Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Ann Oncol ; 31(2): 302-309, 2020 02.
Article in English | MEDLINE | ID: mdl-31959348

ABSTRACT

BACKGROUND: Nivolumab 480 mg every 4 weeks (Q4W) is approved in the European Union, United States, and several other markets across multiple tumor types. Its approval was supported by quantitative efficacy/safety analyses bridging to 3 mg/kg every 2 weeks (Q2W). PATIENTS AND METHODS: The benefit-risk profile of nivolumab 480 mg Q4W relative to 3 mg/kg Q2W was evaluated using population pharmacokinetic modeling and exposure-response (E-R) analyses for safety and efficacy. Pharmacokinetic exposures were predicted for 3203 patients with melanoma, non-small-cell lung cancer (NSCLC), renal cell carcinoma (RCC), squamous cell carcinoma of the head and neck, urothelial carcinoma, or classical Hodgkin lymphoma. Quantitative models analyzed E-R to predict 480-mg Q4W safety across all indications and efficacy for melanoma, NSCLC, and RCC. Intratumoral receptor occupancy (RO) was predicted for parameters representing different tumor types. RESULTS: Time-averaged concentrations for 480 mg Q4W versus 3 mg/kg Q2W were higher during the first 28 days (26.8%) and similar at steady state (5.2%). The maximum concentration (Cmax) after the first dose was higher (110.4%), and the trough concentration at day 28 was lower (-22.1%) with 480 mg Q4W versus 3 mg/kg Q2W. The Cmax achieved with 480 mg Q4W was lower than the previously established safe dose of 10 mg/kg Q2W. The probability of adverse events for key safety end points was similar for 480 mg Q4W and 3 mg/kg Q2W. The predicted overall survival and objective response rates with 480 mg Q4W were comparable to 3 mg/kg Q2W. The predicted high intratumoral RO provided additional evidence to support 480 mg Q4W across tumor types. CONCLUSIONS: The benefit-risk profile for nivolumab 480 mg Q4W was predicted to be similar to that of 3 mg/kg Q2W across tumor types while providing a convenient and flexible option for patients and their caregivers.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Humans , Lung Neoplasms/drug therapy , Nivolumab/adverse effects , Treatment Outcome
2.
Curr Med Res Opin ; 26(2): 397-405, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20001450

ABSTRACT

OBJECTIVE: The relationship between patient characteristics and posaconazole exposures was evaluated in a population pharmacokinetic (PK) model using trial data from neutropenic patients administered oral posaconazole suspension as antifungal prophylaxis. METHODS: Data were analyzed using nonlinear mixed-effects modeling. Covariates were tested using the forward addition, Objective Function (OF) cut-off of 3.84, followed by the backward elimination (OF cut-off 10.88) steps in NONMEM. These covariates included demographics, mucositis, neutropenia, vomiting, diarrhea, proton pump inhibitor (PPI) or H(2)-receptor antagonist usage and baseline bilirubin or baseline gamma-glutamyl transferase (GGT) levels > or =2 x upper limit of normal (ULN). A correlation between posaconazole PK and the occurrence of invasive fungal infection (IFI) was also examined. RESULTS: Statistically significant associations were demonstrated between posaconazole PK and diarrhea, PPI intake, race, and baseline GGT and bilirubin levels. These covariates did not predominate in patients who developed IFI. CONCLUSION: This analysis provides information regarding the correlation of patient covariates with posaconazole exposures estimated in a clinical setting. The results of this analysis agree with previously reported analyses. However, because of the successful prophylaxis and the low number of posaconazole-treated patients with IFI proven or probable (IFIPP), the absence of a statistically significant relationship between IFIPP and exposure may not mean this relationship does not exist. A meta-analysis of several efficacy trials or exploring alternate composite endpoints for efficacy may be needed to answer this question.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Mycoses/prevention & control , Myelodysplastic Syndromes/drug therapy , Neutropenia/chemically induced , Triazoles/pharmacokinetics , Adolescent , Adult , Aged , Aged, 80 and over , Antibiotic Prophylaxis/methods , Antifungal Agents/pharmacokinetics , Antifungal Agents/therapeutic use , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Male , Middle Aged , Mycoses/etiology , Myelodysplastic Syndromes/metabolism , Neutropenia/complications , Neutropenia/metabolism , Population , Single-Blind Method , Triazoles/therapeutic use , Young Adult
3.
Leukemia ; 22(9): 1707-11, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18548095

ABSTRACT

Lonafarnib is an orally bio-available farnesyltransferase inhibitor that prevents farnesylation of specific target proteins including Ras. In a multicenter study, 67 patients with advanced myelodysplastic syndrome (MDS) and chronic myelomonocytic leukemia (CMML) were treated with a continuous oral dose of 200-300 mg of lonafarnib and were evaluated for hematologic, pathologic and pharmacodynamic response. The median age of patients was 70 years (range 44-86). There were 32 patients with MDS (RAEB-20 and RAEB-t-12) and 35 with CMML. Overall 16 (24%) of the patients responded with two patients achieving a complete remission and one a partial response. Responses were seen in 6/32 and 10/35 patients with MDS and CMML, respectively. Of the 19 patients who were platelet transfusion-dependent prior to treatment, 5 (26%) became transfusion-free for a median duration of 185 days. A decrease in the farnesylation of the HDJ-2 protein measured in patient-derived cells was observed in the majority of patients during treatment with lonafarnib, but no clear correlation between changes in farnesylation and clinical effect could be made. Gastrointestinal toxicity was significant with 19% of patients discontinuing therapy due to diarrhea, nausea and/or anorexia. Lonafarnib has demonstrable activity in patients with advanced MDS and CMML.


