 nonexpressors, expressors had a 1.6-fold higher oral tacrolimus clearance and 2.0- to 2.7-fold higher metabolite/parent area under the curve (AUC) ratios for 31-desmethyl tacrolimus (31-DMT), 12-hydroxy tacrolimus, and 13-desmethyl tacrolimus (13-DMT). In addition, the apparent urinary tacrolimus clearance was 36% lower in CYP3A5 expressors as compared with nonexpressors. To explore the mechanism behind this observation, we developed a semiphysiological model of renal tacrolimus disposition and predicted that tacrolimus exposure in the renal epithelium of CYP3A5 expressors is 53% of that for CYP3A5 nonexpressors, when normalized to blood AUC. These data suggest that, at steady state, intrarenal accumulation of tacrolimus and its primary metabolites will depend on the CYP3A5 genotype of the liver and kidneys. This may contribute to interpatient differences in the risk of tacrolimus-induced nephrotoxicity.


Subject(s)
Cytochrome P-450 CYP3A/genetics , Immunosuppressive Agents/pharmacokinetics , Tacrolimus/pharmacokinetics , Adult , Algorithms , Area Under Curve , Biotransformation , DNA/genetics , Female , Genetic Variation , Genotype , Half-Life , Heterozygote , Humans , Immunosuppressive Agents/urine , Kidney/metabolism , Male , Models, Genetic , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Tacrolimus/urine
4.
Article in English | MEDLINE | ID: mdl-23835882

ABSTRACT

Physiological changes in pregnancy, including changes in body composition and metabolic enzyme activity, can alter drug pharmacokinetics. A semi-mechanistic metabolism model was developed to describe the pharmacokinetics of two cytochrome P450 3A (CYP3A) substrates, midazolam and nifedipine, in obstetrics patients. The model parameters were optimized to fit the data of oral midazolam pharmacokinetics in pregnant women, by increasing CYP3A-induced hepatic metabolism 1.6-fold in the model with no change in gut wall metabolism. Fetal metabolism had a negligible effect on maternal plasma drug concentrations. Validation of the model was performed by applying changes in volume of distribution and metabolism, consistent with those observed for midazolam, to the pharmacokinetics parameters of immediate-release nifedipine in healthy volunteers. The predicted steady-state areas under the concentration-time curve (AUCs) for nifedipine were within 15% of the data observed in pregnant women undergoing treatment for preterm labor. This model predicts the pharmacokinetics of two CYP3A substrates in pregnancy, and may be applicable to other CYP3A substrates as well.CPT: Pharmacometrics & Systems Pharmacology (2012) 1, e2; doi:10.1038/psp.2012.5; advance online publication 26 September 2012.

5.
Clin Pharmacol Ther ; 85(6): 607-14, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19295505

ABSTRACT

Glyburide's pharmacokinetics (PK) and pharmacodynamics have not been studied in women with gestational diabetes mellitus (GDM). The objective of this study was to assess steady-state PK of glyburide, as well as insulin sensitivity, beta-cell responsivity, and overall disposition indices after a mixed-meal tolerance test (MMTT) in women with GDM (n = 40), nonpregnant women with type 2 diabetes mellitus (T2DM) (n = 26), and healthy pregnant women (n = 40, MMTT only). At equivalent doses, glyburide plasma concentrations were approximately 50% lower in pregnant women than in nonpregnant subjects. The average umbilical cord/maternal plasma glyburide concentration ratio at the time of delivery was 0.7 +/- 0.4. Insulin sensitivity was approximately fivefold lower in women with GDM as compared with healthy pregnant women. Despite comparable beta-cell responsivity indices, the average beta-cell function corrected for insulin resistance was more than 3.5-fold lower in women with glyburide-treated GDM than in healthy pregnant women. Women with GDM in whom glyburide treatment has failed may benefit from alternative medication or dosage escalation; however, fetal safety should be kept in mind.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetes, Gestational/drug therapy , Glyburide/therapeutic use , Hypoglycemic Agents/therapeutic use , Adult , Area Under Curve , Aryl Hydrocarbon Hydroxylases , Blood Glucose/analysis , Cytochrome P-450 CYP2C9 , Dose-Response Relationship, Drug , Female , Fetal Blood/chemistry , Glyburide/pharmacokinetics , Humans , Hypoglycemic Agents/pharmacokinetics , Insulin Resistance , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Metabolic Clearance Rate , Monte Carlo Method , Pregnancy , Pregnancy Trimester, Third
6.
Drug Metab Dispos ; 37(4): 702-5, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19116263

