Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Med ; 9(14): 4991-5007, 2020 07.
Article in English | MEDLINE | ID: mdl-32436621

ABSTRACT

BACKGROUND: Treatment of patients with solid tumors and KRAS mutations remains disappointing. One option is the combined inhibition of pathways involved in RAF-MEK-ERK and PI3K-AKT-mTOR. METHODS: Patients with relapsed solid tumors were treated with escalating doses of everolimus (E) 2.5-10.0 mg/d in a 14-day run-in phase followed by combination therapy with sorafenib (S) 800 mg/d from day 15. KRAS mutational status was assessed retrospectively in the escalation phase. Extension phase included KRAS-mutated non-small-cell lung cancer (NSCLC) only. Pharmacokinetic analyses were accompanied by pharmacodynamics assessment of E by FDG-PET. Efficacy was assessed by CT scans every 6 weeks of combination. RESULTS: Of 31 evaluable patients, 15 had KRAS mutation, 4 patients were negative for KRAS mutation, and the KRAS status remained unknown in 12 patients. Dose-limiting toxicity (DLT) was not reached. The maximum tolerated dose (MTD) was defined as 7.5 mg/d E + 800 mg/d S due to toxicities at previous dose level (10 mg/d E + 800 mg/d S) including leucopenia/thrombopenia III° and pneumonia III° occurring after the DLT interval. The metabolic response rate in FDG-PET was 17% on day 5 and 20% on day 14. No patient reached partial response in CT scan. Median progression free survival (PFS) and overall survival (OS) were 3.25 and 5.85 months, respectively. CONCLUSIONS: Treatment of patients with relapsed solid tumors with 7.5 mg/d E and 800 mg/d S is safe and feasible. Early metabolic response in FDG-PET was not confirmed in CT scan several weeks later. The combination of S and E is obviously not sufficient to induce durable responses in patients with KRAS-mutant solid tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Everolimus/therapeutic use , Fluorodeoxyglucose F18/metabolism , Lung Neoplasms/drug therapy , Positron-Emission Tomography/methods , Proto-Oncogene Proteins p21(ras)/metabolism , Sorafenib/therapeutic use , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Everolimus/pharmacology , Female , Humans , Male , Middle Aged , Sorafenib/pharmacology
2.
Oncotarget ; 6(36): 38458-68, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26540572

ABSTRACT

Treatment with EGFR kinase inhibitors improves progression-free survival of patients with EGFR-mutant lung cancer. However, all patients with initial response will eventually acquire resistance and die from tumor recurrence. We found that intermittent high-dose treatment with erlotinib induced apoptosis more potently and improved tumor shrinkage significantly than the established low doses. In mice carrying EGFR-mutant xenografts intermittent high-dose treatment (200 mg/kg every other day) was tolerable and prolonged progression-free survival and reduced the frequency of acquired resistance. Intermittent EGFR-targeted high-dose schedules induce more profound as well as sustained target inhibition and may afford enhanced therapeutic efficacy.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Erlotinib Hydrochloride/administration & dosage , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Animals , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Disease-Free Survival , Dose-Response Relationship, Drug , Drug Administration Schedule , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Xenograft Model Antitumor Assays
3.
Breast Care (Basel) ; 9(3): 169-74, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25177258

ABSTRACT

BACKGROUND: Sorafenib was tested for neoadjuvant treatment with an anthracycline/taxane-based chemotherapy in the open-label, multicentre, single-arm phase II study, 'SOFIA'. PATIENTS AND METHODS: INCLUSION CRITERIA WERE: HER2 negative, cT3, cT4 or cT2 cN+, M0 primary breast cancer. Patients received 4 × epirubicin 90 mg/m(2) and cyclophosphamide 600 mg/m(2) (EC) intravenously (i.v.) in 3-weekly cycles followed or preceded by 12 weeks of paclitaxel (Pw) 80 mg/m(2). In cohort 1, sorafenib started at 800 mg daily with chemotherapy. An initial daily sorafenib dose of 200 mg was escalated, based on individual toxicities, every 3 weeks in cohort 2 (starting with EC) and every 2 weeks in cohort 3 (starting with Pw). The primary objective was to identify the most feasible regimen; secondary objectives were safety, pathological complete response (pCR) at surgery and pharmacokinetics. RESULTS: Of the 36 recruited patients, 7/12 patients completed the study in cohort 1 and 24/24 patients in cohorts 2 and 3. The median cumulative sorafenib dose per patient was 37%, 65% and 46% in cohorts 1, 2 and 3, respectively. The main grade 3-4 toxicities were neutropenia and hand-foot syndrome. The pCR (ypT0/is) rate was 27.7%. No pharmacokinetic interaction was observed between sorafenib and epirubicin. CONCLUSION: Sorafenib EC-Pw is feasible if the starting dose is 200 mg, escalated every 3 weeks based on the patients' individual toxicities.

4.
J Clin Pharmacol ; 54(10): 1162-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24782075

ABSTRACT

Diurnal changes in the activity of drug metabolizing enzymes may contribute to the variability in drug disposition and drug effects. The aim of this study was to quantify the circadian rhythmicity exhibited by hepatic CYP3A. A 10 µg/kg intravenous bolus dose, followed by a 30-hour 4 µg/kg/h intravenous infusion of midazolam, used as a probe substrate for hepatic CYP3A activity, was administered to 16 healthy volunteers (8 males and 8 females). Blood samples were drawn hourly for 24 hours after achieving steady state, and plasma concentrations of midazolam and its main metabolite 1-OH midazolam were determined. Population pharmacokinetic analysis was performed using nonlinear mixed effects modeling. One-compartment pharmacokinetic models best described midazolam and 1-OH midazolam pharmacokinetic disposition. An unequivocal but minor diurnal pattern was identified in the midazolam plasma concentration profiles, which was described using a cosine function with a 24-hours period. The fluctuation in the relative CYP3A activity ranged between 10% above average around 15:00, and 10% below average around 03:00. None of the covariates tested had a significant impact on the parameters estimated. Although a diurnal pattern in hepatic CYP3A activity was identified, its magnitude suggests that it is small and without clinical significance for drug therapy.


Subject(s)
Circadian Rhythm/physiology , Cytochrome P-450 CYP3A/metabolism , Midazolam/pharmacokinetics , Models, Biological , Adult , Cytochrome P-450 CYP3A/physiology , Female , Humans , Infusions, Intravenous , Liver/metabolism , Male , Midazolam/analogs & derivatives , Nonlinear Dynamics , Young Adult
5.
Toxicol Mech Methods ; 23(8): 576-83, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23682612

ABSTRACT

Abstract Enzyme kinetic parameters provide essential quantitative information about characterization of individual steps in drug metabolism. Such enzymes are located in a (partially) aqueous environment. For in vitro measurements potential lipophilic substrates regularly require organic solvents to achieve concentrations sufficient for access of the drug to the binding site of the enzyme. However, solvents may interact with the enzymes. In this study, we investigated the effects of methanol, ethanol, acetonitrile and dimethyl sulfoxide (1% to 4%) on the assessment of km, Vmax and Clint for the metabolism of midazolam via CYP3A4 to 1-hydroxymidazolam and the metabolism of caffeine to paraxanthine via CYP1A2 using expressed enzymes in vitro. The presence of acetonitrile proved the highest apparent Vmax value for paraxanthine formation but the lowest values for 1-hydroxymidazolam formation. The km value for midazolam showed no systematic effects of organic solvents, while for caffeine km was up to 8-fold lower for solvent free samples compared to solvent containing samples. The present example suggests that effects of solvents may considerably influence enzyme kinetic parameters beyond a mere change in apparent activity. These effects illustrate a difference for individual enzyme--substrate pairs, solvents, and solvent concentrations. What remains is the determination to which extent these effects compromise in vitro-in vivo extrapolations, and which solvents are most appropriate.


Subject(s)
Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP3A/metabolism , Organic Chemicals/chemistry , Solvents/chemistry , Caffeine/pharmacology , Humans , In Vitro Techniques , Kinetics , Midazolam/pharmacology
6.
Clin Pharmacokinet ; 52(9): 763-81, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23653047

ABSTRACT

BACKGROUND: Accurate predictions of cytochrome P450 (CYP) 3A-mediated drug-drug interactions (DDIs) account for dynamic changes of CYP3A activity at both major expression sites (liver and gut wall) by considering the full pharmacokinetic profile of the perpetrator and the substrate. Physiological-based in vitro-in vivo extrapolation models have become of increasing interest. However, due to discrepancies between the predicted and observed magnitude of DDIs, the role of models fully based on in vivo data is still essential. OBJECTIVE: The primary objective of this study was to develop a coupled dynamic model for the interaction of the CYP3A inhibitor voriconazole and the prototypical CYP3A substrate midazolam. METHODS: Raw concentration data were obtained from a DDI study. Ten subjects were given either no pretreatment (control) or voriconazole twice daily orally. Midazolam was given either intravenously or orally after the last voriconazole dose and during control phases. Data analysis was performed by the population pharmacokinetic approach using non-linear mixed effects modelling (NONMEM 7.2.0). Model evaluation was performed using visual predictive checks and bootstrap analysis. RESULTS: A semiphysiological model was able to describe the pharmacokinetics of midazolam, its major metabolite and voriconazole simultaneously. By considering the temporal disposition of all three substances in the liver and gut wall, a time-varying CYP3A inhibition process was implemented. Only the incorporation of hypothetical enzyme site compartments resulted in an adequate fit, suggesting a sustained inhibitory effect through accumulation. Novel key features of this analysis are the identification of (1) an apparent sustained inhibitory effect by voriconazole due to a proposed quasi accumulation at the enzyme site, (2) a significantly reduced inhibitory potency of intravenous voriconazole for oral substrates, (3) voriconazole as a likely uridine diphosphate glucuronosyltransferase (UGT) 2B inhibitor and (4) considerable sources of interindividual variability. CONCLUSION: The proposed semiphysiological modelling approach generated a mechanistic description of the complex DDI occurring at major CYP3A expression sites and thus may serve as a powerful tool to maximise information acquired from clinical DDI studies. The model has been shown to draw precise and accurate predictions. Therefore, simulations based on this kind of models may be used for various clinical scenarios to improve pharmacotherapy.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors , Midazolam/pharmacokinetics , Models, Biological , Pyrimidines/pharmacokinetics , Triazoles/pharmacokinetics , Antifungal Agents/administration & dosage , Antifungal Agents/blood , Antifungal Agents/pharmacokinetics , Cross-Over Studies , Cytochrome P-450 CYP3A , Drug Interactions , Humans , Liver/enzymology , Male , Midazolam/administration & dosage , Midazolam/blood , Pyrimidines/administration & dosage , Pyrimidines/blood , Triazoles/administration & dosage , Triazoles/blood , Voriconazole
7.
Drug Metab Dispos ; 41(5): 987-93, 2013 May.
Article in English | MEDLINE | ID: mdl-23401474

ABSTRACT

UNLABELLED: This cocktail study evaluated the interaction potential of the oral lavender oil preparation silexan with major P450 (cytochrome P450) enzymes. SUBJECTS AND METHODS: Sixteen healthy male or female Caucasians completed this double-blind, randomized, 2-fold crossover study. Silexan (160 mg) or placebo were administered once daily for 11 days. Additionally, on day 11 of both study periods, 150 mg caffeine (CYP1A2), 125 mg tolbutamide (CYP2C9), 20 mg omeprazole (CYP2C19), 30 mg dextromethorphan-HBr (CYP2D6), and 2 mg midazolam (CYP3A4) were administered orally. Formal interaction was excluded if the 90% confidence interval (CI) for the silexan over placebo ratios for phenotyping metrics (primary: AUC(0-t)) was within a 0.70-1.43 range. RESULTS: According to the AUC(0-t) comparisons, silexan had no relevant effect on CYP1A2, 2C9, 2D6, and 3A4 activity. Secondary phenotyping metrics confirmed this result. Mean ratios for all omeprazole-derived metrics were close to unity. The 90% CI for the AUC(0-t) ratio of omeprazole but not for omeprazole/5-OH-omeprazole plasma ratio 3 hours post-dose or omeprazole/5-OH-omeprazole AUC(0-t) ratio (secondary CYP2C19 metrics) was above the predefined threshold of 1.43, probably caused by the inherent high variability of omeprazole pharmacokinetics. Silexan and the phenotyping drugs were well tolerated. Repeated silexan (160 mg/day) administration has no clinically relevant inhibitory or inducing effects on the CYP1A2, 2C9, 2C19, 2D6, and 3A4 enzymes in vivo.


Subject(s)
Cytochrome P-450 Enzyme System/drug effects , Oils, Volatile/pharmacology , Plant Oils/pharmacology , Acyclic Monoterpenes , Administration, Oral , Area Under Curve , Cross-Over Studies , Double-Blind Method , Drug Interactions , Female , Humans , Lavandula , Limit of Detection , Male , Monoterpenes/blood , Oils, Volatile/administration & dosage , Oils, Volatile/pharmacokinetics , Placebos , Plant Oils/administration & dosage , Plant Oils/pharmacokinetics
8.
Toxicol Mech Methods ; 23(5): 346-51, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23256458

ABSTRACT

OBJECTIVE: Acrylamide (AA), a probable human carcinogen, is present in fried and baked starch-rich food. In vivo, the substance is partly biotransformed to glycidamide (GA), which may account for carcinogenic effects. Existing data suggest an important but not exclusive contribution of CYP2E1 to GA formation. The aim of this project was to derive respective enzyme kinetic parameters for CYP2E1 and to assess a possible role of other important human CYPs for this reaction in vitro. METHODS: AA (0.2-20 mM) was incubated with human liver microsomes (HLM) and human cytochrome P450 enzymes (supersomes™). GA was quantified by a specific LC-MS/MS method. Enzyme kinetic parameters were estimated assuming a single binding site. Furthermore, inhibition experiments were performed with diethyldithiocarbamate (DDC), a potent inhibitor of CYP2E1. RESULTS: The mean ± SD maximum formation rate (Vmax) and Michaelis-Menten constant (Km) for GA formation in HLM were 199 ± 36 pmol GA/mg protein/min and 3.3 ± 0.5 mM, respectively. In AA incubations with supersomes™, only for CYP2E1 measurable GA formation was detected in all tested AA concentrations (Vmax and Km were 5.4 nmol GA/nmol CYP2E1/min and 1.3 mM, respectively). Inhibition constant (IC50) of DDC was 3.1 ± 0.5 µM for HLM and 1.2 ± 0.2 µM for CYP2E1 supersomes™. Therefore, relevant participation of CYPs other than CYP2E1 in the metabolism of AA to GA in humans does not seem likely. CONCLUSION: Our results confirm the major role of CYP2E1 in GA formation from AA, albeit with low affinity and low capacity. Further studies are needed to identify other pathways of GA formation.


Subject(s)
Acrylamide/metabolism , Cytochrome P-450 CYP2E1/metabolism , Humans , In Vitro Techniques , Kinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...