Subject(s)
Leukemia, Myelomonocytic, Chronic/drug therapy , Myelodysplastic Syndromes/drug therapy , Piperidines/administration & dosage , Pyridines/administration & dosage , Adult , Aged , Aged, 80 and over , Drug Monitoring , Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Gastrointestinal Diseases/chemically induced , Humans , Leukemia, Myelomonocytic, Chronic/complications , Maximum Tolerated Dose , Middle Aged , Myelodysplastic Syndromes/complications , Piperidines/toxicity , Pyridines/toxicity , Remission Induction , Treatment Outcome
4.
Int J Clin Pharmacol Ther ; 45(10): 539-47, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17966839

ABSTRACT

OBJECTIVE: The objective was to determine whether food affects the pharmacokinetics and safety of lonafanib, an orally bioavailable farnesyl transferase inhibitor that is under clinical evaluation for the treatment of various hematologic malignancies and solid tumors. METHODS: Two Phase 1 studies were conducted in separate patient populations. A single-dose study was performed in 12 healthy subjects who received lonafarnib 100 mg under fasted and fed conditions. Additionally, a multiple-dose study was performed in 19 patients with advanced cancer who received lonafarnib 200 mg Q 12 H for 28 days under fasted and fed conditions. Nine of the 19 patients completed both treatment cycles and were used for pharmacokinetic assessment. A 2-week washout period separated treatments in each study. Single-dose pharmacokinetics were assessed at various time points up to 48 hours postdose and multiple-dose pharmacokinetics were assessed at Day 15 for 24 hours postdose. RESULTS: The pharmacokinetics of lonafarnib were affected by food during single-dose but not multiple-dose administration. Relative oral bioavailabilities (fed vs. fasted) based on log-transformed maximum plasma concentration (C(max)) and area under the concentration-time curve (AUC) were 48% and 77%, respectively, following single-dose administration, and 87% and 96%, respectively, following multiple-dose administration. Intrasubject variability in the pharmacokinetic parameters was less pronounced after multiple dosing (17%) than that after single dosing (33%) of lonafarnib. Intersubject variability was unaffected by food in either study. In the single-dose study, 7 of the 12 subjects (58%) reported treatment emergent adverse events, the most common being headache. No clinically significant differences in adverse events were seen between fasting and fed states after a single dose administration. Thus, single dose 100 mg lonafarnib was safe and generally well tolerated. In the multiple-dose study, all 19 subjects reported at least one treatment-emergent adverse event. General disorders including fatigue and anorexia, and gastrointestinal disorders including diarrhea, vomiting and nausea, were the most commonly reported adverse events after multiple doses. While gastrointestinal adverse events were reported with equal frequency under both fasting (82%, 14/17) and fed states (83%, 15/18), the incidence of severe gastrointestinal adverse events was higher in fasted (47%, 8/17) vs. fed subjects (22%, 4/18) after multiple-dose administration. CONCLUSION: The administration of food does not affect the pharmacokinetics of lonafanib following multiple-dose administration. We recommend that multiple-dose lonafarnib should be administered with food to enhance tolerability.


Subject(s)
Enzyme Inhibitors/pharmacokinetics , Food-Drug Interactions , Piperidines/pharmacokinetics , Pyridines/pharmacokinetics , Adult , Aged , Analysis of Variance , Area Under Curve , Biological Availability , Cross-Over Studies , Drug Administration Schedule , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Farnesyltranstransferase/antagonists & inhibitors , Female , Humans , Male , Middle Aged , Neoplasms/drug therapy , Neoplasms/metabolism , Piperidines/administration & dosage , Piperidines/adverse effects , Pyridines/administration & dosage , Pyridines/adverse effects
5.
Xenobiotica ; 37(7): 753-69, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17620221

ABSTRACT

The absorption, metabolism and excretion of carbon-14-labeled loratadine (LOR, SCH 29851, Claritin) administered orally to healthy male volunteers were evaluated. Following a single oral 10-mg dose of [(14)C]LOR ( approximately 102 microCi), concentrations of LOR and desloratadine (DL; a pharmacologically active descarboethoxy metabolite of LOR) were determined in plasma. Metabolites in plasma, urine and feces were characterized using a liquid chromatography-mass spectrometry system (LC-MS) connected in line with a flow scintillation analyzer (FSA). Maximum plasma LOR and DL concentrations were achieved at 1.5 h and 1.6 h, respectively; thus, LOR was rapidly absorbed but also rapidly metabolized as indicated by these similar t(max) values. Metabolite profiles of plasma showed that LOR was extensively metabolized via descarboethoxylation, oxidation and glucuronidation. Major circulating metabolites included 3-hydroxy-desloratadine glucuonide (3-OH-DL-Glu), dihydroxy-DL-glucuronides, and several metabolites resulting from descarboethoxylation and oxidation of the piperidine ring. LOR was completely metabolized by 6 h post-dose. LOR-derived radiocarbon was excreted almost equally in the urine (41%) and feces (43%). About 13% of the dose was eliminated in the urine as 3-OH-DL-Glu. DL accounted for less than 2% of the dose recovered in the urine and only trace amounts of LOR were detected. 3-OH-DL was the major fecal metabolite ( approximately 17% of the dose). The combined amount of 5- and 6-hydroxy-DL contributed to an additional 10.7% of the dose in feces. Approximately 5.4% and 2.7% of the dose were excreted in the feces as unchanged drug and DL, respectively.


Subject(s)
Anti-Allergic Agents/pharmacokinetics , Histamine H1 Antagonists, Non-Sedating/pharmacokinetics , Loratadine/pharmacokinetics , Adult , Humans , Male
6.
Xenobiotica ; 37(7): 770-87, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17620222

ABSTRACT

The absorption, metabolism and excretion of desloratadine (DL, Clarinex) were characterized in six healthy male volunteers. Subjects received a single oral 10-mg dose of [(14)C]DL ( approximately 104 microCi). Blood, urine and feces were collected over 240 h. DL was well absorbed; drug-derived radioactivity was excreted in both urine (41%) and feces (47%). With the exception of a single subject, DL was extensively metabolized; the major biotransformation pathway consisted of hydroxylation at the 3 position of the pyridine ring and subsequent glucuronidation (3-OH-DL-glucuronide or M13). In five of the six subjects, DL was slowly eliminated (mean t((1/2)) = 19.5 h) and persisted in the plasma for 48-120 h post-dose. This is in contrast to a t((1/2)) of approximately 110 h and quantifiable plasma DL concentrations for the entire 240-h sampling period in one subject, who was identified phenotypically as a poor metabolizer of DL. This subject also exhibited correspondingly lower amounts of M13 in urine and 3-OH-DL (M40) in feces. Disposition of DL in this subject was characterized by slow absorption, slow metabolism and prolonged elimination. Further clinical studies confirmed the lack of safety issues associated with polymorphism of DL metabolism (Prenner et al. 2006, Expert Opinion on Drug Safety, 5: 211-223).


Subject(s)
Histamine H1 Antagonists, Non-Sedating/pharmacology , Loratadine/analogs & derivatives , Adult , Humans , Loratadine/pharmacokinetics , Male
7.
Int J Clin Pharmacol Ther ; 44(2): 83-92, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16502768

ABSTRACT

OBJECTIVE: To assess the bioequivalence of an ezetimibe/simvastatin (EZE/SIMVA) combination tablet compared to the coadministration of ezetimibe and simvastatin as separate tablets (EZE + SIMVA). METHODS: In this open-label, randomized, 2-part, 2-period crossover study, 96 healthy subjects were randomly assigned to participate in each part of the study (Part I or II), with each part consisting of 2 single-dose treatment periods separated by a 14-day washout. Part I consisted of Treatments A (EZE 10 mg + SIMVA 10 mg) and B (EZE/SIMVA 10/10 mg/mg) and Part II consisted of Treatments C (EZE 10 mg + SIMVA 80 mg) and D (EZE/SIMVA 10/80 mg/mg). Blood samples were collected up to 96 hours post-dose for determination of ezetimibe, total ezetimibe (ezetimibe + ezetimibe glucuronide), simvastatin and simvastatin acid (the most prevalent active metabolite of simvastatin) concentrations. Ezetimibe and simvastatin acid AUC(0-last) were predefined as primary endpoints and ezetimibe and simvastatin acid Cmax were secondary endpoints. Bioequivalence was achieved if 90% confidence intervals (CI) for the geometric mean ratios (GMR) (single tablet/coadministration) of AUC(0-last) and Cmax fell within prespecified bounds of (0.80, 1.25). RESULTS: The GMRs of the AUC(0-last) and Cmax for ezetimibe and simvastatin acid fell within the bioequivalence limits (0.80, 1.25). EZE/ SIMVA and EZE + SIMVA were generally well tolerated. CONCLUSIONS: The lowest and highest dosage strengths of EZE/SIMVA tablet were bioequivalent to the individual drug components administered together. Given the exact weight multiples of the EZE/SIMVA tablet and linear pharmacokinetics of simvastatin across the marketed dose range, bioequivalence of the intermediate tablet strengths (EZE/SIMVA 10/20 mg/mg and EZE/SIMVA 10/40 mg/mg) was inferred, although these dosages were not tested directly. These results indicate that the safety and efficacy profile of EZE + SIMVA coadministration therapy can be applied to treatment with the EZE/SIMVA tablet across the clinical dose range.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Azetidines/pharmacokinetics , Simvastatin/pharmacokinetics , Adolescent , Adult , Analysis of Variance , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/adverse effects , Area Under Curve , Azetidines/administration & dosage , Azetidines/adverse effects , Cross-Over Studies , Drug Combinations , Drug Therapy, Combination , Ezetimibe , Female , Humans , Male , Middle Aged , Reference Values , Simvastatin/administration & dosage , Simvastatin/adverse effects , Tablets , Therapeutic Equivalency , Time Factors , Treatment Outcome
8.
J Clin Pharmacol ; 45(2): 185-92, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15647411

ABSTRACT

Posaconazole is a triazole antifungal in development for the treatment of invasive fungal infections. The authors evaluated the pharmacokinetics and safety of posaconazole in healthy subjects and in those with mild (CL(CR) = 50-80 mL/min), moderate (CL(CR) = 20-49 mL/min), and severe chronic renal disease (CL(CR) <20 mL/min; receiving outpatient hemodialysis) (n = 6/group). Subjects received one 400-mg dose of posaconazole oral suspension with a standardized high-fat breakfast. For hemodialysis-dependent subjects, this dose was given on a nonhemodialysis day, and a second 400-mg dose was given 6 hours before hemodialysis. Blood samples were collected before dose and up to 120 hours postdose. For hemodialysis-dependent subjects following the second dose, additional samples (predialyzed and postdialyzed) were collected before, during, and after dialysis. There was no correlation between posaconazole pharmacokinetics and mild to moderate renal disease; the slopes of the linear regressions for creatinine clearance versus posaconazole AUC, C(max), CL/F, and t1/2 values were not significantly different from zero (P > .130). Mean CL/F values before and during hemodialysis were comparable. Furthermore, the difference in the predialyzed and postdialyzed posaconazole concentrations was only approximately 3%, supporting that posaconazole was not removed by hemodialysis. Protein binding was similar in all groups (approximately 98%) and was unaffected by hemodialysis. Posaconazole was generally well tolerated. One patient had elevated liver function test results that were not present at baseline and were thought to be possibly related to posaconazole. Results of this single-dose study indicate that dosage adjustments for patients with varying degrees of renal disease are not required.