ABSTRACT

The objective of this study was to determine the pharmacokinetic parameters of clonidine during pregnancy compared with previously published data in nonpregnant subjects. Serial blood and urine samples were collected in 17 women during mid to late pregnancy over one steady-state dosing interval to determine clonidine noncompartmental pharmacokinetic parameters (n = 17) and creatinine clearance. In six of these pregnant subjects, maternal and umbilical cord (venous and arterial) plasma samples were collected at the time of delivery for measurement of clonidine concentrations. Clonidine apparent oral clearance was found to be 440 +/- 168 ml/min during pregnancy compared with 245 +/- 72 ml/min as previously reported in nonpregnant subjects (p < 0.0001) (Cunningham et al., 1994). There was a strong correlation (r = 0.82, p < 0.001) between clonidine renal clearance, adjusted for variation in glomerular filtration rate, and urine pH. Umbilical cord to maternal plasma clonidine concentration ratios were 1.0 +/- 0.1 (arterial) and 1.0 +/- 0.1 (venous). In conclusion, clonidine is cleared more rapidly in pregnant women than in nonpregnant subjects. At the time of delivery, the fetus is exposed to similar plasma clonidine concentrations as the mother.


Subject(s)
Adrenergic alpha-Agonists/pharmacokinetics , Clonidine/pharmacokinetics , Hypertension/drug therapy , Pregnancy Complications, Cardiovascular/drug therapy , Adrenergic alpha-Agonists/blood , Adrenergic alpha-Agonists/therapeutic use , Adult , Area Under Curve , Clonidine/blood , Clonidine/therapeutic use , Female , Half-Life , Humans , Hypertension/complications , Pregnancy
7.
Clin Pharmacol Ther ; 84(2): 248-53, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18288078

ABSTRACT

The objectives of the study were to evaluate the effects of pregnancy on CYP3A and P-glycoprotein (P-gp) activities, as measured by disposition of midazolam and digoxin, respectively. Thirteen women received digoxin (0.25 mg p.o.) and midazolam (2 mg p.o.) in random order, separated by 1-2 weeks at 28-32 weeks gestation, and the same order was repeated at 6-10 weeks postpartum. Plasma and urine concentrations were determined by liquid chromatography-mass spectrometry and analyzed by noncompartmental methods. Midazolam CL/F(unbound) (593 +/- 237 l/min vs. 345 +/- 103 l/min; P = 0.007), digoxin CL(Renal, unbound) (272 +/- 45 ml/min vs. 183 +/- 37 ml/min; P < 0.002) and digoxin CL(secretion,) (unbound) (109 +/- 34 ml/min vs. 58 +/- 22 ml/min; P < 0.002) were higher during pregnancy than postpartum. These data are consistent with increased hepatic and/or intestinal CYP3A and renal P-gp activities during pregnancy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cytochrome P-450 CYP3A/metabolism , Digoxin/pharmacokinetics , Midazolam/pharmacokinetics , Postpartum Period/metabolism , Pregnancy/metabolism , Adult , Anesthetics, Intravenous/pharmacokinetics , Anti-Anxiety Agents/pharmacokinetics , Anti-Arrhythmia Agents/pharmacokinetics , Area Under Curve , Cardiotonic Agents/pharmacokinetics , Creatinine/urine , Digoxin/blood , Digoxin/urine , Enzyme Inhibitors/pharmacokinetics , Female , Gas Chromatography-Mass Spectrometry , Genotype , Humans , Hypnotics and Sedatives/pharmacokinetics , Midazolam/blood , Midazolam/urine , Pregnancy Trimester, Third/metabolism
8.
Clin Pharmacol Ther ; 81(4): 547-56, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17329990

ABSTRACT

Amoxicillin is recommended for anthrax prevention in pregnancy. The objective of this study was to evaluate the pharmacokinetics of amoxicillin during pregnancy and postpartum (PP). Sixteen women received amoxicillin during gestation (18-22 weeks (T2) and 30-34 weeks (T3)) as well as 3 months postpartum (PP) to evaluate single-dose pharmacokinetics. Amoxicillin compartmental pharmacokinetic parameters were used to simulate amoxicillin concentration-time profiles following different dosage strategies. Amoxicillin CL(renal) (T2: 24.8+/-6.7 l/h, P<0.001; T3: 24.0+/-3.9 l/h, P<0.001; and PP: 15.3+/-2.6 l/h) and renal CL(secretion) (T2: 280+/-105 ml/min, P<0.002; T3: 259+/-54 ml/min, P<0.001; and PP: 167+/-47 ml/min) were higher during pregnancy than postpartum. Simulations suggest that amoxicillin concentrations adequate to prevent anthrax may be difficult to achieve during pregnancy and postpartum. Increases in amoxicillin CL(renal) and renal CL(secretion) reflect increases in filtration and secretory transport or diminished reabsorption in the kidneys. Amoxicillin may not be an appropriate antibiotic for post-anthrax exposure prophylaxis.


Subject(s)
Amoxicillin/administration & dosage , Amoxicillin/pharmacokinetics , Penicillins/administration & dosage , Penicillins/pharmacokinetics , Pregnancy/metabolism , Adolescent , Adult , Algorithms , Area Under Curve , Computer Simulation , Female , Humans , Middle Aged , Models, Statistical , Monte Carlo Method , Pregnancy Trimester, Second/metabolism , Pregnancy Trimester, Third/metabolism
9.
Ann Pharmacother ; 33(6): 680-2, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10410178

ABSTRACT

OBJECTIVE: To describe a patient with increased tacrolimus concentrations due to a diltiazem drug interaction. CASE SUMMARY: A 68-year-old white man, four months following orthotopic liver transplantation secondary to hepatitis C and Laënnec's cirrhosis, was admitted to the intensive care unit for diarrhea, dehydration, and atrial fibrillation. He was stabilized on oral tacrolimus 8 mg twice daily, with a whole blood tacrolimus trough concentration of 12.9 ng/mL on admission. He was started on a continuous infusion of diltiazem for one day, followed by 30 mg orally every eight hours. Three days after admission, the patient became delirious, confused, and agitated; he was found to have a whole blood tacrolimus trough concentration of 55 ng/mL. The tacrolimus was withheld and diltiazem was discontinued. The tacrolimus concentrations fell over the next three days to 6.7 ng/mL, with a corresponding improvement in his mental status. The oral tacrolimus was restarted at 3 mg twice daily and increased gradually to 5 mg twice daily over the next four days; this produced tacrolimus trough concentrations between 9 and 10 ng/mL. DISCUSSION: Tacrolimus is known to be a substrate for P-glycoprotein and metabolized by CYP3A. Diltiazem inhibits CYP3A, P-glycoprotein, and tacrolimus metabolism in vitro. Although this interaction may have been predictable, this is the first detailed case report describing this clinically significant drug interaction. CONCLUSIONS: Diltiazem can dramatically increase tacrolimus concentrations and result in tacrolimus toxicity. Avoidance of this interaction or careful monitoring of tacrolimus concentrations along with tacrolimus dose reduction is recommended if diltiazem therapy cannot be avoided.


Subject(s)
Calcium Channel Blockers/adverse effects , Diltiazem/adverse effects , Immunosuppressive Agents/pharmacokinetics , Tacrolimus/pharmacokinetics , Aged , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/therapeutic use , Diltiazem/administration & dosage , Diltiazem/therapeutic use , Drug Interactions , Graft Rejection/prevention & control , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Infusions, Intravenous , Liver Transplantation/immunology , Male , Psychoses, Substance-Induced/psychology , Tacrolimus/adverse effects , Tacrolimus/therapeutic use
10.
Transplantation ; 67(5): 707-12, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10096526

