Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cancer Chemother Pharmacol ; 89(1): 117-128, 2022 01.
Article in English | MEDLINE | ID: mdl-34786600

ABSTRACT

PURPOSE: Erdafitinib (JNJ-42756493, BALVERSA) is a tyrosine kinase inhibitor indicated for the treatment of advanced urothelial carcinoma. In this work, a translational model-based approach to inform the choice of the doses in phase 1 trials is illustrated. METHODS: A pharmacokinetic (PK) model was developed to describe the time course of erdafitinib plasma concentrations in mice and rats. Data from multiple xenograft studies in mice and rats were analyzed using the Simeoni tumor growth inhibition (TGI) model. The model parameters were used to derive a range of erdafitinib exposures that might inform the choice of the doses in oncology phase 1 trials. Conversion of exposures to doses was based on preliminary PK assessments from the first-in human (FIH) study. RESULTS: A one-compartment PK disposition model, with linear absorption and dose-dependent clearance, adequately described the PK data in both mice and rats via an allometric scaling approach. The TGI model was able to describe tumor growth dynamics, providing quantitative measurements of erdafitinib antitumor potency in mice and rats. Based on these estimates, ranges of efficacious unbound concentration were identified for erdafitinib in mice (0.642-5.364 µg/L) and rats (0.782-2.565 µg/L). Based on the FIH data, it was possible to transpose exposures into doses and doses of above 4 mg/day provided erdafitinib exposures associated with significant TGI in animals. The findings were in agreement with the results of the FIH trial, in which the first hints of clinical activities were observed at 6 mg. CONCLUSION: The successful modeling exercise of erdafitinib preclinical data showed how translational PK-PD modeling might be a tool to help to inform the choice of the doses in FIH studies.


Subject(s)
Pyrazoles/administration & dosage , Pyrazoles/pharmacokinetics , Quinoxalines/administration & dosage , Quinoxalines/pharmacokinetics , Translational Research, Biomedical/methods , Animals , Clinical Trials, Phase I as Topic , Humans , Mice, Nude , Models, Biological , Pyrazoles/blood , Quinoxalines/blood , Rats , Xenograft Model Antitumor Assays
2.
Eur J Cancer ; 43(12): 1862-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17604156

ABSTRACT

The success rate of clinical drug development is significantly lower in oncology than in other therapeutic areas. Predicting the activity of new compounds in humans from preclinical data could substantially reduce the number of failures. A novel approach for predicting the expected active doses in humans from the first animal studies is presented here. The method relies upon a PK/PD model of tumour growth inhibition in xenografts, which provides parameters describing the potency of the tested compounds. Anticancer drugs, currently used in the clinic, were evaluated in xenograft models and their potency parameters were estimated. A good correlation was obtained between these parameters and the exposures sustained at the therapeutically relevant dosing regimens. Based on the corresponding regression equation and the potency parameters estimated in the first preclinical studies, the therapeutically active concentrations of new compounds can be estimated. An early knowledge of level of exposure or doses to be reached in humans will improve the risk evaluation and decision making processes in anticancer drug development.


Subject(s)
Antineoplastic Agents/administration & dosage , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , Mice , Mice, Nude , Random Allocation , Xenograft Model Antitumor Assays
3.
Eur J Med Chem ; 41(7): 795-808, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16644065

ABSTRACT

The site of biotransformation, the extent and rate of metabolism and the number of active metabolic pathways are among the most important characteristics of the pharmacokinetics of a drug. The catalytic activity of drug metabolizing enzymes is likely the most influential determinant of the pharmacokinetic variability. Metabolic stability is the prerequisite for sustaining the therapeutically relevant concentrations. Metabolic inhibition and induction can give rise to clinically important drug-drug interactions. A variety of computational approaches are currently available for predicting different cytochrome P450 (CYP)-related metabolism endpoints. The present review will describe these approaches and their impact on drug development process. Indications on the available software for the implementation will also be given.


Subject(s)
Computational Biology/methods , Cytochrome P-450 Enzyme System/metabolism , Drug Evaluation, Preclinical/methods , Animals , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Ligands , Substrate Specificity
4.
Math Biosci ; 200(2): 127-51, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16516246

ABSTRACT

A mathematical model for describing the cancer growth dynamics in response to anticancer agents administration in xenograft models is discussed. The model consists of a system of ordinary differential equations involving five parameters (three for describing the untreated growth and two for describing the drug action). Tumor growth in untreated animals is modelled by an exponential growth followed by a linear growth. In treated animals, tumor growth rate is decreased by an additional factor proportional to both drug concentration and proliferating cells. The mathematical analysis conducted in this paper highlights several interesting properties of this tumor growth model. It suggests also effective strategies to design in vivo experiments in animals with potential saving of time and resources. For example, the drug concentration threshold for the tumor eradication, the delay between drug administration and tumor regression, and a time index that measures the efficacy of a treatment are derived and discussed. The model has already been employed in several drug discovery projects. Its application on a data set coming from one of these projects is discussed in this paper.


Subject(s)
Antineoplastic Agents/pharmacology , Models, Biological , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Xenograft Model Antitumor Assays/methods , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Cell Growth Processes/drug effects , Cell Line, Tumor , Humans , Mice , Mice, Nude , Neoplasms, Experimental/metabolism , Tumor Burden
5.
Cancer Res ; 55(10): 2090-6, 1995 May 15.
Article in English | MEDLINE | ID: mdl-7743508

ABSTRACT

Methoxymorpholinyldoxorubicin (FCE 23762) is a novel, highly lipophilic doxorubicin analogue. It possesses potent in vitro and in vivo antitumor activity including efficacy in multidrug-resistant tumor cell lines. It is also metabolically activated in vivo resulting in an 80-fold increase in potency over the parent drug. In this phase I study the drug was administered by i.v. bolus injection at 3-week intervals. Fifty-three patients with refractory solid tumors were treated; 133 courses of FCE 23762 were administered at doses ranging from 30 to 2250 micrograms/m2. The dose limiting toxicity was reversible myelo-suppression (granulocytopenia and thrombocytopenia), demonstrating a delayed nadir and recovery in comparison to doxorubicin. Other toxicities included transient elevation of hepatic transaminases, delayed and prolonged nausea and vomiting, mucositis, anorexia, fatigue, and diarrhea. Heavily pretreated patients demonstrated more myelosuppression than previously untreated patients at 1250 micrograms/m2. No cardiotoxicity was observed. Four objective tumor responses were seen: one complete response in a patient with pelvic recurrence of cervical cancer; one partial response in a patient with cutaneous and lymph gland metastases from head and neck cancer; and two minor responses in patients with liver metastases from colorectal cancer. Plasma concentrations of FCE 23762 and its 13-dihydro metabolite, FCE 26176, were measured in 20 patients at doses > or = 675 micrograms/m2, using HPLC with fluorescence detection. The area under the plasma concentration-time curve ranged from 30 to 80 ng/h/ml; plasma data suggested linear kinetics in the range of tested doses (although there was considerable interpatient variability). The maximum tolerated dose defined in this study using this schedule is 1500 micrograms/m2. A safe phase II dose for previously untreated patients using this schedule is 1250 micrograms/m2; however, this may actually be below the optimal dose for this patient population.


Subject(s)
Doxorubicin/analogs & derivatives , Neoplasms/drug therapy , Adult , Aged , Doxorubicin/adverse effects , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Female , Humans , Liver/drug effects , Male , Middle Aged , Neoplasms/metabolism , Neutropenia/chemically induced , Thrombocytopenia/chemically induced
6.
Clin Cancer Res ; 4(1): 53-9, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9516952

ABSTRACT

Tallimustine [PNU 152241 (FCE 24517)] is a synthetic derivative of the DNA minor groove binder distamycin A, in which the NH2-terminal formyl group is substituted by benzoyl mustard. In this Phase I clinical trial, patients with advanced solid tumors received i.v. bolus injections of tallimustine daily for 3 consecutive days. Patients were treated at six dosage levels of 33.3 micrograms/m2/day to 250 micrograms/m2/day for 3 consecutive days, with courses of therapy repeated every 28 days. Detailed pharmacokinetic blood sampling was performed during the first 3 days of the first course of tallimustine. The plasma samples were analyzed by high-performance liquid chromatography with UV detection. Forty-eight eligible patients were treated at all six dosage levels. The dominant dose-related toxicity of tallimustine was neutropenia, becoming dose limiting at 250 micrograms/m2/day. At this dosage level, one patient experienced febrile neutropenia, and a second patient died on study of indeterminate cause. Thrombocytopenia was not observed, and only 10 patients developed anemia < 8.0 gm/dl. Sporadic elevation of liver enzymes or bilirubin was observed but was not dose related. Pharmacokinetic analysis gave reliable results for 33 patients. For most patients, analysis of the data best fit a three-exponential model. Dose-related increases in areas under the concentration-time curve and end-of-infusion concentrations were observed. There was no significant plasma accumulation of tallimustine over the 3 days of administration. The terminal half-life of tallimustine in individual patients ranged from 6.83 to 39.02 h following the last dose. In summary, the recommended Phase II dosage for tallimustine is 200 micrograms/m2/day for 3 consecutive days every 28 days. Neutropenia is the principal toxicity of this agent at this dosage and schedule.


Subject(s)
Antineoplastic Agents/adverse effects , Distamycins/adverse effects , Neoplasms/drug therapy , Nitrogen Mustard Compounds/adverse effects , Adult , Aged , Distamycins/administration & dosage , Distamycins/pharmacokinetics , Female , Humans , Male , Middle Aged , Nitrogen Mustard Compounds/administration & dosage , Nitrogen Mustard Compounds/pharmacokinetics
7.
Clin Cancer Res ; 7(12): 3928-33, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751484

ABSTRACT

Our aim was to establish, in patients with solid tumors, the dose-limiting toxicity, maximum tolerated dose (MTD), and pharmacology of PNU-145156E, a new sulfonated distamycin A derivative that blocked circulating angiogenesis-promoting growth factors in animal studies and exhibited an antitumor effect in murine solid tumors. In a Phase I study, PNU-145156E was administered i.v. every 6 weeks. Included were patients with solid tumors; an Eastern Cooperative Oncology Group performance score

Subject(s)
Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/pharmacokinetics , Distamycins/adverse effects , Distamycins/pharmacokinetics , Neoplasms/drug therapy , Adult , Aged , Angiogenesis Inhibitors/blood , Area Under Curve , Blood Coagulation/drug effects , Distamycins/blood , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Male , Metabolic Clearance Rate , Middle Aged , Neoplasms/blood , Patient Selection
8.
Clin Pharmacol Ther ; 66(3): 282-7, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10511064

ABSTRACT

BACKGROUND: Reboxetine [(R,S)-2[(R,S)-alpha-(2-ethoxyphenoxy)benzyl]morpholine methanesulfonate] is a racemic compound that consists of equal proportions of R,R- and S,S-enantiomers. This study investigated the hemodynamic effects of reboxetine and the R,R-enantiomer compared with placebo in volunteers. The pharmacokinetics of reboxetine and its enantiomers were also investigated in the study. METHODS: Nine healthy, male volunteers received single doses of 4 mg reboxetine, 2 mg R,R-enantiomer, and placebo at weekly intervals. Reboxetine and the R,R-enantiomer were well tolerated in all volunteers. RESULTS: The heart rates of patients in the supine and standing positions were increased after reboxetine administration compared with the R,R-enantiomer (P < .05, except supine heart rate at 6 hours) and placebo (P < .05). Supine systolic and diastolic blood pressure was also increased by 3 +/- 4 and 1 +/- 4 mm Hg, respectively, after reboxetine compared with R,R-enantiomer (-2 +/- 4 and -4 +/- 3 mm Hg) and placebo (-4 +/- 4 and -4 +/- 4 mm Hg) administration. The systolic and diastolic blood pressure measurements for subjects while standing did not differ significantly among treatments. There was no significant difference between the maximum plasma concentration, mean time to maximum plasma concentration, plasma half-life, or area under the plasma concentration-time curve (AUC) of the R,R-enantiomer after reboxetine or R,R-enantiomer administration. The ratio of the mean AUC values for the R,R- and S,S-enantiomers was 2.1. CONCLUSION: These findings suggest that the S,S-enantiomer is responsible for the hemodynamic effects of reboxetine in humans. Increases in supine blood pressure after reboxetine administration may be interpreted as regression to the mean value and not caused by any treatment effect.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacology , Blood Pressure/drug effects , Heart Rate/drug effects , Morpholines/pharmacology , Adrenergic Uptake Inhibitors/pharmacokinetics , Adult , Area Under Curve , Double-Blind Method , Electrocardiography/drug effects , Half-Life , Humans , Male , Morpholines/pharmacokinetics , Posture , Reboxetine , Reference Values , Stereoisomerism , Time Factors
9.
J Clin Pharmacol ; 40(5): 482-7, 2000 May.
Article in English | MEDLINE | ID: mdl-10806601

ABSTRACT

Reboxetine is a new selective norepinephrine reuptake inhibitor (selective NRI) for the short- and long-term treatment of depression that is effective and well tolerated at a dose of 8 to 10 mg/day. This study assessed the pharmacokinetics of reboxetine in volunteers with renal impairment. A single 4 mg dose of reboxetine was administered to a total of 18 volunteers with mild (n = 6), moderate (n = 6), or severe (n = 6) renal impairment (creatinine clearance: 56-64, 26-51, and 9-19 ml/min, respectively), and reboxetine concentrations were measured in plasma by HPLC. Mean AUC infinity increased by 43% (mild vs. severe; p < 0.01) as renal function declined, while renal clearance and total urinary excretion of unchanged reboxetine decreased by 67% and 62%, respectively (mild vs. severe; p < 0.01 for both parameters). tmax and t1/2 were not significantly different between groups. In comparison with historical data from young healthy volunteers, AUC infinity and t1/2 are at least doubled in volunteers with renal impairment, while CLr is halved. This pharmacokinetic study has shown that increasing renal dysfunction leads to increasing systemic exposure to reboxetine, particularly in severe renal insufficiency, although reboxetine was well tolerated by all volunteers. Thus, a reduction of the starting dose of reboxetine to 2 mg twice daily would be prudent in patients with renal dysfunction.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacokinetics , Morpholines/pharmacokinetics , Renal Insufficiency/metabolism , Adult , Aged , Analysis of Variance , Area Under Curve , Creatinine/metabolism , Female , Humans , Male , Metabolic Clearance Rate , Middle Aged , Morpholines/blood , Morpholines/urine , Reboxetine
10.
Eur Neuropsychopharmacol ; 7 Suppl 1: S23-35; discussion S71-3, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9169308

ABSTRACT

The pharmacokinetics and metabolism of reboxetine, a selective noradrenaline reuptake inhibitor, in humans and animal models are reviewed here. Reboxetine has potent antidepressant activity, low affinity for alpha-adrenergic and muscarinic receptors and low toxicity in animals. It is a mixture of (R,R) and (S,S) enantiomer, the latter being more potent but no qualitative differences in pharmacodynamic properties are observed between the two. Humans rapidly absorb reboxetine (tmax about 2 h) with a terminal half-life of elimination (t1/2) of 13 h, allowing twice-daily administration. Animal models also rapidly absorb reboxetine (tmax 0.5-2 h) but t1/2 was 1-2 h. Food does not affect bioavailability. There were no major inter-species differences in the metabolic profile of reboxetine. Elimination is principally renal in humans and monkeys. Reboxetine has linear pharmacokinetics in young, healthy males for single doses of 1-5 mg and in elderly, female depressed patients (up to 4 mg b.i.d.). Multiple dosing, gender or liver insufficiency had no significant effects on the pharmacokinetics. Elderly (particularly frail elderly) patients and patients with severe renal impairment may need dose reduction. Reboxetine shows no clinically relevant interaction with lorazepam and has no inhibitory effects on the major enzymes involved in drug metabolism. It may be possible to use reboxetine in combination with monoamine oxidase inhibitors as it has no inhibitory effect on this enzyme; in addition, it may protect patients against tyramine-induced reactions. In conclusion, reboxetine seems to be an antidepressant with negligible interference with the pharmacokinetics of other drugs thus fewer drug-drug interactions are expected.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacokinetics , Antidepressive Agents/pharmacokinetics , Morpholines/pharmacokinetics , Age Factors , Animals , Area Under Curve , Biological Availability , Brain/metabolism , Food-Drug Interactions , Humans , Kidney Diseases/metabolism , Liver/drug effects , Liver/enzymology , Liver Diseases/metabolism , Morpholines/urine , Reboxetine , Stereoisomerism
11.
J Pharm Sci ; 83(10): 1421-4, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7884663

ABSTRACT

The effect of formulation on the urinary pharmacokinetics, pharmacodynamics, and relative bioavailability of cabergoline was investigated. Twelve healthy female volunteers, aged 23-35 years, were treated, according to an open, randomized, crossover design, with cabergoline (1-mg single oral dose) both as tablets and as a solution. The two administrations were separated by a 4-week wash-out period. Cabergoline and prolactin were measured in urine and plasma, respectively, by specific radioimmunoassays. Blood samples were collected before and up to 30 days after dosing. Urine was collected before and up to 8 days after dosing. Cabergoline elimination half-lives calculated from urinary data were 68 and 63 h after administration of the tablets and the solution, respectively. Urinary excretion of unchanged cabergoline accounted, on average, for 1.92% (range, 0.14-3.26) and 1.80% (range, 0.67-3.09) of the dose after administration of the tablets and the aqueous solution, respectively. Relative bioavailability of tablets vs solution was 99% (geometric mean with the 90% confidence intervals of 68-144%). Prolactin levels in 10 out of 12 subjects fell below the detection limit of the assay (1.5 micrograms/L) after both treatments. The mean maximum prolactin decrease (ca. 70%) was achieved by 2 or 3 h after dosing; the effect persisted up to 9 days, being completely exhausted 23-28 days after dosing. The analysis of variance performed on the pharmacodynamic effects of the two cabergoline formulations indicated that the percent decreases of plasma prolactin levels were not significantly different for tablets and solution. These results indicate that the pharmacodynamics and relative bioavailability of cabergoline are not influenced by formulation, as tablets or solution.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Dopamine Agonists/pharmacology , Dopamine Agonists/pharmacokinetics , Ergolines/pharmacology , Ergolines/pharmacokinetics , Adult , Antineoplastic Agents/administration & dosage , Biological Availability , Cabergoline , Chemistry, Pharmaceutical , Cross-Over Studies , Dopamine Agonists/administration & dosage , Ergolines/administration & dosage , Female , Humans , Solutions , Tablets
12.
Mutat Res ; 348(4): 187-92, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8544872

ABSTRACT

The rate of ribosomal gene activity was evaluated by silver staining of the Nucleolus Organisers (NOs) in cultured CHO-K1 cells after a 12 h pulse with two demethylating agents (L-ethionine and 5-azacytidine). Silver staining of the NOs was measured every 24 h, from 24 up to 110 h after seeding. The purpose was to test the hypothesis that drug-induced demethylation is associated to heritable modifications of rDNA activity. Ribosomal gene activity was shown to be significantly increased by both agents. The increase persisted throughout the experiments, thereby suggesting the heritability of this epigenetic modification. The analysis of heritable DNA damage or modification is an important task in studying the risk of cancer onset and the mechanisms of cancer induction. In these studies two main results were obtained: (i) heritable DNA variations can be induced by both mutational and epigenetic changes; (ii) the modified end-point was not negatively selected.


Subject(s)
Antimetabolites/toxicity , DNA, Ribosomal/drug effects , Gene Expression Regulation, Neoplastic , Germ-Line Mutation , Mutagens/toxicity , Animals , Azacitidine/metabolism , Azacitidine/toxicity , CHO Cells , Chromosome Deletion , Clone Cells , Cricetinae , DNA, Ribosomal/metabolism , Ethionine/metabolism , Ethionine/toxicity , Methylation/drug effects , Mitotic Index/drug effects , Mutagens/metabolism , Nucleolus Organizer Region/drug effects , Nucleolus Organizer Region/metabolism , Silver Staining
13.
Mutat Res ; 347(3-4): 129-33, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7565903

ABSTRACT

Background radiation is likely to constitute one of the factors involved in biological evolution since radiations are able to affect biological processes. Therefore, it is possible to hypothesize that organisms are adapted to environmental background radiation and that this adaptation could increase their ability to respond to the harmful effects of ionizing radiations. In fact, adaptive responses to alkylating agents and to low doses of ionizing radiation have been found in many organisms. In order to test for effects of adaptation, cell susceptibility to treatments with high doses of radiomimetic chemical agents has been studied by growing them in a reduced environmental radiation background. The experiment has been performed by culturing yeast cells (Saccharomyces cerevisiae D7) in parallel in a standard background environment and in the underground Gran Sasso National Laboratory, with reduced environmental background radiation. After a conditioning period, yeast cells were exposed to recombinogenic doses of methyl methanesulfonate. The yeast cells grown in the Gran Sasso Laboratory showed a higher frequency of radiomimetic induced recombination as compared to those grown in the standard environment. This suggests that environmental radiation may act as a conditioning agent.


Subject(s)
Background Radiation , Methyl Methanesulfonate/pharmacology , Mutagens/pharmacology , Recombination, Genetic/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/radiation effects , Biological Evolution , Dose-Response Relationship, Radiation , Geography , Geological Phenomena , Geology , Italy , Saccharomyces cerevisiae/growth & development
14.
Int J Clin Pharmacol Ther ; 38(5): 254-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10839469

ABSTRACT

OBJECTIVES: To examine the pharmacokinetic characteristics of the selective norepinephrine reuptake inhibitor, reboxetine, in elderly patients with depression. PATIENTS: Twelve female inpatients (mean age 80 +/- 4 years) with major depressive or dysthymic disorder were enrolled in a 4-week uncontrolled study of oral reboxetine 2-8 mg/day. METHODS: After a one-week washout period, patients were randomized into two groups (groups A and B, n = 6/group). Reboxetine was given twice daily, starting with 2 mg/day during week 1 and increasing by 2 mg/day each week to 8 mg/day in week 4. Pharmacokinetic evaluations were carried out at two dosage levels in each group: at the end of weeks 1 and 3 in group A (2 and 6 mg/day), and at the end of weeks 2 and 4 in group B (4 and 8 mg/day). Blood and urine samples were taken for determination of reboxetine pharmacokinetics. RESULTS: Reboxetine displayed linear pharmacokinetics, with dose-proportional changes, in elderly depressed patients. Mean total urinary recovery ranged from 4.06 to 6.17%. The mean area under the plasma concentration-time curve (AUCtau) and the maximum plasma drug concentration (Cmax) showed considerable variation between patients; at a dosage of 4 mg/day, AUCtau was 1,466-6,866 ngxh/ml and Cmax ranged from 169 to 663 ng/ml. CONCLUSIONS: The pharmacokinetics of reboxetine are linear across the dosage range of 2-8 mg/day in elderly depressed patients, although Cmax and AUCtau values are higher (and more variable) than in young adults. These results support the use of a lower starting dose (4 mg/day) of reboxetine in the elderly.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacokinetics , Depressive Disorder/metabolism , Morpholines/pharmacokinetics , Adrenergic Uptake Inhibitors/administration & dosage , Aged , Aged, 80 and over , Area Under Curve , Dose-Response Relationship, Drug , Female , Humans , Morpholines/administration & dosage , Reboxetine
15.
Therapie ; 48(1): 23-6, 1993.
Article in English | MEDLINE | ID: mdl-8356541

ABSTRACT

The aim of the present study was to evaluate the plasma pharmacokinetics of acipimox and of its N-deoxy metabolite (5-methylpyrazine-2-carboxylic acid, MPCA) following single and repeated administration of 250 mg acipimox (thrice daily, for 6 days) to ten healthy volunteers. Mean maximum concentration, the corresponding time, area under the curve extrapolated to infinity and elimination half-life values of acipimox after single administration were equal to 5.74 micrograms/ml (range 2.56-8.38 micrograms/ml), 1.7 h (1-3 h), 16.99 micrograms/ml.h (11.28-22.17 micrograms/ml.h) and 1.15 h (0.79-1.48 h), respectively. Mean area under the curve over one dosing interval (8 h) and elimination half-life values of acipimox after repeated dosing were not significantly different from the corresponding values after the single dose. No significant accumulation was observed following the repeated treatment, since the mean accumulation ratio was 1.08 (range 0.74-1.52). The mean maximum concentration and corresponding time values in the 7 out of 10 subjects with detectable metabolite levels after the single dose were 0.19 microgram/ml (0.10-0.34 microgram/ml) and 6.7 h (3-12 h), respectively, whilst after the repeated treatment, detectable concentrations of the metabolite were observed in all subjects, the mean maximum concentration value being equal to 0.48 micrograms/ml (0.11-1.19 microgram/ml). The average ratio of the parent/metabolite area under the curve values (8 h) after repeated dosing was equal to 14 (range 2-56). Inter-subject variability in the extent of metabolite formation was very high.


Subject(s)
Hypolipidemic Agents/pharmacokinetics , Pyrazines/pharmacokinetics , Administration, Oral , Adult , Drug Administration Schedule , Drug Evaluation , Female , Humans , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/blood , Male , Pyrazines/administration & dosage , Pyrazines/blood , Volunteers
16.
Clin Pharmacol Ther ; 91(5): 863-71, 2012 May.
Article in English | MEDLINE | ID: mdl-22472989

ABSTRACT

Many difficulties may arise during the modeling of the time course of Hamilton Rating Scale for Depression (HAM D)scores in clinical trials for the evaluation of antidepressant drugs: (i) flexible designs, used to increase the chance of selecting more efficacious doses, (ii) dropout events, and (iii) adverse effects related to the experimental compound.It is crucial to take into account all these factors when designing an appropriate model of the HAM D time course and to obtain a realistic description of the dropout process. In this work, we propose an integrated approach to the modeling of a double-blind, flexible-dose, placebo-controlled, phase II depression trial that comprises response,tolerability, and dropout. We investigate three different dropout mechanisms in terms of informativeness. Goodness of fit is quantitatively assessed with respect to response (HAM D score) and dropout data. We show that dropout is a complex phenomenon that may be influenced by HAM D evolution, dose changes, and occurrence of drug-related adverse effects.


Subject(s)
Antidepressive Agents/administration & dosage , Depression/drug therapy , Patient Dropouts , Antidepressive Agents/adverse effects , Double-Blind Method , Humans , Psychiatric Status Rating Scales , Research Design
18.
Eur J Cancer ; 45(18): 3336-46, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19854637

ABSTRACT

In clinical oncology, combination regimens may result in a synergistic, additive or antagonistic interaction (i.e. the effect of the combination is greater, similar or smaller than the sum of the effects of the individual compounds). For this reason, during the drug development process, in vivo pre-clinical studies are performed to assess the interaction of anticancer agents given in combination. Starting from a widely used single compound PK/PD model, a new additivity model able to predict the tumour growth inhibition in xenografted mice after the administration of compounds in combination was developed, under the assumption of a pharmacodynamic null interaction. By comparing the predicted curves with actual tumour weight data, possible departures from additivity can be immediately ascertained by visual inspection; a statistical procedure based on a chi(2) test has also been developed for this aim. The advantages of the proposed approach in comparison to other modelling methodologies are discussed and its application to four combination studies is presented.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neoplasms/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Drug Interactions , Mice , Models, Biological , Neoplasm Transplantation , Neoplasms/drug therapy , Xenograft Model Antitumor Assays
19.
Breast Cancer Res Treat ; 111(2): 377-88, 2008 Sep.
Article in English | MEDLINE | ID: mdl-17952589

ABSTRACT

PURPOSE: Raloxifene is a second-generation selective estrogen receptor modulator that reduces the incidence of breast cancer in postmenopausal women. Exemestane, a steroidal aromatase inhibitor, decreases contralateral new breast cancers in postmenopausal women when taken in the adjuvant setting. Preclinical evidence suggests a rationale for coadministration of these agents to achieve complete estrogen blockade. EXPERIMENTAL DESIGN: We tested the safety and tolerability of combination exemestane and raloxifene in 11 postmenopausal women with a history of hormone receptor-negative breast cancer. Patients were randomized to either raloxifene (60 mg PO daily) or exemestane (25 mg PO daily) for 2 weeks. Patients then initiated combination therapy at the same dose levels for a minimum of 1 year. Pharmacokinetic and pharmacodynamic data for plasma estrogens, raloxifene, exemestane, and their metabolites were collected at the end of single-agent therapy and during combination therapy. RESULTS: Plasma concentration-time profiles for each drug were unchanged with monotherapy versus combination therapy. Raloxifene did not affect plasma estrogen levels. Plasma estrogen concentrations were suppressed below the lower limit of detection by exemestane as monotherapy and when administered in combination with raloxifene. The most common adverse events of any grade included arthralgias, hot flashes, vaginal dryness and myalgias. CONCLUSIONS: In this small study, coadministration of raloxifene and exemestane did not affect the pharmacokinetics or pharmacodynamics of either agent to a significant degree in postmenopausal women. The combination of estrogen receptor blockade and suppression of estrogen synthesis is well tolerated and warrants further investigation.


Subject(s)
Androstadienes/pharmacokinetics , Aromatase Inhibitors/pharmacokinetics , Breast Neoplasms/metabolism , Raloxifene Hydrochloride/pharmacokinetics , Selective Estrogen Receptor Modulators/pharmacokinetics , Aged , Androstadienes/adverse effects , Drug Therapy, Combination , Estradiol/blood , Estrone/blood , Female , Humans , Middle Aged , Postmenopause , Raloxifene Hydrochloride/adverse effects
20.
Biopharm Drug Dispos ; 20(1): 53-7, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10086838

ABSTRACT

The absolute bioavailability of reboxetine enantiomers was assessed in six male and six female volunteers. In a two-way crossover study, subjects received 1.0 mg reboxetine orally and 0.3 mg reboxetine as an intravenous bolus. The R,R(-) and S,S(+) enantiomers in serial plasma and urine samples were determined by a validated LC-MS-MS method. There were no significant differences between treatments for clearance or dose-corrected AUC(0-infinity) values. The absolute bioavailability was 0.919 and 1.02 for R,R(-) reboxetine and S,S(+) reboxetine, respectively. A secondary objective of the study was to assess gender effects on pharmacokinetics of the enantiomers. Significant differences in volume of distribution between genders were observed, but differences in weight-corrected volumes were not significant. Weight-corrected systemic clearance and oral clearance tended to be lower in males, but this difference reached statistical significance only for weight-corrected oral clearance of R,R(-) reboxetine. C(max) after oral administration was 40 and 48% higher in women than men for R,R(-) reboxetine and S,S(+) reboxetine, respectively. These results indicate that reboxetine enantiomers are well absorbed after oral administration and that little first-pass metabolism occurs. There are no clinically significant effects of gender on the pharmacokinetics of reboxetine enantiomers.


Subject(s)
Antidepressive Agents/pharmacokinetics , Morpholines/pharmacokinetics , Administration, Oral , Adrenergic Uptake Inhibitors/administration & dosage , Adrenergic Uptake Inhibitors/blood , Adrenergic Uptake Inhibitors/pharmacokinetics , Adult , Antidepressive Agents/administration & dosage , Antidepressive Agents/blood , Area Under Curve , Biological Availability , Cross-Over Studies , Electrocardiography , Female , Half-Life , Humans , Injections, Intravenous , Male , Metabolic Clearance Rate , Morpholines/administration & dosage , Morpholines/blood , Reboxetine , Regression Analysis , Sex Characteristics , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL