Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Target Oncol ; 15(6): 803, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33108585

ABSTRACT

The article Phase Ib Trial of the PI3K Inhibitor Copanlisib Combined with the Allosteric MEK Inhibitor Refametinib in Patients with Advanced Cancer.

2.
Target Oncol ; 15(2): 163-174, 2020 04.
Article in English | MEDLINE | ID: mdl-32314268

ABSTRACT

BACKGROUND: Dual inhibition of PI3K and MAPK signaling is conceptually a promising anticancer therapy. OBJECTIVE: This phase 1b trial investigated the safety, maximum tolerated dose (MTD), recommended phase II dose, pharmacokinetics, tumor response, fluorodeoxyglucose positron emission tomography (FDG-PET) pharmacodynamics, and biomarker explorations for the combination of pan-PI3K inhibitor copanlisib and allosteric MEK inhibitor refametinib in patients with advanced solid tumors. PATIENTS AND METHODS: This was an adaptive trial with eight dose cohorts combining dose escalation and varying schedules in repeated 28-day cycles. Patients received copanlisib (0.2-0.8 mg/kg intravenously) intermittently (days 1, 8, 15) or weekly (days 1, 8, 15, 22) each cycle, and refametinib (30-50 mg twice daily orally) continuously or 4 days on/3 days off. Patients with KRAS, NRAS, BRAF, or PI3KCA mutations were eligible for the expansion cohort. RESULTS: In the dose-escalation (n = 49) and expansion (n = 15) cohorts, the most common treatment-emergent adverse events included diarrhea (59.4%), nausea, acneiform rash, and fatigue (51.6% each). Dose-limiting toxicities included oral mucositis (n = 4), increased alanine aminotransferase/aspartate aminotransferase (n = 3), rash acneiform, hypertension (n = 2 each), and diarrhea (n = 1). MTD was copanlisib 0.4 mg/kg weekly and refametinib 30 mg twice daily. No pharmacokinetic interactions were identified. Decreased tumor FDG uptake and MEK-ERK signaling inhibition were demonstrated during treatment. Best response was stable disease (n = 21); median treatment duration was 6 weeks. CONCLUSIONS: Despite sound rationale and demonstrable pharmacodynamic tumor activity in relevant tumor populations, a dose and schedule could not be identified for this drug combination that was both tolerable and offered clear efficacy in the population assessed. CLINICALTRIALS. GOV IDENTIFIER: NCT01392521.

3.
Mol Cancer Ther ; 19(2): 468-478, 2020 02.
Article in English | MEDLINE | ID: mdl-31619463

ABSTRACT

The PI3K inhibitor copanlisib has efficacy and manageable safety in patients with indolent lymphoma and solid tumors. Pharmacodynamic effects relative to copanlisib dose and plasma exposure were evaluated. Patients with lymphoma or solid tumors received copanlisib 0.4 or 0.8 mg/kg on days 1, 8, and 15 of a 28-day cycle. Primary variables were maximum changes in phosphorylated AKT (pAKT) levels in platelet-rich plasma (PRP) and plasma glucose. Other evaluations included PI3K signaling markers and T-lymphocytes in paired tumor biopsies, the relationship between estimated plasma exposure and pharmacodynamic markers, response, and safety. Sixty-three patients received copanlisib. PRP pAKT levels showed sustained reductions from baseline following copanlisib [median inhibition: 0.4 mg/kg, 73.8% (range -94.9 to 144.0); 0.8 mg/kg, 79.6% (range -96.0 to 408.0)]. Tumor pAKT was reduced versus baseline with copanlisib 0.8 mg/kg in paired biopsy samples (P < 0.05). Dose-related transient plasma glucose elevations were observed. Estimated copanlisib plasma exposure significantly correlated with changes in plasma pAKT and glucose metabolism markers. There were two complete responses and six partial responses; seven of eight responders received copanlisib 0.8 mg/kg. Adverse events (all grade) included hyperglycemia (52.4%), fatigue (46.0%), and hypertension (41.3%). Copanlisib demonstrated dose-dependent pharmacodynamic evidence of target engagement and PI3K pathway modulation/inhibition in tumor and immune cells. Results support the use of copanlisib 0.8 mg/kg (or flat-dose equivalent of 60 mg) in solid tumors and lymphoma, and provide a biomarker hypothesis for studies of copanlisib combined with immune checkpoint inhibitors (NCT03711058).


Subject(s)
Biopsy/methods , Lymphoma/drug therapy , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/therapeutic use , Pyrimidines/therapeutic use , Quinazolines/therapeutic use , Cell Line, Tumor , Female , Humans , Lymphoma/surgery , Male , Neoplasms/surgery , Phosphatidylinositol 3-Kinases/pharmacology , Pyrimidines/pharmacology , Quinazolines/pharmacology
4.
Cancer Chemother Pharmacol ; 80(3): 535-544, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28714036

ABSTRACT

PURPOSE: To determine the pharmacokinetics of radiolabeled copanlisib (BAY 80-6946) in healthy male volunteers and to investigate the disposition and biotransformation of copanlisib. METHODS: A single dose of 12 mg copanlisib containing 2.76 MBq [14C]copanlisib was administered as a 1-h intravenous infusion to 6 volunteers with subsequent sampling up to 34 days. Blood, plasma, urine and feces were collected to monitor total radioactivity, parent compound and metabolites. RESULTS: Copanlisib treatment was well tolerated. Copanlisib was rapidly distributed throughout the body with a volume distribution of 1870 L and an elimination half-life of 52.1-h (range 40.4-67.5-h). Copanlisib was the predominant component in human plasma (84% of total radioactivity AUC) and the morpholinone metabolite M1 was the only circulating metabolite (about 5%). Excretion of drug-derived radioactivity based on all 6 subjects was 86% of the dose within a collection interval of 20-34 days with 64% excreted into feces as major route of elimination and 22% into urine. Unchanged copanlisib was the main component excreted into urine (15% of dose) and feces (30% of dose). Excreted metabolites (41% of dose) of copanlisib resulted from oxidative biotransformation. CONCLUSIONS: Copanlisib was eliminated predominantly in the feces compared to urine as well as by hepatic biotransformation, suggesting that the clearance of copanlisib would more likely be affected by hepatic impairment than by renal dysfunction. The dual mode of elimination via unchanged excretion of copanlisib and oxidative metabolism decreases the risk of clinically relevant PK-related drug-drug interactions.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use , Quinazolines/therapeutic use , Administration, Intravenous , Class I Phosphatidylinositol 3-Kinases/pharmacokinetics , Class I Phosphatidylinositol 3-Kinases/therapeutic use , Healthy Volunteers , Humans , Male , Middle Aged , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Quinazolines/administration & dosage , Quinazolines/pharmacokinetics
5.
Clin Cancer Res ; 20(7): 1900-9, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24523439

ABSTRACT

PURPOSE: This phase I trial was undertaken to determine the maximum tolerated dose (MTD), dose-limiting toxicities (DLT), safety, tolerability, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity of the novel smoothened inhibitor sonidegib (LDE225), a potent inhibitor of hedgehog signaling, in patients with advanced solid tumors. EXPERIMENTAL DESIGN: Oral sonidegib was administered to 103 patients with advanced solid tumors, including medulloblastoma and basal cell carcinoma (BCC), at doses ranging from 100 to 3,000 mg daily and 250 to 750 mg twice daily, continuously, with a single-dose pharmacokinetics run-in period. Dose escalations were guided by a Bayesian logistic regression model. Safety, tolerability, efficacy, pharmacokinetics, and biomarkers in skin and tumor biopsies were assessed. RESULTS: The MTDs of sonidegib were 800 mg daily and 250 mg twice daily. The main DLT of reversible grade 3/4 elevated serum creatine kinase (18% of patients) was observed at doses ≥ the MTD in an exposure-dependent manner. Common grade 1/2 adverse events included muscle spasm, myalgia, gastrointestinal toxicities, increased liver enzymes, fatigue, dysgeusia, and alopecia. Sonidegib exposure increased dose proportionally up to 400 mg daily, and displayed nonlinear pharmacokinetics at higher doses. Sonidegib exhibited exposure-dependent reduction in GLI1 mRNA expression. Tumor responses observed in patients with medulloblastoma and BCC were associated with evidence of hedgehog pathway activation. CONCLUSIONS: Sonidegib has an acceptable safety profile in patients with advanced solid tumors and exhibits antitumor activity in advanced BCC and relapsed medulloblastoma, both of which are strongly associated with activated hedgehog pathway, as determined by gene expression.


Subject(s)
Biphenyl Compounds/administration & dosage , Carcinoma, Basal Cell/drug therapy , Medulloblastoma/drug therapy , Pyridines/administration & dosage , Receptors, G-Protein-Coupled/antagonists & inhibitors , Administration, Oral , Adult , Aged , Aged, 80 and over , Biphenyl Compounds/adverse effects , Biphenyl Compounds/pharmacokinetics , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Maximum Tolerated Dose , Medulloblastoma/genetics , Medulloblastoma/pathology , Middle Aged , Neoplasm Staging , Pyridines/adverse effects , Pyridines/pharmacokinetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Smoothened Receptor , Transcription Factors/biosynthesis , Zinc Finger Protein GLI1
6.
Haematologica ; 97(5): 731-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22315495

ABSTRACT

BACKGROUND: This study evaluates the correlation between imatinib trough plasma concentrations (C(min)) and clinical response and safety in patients with newly diagnosed Philadelphia chromosome-positive chronic myeloid leukemia in chronic phase in the Tyrosine Kinase Inhibitor OPtimization and Selectivity (TOPS) trial. DESIGN AND METHODS: Patients were randomized 1:2 to 400 mg/day or 800 mg/day imatinib. Imatinib C(min) levels were collected at pre-dose before treatment, and at the end of months 1 (day 29), 6, 9, and 12. RESULTS: Imatinib C(min) were stable over time in the 400 mg/day dose arm, but showed a slight decrease in the 800 mg/day arm due to dose adjustments between months 1-6. The overall median imatinib C(min) levels were 1040, 1200, 1935, and 2690 ng/mL for the actual 300, 400, 600, and 800 mg/day doses, respectively. The rates of major molecular response (MMR) at 3, 6, 9, and 12 months, and complete cytogenetic response (CCyR) at 6 and 12 months were significantly lower among patients with the lowest imatinib C(min) levels at Day 29 (<1165 ng/mL, 25th percentile). There was an apparent association between high imatinib C(min) and the occurrence of grade 3/4 neutropenia and all-grade rash, diarrhea, arthralgia/myalgia, and all-cause edema. Conclusions Imatinib C(min) levels were relatively stable over time and proportional to the dose administered. Patients with an imatinib C(min) above 1165 ng/mL on Day 29 achieved MMR faster and had higher MMR and CCyR rates at 12 months. There appeared to be an association between imatinib C(min) and the frequency of some adverse events. This trial was registered at http://www.clinicaltrials.gov as NCT00124748.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myeloid, Chronic-Phase/drug therapy , Piperazines/blood , Piperazines/therapeutic use , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/blood , Pyrimidines/therapeutic use , Adolescent , Adult , Aged , Benzamides , Female , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Leukemia, Myeloid, Chronic-Phase/blood , Male , Maximum Tolerated Dose , Middle Aged , Piperazines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Remission Induction , Tissue Distribution , Treatment Outcome , Young Adult
7.
J Ocul Pharmacol Ther ; 25(4): 335-44, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19492955

ABSTRACT

PURPOSE: Studies were conducted to evaluate the ocular penetration and systemic exposure to besifloxacin, a fluoroquinolone antibiotic, following topical ocular administration to animals and humans. METHODS: Besifloxacin ophthalmic suspension (0.6%) was administered as a topical ocular instillation to pigmented rabbits, cynomolgus monkeys, and human subjects. At predetermined intervals after dosing, samples of ocular tissues and plasma were collected and analyzed for besifloxacin levels using HPLC/MS/MS methods. RESULTS: Besifloxacin demonstrated good ocular penetration in rabbits and monkeys, with rapid absorption and sustained concentrations observed in anterior ocular tissues through 24 h after a single administration. Maximum besifloxacin concentrations in conjunctiva, cornea, and aqueous humor of monkeys were 6.43 microg/g, 2.10 microg/g, and 0.796 microg/mL, respectively, after a single topical dose, and concentrations declined in these tissues with an apparent half-life of 5-14 h. Following a single topical ocular administration to humans, the maximum besifloxacin concentration in tears was 610 microg/g with concentrations decreasing to approximately 1.6 microg/g at 24 h. The resulting pharmacokinetic parameters for besifloxacin in human tears were evaluated relative to the MIC(90) values (microg/mL) for besifloxacin against Streptococcus pneumoniae (0.125), Staphylococcus aureus (0.25), Staphylococcus epidermidis (0.5), and Haemophilus influenzae (0.06). Following a single topical administration, the C(max)/MIC(90) ratios for besifloxacin in human tears were > or =1,220, and the AUC((0-24))/MIC(90) ratios were > or =2,500 for these relevant ocular pathogens. Following repeated 3-times daily (TID) topical ocular administration to human subjects with clinically diagnosed bacterial conjunctivitis, maximum besifloxacin concentrations in plasma were less than 0.5 ng/mL, on average. CONCLUSIONS: Taken together, the results of the current investigation provide a PK/PD-based rationale that supports the use of besifloxacin for the safe and effective treatment of ocular infections.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Azepines/pharmacokinetics , Fluoroquinolones/pharmacokinetics , Tears/metabolism , Administration, Topical , Adult , Animals , Anti-Bacterial Agents/administration & dosage , Aqueous Humor/metabolism , Area Under Curve , Azepines/administration & dosage , Chromatography, High Pressure Liquid/methods , Conjunctiva/metabolism , Cornea/metabolism , Drug Administration Schedule , Female , Fluoroquinolones/administration & dosage , Half-Life , Humans , Macaca fascicularis , Male , Microbial Sensitivity Tests , Ophthalmic Solutions/administration & dosage , Rabbits , Species Specificity , Tandem Mass Spectrometry/methods , Tissue Distribution , Young Adult
8.
J Pharm Biomed Anal ; 49(5): 1241-9, 2009 Jul 12.
Article in English | MEDLINE | ID: mdl-19304431

ABSTRACT

We have developed and validated an accurate, sensitive, and robust LC-MS/MS method that determines the concentration of CRx-102 (the combination of prednisolone and dipyridamole) in human plasma. In this method, prednisolone, dipyridamole, and the combined internal standards (IS) prednisolone-d(6) (IS for prednisolone) and dipyridamole-d(20) (IS for dipyridamole) were extracted from 100 microL human EDTA plasma using methylbutyl ether. Calibration curves were linear over a concentration range of 0.4-200 ng/mL for prednisolone and 5-3000 ng/mL for dipyridamole. The analytes were quantitatively determined using tandem mass spectrometry operated in positive electrospray ionization in a multiple reaction monitoring (MRM) mode. This validated method has been used successfully in clinical pharmacokinetic studies of CRx-102 in healthy volunteers.


Subject(s)
Chromatography, Liquid/methods , Dipyridamole/blood , Prednisolone/blood , Tandem Mass Spectrometry/methods , Area Under Curve , Calibration , Dipyridamole/chemistry , Dipyridamole/pharmacokinetics , Drug Combinations , Drug Stability , Freezing , Half-Life , Humans , Metabolic Clearance Rate , Molecular Structure , Prednisolone/chemistry , Prednisolone/pharmacokinetics , Quality Control , Reference Standards , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Mass, Electrospray Ionization/methods , Temperature , Time Factors
9.
Article in English | MEDLINE | ID: mdl-18417430

ABSTRACT

A rapid and sensitive method was developed using high-performance liquid chromatography coupled with tandem mass spectrometry (LC/MS/MS) for the quantification of besifloxacin in human tears using sparfloxacin as the internal standard (IS). Besifloxacin was extracted from human tear samples using an ammonium formate buffer at pH 3.25. The method was validated over a concentration range of 2-2000 ng/mL, with a total run time of less than 4 min. The overall intra- and inter-day precision for this method was less than 6%. The method was used to measure besifloxacin concentrations in tear samples collected after topical ocular administration to humans; besifloxacin concentrations were 610+/-540 microg/g (15 min) and 1.60+/-2.28 microg/g (24h).


Subject(s)
Anti-Bacterial Agents/analysis , Azepines/analysis , Chromatography, High Pressure Liquid/methods , Fluoroquinolones/analysis , Tandem Mass Spectrometry/methods , Tears/chemistry , Anti-Bacterial Agents/pharmacokinetics , Azepines/pharmacokinetics , Calibration , Fluoroquinolones/pharmacokinetics , Humans , Reference Standards , Reproducibility of Results , Sensitivity and Specificity
10.
Drug Metab Dispos ; 31(5): 548-58, 2003 May.
Article in English | MEDLINE | ID: mdl-12695342

ABSTRACT

Human cytochrome P450 3A4 (CYP3A4) is the most abundant hepatic and intestinal phase I drug-metabolizing enzyme, and participates in the oxidative metabolism of approximately 50% of drugs on the market. In the present study, a transgenic-CYP3A4 (Tg-CYP3A4) mouse model that expresses CYP3A4 in the intestine and is phenotypically normal was generated, which was genotyped by both polymerase chain reaction and Southern blotting. Intestinal microsomes prepared from Tg-CYP3A4 mice metabolized midazolam (MDZ) to 1'-hydroxymidazolam about 2 times, and to 4-hydroxymidazolam around 3 times faster than that from wild-type (WT) mice. These increased MDZ hydroxylation activities were completely inhibited by an anti-CYP3A4 monoclonal antibody. The time course of plasma MDZ and its metabolite concentrations was measured after intravenous (0.25 mg/kg) and oral (2.5 mg/kg) administration of MDZ, and pharmacokinetic parameters were estimated by fitting to a noncompartmental model. Pretreatment with ketoconazole increased orally dosed MDZ maximum plasma concentration (C(max)), time of the maximum concentration, area under the plasma concentration-time curve from zero to infinity (AUC(0- infinity)), and elimination half-life (t(1/2)) to 3.2-, 1.7-, 7.7-, 2-fold, and decreased MDZ apparent oral clearance about 8-fold in Tg-CYP3A4 mice. The ratios of MDZ C(max), AUC(0- infinity), t(1/2) and bioavailability between Tg-CYP3A4 and WT mice after the oral dose of MDZ were 0.3, 0.6, 0.5, and 0.5, respectively. These results suggest that this Tg-CYP3A4 mouse would be an appropriate in vivo animal model for the evaluation of human intestine CYP3A4 metabolism of drug candidates and potential food-drug and drug-drug interactions in preclinical drug development.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Intestine, Small/metabolism , Midazolam/analogs & derivatives , Midazolam/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Biological Availability , Blotting, Southern , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Dexamethasone/pharmacology , Drug Interactions , Enzyme Inhibitors/pharmacology , Half-Life , Humans , In Vitro Techniques , Ketoconazole/pharmacology , Male , Mice , Mice, Transgenic , Microsomes/drug effects , Microsomes/metabolism , Midazolam/blood , Midazolam/metabolism , Polymerase Chain Reaction , Rifampin/pharmacology , Time Factors
11.
J Pharmacol Exp Ther ; 304(2): 539-46, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12538805

ABSTRACT

Tryptamine is a trace amine in mammalian central nervous system that interacts with the trace amine TA(2) receptor and is now thought to function as a neurotransmitter or neuromodulator. It had been reported that deamination of tryptamine to tryptophol was mediated by CYP2D6, a cytochrome P450 that is expressed in human brain, suggesting that tryptamine may be an endogenous substrate for this polymorphic enzyme. We were unable to confirm this report and have reinvestigated tryptamine metabolism in human liver microsomes (HLM) and in microsomes expressing recombinant human cytochrome P450 and monoamine oxidase (MAO) isozymes. Tryptamine was oxidized to indole-3-acetaldehyde by HLM and recombinant human MAO-A in the absence of NADPH, and indole-3-acetaldehyde was further reduced to tryptophol by aldehyde reductase in HLM in the presence of NADPH. Steady-state kinetic parameters were estimated for each reaction step by HLM and MAO-A. The CYP2D6 substrates bufuralol and debrisoquine showed strong inhibition of both tryptophol production from tryptamine in HLM and the formation of indole-3-acetaldehyde from tryptamine catalyzed by recombinant MAO-A. Anti-CYP2D6 monoclonal antibody did not inhibit these reactions. Pargyline, a nonselective MAO inhibitor, did not show cross inhibition to debrisoquine 4-hydroxylation and dextromethorphan O-demethylation by HLM and recombinant CYP2D6 enzyme. This is the first unequivocal report of the selective conversion of tryptamine to tryptophol by MAO-A. CYP2D6 does not contribute to this reaction.


Subject(s)
Cytochrome P-450 Enzyme System/physiology , Monoamine Oxidase/physiology , Tryptamines/metabolism , Cytochrome P-450 CYP2D6/metabolism , Deamination/drug effects , Humans , Isoenzymes/physiology , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Monoamine Oxidase Inhibitors/pharmacology
12.
J Pharmacol Exp Ther ; 301(3): 1025-32, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12023534

ABSTRACT

A panel of 15 recombinant cytochromes P450 expressed in human B-lymphoblastoid cells was used to study debrisoquine 4-hydroxylation. Both CYP2D6 and CYP1A1 carried out the reaction. The apparent K(m) (micromolar) and V(max) (picomoles per minute per picomole of P450) for CYP2D6 were 12.1 and 18.2 and for CYP1A1 were 23.1 and 15.2, respectively. CYP1A1 debrisoquine 4-hydroxylase was inhibited by the CYP1A1 inhibitor alpha-naphthoflavone and the CYP1A1 substrate 7-ethoxyresorufin. Additionally and surprisingly, this reaction was also inhibited by quinidine and quinine, with respective IC(50) values of 1.38 +/- 0.10 and 3.31 +/- 0.14 microM, compared with those for CYP2D6 debrisoquine 4-hydroxylase of 0.018 +/- 0.05 and 3.75 +/- 2.07 microM, respectively. Anti-CYP1A1 monoclonal antibody (mAb) 1-7-1 abolished CYP1A1 debrisoquine hydroxylase and anti-CYP2D6 mAb 50-1-3 eradicated CYP2D6 debrisoquine 4-hydroxylase. Three further CYP2D6-specific reactions were tested: dextromethorphan O-demethylation, bufuralol 1'-hydroxylation, and sparteine dehydrogenation. The CYP2D6 specificity, judged by the CYP2D6/CYP1A1 activity ratios was 18.5, 7.0, 6.0, and 1.6 for dextromethorphan, bufuralol, sparteine, and debrisoquine, respectively. Thus, debrisoquine is not a specific CYP2D6 substrate and quinidine is not a specific CYP2D6 inhibitor. These findings have significant implications for the conduct of in vitro drug metabolism inhibition studies and underscore the fallacy of "specific chemical inhibitors" of a supergene family of enzymes that have overlapping substrate specificities. The use of highly specific mAbs in such studies is mandated. It is unclear as yet whether these findings have implications for the relationship between CYP2D6 genotype and in vivo debrisoquine 4-hydroxylase activity.


Subject(s)
Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP2D6 Inhibitors , Cytochrome P-450 CYP2D6/metabolism , Debrisoquin/metabolism , Enzyme Inhibitors/pharmacology , Quinidine/pharmacology , Quinine/pharmacology , Adrenergic Agents/chemistry , Adrenergic Agents/metabolism , Antimalarials/pharmacology , Debrisoquin/chemistry , Dose-Response Relationship, Drug , Humans , Hydroxylation/drug effects , Microsomes/drug effects , Microsomes/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...