ABSTRACT

BACKGROUND: Mycophenolate mofetil (MMF) has been shown to have promise in short-term liver transplantation graft rescue studies. The purpose of this study was to evaluate the long-term efficacy and safety of MMF in liver transplant patients who had failed cyclosporine (CsA)-based conventional immunosuppression. METHODS: Nineteen orthotopic liver allograft recipients were converted from azathioprine to MMF in combination with CsA and prednisone in this prospective, open-labeled, single-center, graft rescue, pilot study. Six patients were taken off CsA when MMF was initiated. A 4-year patient follow-up is reported here. Patients were considered to have failed CsA-based immunosuppression either for refractory rejection, chronic rejection, or severe CsA neurologic toxicity. RESULTS: Twelve patients had complete histologic resolution, two had partial resolution, and three had worsening of their rejection. Thirteen patients had a complete biochemical response; one had a partial response and four had worsening of their rejection. Two patients had no histologic and one no biochemical follow-up. Of the six patients treated with MMF and prednisone alone, four had complete resolution of rejection without recurrence. The majority of adverse reactions were gastrointestinal [nausea and/or vomiting (n=5); diarrhea (n=8); gastritis, duodenitis, or esophagitis (n=4); and ulcers (n=2)] or bone marrow suppressive [leukopenia (n=9), anemia (n=6), and thrombocytopenia (n=5)]. CONCLUSIONS: MMF seems to be an effective alternative immunosuppressive in patients failing CsA-based conventional therapy. MMF may be of particular benefit in patients who do not tolerate CsA or tacrolimus. The long-term safety profile is similar to that of other immunosuppressives.


Subject(s)
Graft vs Host Disease/prevention & control , Immunosuppressive Agents/therapeutic use , Liver Transplantation/immunology , Mycophenolic Acid/analogs & derivatives , Adult , Cytomegalovirus Infections/immunology , Female , Follow-Up Studies , Gastrointestinal Diseases/chemically induced , Humans , Immunosuppressive Agents/adverse effects , Male , Middle Aged , Mycophenolic Acid/adverse effects , Mycophenolic Acid/therapeutic use , Neoplasms/immunology , Treatment Outcome
11.
J Clin Pharmacol ; 39(1): 91-6, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9987705

ABSTRACT

Tacrolimus is a marketed immunosuppressant used in liver and kidney transplantation. It is subject to extensive metabolism by CYP3A4 and is a substrate for P-glycoprotein-mediated transport. A pharmacokinetic interaction with rifampin, an antituberculosis agent and potent inducer of CYP3A4 and P-glycoprotein, and tacrolimus was evaluated in six healthy male volunteers. Tacrolimus was administered at doses of 0.1 mg/kg orally and 0.025 mg/kg/4 hours intravenously. The pharmacokinetics of tacrolimus were obtained from serial blood samples collected over 96 hours, after single oral and intravenous administration prior to and during an 18-day concomitant rifampin dosing phase. Coadministration of rifampin significantly increased tacrolimus clearance (36.0 +/- 8.1 ml/hr/kg vs. 52.8 +/- 9.6 ml/hr/kg; p = 0.03) and decreased tacrolimus bioavailability (14.4% +/- 5.7% vs. 7.0% +/- 2.7%; p = 0.03). Rifampin appears to induce both intestinal and hepatic metabolism of tacrolimus, most likely through induction of CYP3A and P-glycoprotein in the liver and small bowel.


Subject(s)
Enzyme Inhibitors/pharmacology , Immunosuppressive Agents/pharmacokinetics , Rifampin/pharmacology , Tacrolimus/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Biological Availability , Drug Interactions , Humans , Injections, Intravenous , Intestinal Mucosa/metabolism , Intestines/drug effects , Liver/drug effects , Liver/metabolism , Male , Metabolic Clearance Rate/drug effects , Tacrolimus/blood
12.
Ann Oncol ; 9(10): 1123-6, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9834826

ABSTRACT

Tamoxifen, a non-steroidal anti-estrogen, has been used successfully for a decade as post-operative adjuvant therapy for breast cancer. Tamoxifen is generally well tolerated with few side effects, especially at the typical dose of 10 mg twice daily. However, hepatic effects have been reported after tamoxifen administration and are usually found to be cholestatic in nature. Although previous reports concentrate on tamoxifen as a probable cause of drug-induced hepatotoxicity, very little attention has been focused on the use of tamoxifen in patients with pre-existing liver dysfunction and the possible need for dose adjustment. We present the case of a 48-year-old woman with an acute exacerbation of her pre-existing liver dysfunction and subsequent elevations of tamoxifen blood levels after approximately one year of tamoxifen therapy for adjuvant treatment of breast cancer. Tamoxifen dosing was adjusted based on serum levels.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Chemical and Drug Induced Liver Injury/etiology , Liver Diseases/complications , Tamoxifen/therapeutic use , Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Agents, Hormonal/pharmacokinetics , Breast Neoplasms/complications , Breast Neoplasms/metabolism , Chemical and Drug Induced Liver Injury/blood , Female , Humans , Liver Diseases/blood , Liver Function Tests , Middle Aged , Tamoxifen/adverse effects , Tamoxifen/pharmacokinetics
13.
Clin Pharmacol Ther ; 62(1): 41-9, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9246018

ABSTRACT

OBJECTIVE: To quantitate the effect of ketoconazole, an azole antifungal agent and potent inhibitor of CYP3A4 and P-glycoprotein, on the bioavailability of tacrolimus, a substrate of the CYP3A system and of P-glycoprotein. SUBJECTS AND METHODS: The pharmacokinetics of tacrolimus were studied in six healthy volunteers (two women and four men) in a four-dose study after each received single doses of tacrolimus alone (0.1 mg/kg orally and 0.025 mg/kg intravenously) and with coadministered ketoconazole (200 mg orally at bedtime for 12 days). The dose of tacrolimus was reduced during the ketoconazole phase (0.04 mg/kg orally; 0.01 mg/kg intravenously). Ketoconazole and tacrolimus doses were separated by approximately 10 hours. Whole blood tacrolimus concentrations were determined by enzyme-linked immunosorbent assay. Estimated pharmacokinetic parameters in whole blood (mean +/- SD) before and with ketoconazole were calculated with noncompartmental techniques. RESULTS: Coadministration of ketoconazole did not consistently affect tacrolimus clearance (55.6 +/- 16.7 ml/hr/kg versus 42.5 +/- 7.6 ml/hr/kg), and steady-state volume of distribution was unchanged (0.99 +/- 0.26 L/kg versus 0.93 +/- 0.25 L/kg). However, a significant increase in tacrolimus bioavailability (14% +/- 5% versus 30% +/- 8%; p < 0.01) was observed with coadministered ketoconazole. Hepatic bioavailability was unchanged by the presence of ketoconazole (96% +/- 1% versus 97% +/- 1%). CONCLUSIONS: Because ketoconazole did not alter hepatic bioavailability and because 10 hours separated administration times of the drugs, it appears that the marked increase in tacrolimus bioavailability can be explained by ketoconazole having a local inhibitory effect on tacrolimus gut metabolism or on intestinal P-glycoprotein activity.


Subject(s)
Antifungal Agents/pharmacology , Immunosuppressive Agents/pharmacokinetics , Ketoconazole/pharmacology , Tacrolimus/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Administration, Oral , Adult , Biological Availability , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Female , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/blood , Male , Mixed Function Oxygenases/antagonists & inhibitors , Tacrolimus/administration & dosage , Tacrolimus/blood
14.
Clin Pharmacol Ther ; 59(3): 297-303, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8653992

ABSTRACT

We evaluated the effect of water-soluble vitamin E (d-alpha-tocopheryl polyethylene glycol 1000 succinate [TPGS]; Liqui-E) on the oral pharmacokinetics of the cyclosporine, a poorly available (approximately 30%) drug, in healthy volunteers. Ten healthy subjects were given two doses of oral cyclosporine (10mg/kg) separated by a 7-day washout period. Oral TPGS (2.6 IU/kg) was administered concomitantly with one of the cyclosporine doses in a randomized order. A significant increase was observed in area under the blood concentration-time curve (AUC;mean +/ SD) with concomitant TPGS administration (3908 +/- 2601 versus 6296 +/- 5102 ng x hr/ml). Significant decreases were observed in apparent oral clearance (0.24 +/- 0.14 versus 0.15 +/- 0.08 L/hr/kg) and apparent oral steady-state volume of distribution (1.57 +/- 0.95 versus 1.07 +/- 0.73 L/kg). No significant changes were observed in the ratios of metabolites to parent drug AUC values. The comparable relative decreases in apparent oral clearance (38%) and apparent oral steady-state volume of distribution (30%) with TPGS are most likely explained by enhanced absorption, decreased counter transport back into the intestine by P-glycoprotein, or some unknown mechanism by which cyclosporine is protected from metabolism in the gut, thereby increasing bioavailability.


Subject(s)
Cyclosporine/pharmacokinetics , Immunosuppressive Agents/pharmacokinetics , Vitamin E/pharmacology , Administration, Oral , Adult , Cross-Over Studies , Cyclosporine/administration & dosage , Female , Humans , Immunosuppressive Agents/administration & dosage , Male , Reference Values , Solubility , Water
15.
Clin Pharmacol Ther ; 58(5): 492-7, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7586942

ABSTRACT

The low and variable bioavailability of cyclosporine has been attributed to poor absorption. However, recent studies have suggested that intestinal first-pass metabolism exerts a significant effect on bioavailability. We describe theory and methods to differentiate the contribution from oral absorption and intestinal and hepatic metabolism to overall cyclosporine bioavailability. Analysis of data from previous studies in our laboratories shows that in the absence of intestinal metabolism, cyclosporine absorption from its presently available dosage form averages at least 65% +/- 12% in healthy volunteers and 77% +/- 19% in kidney transplant patients. Analysis also suggests that the extraction ratio for cyclosporine in the gut is approximately twice the hepatic extraction and that cyclosporine absorption does not present a problem, with an average of 86% of the drug absorbed intact from its commercially available product in healthy volunteers. The boundary condition analysis described should have broad application in the differentiation of factors responsible for poor bioavailability.


Subject(s)
Cyclosporine/pharmacokinetics , Immunosuppressive Agents/pharmacokinetics , Liver/metabolism , Administration, Oral , Biological Availability , Cyclosporine/metabolism , Drug Interactions , Erythromycin/pharmacology , Humans , Immunosuppressive Agents/metabolism , Injections, Intravenous , Intestinal Absorption/drug effects , Intestinal Mucosa/metabolism , Ketoconazole/pharmacology , Kidney Transplantation , Liver/drug effects , Protein Synthesis Inhibitors/pharmacology , Rifampin/pharmacology
16.
Clin Pharmacol Ther ; 58(1): 15-9, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7628178

ABSTRACT

The pharmacokinetics of cyclosporine were studied in the blood of five normal healthy volunteers (two men and three women) after each received oral and intravenous cyclosporine alone and with concomitant oral ketoconazole. Administration of ketoconazole caused a significant decrease in intravenous cyclosporine clearance (0.18 +/- 0.05 L/kg/hr versus 0.32 +/- 0.09 L/hr/kg) and a significant increase in cyclosporine oral bioavailability (56.4% +/- 11.7% versus 22.4% +/- 4.8%) compared with values before ketoconazole administration. Steady-state volume of distribution for intravenously administered cyclosporine was unchanged (1.26 +/- 0.44 L/kg versus 1.10 +/- 0.27 L/kg). Hepatic bioavailability (1 - hepatic extraction ratio) calculated for intravenous cyclosporine increased by 11% in the presence of ketoconazole (86.3% +/- 3.7% versus 75.2% +/- 6.6% without ketoconazole), which accounts for only one third of the observed increase in cyclosporine oral bioavailability. Because it is unlikely that ketoconazole had a significant effect on either cyclosporine absorption or hepatic blood flow, the increase in cyclosporine bioavailability observed in this study is most likely explained by inhibition of gastrointestinal cytochrome P450 enzymes.


Subject(s)
Cyclosporine/metabolism , Intestinal Absorption/drug effects , Ketoconazole/pharmacology , Administration, Oral , Biological Availability , Cyclosporine/pharmacokinetics , Drug Administration Schedule , Drug Interactions , Female , Humans , Infusions, Intravenous , Liver/metabolism , Male
17.
Clin Pharmacol Ther ; 57(3): 281-90, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7535213

ABSTRACT

OBJECTIVE: To characterize the pharmacokinetics of the immunosuppressive agent tacrolimus (FK 506) in liver transplant patients. METHODS: Patients (n = 16) were assessed during and after 1- to 3-day intravenous infusions followed by a 2-week course of oral dose therapy. Plasma and whole blood data were fitted simultaneously with equations accounting for nonlinear drug binding by red blood cells to generate clearance (CL) and volume of distribution (V). RESULTS: The maximum blood/plasma ratio of tacrolimus was 55.5 +/- 26.8 (SD) and half-life averaged 12.1 +/- 4.7 hours. The CL and V were relatively high based on plasma concentrations (CL, 1.7 L/hr/kg; V, 30 L/kg) and low based on whole blood (CL, 54 ml/hr/kg; V, 0.9 L/kg), with moderate variability (coefficient of variation, 34% to 49%) among the patients. Correlations of plasma CL and V with maximum blood/plasma ratios (ranging from 13 to 114) were strong (r = 0.65 and r = 0.73). Blood binding affects the disposition of tacrolimus, and plasma concentrations are indirectly and inversely related to red cell binding. The oral dose data for tacrolimus yielded a brief absorption lag time (tlag, 0.39 hour), a variable first-order absorption rate constant (ka, 4.5 +/- 3.0 hr-1), and consistent bioavailability (F, 25% +/- 10%). The area under the concentration-time curve versus 12-hour minimum concentration relationships for both whole blood and plasma were nearly linear, confirming the utility of trough values for monitoring drug exposure. CONCLUSION: This study provides pharmacokinetic guidelines for the use of tacrolimus in patients undergoing hepatic transplantation. Nonlinear blood binding is a major source of interpatient variation in the disposition of tacrolimus.


Subject(s)
Liver Transplantation , Tacrolimus/pharmacokinetics , Administration, Oral , Biological Availability , Half-Life , Humans , Infusions, Intravenous , Tacrolimus/administration & dosage , Tacrolimus/blood
18.
Ann Pharmacother ; 27(10): 1193-4, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8251685

ABSTRACT

OBJECTIVE: To report the apparent development of endstage liver disease secondary to nitrofurantoin administration. PATIENT: A 40-year-old woman developed hepatic failure after receiving nitrofurantoin 200 mg po bid for one month for prophylaxis against urinary-tract infections. Symptoms first occurred after two weeks of nitrofurantoin therapy. Other causes of hepatic failure (e.g., viral infection, autoimmune disorder, concomitant medications, cancer, Wilson's disease, ethanol abuse, pregnancy) were ruled out. CONCLUSIONS: Acute and chronic hepatic injury has previously been reported with the use of nitrofurantoin. The proposed mechanism may be immunoallergic or metabolic in origin. On evaluating concomitant diseases and medications in our patient as well as the temporal sequence of her signs and symptoms, it appears that she developed hepatic failure secondary to the use of nitrofurantoin, ultimately necessitating orthotopic liver transplantation. The potential for the development of such a severe adverse reaction warrants careful evaluation of symptoms that may potentially be caused by hepatic damage as well as immediate discontinuation of nitrofurantoin in patients presenting with jaundice.


Subject(s)
Hepatic Encephalopathy/chemically induced , Liver Transplantation , Nitrofurantoin/adverse effects , Adult , Female , Hepatic Encephalopathy/surgery , Humans , Nitrofurantoin/therapeutic use , Urinary Tract Infections/drug therapy
19.
Clin Pharmacol Ther ; 52(5): 453-7, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1424418

ABSTRACT

The pharmacokinetics of cyclosporine was studied in six healthy volunteers after administration of the drug orally (10 mg/kg) and intravenously (3 mg/kg) with and without concomitant rifampin administration. Both blood and plasma (separated at 37 degrees C) samples were analyzed for cyclosporine concentration. For blood and plasma, respectively, clearances of cyclosporine were calculated to be 0.30 and 0.55 L/hr/kg, values for volume of distribution at steady state were 1.31 and 1.68 L/kg, and bioavailabilities were 27% and 33% during the pre-rifampin phase. Post-rifampin phase clearances of cyclosporine were 0.42 and 0.79 L/hr/kg, values for volume of distribution at steady state were 1.36 and 1.35 L/kg, and bioavailabilities were 10% and 9% for blood and plasma, respectively. Rifampin not only induces the hepatic metabolism of cyclosporine but also decreases its bioavailability to a greater extent than would be predicted by the increased metabolism. The decreased bioavailability most probably can be explained by an induction of intestinal cytochrome P450 enzymes, which appears to be markedly greater than the induction of hepatic metabolism.


Subject(s)
Cyclosporine/pharmacokinetics , Liver/metabolism , Rifampin/pharmacology , Administration, Oral , Adult , Biological Availability , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Enzyme Induction , Female , Humans , Infusions, Intravenous , Intestines/enzymology , Liver/enzymology , Male , Middle Aged , Reference Values , Rifampin/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...