Subject(s)
Kidney Failure, Chronic/drug therapy , Treatment Outcome , Triazoles/pharmacokinetics , Administration, Oral , Alanine Transaminase/drug effects , Alanine Transaminase/metabolism , Area Under Curve , Aspartate Aminotransferases/drug effects , Aspartate Aminotransferases/metabolism , Biological Availability , Creatinine/metabolism , Dietary Fats/administration & dosage , Dietary Fats/pharmacokinetics , Drug Administration Schedule , Eating , Female , Half-Life , Humans , Kidney Failure, Chronic/epidemiology , Liver Function Tests/adverse effects , Male , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Middle Aged , Protein Binding/drug effects , Renal Dialysis/methods , Suspensions , Triazoles/administration & dosage , Triazoles/metabolism
9.
Int J Clin Pharmacol Ther ; 42(9): 512-8, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15487810

ABSTRACT

OBJECTIVE: Ezetimibe is a novel lipid-lowering drug that prevents intestinal absorption of dietary and biliary cholesterol leading to significant reduction in total-C, LDL-C, Apo B, and TG and increases in HDL-C in patients with hypercholesterolemia. Gemfibrozil, a fibric acid derivative, is an effective lipid-modulating agent that increases serum high-density lipoprotein cholesterol and decreases serum TG. The objective of this study was to evaluate the potential for a pharmacokinetic (PK) interaction between ezetimibe and gemfibrozil. METHODS: This was a randomized, open-label, 3-way crossover, multiple-dose study in 12 healthy adult male volunteers. All subjects received the following 3 treatments orally for 7 days: ezetimibe 10 mg once daily, gemfibrozil 600 mg every 12 hours, and ezetimibe 10 mg once daily plus gemfibrozil 600 mg every 12 hours. A washout period of > or = 7 days separated the 3 treatments. In each treatment, blood samples were collected on day 7 to assess the steady-state PK of ezetimibe and gemfibrozil. The oral bioavailability of ezetimibe coadministered with gemfibrozil relative to each drug administered alone was evaluated with an analysis-of-variance model. RESULTS: Ezetimibe was rapidly absorbed and extensively conjugated to its glucuronide metabolite. Ezetimibe did not alter the bioavailability (based on AUC) of gemfibrozil. The mean AUC0-12 of gemfibrozil was 74.7 and 74.1 microg h/ml with and without ezetimibe coadministration, respectively (log-transformed geometric mean ratio (GMR) = 99.2; 90% confidence interval (CI) = 92 - 107%). Conversely, gemfibrozil significantly (p < 0.05) increased the plasma concentrations of ezetimibe and total ezetimibe (i.e. ezetimibe plus ezetimibe-glucuronide). Exposure to ezetimibe and total ezetimibe was increased approximately 1.4-fold and 1.7-fold, respectively (CI = 109 - 173% for ezetimibe and 142 - 190% for total ezetimibe), however, this increase was not considered to be clinically relevant. Ezetimibe and gemfibrozil administered alone or concomitantly for 7 days was well tolerated. CONCLUSIONS: The coadministration of ezetimibe and gemfibrozil in patients is unlikely to cause a clinically significant drug interaction. The coadministration of these agents is a promising approach for patients with mixed dyslipidemia. Additional clinical studies are warranted.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Azetidines/pharmacokinetics , Gemfibrozil/pharmacokinetics , Administration, Oral , Adolescent , Adult , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/adverse effects , Area Under Curve , Azetidines/administration & dosage , Azetidines/adverse effects , Cross-Over Studies , Drug Administration Schedule , Drug Interactions , Ezetimibe , Gemfibrozil/administration & dosage , Gemfibrozil/adverse effects , Humans , Hyperlipidemias/drug therapy , Male , Middle Aged , Reference Values
10.
Xenobiotica ; 34(5): 487-500, 2004 May.
Article in English | MEDLINE | ID: mdl-15370964

ABSTRACT

1. Temozolomide, an imidazotetrazine derivative, is a cytotoxic alkylating agent of broad-spectrum antitumour activity. The absorption, metabolism, distribution and excretion of temozolomide have been investigated in male and female Sprague-Dawley and Long-Evans rats following single oral or intravenous dose administration of 200 mg m(-2) non-radiolabelled or (14)C-radiolabelled temozolomide. The distribution of (14)C-temozolomide was also evaluated by whole-body autoradiography in male Sprague-Dawley rats. Plasma concentrations of temozolomide and its active metabolite 3-methyl-(triazen-1-yl)imidazole-4-carboxamide (MTIC) were determined by high-performance liquid chromatography (HPLC) with ultraviolet detection. Plasma, urine and faeces were profiled by HPLC with radiochemical detection. 2. Temozolomide was rapidly and extensively (>90%) absorbed and widely distributed in tissues. The distribution pattern of radioactivity was gender independent. Penetration into the brain following oral or intravenous administration was 35-39% based on the brain/plasma AUC ratio. 3. Following intravenous or oral administration, temozolomide was primarily eliminated renally (75-85% of the dose) as either unchanged drug, a carboxylic acid analogue, AIC (a degradation product) and a highly polar unidentified peak. Biliary excretion was minimal (1.4-1.6%). The pharmacokinetics (oral versus intravenous) were similar and gender independent. The absolute oral availability was 96-100%. Temozolomide was rapidly eliminated (t(1/2) = 1.2 h) and converted to MTIC. 4. Systemic exposure to MTIC was about 2% that of temozolomide. Overall, the disposition of temozolomide in rats was similar to that observed in humans.


Subject(s)
Dacarbazine/analogs & derivatives , Dacarbazine/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/pharmacokinetics , Autoradiography , Brain/metabolism , Carbon Radioisotopes , Dacarbazine/administration & dosage , Dacarbazine/metabolism , Female , Injections, Intravenous , Male , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/metabolism , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Temozolomide , Tissue Distribution
11.
Br J Cancer ; 88(7): 1004-11, 2003 Apr 07.
Article in English | MEDLINE | ID: mdl-12671695

ABSTRACT

Temozolomide, an oral DNA methylator that inactivates the DNA repair enzyme O(6)-alkylguanine-DNA alkyltransferase (AGAT), has demonstrated anticancer activity on protracted schedules. Protracted schedules may lead to an 'autoenhancement' of temozolomide's inherent cytotoxic potential by cumulative reduction of the cell's capacity for AGAT-mediated DNA repair and resistance. This study was undertaken to characterise AGAT inactivation and regeneration in the peripheral blood mononuclear cells (PBMCs) of patients treated on two protracted temozolomide schedules. O(6)-alkyl guanine-DNA alkyltransferase activity was measured in the PBMCs of patients treated on two phase I protracted temozolomide studies. Patients were treated daily for either 7 days every 2 weeks (Schedule A) or 21 days every 4 weeks (Schedule B). The effects of various temozolomide doses (75-175 mg m(-2)), treatment duration (7-21 days), and temozolomide plasma levels on AGAT inactivation and regeneration, as well as the relation between AGAT inactivation and toxicity, were studied. O(6)-alkyl guanine-DNA alkyltransferase activity in PBMCs was measured serially in 52 patients. Marked inactivation of AGAT occurred following 7 days of temozolomide treatment, with mean AGAT activity decreasing by 72% (P<0.0001). Similarly, mean AGAT activity decreased by 63 and 73% after 14 and 21 days of treatment, respectively (P<0.001 for both comparisons). O(6)-alkyl guanine-DNA alkyltransferase inactivation was greater after 7 days of treatment with higher doses of temozolomide than lower doses and remained markedly reduced 7 days post-treatment. However, AGAT inactivation following temozolomide treatment for 14 and 21 days was similar at all doses. On the continuous 21-day schedule, AGAT inactivation was significantly greater in patients who experienced severe thrombocytopenia than those who did not (90.3+/-5.5 vs 72.5+/-16.1%, P<0.045). In conclusion, protracted administration of temozolomide, even at relatively low daily doses, leads to significant and prolonged depletion of AGAT activity, which may enhance the antitumour activity of the agent.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Dacarbazine/analogs & derivatives , Dacarbazine/administration & dosage , Enzyme Inhibitors/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Dacarbazine/pharmacology , Dose-Response Relationship, Drug , Drug Administration Schedule , Humans , Leukocytes, Mononuclear/enzymology , Temozolomide
12.
Eur J Cancer ; 38(17): 2272-8, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12441264

ABSTRACT

A single-agent dose-escalating phase I study on the farnesyl transferase inhibitor SCH 66336 was performed to determine the safety profile and recommended dose for phase II studies. Plasma pharmacokinetics were determined as well as the SCH 66336-induced inhibition of farnesyl protein transferase in vivo. SCH 66336 was given orally once daily (OD) without interruption to patients with histologically-confirmed solid tumours. Routine antiemetics were not prescribed. 12 patients were enrolled into the study. Dose levels studied were 300 mg (6 patients) and 400 mg (6 patients) OD. Pharmacokinetic sampling was performed on days 1 and 15. Although at 400 mg OD only 1 patient had a grade 3 diarrhoea, 3 out of 6 patients interrupted treatment early due to a combination of various grade 1-3 toxicities (diarrhoea, uremiacreatinine, asthenia, vomiting, weight loss) indicating that this dose was not tolerable for a prolonged period of time. At 300 mg OD, the same pattern of toxicities was observed, but all were grade 1-2. Therefore, this dose can be recommended for phase II studies. Pharmacokinetic analysis showed that peak plasma concentrations as well as the AUCs were dose-related, with increased parameters at day 15 compared with day 1, indicating some accumulation upon multiple dosing. Plasma half-life ranged from 5 to 9 h and appeared to increase with increasing dose. Steady state plasma concentrations were attained by day 14. A large volume of distribution at steady state suggested extensive distribution outside the plasma compartment. There is evidence of inhibition of protein prenylation in some patients after OD oral administration of SCH 66336. SCH 66336 can be safely administered using a continuous oral OD dosing regimen. The recommended dose for phase II studies using this regimen is 300 mg OD.


Subject(s)
Antineoplastic Agents/administration & dosage , Enzyme Inhibitors/administration & dosage , Neoplasms/drug therapy , Piperidines/administration & dosage , Pyridines/administration & dosage , Administration, Oral , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Drug Resistance, Neoplasm , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Female , Follow-Up Studies , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/metabolism , Piperidines/adverse effects , Piperidines/pharmacokinetics , Pyridines/adverse effects , Pyridines/pharmacokinetics
13.
Ann Oncol ; 13(7): 1067-71, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12176785

ABSTRACT

BACKGROUND: ras genes encode Ras proteins that are important for signal transduction in cancer cells. Farnesyl protein transferase (FPTase) is an enzyme that is responsible for a critical post-translational modification of Ras. PATIENTS AND METHODS: We report the results of a phase II trial of SCH 66336, an FPTase inhibitor, in patients with metastatic colorectal cancer. This is the first reported experience of an FPTase inhibitor in this disease. All patients were considered refractory to first- and second-line therapy. A total of 21 evaluable patients were treated with a starting dose of 200 mg b.i.d. given continuously. RESULTS: The major side-effects were fatigue (grade 1 in 42%, grade 2 in 42% and grade 3 in 14%), diarrhea (grade 1 in 23% and grade 3 in 42%) and nausea (grade 2 in 16%). Elevations in serum creatinine (grade 2 or 3) were observed in 19% of patients and appeared to be related to dehydration induced by diarrhea. Significant hematological toxicity was not observed (only grade 1 thrombocytopenia in 19% and grade 2 or 3 anemia in 28%). Pharmacological studies revealed adequate mean pre-dose plasma concentrations in this group of patients on day 15 of therapy. No objective responses were observed, although stable disease was seen in three patients for several months. Administration of SCH 66336 was accompanied by gastrointestinal toxicity. CONCLUSIONS: Future development of this compound cannot be recommended as monotherapy in this disease.


Subject(s)
Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Enzyme Inhibitors/administration & dosage , Fluorouracil/administration & dosage , Piperidines/administration & dosage , Pyridines/administration & dosage , Administration, Oral , Adult , Aged , Aged, 80 and over , Biopsy, Needle , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Resistance , Female , Follow-Up Studies , Humans , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Male , Middle Aged , Neoplasm Staging , Survival Analysis , Treatment Outcome
14.
J Clin Pharmacol ; 41(9): 943-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11549098

ABSTRACT

Ezetimibe is a novel selective inhibitor of intestinal cholesterol absorption, which has been shown to significantly decrease low-density lipoprotein cholesterol (LDL-C). In this article, the relationship between plasma ezetimibe concentrations and lowering of LDL-C is determined using Emax and regression models. Data from two phase II double-blind placebo-controlled studies (n = 232 and 177) were used in which daily doses of ezetimibe ranging from 0.25 to 10 mg were administered for 12 weeks. Ezetimibe concentrations correlated significantly with percentage change in LDL-C from baseline (%LDL-C). Reductions in %LDL-C of 10%, 15%, and 20% were achieved with concentrations in the ranges 0 to 2, 2 to 15, and > 15 ng/ml, respectively, as compared with placebo. To achieve > 15% reduction in LDL-C, patients need to maintain trough concentrations > 15 ng/ml, taking plasma concentrations as a surrogate for concentrations at the enterocyte. Based on the doses administered, the 10 mg dose had the highest likelihood of sustaining such concentrations, confirming that a daily 10 mg dose of ezetimibe is an optimal therapeutic dose in the treatment of hypercholesterolemia.


Subject(s)
Anticholesteremic Agents/blood , Anticholesteremic Agents/therapeutic use , Azetidines/blood , Azetidines/therapeutic use , Cholesterol, LDL/blood , Hypercholesterolemia/drug therapy , Adolescent , Adult , Aged , Algorithms , Anticholesteremic Agents/adverse effects , Azetidines/adverse effects , Chromatography, Liquid , Dose-Response Relationship, Drug , Ezetimibe , Female , Humans , Hypercholesterolemia/blood , Linear Models , Male , Mass Spectrometry , Middle Aged
15.
Clin Ther ; 23(6): 871-85, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11440287

ABSTRACT

BACKGROUND: Ezetimibe, a selective inhibitor of intestinal cholesterol absorption, is in clinical development for the treatment of hypercholesterolemia. It is rapidly absorbed and glucuronidated in the intestine. The parent compound and its conjugated metabolite undergo enterohepatic recirculation, resulting in multiple peaks in the plasma concentration-time profile. OBJECTIVE: The purpose of this study was to develop a population pharmacokinetic (PPK) model for ezetimibe that incorporates enterohepatic recirculation. METHODS: A population compartment model incorporating input from the gallbladder, consistent with food intake, was developed to account for enterohepatic recirculation. The amount recycled was allowed to vary within a subject and between subjects, accommodating variability in bile secretion. The data used consisted of 90 profiles from healthy subjects who received single or multiple doses of ezetimibe 10 or 20 mg. Modeling was carried out using a nonlinear mixed-effect function in the S-PLUS statistical program. RESULTS: The amount of ezetimibe recycled into the central compartment was estimated to be approximately 17% to 20% of the total amount absorbed, independent of the volume of distribution. The intersubject coefficient of variation was 46% to 80% in the absorption rate constant, 27% in the distribution phase, and approximately 50% in the volume of distribution. CONCLUSIONS: PPK models adapted for enterohepatic recirculation allowed a formal assessment of the magnitude and frequency of the enterohepatic recirculation process, and the associated intersubject and intrasubject variability in healthy subjects. The PPK approach also helped to assess the correlation between the observed maximum or minimum (24 hours postdose) concentration with the model-based area under the curve, confirming the appropriateness of the former measures as a surrogate of drug exposure for a possible correlation with pharmacodynamics.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Azetidines/pharmacokinetics , Enterohepatic Circulation , Adult , Algorithms , Anticholesteremic Agents/blood , Area Under Curve , Azetidines/blood , Ezetimibe , Female , Half-Life , Humans , Male , Models, Biological
16.
J Pharm Biomed Anal ; 24(3): 461-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11199225

ABSTRACT

Temozolomide (SCH 52365; TEMODAL) is an antineoplastic agent with activity against a broad spectrum of murine tumors. This compound is currently marketed in the European Union for the treatment of patients with glioblastoma multiforme and anaplastic astrocytoma, which are serious and aggressive types of brain cancers. It has been postulated that temozolomide exerts its in vivo activity via the decomposition product MTIC, which is believed to alkylate nucleophiles, and in the process is converted to AIC. A high-performance liquid chromatographic (HPLC) method was developed and validated for the analysis of temozolomide in human plasma. The determination of temozolomide involved extraction with ethyl acetate followed by separation on a reversed phase C-18 column and quantification by UV absorbance at 316 nm. The calibration curve was linear over a concentration range of 0.1-20 microg/ml. The limit of quantitation was 0.1 microg/ml, where the coefficient of variation (CV) was 0% and the bias was 10.0%. The method was precise with a coefficient of variation ranging from 2.5 to 6.9% and accurate with a bias ranging from 5.0 to 10.0%. Temozolomide was unstable at 37 degrees C in human plasma with a degradation t1/2 of 15 min; however, it was stable at 4 degrees C for at least 30 min. Temozolomide was stable in acidified human plasma (pH < 4) for at least 24 h at 25 degrees C, and for at least 30 days at -20 degrees C. Moreover, temozolomide was stable in acidified human plasma after being subjected to three freeze thaw cycles. The assay was shown to be specific, accurate, precise, and reliable for use in pharmacokinetic studies.


Subject(s)
Antineoplastic Agents/blood , Chromatography, High Pressure Liquid/methods , Dacarbazine/analogs & derivatives , Dacarbazine/blood , Antineoplastic Agents/pharmacokinetics , Dacarbazine/pharmacokinetics , Humans , Reference Standards , Reproducibility of Results , Spectrophotometry, Ultraviolet , Temozolomide
17.
J Clin Oncol ; 19(4): 1167-75, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11181683

ABSTRACT

PURPOSE: A single-agent dose-escalating phase I and pharmacokinetic study on the farnesyl transferase inhibitor SCH 66336 was performed to determine the safety profile, maximum-tolerated dose, and recommended dose for phase II studies. Plasma and urine pharmacokinetics were determined. PATIENTS AND METHODS: SCH 66336 was given orally bid without interruption to patients with histologically or cytologically confirmed solid tumors. Routine antiemetics were not prescribed. RESULTS: Twenty-four patients were enrolled onto the study. Dose levels studied were 25, 50, 100, 200, 400, and 300 mg bid. Pharmacokinetic sampling was performed on days 1 and 15. At 400 mg bid, the dose-limiting toxicity (DLT) consisted of grade 4 vomiting, grade 4 neutropenia and thrombocytopenia, and the combination of grade 3 anorexia and diarrhea with reversible grade 3 plasma creatinine elevation. After dose reduction, at 300 mg bid, the DLTs consisted of grade 4 neutropenia, grade 3 neurocortical toxicity, and the combination of grade 3 fatigue with grade 2 nausea and diarrhea. The recommended dose for phase II studies is 200 mg bid, which was found feasible for prolonged periods of time. Pharmacokinetic analysis showed a greater than dose-proportional increase in drug exposure and peak plasma concentrations, with increased parameters at day 15 compared with day 1, indicating some accumulation on multiple dosing. Plasma half-life ranged from 4 to 11 hours and seemed to increase with increasing doses. Steady-state plasma concentrations were attained at days 7 through 14. A large volume of distribution at steady-state indicated extensive distribution outside the plasma compartment. CONCLUSION: SCH 66336 can be administered safely using a continuous oral bid dosing regimen. The recommended dose for phase II studies using this regimen is 200 mg bid.


Subject(s)
Antineoplastic Agents/therapeutic use , Piperidines/therapeutic use , Pyridines/therapeutic use , Administration, Oral , Adult , Aged , Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase , Female , Humans , Male , Middle Aged , Piperidines/pharmacokinetics , Pyridines/pharmacokinetics
18.
J Clin Oncol ; 18(1): 158-66, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10623706

ABSTRACT

PURPOSE: To compare, in 305 patients with advanced metastatic melanoma, temozolomide and dacarbazine (DTIC) in terms of overall survival, progression-free survival (PFS), objective response, and safety, and to assess health-related quality of life (QOL) and pharmacokinetics of both drugs and their metabolite, 5-(3-methyltriazen-1-yl)imidazole-4-carboximide (MTIC). PATIENTS AND METHODS: Patients were randomized to receive either oral temozolomide at a starting dosage of 200 mg/m(2)/d for 5 days every 28 days or intravenous (IV) DTIC at a starting dosage of 250 mg/m(2)/d for 5 days every 21 days. RESULTS: In the intent-to-treat population, median survival time was 7.7 months for patients treated with temozolomide and 6.4 months for those treated with DTIC (hazards ratio, 1.18; 95% confidence interval [CI], 0.92 to 1.52). Median PFS time was significantly longer in the temozolomide-treated group (1.9 months) than in the DTIC-treated group (1.5 months) (P =.012; hazards ratio, 1.37; 95% CI, 1.07 to 1.75). No major difference in drug safety was observed. Temozolomide was well tolerated and produced a noncumulative, transient myelosuppression late in the 28-day cycle. The most common nonhematologic toxicities were mild to moderate nausea and vomiting, which were easily managed. Temozolomide therapy improved health-related QOL; more patients showed improvement or maintenance of physical functioning at week 12. Systemic exposure (area under the curve) to the parent drug and the active metabolite, MTIC, was higher after treatment with oral temozolomide than after IV administration of DTIC. CONCLUSION: Temozolomide demonstrates efficacy equal to that of DTIC and is an oral alternative for patients with advanced metastatic melanoma.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Melanoma/drug therapy , Adult , Aged , Aged, 80 and over , Biological Availability , Consumer Product Safety , Dacarbazine/pharmacokinetics , Disease-Free Survival , Female , Humans , Male , Melanoma/mortality , Melanoma/pathology , Neoplasm Metastasis , Quality of Life , Regression Analysis , Survival Rate
19.
J Clin Oncol ; 17(8): 2604-13, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10561328

ABSTRACT

PURPOSE: To determine the principal toxicities, characterize the pharmacokinetics (PKs) and pharmacodynamics (PDs) of temozolomide (TMZ) on a daily-for-5-days schedule, and recommend a dose for subsequent disease-directed studies in both minimally pretreated (MP) and heavily pretreated (HP) patients. PATIENTS AND METHODS: Patients received TMZ as a single oral dose daily for 5 consecutive days every 28 days. TMZ doses were escalated from 100 to 150, and 150 to 200 mg/m(2)/d in separate cohorts of MP and HP patients. PK plasma was sampled on days 1 and 5. TMZ concentrations were analyzed and pertinent PK parameters were related to the principal toxicities of TMZ in PD analyses. RESULTS: Twenty-four patients were treated with 85 courses of TMZ. Thrombocytopenia and neutropenia were the principal dose-limiting toxicities (DLTs) of TMZ on this schedule. The cumulative rate of severe myelosuppressive effects was unacceptably high at TMZ doses exceeding 150 mg/m(2)/d in both MP and HP patients. TMZ was absorbed rapidly with maximum concentrations achieved in 0.90 hours, on average, and elimination was rapid, with a half-life and systemic clearance rate (Cl(S/F)) averaging 1.8 hours and 115 mL/min/m(2), respectively. When clearance was normalized to body-surface area (BSA), interpatient variability in Cl(S/F) was reduced from 20% to 13% on day 1 and from 16% to 10% on day 5. Patients who experienced DLT had significantly higher maximum drug concentration( )(median 16 v 9.5 microg/mL, P =. 0084) and area under the concentration-time curve (median 36 v 23 microg-h/mL, P =.0019) values on day 5. CONCLUSION: Prior myelosuppressive therapy was not a determinant of toxicity. TMZ 150 mg/m(2)/d administered as a single oral dose daily for 5 days every 4 weeks is well tolerated by MP and HP patients, with higher doses resulting in unacceptably high rates of severe hematologic toxicity. TMZ doses should be individualized according to BSA rather than use of a prespecified oral dose for all individuals. TMZ is an optimal agent to develop in combination with other cytotoxic, biologic, and targeted therapeutics for patients with relevant malignancies.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Dacarbazine/analogs & derivatives , Neoplasms/drug therapy , Neutropenia/chemically induced , Administration, Oral , Adult , Aged , Antineoplastic Agents, Alkylating/pharmacokinetics , Area Under Curve , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Dacarbazine/pharmacokinetics , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Male , Metabolic Clearance Rate , Middle Aged , Neoplasms/pathology , Temozolomide
20.
Br J Cancer ; 81(6): 1022-30, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10576660

ABSTRACT

Temozolomide, an oral cytotoxic agent with approximately 100% bioavailability after one administration, has demonstrated schedule-dependent clinical activity against highly resistant cancers. Thirty patients with minimal prior chemotherapy were enrolled in this phase I trial to characterize the drug's safety, pharmacokinetics and anti-tumour activity, as well as to assess how food affects oral bioavailability. To determine dose-limiting toxicities (DLT) and the maximum tolerated dose (MTD), temozolomide 100-250 mg m(-2) was administered once daily for 5 days every 28 days. The DLT was thrombocytopenia, and the MTD was 200 mg m(-2) day(-1). Subsequently, patients received the MTD to study how food affects the oral bioavailability of temozolomide. When given orally once daily for 5 days, temozolomide was well tolerated and produced a non-cumulative, transient myelosuppression. The most common non-haematological toxicities were mild to moderate nausea and vomiting. Clinical activity was observed against several advanced cancers, including malignant glioma and metastatic melanoma. Temozolomide demonstrated linear and reproducible pharmacokinetics and was rapidly absorbed (mean Tmax approximately 1 h) and eliminated (mean t1/2 = 1.8 h). Food produced a slight reduction (9%) in absorption of temozolomide. Temozolomide 200 mg m(-2) day(-1) for 5 days, every 28 days, is recommended for phase II studies.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Administration, Oral , Adult , Aged , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Biological Availability , Brain Neoplasms/metabolism , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Dacarbazine/pharmacokinetics , Dose-Response Relationship, Drug , Eating , Female , Glioma/metabolism , Humans , Male , Melanoma/metabolism , Middle Aged , Neoplasms/drug therapy , Skin Neoplasms/metabolism , Temozolomide , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL