Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Clin Pharmacol ; 63(6): 721-731, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36854991

RESUMEN

Golimumab was recently evaluated in a phase 2a, randomized, double-blind, placebo-controlled, multicenter study (T1GER study) for safety and efficacy in children and young adults with newly diagnosed type 1 diabetes (T1D). Golimumab showed significant treatment effect where endogenous insulin production was preserved and clinical and metabolic parameters were improved. The objective of this report was to evaluate pharmacokinetic (PK) and pharmacodynamic data from the T1GER study by developing a population pharmacokinetic (PopPK) model and performing exposure-response (ER) analyses. The PopPK model was developed using data from the T1D study and 2 other pediatric studies with golimumab in ulcerative colitis and in polyarticular juvenile idiopathic arthritis. A 1-compartment model with first-order absorption and elimination was applied to describe the concentration-time profiles. Typical parameters normalized to the values in subjects with a standard weight of 70 kg were apparent clearance, 0.850 L/day; apparent volume of distribution, 16.0 L; and absorption rate constant, 1.01/day. From the ER analyses, no clear trends were observed for changes in both C-peptide area under the concentration-time curve and hemoglobin A1c levels for the relatively narrow exposure ranges following the body surface area-based dosing regimen used in this study. In conclusion, the developed PopPK model was able to adequately describe the observed PK of golimumab in patients with T1D. Although golimumab showed significant treatment effect, the ER analyses did not show clear trends within the active treatment group, which may indicate that the exposure from this T1D-specific dosing regimen was at the plateau of the ER curve.


Asunto(s)
Artritis Juvenil , Diabetes Mellitus Tipo 1 , Humanos , Niño , Adulto Joven , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Anticuerpos Monoclonales/farmacocinética , Artritis Juvenil/tratamiento farmacológico , Insulina
2.
Clin Transl Sci ; 15(12): 3000-3011, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36204820

RESUMEN

The aims of this work were to develop a population pharmacokinetic (PK) model for chimeric antigen receptor (CAR) transgene after single intravenous infusion administration of ciltacabtagene autoleucel in adult patients with relapsed or refractory multiple myeloma. CAR transgene level in blood were measured by quantitative polymerase chain reaction (qPCR) from 97 subjects in a phase Ib/II CARTITUDE-1 study (NCT03548207), with a targeted cilta-cel dose of 0.75 × 106 (range 0.5-1.0 × 106 ) CAR positive viable T-cells per kg body weight. The population PK model development was primarily guided by the current mechanistic understanding of CAR-T kinetics and the principles of building a parsimonious model. Cilta-cel PK was adequately described by a two-compartment model (with a fast and a slow apparent decline rate from each compartment, respectively) and a chain of four transit compartments with a lag time empirically representing the process from infused CAR-T cell to measurable CAR transgene. No apparent relationship was observed between cilta-cel dose (i.e., the actual number of CAR positive viable T-cells infused), given the narrow dose range, and the observed transgene level. Based on covariate search and subgroup analysis of maximum systemic CAR transgene level (Cmax ) and area under curve from the first dose to day 28 (AUC0-28d ), none of the investigated subjects' demographics, baseline characteristics, and manufactured product characteristics had significant effects on cilta-cel PK. The developed model is deemed robust and adequate for enabling subsequent exposure-safety and exposure-efficacy analyses.


Asunto(s)
Mieloma Múltiple , Receptores Quiméricos de Antígenos , Adulto , Humanos , Mieloma Múltiple/tratamiento farmacológico , Inmunoterapia Adoptiva/efectos adversos , Receptores Quiméricos de Antígenos/genética , Linfocitos T
3.
CPT Pharmacometrics Syst Pharmacol ; 11(8): 1111-1121, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35665486

RESUMEN

Rivaroxaban is approved in various regions for the treatment of acute venous thromboembolism (VTE) in children aged between 0 and 18 years and was recently investigated for thromboprophylaxis in children aged between 2 and 8 years (with body weights <30 kg) with congenital heart disease who had undergone the Fontan procedure. In the absence of clinical data, rivaroxaban doses for thromboprophylaxis in post-Fontan children aged 9 years and older or ≥30 kg were derived by a bridging approach that used physiologically-based pharmacokinetic (PBPK) and population pharmacokinetic (popPK) models based on pharmacokinetic (PK) data from 588 pediatric patients and from adult patients who received 10 mg once daily for thromboprophylaxis after major orthopedic surgeries as a reference. Both models showed a tendency toward underestimating rivaroxaban exposure in post-Fontan patients aged between 2 and 5 years but accurately described rivaroxaban PK in post-Fontan patients aged between 5 and 8 years. Under the assumption that hepatic function is not impaired in post-Fontan patients, PBPK and popPK simulations indicated that half of the rivaroxaban doses for the same body weight given to pediatric patients treated for acute VTE would yield in pediatric post-Fontan patients exposures similar to the exposure observed in adult patients receiving 10 mg rivaroxaban once daily for thromboprophylaxis. Simulation-derived doses (7.5 mg rivaroxaban once daily for body weights 30-<50 kg and 10 mg once daily for body weights ≥50 kg) were therefore included in the recent US label of rivaroxaban for thromboprophylaxis in children aged 2 years and older with congenital heart disease who have undergone the Fontan procedure.


Asunto(s)
Cardiopatías Congénitas , Tromboembolia Venosa , Adolescente , Adulto , Anticoagulantes , Peso Corporal , Niño , Preescolar , Cardiopatías Congénitas/inducido químicamente , Cardiopatías Congénitas/tratamiento farmacológico , Cardiopatías Congénitas/cirugía , Humanos , Lactante , Recién Nacido , Rivaroxabán , Tromboembolia Venosa/etiología , Tromboembolia Venosa/prevención & control
4.
J Clin Pharmacol ; 62(2): 220-231, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34524700

RESUMEN

Thrombosis remains an important complication for children with single-ventricle physiology following the Fontan procedure, and effective thromboprophylaxis is an important unmet medical need. To obviate conventional dose-finding studies and expedite clinical development, a rivaroxaban dose regimen for this indication was determined using a model-informed drug development approach. A physiologically based pharmacokinetic rivaroxaban model was used to predict a pediatric dosing regimen that would produce drug exposures similar to that of 10 mg once daily in adults. This regimen was used in an open-label, multicenter phase III study, which investigated the use of rivaroxaban for thromboprophylaxis in post-Fontan patients 2 to 8 years of age. The pharmacokinetics (PK) of rivaroxaban was assessed in part A (n = 12) and in part B (n = 64) of the UNIVERSE study. The safety and efficacy in the rivaroxaban group were compared to those in the acetylsalicylic acid group for 12 months. Pharmacodynamic end points were assessed in both parts of the study. Rivaroxaban exposures achieved in parts A and B were similar to the adult reference exposures. Prothrombin time also showed similarity to the adult reference. Exposure-response analysis did not identify a quantitative relationship between rivaroxaban exposures and efficacy/safety outcomes within the observed exposure ranges. A body weight-based dose regimen selected by physiologically based pharmacokinetic modeling was shown in the UNIVERSE study to be appropriate for thromboprophylaxis in the post-Fontan pediatric population. Model-based dose selection can support pediatric drug development and bridge adult dose data to pediatrics, thereby obviating the need for dose-finding studies in pediatric programs.


Asunto(s)
Anticoagulantes/administración & dosificación , Anticoagulantes/farmacología , Rivaroxabán/administración & dosificación , Rivaroxabán/farmacología , Trombosis/prevención & control , Anticoagulantes/farmacocinética , Área Bajo la Curva , Pesos y Medidas Corporales , Niño , Preescolar , Femenino , Procedimiento de Fontan/métodos , Humanos , Masculino , Modelos Biológicos , Tiempo de Tromboplastina Parcial , Estudios Prospectivos , Tiempo de Protrombina , Rivaroxabán/farmacocinética
5.
Clin Transl Sci ; 15(2): 464-476, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34581012

RESUMEN

It is known that interleukin-6 (IL-6) can significantly modulate some key drug-metabolizing enzymes, such as phase I cytochrome P450s (CYPs). In this study, a physiologically-based pharmacokinetic (PBPK) model was developed to assess CYPs mediated therapeutic protein drug interactions (TP-DIs) in patients with immune-mediated inflammatory diseases (IMIDs) with elevated systemic IL-6 levels when treated by anti-IL-6 therapies. Literature data of IL-6 levels in various diseases were incorporated in SimCYP to construct respective virtual patient populations. The modulation effects of systemic IL-6 level and local IL-6 level in the gastrointestinal tract (GI) on CYPs activities were assessed. Upon blockade of the IL-6 signaling pathway by an anti-IL-6 treatment, the area under plasma concentration versus time curves (AUCs) of S-warfarin, omeprazole, and midazolam were predicted to decrease by up to 40%, 42%, and 46%, respectively. In patients with Crohn's disease and ulcerative colitis treated with an anti-IL-6 therapy, the lowering of the elevated IL-6 levels in the local GI tissue were predicted to result in further decreases in AUCs of those CYP substrates. The propensity of TP-DIs under comorbidity conditions, such as in patients with cancer with IMID, were also explored. With further validation with relevant clinical data, this PBPK model may provide an in silico way to quantify the magnitude of potential TP-DI in patients with elevated IL-6 levels when an anti-IL-6 therapeutic is used with concomitant small-molecule drugs. This model may be further adapted to evaluate the CYP modulation effect by other therapeutic modalities, which would significantly alter levels of proinflammatory cytokines during the treatment period.


Asunto(s)
Sistema Enzimático del Citocromo P-450 , Midazolam , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Humanos , Midazolam/farmacocinética , Modelos Biológicos , Omeprazol , Warfarina
6.
Clin Pharmacol Ther ; 109(1): 131-139, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32865226

RESUMEN

Ustekinumab (STELARA) is a human monoclonal antibody against interleukins-12 and -23 for the treatment of adult and adolescent (≥ 12 to < 18 years of age) patients with moderate-to-severe plaque psoriasis. A phase III study was recently completed in pediatric patients (≥ 6 to < 12 years of age) with psoriasis. The objectives of the current analysis were to develop a population pharmacokinetic (PK) model and a joint longitudinal exposure-response model using ordered categorial end points derived from Psoriasis Area and Severity Index (PASI) and Physician's Global Assessment (PGA) scores (namely a joint PASI response criteria (PRC) and PGA model) to characterize the PK and exposure-response relationship of ustekinumab in pediatric patients with psoriasis. The developed pediatric models reasonably predicted the PK of ustekinumab, as well as the PRC and PGA clinical response in pediatric patients with psoriasis. In addition, the joint PRC and PGA modeling framework was able to adequately extrapolate clinical response in pediatric patients using data collected from clinical studies in adult patients with psoriasis.


Asunto(s)
Psoriasis/tratamiento farmacológico , Ustekinumab/uso terapéutico , Adolescente , Adulto , Anciano , Anticuerpos Monoclonales/uso terapéutico , Niño , Esquema de Medicación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pediatría/métodos , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Adulto Joven
8.
J Clin Pharmacol ; 59(5): 668-676, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30536675

RESUMEN

Dacogen, the formulated product of the pharmaceutically active agent decitabine (5 aza-2'-deoxycytidine), is approved for treatment of myelodysplastic syndromes (MDSs) and acute myeloid leukemia (AML). The current analysis was performed to characterize the pharmacokinetics of decitabine in pediatric patients with AML and evaluate their consistency with the PK in adult patients. A population pharmacokinetic model was developed by pooling decitabine concentration-time data from 5 adult (AML and MDS) and 2 pediatric (AML) studies. A total of 840 concentration-time data points obtained from 71 adults and 28 pediatric subjects (1 to 16 years old) were available for analysis. A 2-compartment linear pharmacokinetic (PK) model with allometric scaling using body surface area accounting for body size adequately described the PK of decitabine. After accounting for body size, decitabine pharmacokinetics were not affected by age, sex, race, dosing regimen, renal function (creatinine clearance), bilirubin, or disease type (AML or MDS) and all PK parameters (including clearance, steady-state volume of distribution, maximum concentration, time to reach maximal concentration, and terminal half-life) were comparable between adult and pediatric patients. Simulated concentration-time profiles using the final population PK model suggested that decitabine exposure at steady state was similar in adults and pediatrics for a 20 mg/m2 decitabine dose administered as a 1-hour infusion once daily. The current analysis suggests that decitabine PK is similar in pediatric AML patients and a combined adult AML and MDS population.


Asunto(s)
Decitabina/farmacocinética , Leucemia Mieloide Aguda/metabolismo , Adolescente , Adulto , Niño , Preescolar , Simulación por Computador , Esquema de Medicación , Femenino , Humanos , Lactante , Leucemia Mieloide Aguda/tratamiento farmacológico , Masculino , Modelos Biológicos
9.
J Clin Pharmacol ; 58(4): 485-493, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29193123

RESUMEN

Gefitinib is a selective inhibitor of epidermal growth factor receptor (EGFR) tyrosine kinase and is used for the treatment of non-small-cell lung cancer (NSCLC) with activating EGFR mutations. Gefitinib is metabolized by CYP2D6 and CYP3A4. This analysis compared the systemic exposure of gefitinib after oral administration in CYP2D6 ultrarapid metabolizers (UM) vs extensive metabolizers (EM). Physiologically based pharmacokinetic (PBPK) modeling was conducted using a population-based simulator. The model was calibrated using itraconazole-gefitinib clinical drug-drug interaction data and validated with gefitinib data in CYP2D6 EM vs poor metabolizers (PM). System components of the PBPK model were evaluated using published clinical metoprolol pharmacokinetic data in CYP2D6 UM, EM, and PM. Our PBPK model predicted a gefitinib geometric least-squares mean area under the plasma concentration-time curve (AUC) from time 0 to 264 hours (AUC(0-264) ) ratio in the presence vs absence of itraconazole of 1.85, similar to the ratio of 1.78 from clinical study data. Predicted and observed metoprolol geometric least-squares mean AUC(0-24) ratios in UM vs EM were also similar (0.46 and 0.55, respectively), suggesting that system components related to CYP2D6 in the PBPK model were properly established. In addition, the PBPK model also captured gefitinib pharmacokinetic profiles in CYP2D6 polymorphic populations. The final PBPK model predicted a decrease in gefitinib AUC of ∼39% in CYP2D6 UM vs EM. Such changes in exposure will have limited impact as the reduced exposure is still above gefitinib's in vitro IC90 for EGFR activating mutations in NSCLC patients.


Asunto(s)
Antineoplásicos/farmacocinética , Citocromo P-450 CYP2D6/metabolismo , Gefitinib/farmacocinética , Modelos Biológicos , Inhibidores de Proteínas Quinasas/farmacocinética , Adolescente , Adulto , Anciano , Antineoplásicos/sangre , Estudios Cruzados , Citocromo P-450 CYP2D6/genética , Interacciones Farmacológicas , Femenino , Gefitinib/sangre , Genotipo , Humanos , Itraconazol/farmacología , Masculino , Metoprolol/farmacología , Persona de Mediana Edad , Inhibidores de Proteínas Quinasas/sangre , Adulto Joven
10.
Clin Pharmacol Ther ; 104(1): 188-200, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29027194

RESUMEN

The accuracy of physiologically based pharmacokinetic (PBPK) model prediction in children, especially those younger than 2 years old, has not been systematically evaluated. The aim of this study was to characterize the pediatric predictive performance of the PBPK approach for 10 drugs extensively metabolized by CYP1A2 (theophylline), CYP2C8 (desloratidine, montelukast), CYP2C9 (diclofenac), CYP2C19 (esomeprazole, lansoprazole), CYP2D6 (tramadol), and CYP3A4 (itraconazole, ondansetron, sufentanil). Model performance in children was evaluated by comparing simulated plasma concentration-time profiles with observed clinical results for each drug and age group. PBPK models reasonably predicted the pharmacokinetics of desloratadine, diclofenac, itraconazole, lansoprazole, montelukast, ondansetron, sufentanil, theophylline, and tramadol across all age groups. Collectively, 58 out of 67 predictions were within 2-fold and 43 out of 67 predictions within 1.5-fold of observed values. Developed PBPK models can reasonably predict exposure in children age 1 month and older for an array of predominantly CYP metabolized drugs.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Modelos Biológicos , Preparaciones Farmacéuticas/metabolismo , Farmacocinética , Acetatos/metabolismo , Acetatos/farmacocinética , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacocinética , Antiasmáticos/metabolismo , Antiasmáticos/farmacocinética , Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/farmacocinética , Antifúngicos/metabolismo , Antifúngicos/farmacocinética , Broncodilatadores/metabolismo , Broncodilatadores/farmacocinética , Niño , Preescolar , Ciclopropanos , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP2C19/metabolismo , Citocromo P-450 CYP2C8/metabolismo , Citocromo P-450 CYP2C9/metabolismo , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP3A/metabolismo , Diclofenaco/metabolismo , Diclofenaco/farmacocinética , Esomeprazol/metabolismo , Esomeprazol/farmacocinética , Antagonistas de los Receptores Histamínicos H1 no Sedantes/metabolismo , Antagonistas de los Receptores Histamínicos H1 no Sedantes/farmacocinética , Humanos , Lactante , Recién Nacido , Itraconazol/metabolismo , Itraconazol/farmacocinética , Lansoprazol/metabolismo , Lansoprazol/farmacocinética , Loratadina/análogos & derivados , Loratadina/metabolismo , Loratadina/farmacocinética , Ondansetrón/metabolismo , Ondansetrón/farmacocinética , Inhibidores de la Bomba de Protones/metabolismo , Inhibidores de la Bomba de Protones/farmacocinética , Quinolinas/metabolismo , Quinolinas/farmacocinética , Antagonistas de la Serotonina/metabolismo , Antagonistas de la Serotonina/farmacocinética , Sufentanilo/metabolismo , Sufentanilo/farmacocinética , Sulfuros , Teofilina/metabolismo , Teofilina/farmacocinética
11.
J Clin Pharmacol ; 57(10): 1258-1267, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28581633

RESUMEN

Zolmitriptan is a serotonin (5-HT) 1B/1D receptor agonist effective for the treatment of migraine. This analysis aimed to develop a population pharmacokinetic (PK) model for zolmitriptan and its active metabolite in adults and adolescents and provide appropriate dosing regimens to be used in clinical trials for children 6-11 years old. The data from a single-dose clinical study of 5.0-mg zolmitriptan nasal spray (ZNS) conducted in adult and adolescent patients with migraine between migraine attacks was applied. Similar plasma concentration profiles of zolmitriptan and its metabolite, 183C91, were observed in adults and adolescents. A 1-compartment model with first-order absorption and first-order elimination reasonably described the zolmitriptan PK. With a portion of elimination of zolmitriptan being treated as the conversion from zolmitriptan to 183C91, the disposition of 183C91 was described by a 1-compartment model with first-order elimination. The estimated typical apparent volume of distribution and clearance of zolmitriptan were 136 L and 121 L/h, respectively, with 56.5% and 42.6% between-subject variability, respectively. Based on the simulation results with the final population PK model, a body weight-based dosing scheme of 5.0 and 2.5 mg ZNS in children greater than and less than 50 kg is recommended to achieve exposures similar to those of the adult and adolescent population administered 5.0 mg ZNS. The recommended doses for children to achieve exposure similar to that observed in adults given 2.5 mg ZNS are 2.5 mg (≥50 kg) and 1.0 mg (15-50 kg). These dosing regimens could be used in future clinical trials.


Asunto(s)
Modelos Biológicos , Oxazolidinonas/administración & dosificación , Oxazolidinonas/farmacocinética , Agonistas del Receptor de Serotonina 5-HT1/administración & dosificación , Agonistas del Receptor de Serotonina 5-HT1/farmacocinética , Triptaminas/administración & dosificación , Triptaminas/farmacocinética , Administración Intranasal , Adolescente , Adulto , Peso Corporal , Niño , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Persona de Mediana Edad , Trastornos Migrañosos/sangre , Trastornos Migrañosos/tratamiento farmacológico , Trastornos Migrañosos/metabolismo , Oxazoles/sangre , Oxazolidinonas/sangre , Oxazolidinonas/uso terapéutico , Agonistas del Receptor de Serotonina 5-HT1/sangre , Agonistas del Receptor de Serotonina 5-HT1/uso terapéutico , Triptaminas/sangre , Triptaminas/uso terapéutico , Adulto Joven
12.
Biopharm Drug Dispos ; 38(6): 389-393, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28255999

RESUMEN

Itopride, a substrate of FMO3, has been used for the symptomatic treatment of various gastrointestinal disorders. Physiologically based pharmacokinetic (PBPK) modeling was applied to evaluate the impact of FMO3 polymorphism on itopride pharmacokinetics (PK). The Asian populations within the Simcyp simulator were updated to incorporate information on the frequency, activity and abundance of FMO3 enzyme with different phenotypes. A meta-analysis of relative enzyme activities suggested that FMO3 activity in subjects with homozygous Glu158Lys and Glu308Gly mutations (Lys158 and Gly308) in both alleles is ~47% lower than those carrying two wild-type FMO3 alleles. Individuals with homozygous Lys158 and Gly308 mutations account for about 5% of the total population in Asian populations. A CLint of 9 µl/min/pmol was optimised for itopride via a retrograde approach as human liver microsomal results would under-predict its clearance by ~7.9-fold. The developed itopride PBPK model was first verified with three additional clinical studies in Korean and Japanese subjects resulting in a predicted clearance of 52 to 69 l/h, which was comparable to those observed (55 to 88 l/h). The model was then applied to predict plasma concentration-time profiles of itopride in Chinese subjects with wild type or homozygous Lys158 and Gly308 FMO3 genotypes. The ratios of predicted to observed AUC of itopride in subjects with each genotype were 1.23 and 0.94, respectively. In addition, the results also suggested that for FMO3 metabolised drugs with a safety margin of 2 or more, proactive genotyping FMO3 to exclude subjects with homozygous Lys158/Gly308 alleles may not be necessary.


Asunto(s)
Benzamidas/farmacocinética , Compuestos de Bencilo/farmacocinética , Modelos Biológicos , Oxigenasas/genética , Oxigenasas/metabolismo , Adulto , Pueblo Asiatico/genética , Benzamidas/sangre , Compuestos de Bencilo/sangre , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo Genético , Adulto Joven
13.
J Clin Pharmacol ; 57(3): 336-344, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27530649

RESUMEN

Aztreonam is a monocyclic ß-lactam antibiotic often used to treat infections caused by Enterobacteriaceae or Pseudomonas aeruginosa. Despite the long history of clinical use, population pharmacokinetic modeling of aztreonam in renally impaired patients is not yet available. The aims of this study were to assess the impact of renal impairment on aztreonam exposure and to evaluate dosing regimens for patients with renal impairment. A population model describing aztreonam pharmacokinetics following intravenous administration was developed using plasma concentrations from 42 healthy volunteers and renally impaired patients from 2 clinical studies. The final pharmacokinetic model was used to predict aztreonam plasma concentrations and evaluate the probability of pharmacodynamic target attainment (PTA) in patients with different levels of renal function. A 2-compartment model with first-order elimination adequately described aztreonam pharmacokinetics. The population mean estimates of aztreonam clearance, intercompartmental clearance, volume of distribution of the central compartment, and volume of distribution of the peripheral compartment were 4.93 L/h, 9.26 L/h, 7.43 L, and 6.44 L, respectively. Creatinine clearance and body weight were the most significant variables to explain patient variability in aztreonam clearance and volume of distribution, respectively. Simulations using the final pharmacokinetic model resulted in a clinical susceptibility break point of 4 and 8 mg/L, respectively, based on the clinical use of 1- and 2-g loading doses with the same or reduced maintenance dose every 8 hours for various renal deficiency patients. The population pharmacokinetic modeling and PTA estimation support adequate PTAs (>90% PTA) from the aztreonam label for dose adjustment of aztreonam in patients with moderate and severe renal impairment.


Asunto(s)
Antibacterianos/farmacocinética , Aztreonam/farmacocinética , Método de Montecarlo , Insuficiencia Renal/metabolismo , Adulto , Factores de Edad , Antibacterianos/administración & dosificación , Aztreonam/administración & dosificación , Estatura , Peso Corporal , Ensayos Clínicos como Asunto , Creatinina/sangre , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Modelos Biológicos
14.
J Med Chem ; 57(6): 2643-56, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24568657

RESUMEN

Species of Candida, primarily C. albicans and with increasing prevalence, C. glabrata, are responsible for the majority of fungal bloodstream infections that cause morbidity, especially among immune compromised patients. While the development of new antifungal agents that target the essential enzyme, dihydrofolate reductase (DHFR), in both Candida species would be ideal, previous attempts have resulted in antifolates that exhibit inconsistencies between enzyme inhibition and antifungal properties. In this article, we describe the evaluation of pairs of propargyl-linked antifolates that possess similar physicochemical properties but different shapes. All of these compounds are effective at inhibiting the fungal enzymes and the growth of C. glabrata; however, the inhibition of the growth of C. albicans is shape-dependent with extended para-linked compounds proving more effective than compact, meta-linked compounds. Using crystal structures of DHFR from C. albicans and C. glabrata bound to lead compounds, 13 new para-linked compounds designed to inhibit both species were synthesized. Eight of these compounds potently inhibit the growth of both fungal species with three compounds displaying dual MIC values less than 1 µg/mL. Analysis of the active compounds shows that shape and distribution of polar functionality is critical in achieving dual antifungal activity.


Asunto(s)
Antifúngicos , Candida albicans/efectos de los fármacos , Candida glabrata/efectos de los fármacos , Antagonistas del Ácido Fólico/síntesis química , Antagonistas del Ácido Fólico/farmacología , Morfinanos/síntesis química , Morfinanos/farmacología , Cromatografía Líquida de Alta Presión , Cristalización , Cristalografía por Rayos X , Antagonistas del Ácido Fólico/química , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Conformación Molecular , Morfinanos/química , NADP/química , Solubilidad , Relación Estructura-Actividad , Tetrahidrofolato Deshidrogenasa/efectos de los fármacos , Difracción de Rayos X
15.
ACS Med Chem Lett ; 4(8): 757-61, 2013 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-24900743

RESUMEN

Natural products have long been recognized as a rich source of potent therapeutics but further development is often limited by high structural complexity and high molecular weight. In contrast, at the core of the thujaplicins is a lead-like tropolone scaffold characterized by relatively low molecular weight, ample sites for diversification, and metal-binding functionality poised for targeting a range of metalloenzyme drug targets. Here, we describe the development of this underutilized scaffold for the discovery of tropolone derivatives that function as isozyme-selective inhibitors of the validated anticancer drug target, histone deacetylase (HDAC). Several monosubstituted tropolones display remarkable levels of selectivity for HDAC2 and potently inhibit the growth of T-cell lymphocyte cell lines. The tropolones represent a new chemotype of isozyme-selective HDAC inhibitors.

16.
Drug Metab Dispos ; 40(10): 2002-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22815313

RESUMEN

Propargyl-linked antifolates that target dihydrofolate reductase are potent inhibitors of several species of pathogenic bacteria and fungi. This novel class of antifolates possesses a relatively uncommon acetylenic linker designed to span a narrow passage in the enzyme active site and join two larger functional domains. Because the use of alkyne functionality in drug molecules is limited, it was important to evaluate some key physicochemical properties of these molecules and specifically to assess the overall stability of the acetylene. Herein, we report studies on four compounds from our lead series that vary specifically in the environment of the alkyne. We show that the compounds are soluble, chemically stable in water, as well as simulated gastric and intestinal fluids with half-lives of approximately 30 min after incubation with mouse liver microsomes. Their primary in vitro route of metabolism involves oxidative transformations of pendant functionality with little direct alteration of the alkyne. Identification of several major metabolites indicated the formation of N-oxides; the rate of formation of these oxides was highly influenced by branching substitutions around the propargyl linker. On the basis of the lessons of these metabolic studies, a more advanced inhibitor was designed, synthesized, and shown to have increased (t(1/2) = 65 min) metabolic stability while maintaining potent enzyme inhibition.


Asunto(s)
Alquinos/metabolismo , Diseño de Fármacos , Antagonistas del Ácido Fólico/metabolismo , Microsomas Hepáticos/metabolismo , Alquinos/química , Alquinos/farmacología , Animales , Biotransformación , Estabilidad de Medicamentos , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/farmacología , Semivida , Cinética , Ratones , Modelos Moleculares , Estructura Molecular , Oxidación-Reducción , Solubilidad , Relación Estructura-Actividad , Tetrahidrofolato Deshidrogenasa/metabolismo
17.
Mol Imaging Biol ; 13(3): 416-423, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20574851

RESUMEN

PURPOSE: The purpose of this study was to demonstrate a novel protein-based magnetic resonance imaging (MRI) contrast agent that has the capability of targeting prostate cancer and which provides high-sensitivity MR imaging in tumor cells and mouse models. PROCEDURE: A fragment of gastrin-releasing peptide (GRP) was fused into a protein-based MRI contrast agent (ProCA1) at different regions. MR imaging was obtained in both tumor cells (PC3 and H441) and a tumor mouse model administrated with ProCA1.GRP. RESULTS: PC3 and DU145 cells treated with ProCA1.GRPs exhibited enhanced signal in MRI. Intratumoral injection of ProCA1.GRP in a PC3 tumor model displayed enhanced MRI signal. The contrast agent was retained in the PC3 tumor up to 48 h post-injection. CONCLUSIONS: Protein-based MRI contrast agent with tumor targeting modality can specifically target GRPR-positive prostate cancer. Intratumoral injection of the ProCA1 agent in the prostate cancer mouse model verified the targeting capability of ProCA1.GRP and showed a prolonged retention time in tumors.


Asunto(s)
Proteínas Portadoras , Medios de Contraste , Imagen por Resonancia Magnética/métodos , Imagen Molecular/métodos , Neoplasias de la Próstata/diagnóstico , Animales , Medios de Contraste/química , Péptido Liberador de Gastrina , Masculino , Ratones , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...