Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 809
Filtrar
1.
Eur J Clin Pharmacol ; 80(8): 1219-1227, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38691139

RESUMEN

OBJECTIVES: To describe the pharmacokinetic (PK) characteristics of nirmatrelvir/ritonavir in renal transplant recipients and explore the potential factors that related to the PK variance of nirmatrelvir/ritonavir and its interaction with calcineurin inhibitor (CNI). METHODS: Renal transplant recipients treated with CNI and nirmatrelvir/ritonavir were prospectively enrolled. Steady-state plasma concentrations of nirmatrelvir/ritonavir were determined by high-performance liquid chromatography-tandem mass spectrometry, and the PK parameters were calculated using non-compartmental analysis. Spearman correlation analysis was used for exploring influencing factors. RESULTS: A total of eight recipients were enrolled; for nirmatrelvir and ritonavir, AUC/dose was 0.24179 ± 0.14495 and 0.06196 ± 0.03767 µg·h·mL-1·mg-1. Red blood cell (RBC), hematocrit (Ht), hemoglobins (Hb), and creatinine clearance (Ccr) were negatively correlated with AUC/dose of nirmatrelvir, while Ccr, CYP3A5 genotype, and CYP3A4 genotype were related to the AUC/dose of ritonavir. Ccr was negatively correlated with the C0/dose of tacrolimus (TAC) after termination of nirmatrelvir/ritonavir (rs = -0.943, p = 0.008). CONCLUSIONS: The PK characteristics of nirmatrelvir/ritonavir vary greatly among renal transplant recipients. Factors including Ccr and CYP3A5 genotype were related to the in vivo exposure of nirmatrelvir/ritonavir. During the whole process before and after nirmatrelvir/ritonavir therapy, it is recommended to adjust the CNI basing on renal function to avoid CNI toxicity exposure.


Asunto(s)
Inhibidores de la Calcineurina , Interacciones Farmacológicas , Trasplante de Riñón , Ritonavir , Humanos , Ritonavir/farmacocinética , Ritonavir/farmacología , Masculino , Inhibidores de la Calcineurina/farmacocinética , Inhibidores de la Calcineurina/farmacología , Inhibidores de la Calcineurina/administración & dosificación , Femenino , Persona de Mediana Edad , Adulto , Citocromo P-450 CYP3A/metabolismo , Citocromo P-450 CYP3A/genética , Inmunosupresores/farmacocinética , Inmunosupresores/administración & dosificación , Estudios Prospectivos , Tacrolimus/farmacocinética , Tacrolimus/administración & dosificación , Tacrolimus/farmacología , Genotipo , Área Bajo la Curva , Receptores de Trasplantes
2.
PLoS Med ; 20(11): e1004303, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37988391

RESUMEN

BACKGROUND: The current World Health Organization (WHO) pediatric tuberculosis dosing guidelines lead to suboptimal drug exposures. Identifying factors altering the exposure of these drugs in children is essential for dose optimization. Pediatric pharmacokinetic studies are usually small, leading to high variability and uncertainty in pharmacokinetic results between studies. We pooled data from large pharmacokinetic studies to identify key covariates influencing drug exposure to optimize tuberculosis dosing in children. METHODS AND FINDINGS: We used nonlinear mixed-effects modeling to characterize the pharmacokinetics of rifampicin, isoniazid, and pyrazinamide, and investigated the association of human immunodeficiency virus (HIV), antiretroviral therapy (ART), drug formulation, age, and body size with their pharmacokinetics. Data from 387 children from South Africa, Zambia, Malawi, and India were available for analysis; 47% were female and 39% living with HIV (95% on ART). Median (range) age was 2.2 (0.2 to 15.0) years and weight 10.9 (3.2 to 59.3) kg. Body size (allometry) was used to scale clearance and volume of distribution of all 3 drugs. Age affected the bioavailability of rifampicin and isoniazid; at birth, children had 48.9% (95% confidence interval (CI) [36.0%, 61.8%]; p < 0.001) and 64.5% (95% CI [52.1%, 78.9%]; p < 0.001) of adult rifampicin and isoniazid bioavailability, respectively, and reached full adult bioavailability after 2 years of age for both drugs. Age also affected the clearance of all drugs (maturation), children reached 50% adult drug clearing capacity at around 3 months after birth and neared full maturation around 3 years of age. While HIV per se did not affect the pharmacokinetics of first-line tuberculosis drugs, rifampicin clearance was 22% lower (95% CI [13%, 28%]; p < 0.001) and pyrazinamide clearance was 49% higher (95% CI [39%, 57%]; p < 0.001) in children on lopinavir/ritonavir; isoniazid bioavailability was reduced by 39% (95% CI [32%, 45%]; p < 0.001) when simultaneously coadministered with lopinavir/ritonavir and was 37% lower (95% CI [22%, 52%]; p < 0.001) in children on efavirenz. Simulations of 2010 WHO-recommended pediatric tuberculosis doses revealed that, compared to adult values, rifampicin exposures are lower in most children, except those younger than 3 months, who experience relatively higher exposure for all drugs, due to immature clearance. Increasing the rifampicin doses in children older than 3 months by 75 mg for children weighing <25 kg and 150 mg for children weighing >25 kg could improve rifampicin exposures. Our analysis was limited by the differences in availability of covariates among the pooled studies. CONCLUSIONS: Children older than 3 months have lower rifampicin exposures than adults and increasing their dose by 75 or 150 mg could improve therapy. Altered exposures in children with HIV is most likely caused by concomitant ART and not HIV per se. The importance of the drug-drug interactions with lopinavir/ritonavir and efavirenz should be evaluated further and considered in future dosing guidance. TRIAL REGISTRATION: ClinicalTrials.gov registration numbers; NCT02348177, NCT01637558, ISRCTN63579542.


Asunto(s)
Infecciones por VIH , Tuberculosis , Adulto , Recién Nacido , Niño , Humanos , Femenino , Lactante , Preescolar , Adolescente , Masculino , Ritonavir/farmacocinética , Ritonavir/uso terapéutico , Lopinavir/farmacocinética , Lopinavir/uso terapéutico , Rifampin , Isoniazida/uso terapéutico , Isoniazida/farmacocinética , Pirazinamida/farmacocinética , Antituberculosos , Tuberculosis/tratamiento farmacológico , Tuberculosis/epidemiología , Infecciones por VIH/tratamiento farmacológico , VIH
3.
PLoS Pathog ; 17(3): e1009382, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33730100

RESUMEN

Mosquitoes are vectors of major diseases such as dengue fever and malaria. Mass drug administration of endectocides to humans and livestock is a promising complementary approach to current insecticide-based vector control measures. The aim of this study was to establish an insect model for pharmacokinetic and drug-drug interaction studies to develop sustainable endectocides for vector control. Female Aedes aegypti mosquitoes were fed with human blood containing either ivermectin alone or ivermectin in combination with ketoconazole, rifampicin, ritonavir, or piperonyl butoxide. Drug concentrations were quantified by LC-MS/MS at selected time points post-feeding. Primary pharmacokinetic parameters and extent of drug-drug interactions were calculated by pharmacometric modelling. Lastly, the drug effect of the treatments was examined. The mosquitoes could be dosed with a high precision (%CV: ≤13.4%) over a range of 0.01-1 µg/ml ivermectin without showing saturation (R2: 0.99). The kinetics of ivermectin were characterised by an initial lag phase of 18.5 h (CI90%: 17.0-19.8 h) followed by a slow zero-order elimination rate of 5.5 pg/h (CI90%: 5.1-5.9 pg/h). By contrast, ketoconazole, ritonavir, and piperonyl butoxide were immediately excreted following first order elimination, whereas rifampicin accumulated over days in the mosquitoes. Ritonavir increased the lag phase of ivermectin by 11.4 h (CI90%: 8.7-14.2 h) resulting in an increased exposure (+29%) and an enhanced mosquitocidal effect. In summary, this study shows that the pharmacokinetics of drugs can be investigated and modulated in an Ae. aegypti animal model. This may help in the development of novel vector-control interventions and further our understanding of toxicology in arthropods.


Asunto(s)
Aedes/efectos de los fármacos , Insecticidas/farmacocinética , Ivermectina/farmacocinética , Animales , Inhibidores del Citocromo P-450 CYP3A/farmacocinética , Interacciones Farmacológicas/fisiología , Humanos , Modelos Animales , Control de Mosquitos/métodos , Mosquitos Vectores/efectos de los fármacos , Ritonavir/farmacocinética
4.
Pharm Res ; 40(8): 1927-1938, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37231296

RESUMEN

PURPOSE: PAXLOVID™ is nirmatrelvir tablets co-packaged with ritonavir tablets. Ritonavir is used as a pharmacokinetics (PK) enhancer to reduce metabolism and increase exposure of nirmatrelvir. This is the first disclosure of Paxlovid physiologically-based pharmacokinetic (PBPK) model. METHODS: Nirmatrelvir PBPK model with first-order absorption kinetics was developed using in vitro, preclinical, and clinical data of nirmatrelvir in the presence and absence of ritonavir. Clearance and volume of distribution were derived from nirmatrelvir PK obtained using a spray-dried dispersion (SDD) formulation where it is considered to be dosed as an oral solution, and absorption is near complete. The fraction of nirmatrelvir metabolized by CYP3A was estimated based on in vitro and clinical ritonavir drug-drug interaction (DDI) data. First-order absorption parameters were established for both SDD and tablet formulation using clinical data. Nirmatrelvir PBPK model was verified with both single and multiple dose human PK data, as well as DDI studies. Simcyp® first-order ritonavir compound file was also verified with additional clinical data. RESULTS: The nirmatrelvir PBPK model described the observed PK profiles of nirmatrelvir well with predicted AUC and Cmax values within ± 20% of the observed. The ritonavir model performed well resulting in predicted values within twofold of observed. CONCLUSIONS: Paxlovid PBPK model developed in this study can be applied to predict PK changes in special populations, as well as model the effect of victim and perpetrator DDI. PBPK modeling continues to play a critical role in accelerating drug discovery and development of potential treatments for devastating diseases such as COVID-19. NCT05263895, NCT05129475, NCT05032950 and NCT05064800.


Asunto(s)
COVID-19 , Ritonavir , Humanos , Ritonavir/farmacocinética , Simulación por Computador , Cinética , Interacciones Farmacológicas , Modelos Biológicos
5.
Xenobiotica ; 52(6): 541-554, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36083110

RESUMEN

The pharmacokinetics, elimination, and metabolism of fostemsavir (FTR), a prodrug of the HIV-1 attachment inhibitor temsavir (TMR), were investigated in healthy volunteers. FTR was administered with and without ritonavir (RTV), a protease inhibitor previously shown to boost TMR exposures. In vitro studies were also used to identify the enzymes responsible for the metabolism of TMR.Total recovery of the administered dose ranged from 78% to 89%. Approximately 44% to 58% of the dose was excreted in urine, 20%-36% in faeces, and 5% in bile, as TMR and metabolites. RTV had no effect on the recovery of radioactivity in any matrix.Compared to FTR alone, pre-treatment of subjects with RTV increased the exposure of TMR by ∼66% and reduced the exposure of plasma total radioactivity by ∼68%.The major route of TMR elimination was through biotransformation. TMR, M28 (N-dealkylation), and M4 (amide hydrolysis) were the major circulating components in plasma. Pre-treatment with RTV increased the amount of TMR present, decreased the amount of circulating M28, and M4 was unchanged.CYP3A4 metabolism accounted for 21% of the dose, forming multiple oxidative metabolites. This pathway was inhibited by coadministration of RTV.


Asunto(s)
Fármacos Anti-VIH , Profármacos , Administración Oral , Amidas , Fármacos Anti-VIH/farmacocinética , Citocromo P-450 CYP3A , Heces , Voluntarios Sanos , Humanos , Masculino , Organofosfatos , Piperazinas , Inhibidores de Proteasas , Ritonavir/farmacocinética
6.
Paediatr Anaesth ; 32(10): 1091-1099, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35842922

RESUMEN

The protease inhibitor, ritonavir, is a strong inhibitor of CYP 3A. The drug is used for management of the human immunovirus and is currently part of an oral antiviral drug combination (nirmatrelvir-ritonavir) for the early treatment of SARS-2 COVID-19-positive patients aged 12 years and over who have recognized comorbidities. The CYP 3A enzyme system is responsible for clearance of numerous drugs used in anesthesia (e.g., alfentanil, fentanyl, methadone, rocuronium, bupivacaine, midazolam, ketamine). Ritonavir will have an impact on drug clearances that are dependent on ritonavir concentration, anesthesia drug intrinsic hepatic clearance, metabolic pathways, concentration-response relationship, and route of administration. Drugs with a steep concentration-response relationship (ketamine, midazolam, rocuronium) are mostly affected because small changes in concentration have major changes in effect response. An increase in midazolam concentration is observed after oral administration because CYP 3A in the gastrointestinal wall is inhibited, causing a large increase in relative bioavailability. Fentanyl infusion may be associated with a modest increase in plasma concentration and effect, but the large between subject variability of pharmacokinetic and pharmacodynamic concentration changes suggests it will have little impact on an individual patient, especially when used with adverse effect monitoring. It has been proposed that drugs that have no or only a small metabolic pathway involving the CYP 3A enzyme be used during anesthesia, for example, propofol, atracurium, remifentanil, and the volatile agents. That anesthesia approach denies children of drugs with considerable value. It is better that the inhibitory changes in clearance of these drugs are understood so that rational drug choices can be made to tailor drug use to the individual patient. Altered drug dose, anticipation of duration of effect, timing of administration, use of reversal agents and perioperative monitoring would better behoove children undergoing anesthesia.


Asunto(s)
Anestesia , Tratamiento Farmacológico de COVID-19 , Ketamina , Alfentanilo , Antivirales , Niño , Citocromo P-450 CYP3A/metabolismo , Interacciones Farmacológicas , Inhibidores Enzimáticos , Humanos , Midazolam , Inhibidores de Proteasas/farmacología , Ritonavir/farmacocinética , Rocuronio
7.
AAPS PharmSciTech ; 23(6): 177, 2022 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-35750994

RESUMEN

Efavirenz (EFV) with a booster dose of ritonavir (RTV) (EFV-RTV) inhibits the metabolism of EFV and improves its bioavailability. However, inadequate organ perfusion with surface permeability glycoprotein (P-gp) efflux sustains the viable HIV. Hence, the present investigations were aimed to evaluate the pharmacokinetics and tissue distribution efficiency of EFV by encapsulating it into PEGyalated PAMAM (polyamidoamine) G4 dendrimers with a booster dose of RTV (PPG4ER). The entrapment efficiency of PEGylated PAMAM G4 dendrimers was found to be 94% and 92.12% for EFV and RTV respectively with a zeta potential of 0.277 mV. The pharmacokinetics and tissue distribution behavior of EFV within PPG4ER was determined by developing and validating a simple, sensitive, and reliable bioanalytical method of RP-HPLC. The developed bioanalytical method was very sensitive with a quantification limit of 18.5 ng/ml and 139.2 ng/ml for EFV and RTV, respectively. The comparative noncompartmental pharmacokinetic parameters of EFV were determined by administrating a single intraperitoneal dose of EFV, EFV-RTV, and PPG4ER to Wistar rats. The PPG4ER produced prolonged release of EFV with a mean residential time (MRT) of 24 h with Cmax 7.68 µg/ml in plasma against EFV-RTV with MRT 11 h and Cmax 3.633 µg/ml. The PPG4ER was also detected in viral reservoir tissues (lymph node and spleen) for 3-4 days, whereas free EFV and EFV-RTV were cleared within 72 h. The pharmacokinetic data including Cmax, t1/2, AUCtot, and MRT were significantly improved in PPG4ER as compared with single EFV and EFV-RTV. This reveals that the PPG4ER has great potential to target the virus harbors tissues and improve bioavailability.


Asunto(s)
Fármacos Anti-VIH , Dendrímeros , Infecciones por VIH , Alquinos , Animales , Fármacos Anti-VIH/farmacocinética , Benzoxazinas , Disponibilidad Biológica , Ciclopropanos , Infecciones por VIH/tratamiento farmacológico , Polietilenglicoles/uso terapéutico , Ratas , Ratas Wistar , Ritonavir/farmacocinética
8.
Mol Pharm ; 18(11): 4079-4089, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34613730

RESUMEN

The solution behavior and membrane transport of multidrug formulations were herein investigated in a biorelevant medium simulating fasted conditions. Amorphous multidrug formulations were prepared by the solvent evaporation method. Combinations of atazanavir (ATV) and ritonavir (RTV) and felodipine (FDN) and indapamide (IPM) were prepared and stabilized by a polymer for studying their dissolution (under non-sink conditions) and membrane transport in fasted state simulated intestinal fluid (FaSSIF). The micellar solubilization by FaSSIF enhanced the amorphous solubility of the drugs to different extents. Similar to buffer, the maximum achievable concentration of drugs in combination was reduced in FaSSIF, but the extent of reduction was affected by the degree of FaSSIF solubilization. Dissolution studies of ATV and IPM revealed that the amorphous solubility of these two drugs was not affected by FaSSIF solubilization. In contrast, RTV was significantly affected by FaSSIF solubilization with a 30% reduction in the maximum achievable concentration upon combination to ATV, compared to 50% reduction in buffer. This positive deviation by FaSSIF solubilization was not reflected in the mass transport-time profiles. Interestingly, FDN concentrations remain constant until the amount of IPM added was over 1000 µg/mL. No decrease in the membrane transport of FDN was observed for a 1:1 M ratio of FDN-IPM combination. This study demonstrates the importance of studying amorphous multidrug formulations under physiologically relevant conditions to obtain insights into the performance of these formulations after oral administration.


Asunto(s)
Líquidos Corporales/química , Química Farmacéutica/métodos , Administración Oral , Sulfato de Atazanavir/administración & dosificación , Sulfato de Atazanavir/química , Sulfato de Atazanavir/farmacocinética , Membrana Celular/metabolismo , Combinación de Medicamentos , Felodipino/administración & dosificación , Felodipino/química , Felodipino/farmacocinética , Indapamida/administración & dosificación , Indapamida/química , Indapamida/farmacocinética , Intestinos , Membranas Artificiales , Ritonavir/administración & dosificación , Ritonavir/química , Ritonavir/farmacocinética , Solubilidad
9.
Eur J Clin Pharmacol ; 77(3): 389-397, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33048175

RESUMEN

OBJECTIVE: To develop a population pharmacokinetic model for lopinavir boosted by ritonavir in coronavirus disease 2019 (Covid-19) patients. METHODS: Concentrations of lopinavir/ritonavir were assayed by an accredited LC-MS/MS method. The population pharmacokinetics of lopinavir was described using non-linear mixed-effects modeling (NONMEM version 7.4). After determination of the base model that better described the data set, the influence of covariates (age, body weight, height, body mass index (BMI), gender, creatinine, aspartate aminotransferase (AST), alanine aminotransferase (ALT), C reactive protein (CRP), and trough ritonavir concentrations) was tested on the model. RESULTS: From 13 hospitalized patients (4 females, 9 males, age = 64 ± 16 years), 70 lopinavir/ritonavir plasma concentrations were available for analysis. The data were best described by a one-compartment model with a first-order input (KA). Among the covariates tested on the PK parameters, only the ritonavir trough concentrations had a significant effect on CL/F and improved the fit. Model-based simulations with the final parameter estimates under a regimen lopinavir/ritonavir 400/100 mg b.i.d. showed a high variability with median concentration between 20 and 30 mg/L (Cmin/Cmax) and the 90% prediction intervals within the range 1-100 mg/L. CONCLUSION: According to the estimated 50% effective concentration of lopinavir against SARS-CoV-2 virus in Vero E6 cells (16.7 mg/L), our model showed that at steady state, a dose of 400 mg b.i.d. led to 40% of patients below the minimum effective concentration while a dose of 1200 mg b.i.d. will reduce this proportion to 22%.


Asunto(s)
Antivirales/farmacocinética , COVID-19/metabolismo , Lopinavir/farmacocinética , Ritonavir/farmacocinética , Anciano , Anciano de 80 o más Años , Animales , Antivirales/uso terapéutico , Índice de Masa Corporal , Chlorocebus aethiops , Simulación por Computador , Combinación de Medicamentos , Femenino , Humanos , Lopinavir/uso terapéutico , Masculino , Persona de Mediana Edad , Modelos Biológicos , Población , Ritonavir/uso terapéutico , Análisis de Supervivencia , Distribución Tisular , Células Vero , Tratamiento Farmacológico de COVID-19
10.
Pharmacogenet Genomics ; 30(3): 45-53, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32106141

RESUMEN

OBJECTIVE: In AIDS Clinical Trials Group study A5316, efavirenz lowered plasma concentrations of etonogestrel and ethinyl estradiol, given as a vaginal ring, while atazanavir/ritonavir increased etonogestrel and lowered ethinyl estradiol concentrations. We characterized the pharmacogenetics of these interactions. METHODS: In A5316, women with HIV enrolled into control (no antiretrovirals), efavirenz [600 mg daily with nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs)], and atazanavir/ritonavir (300/100 mg daily with NRTIs) groups. On day 0, a vaginal ring was inserted, releasing etonogestrel/ethinyl estradiol 120/15 µg/day. Intensive plasma sampling for antiretrovirals was obtained on days 0 and 21, and single samples for etonogestrel and ethinyl estradiol on days 7, 14, and 21. Seventeen genetic polymorphisms were analyzed. RESULTS: The 72 participants in this analysis included 25, 24 and 23 in the control, efavirenz, and atazanavir/ritonavir groups, respectively. At day 21 in the efavirenz group, CYP2B6 genotype was associated with increased plasma efavirenz exposure (P = 3.2 × 10), decreased plasma concentrations of etonogestrel (P = 1.7 × 10), and decreased ethinyl estradiol (P = 6.7 × 10). Compared to controls, efavirenz reduced median etonogestrel concentrations by at least 93% in CYP2B6 slow metabolizers versus approximately 75% in normal and intermediate metabolizers. Efavirenz reduced median ethinyl estradiol concentrations by 75% in CYP2B6 slow metabolizers versus approximately 41% in normal and intermediate metabolizers. CONCLUSION: CYP2B6 slow metabolizer genotype worsens the pharmacokinetic interaction of efavirenz with hormonal contraceptives administered by vaginal ring. Efavirenz dose reduction in CYP2B6 slow metabolizers may reduce, but will likely not eliminate, this interaction.


Asunto(s)
Fármacos Anti-VIH/uso terapéutico , Sulfato de Atazanavir/uso terapéutico , Benzoxazinas/uso terapéutico , Anticonceptivos Femeninos/sangre , Agentes Anticonceptivos Hormonales/sangre , Ritonavir/uso terapéutico , Adulto , Alquinos , Sulfato de Atazanavir/farmacocinética , Benzoxazinas/farmacocinética , Anticonceptivos Femeninos/administración & dosificación , Anticonceptivos Femeninos/farmacocinética , Agentes Anticonceptivos Hormonales/administración & dosificación , Agentes Anticonceptivos Hormonales/farmacocinética , Dispositivos Anticonceptivos Femeninos , Ciclopropanos , Citocromo P-450 CYP2B6/genética , Desogestrel/sangre , Desogestrel/farmacocinética , Interacciones Farmacológicas , Etinilestradiol/sangre , Etinilestradiol/farmacocinética , Femenino , Estudios de Asociación Genética , Genotipo , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Humanos , Persona de Mediana Edad , Farmacogenética , Polimorfismo Genético , Polimorfismo de Nucleótido Simple , Ritonavir/farmacocinética , Vagina
11.
Am J Transplant ; 20(7): 1896-1901, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32337790

RESUMEN

The current coronavirus disease 2019 (COVID-19) pandemic requires extra attention for immunocompromised patients, including solid organ transplant recipients. We report on a case of a 35-year-old renal transplant recipient who suffered from a severe COVID-19 pneumonia. The clinical course was complicated by extreme overexposure to the mammalian target of rapamycin inhibitor everolimus, following coadministration of chloroquine and lopinavir/ritonavir therapy. The case is illustrative for dilemmas that transplant professionals may face in the absence of evidence-based COVID-19 therapy and concurrent pressure for exploration of experimental pharmacological treatment options. However, the risk-benefit balance of experimental or off-label therapy may be weighed differently in organ transplant recipients than in otherwise healthy COVID-19 patients, owing to their immunocompromised status and potential drug interactions with immunosuppressive therapy. With this case report, we aimed to achieve increased awareness and improved management of drug-drug interactions associated with the various treatment options for COVID-19 in renal transplant patients.


Asunto(s)
Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/terapia , Everolimus/farmacocinética , Fallo Renal Crónico/complicaciones , Trasplante de Riñón , Neumonía Viral/complicaciones , Neumonía Viral/terapia , Receptores de Trasplantes , Adulto , Antivirales/administración & dosificación , Antivirales/farmacocinética , Betacoronavirus , COVID-19 , Cloroquina/administración & dosificación , Cloroquina/farmacocinética , Combinación de Medicamentos , Interacciones Farmacológicas , Everolimus/administración & dosificación , Humanos , Huésped Inmunocomprometido , Inmunosupresores/administración & dosificación , Inmunosupresores/farmacocinética , Fallo Renal Crónico/cirugía , Lopinavir/administración & dosificación , Lopinavir/farmacocinética , Masculino , Países Bajos , Pandemias , Radiografía Torácica , Ritonavir/administración & dosificación , Ritonavir/farmacocinética , SARS-CoV-2 , Resultado del Tratamiento
12.
Artículo en Inglés | MEDLINE | ID: mdl-32071055

RESUMEN

In children requiring lopinavir coformulated with ritonavir in a 4:1 ratio (lopinavir-ritonavir-4:1) and rifampin, adding ritonavir to achieve a 4:4 ratio with lopinavir (LPV/r-4:4) overcomes the drug-drug interaction. Possible drug-drug interactions within this regimen may affect abacavir concentrations, but this has never been studied. Children weighing <15 kg needing rifampin and LPV/r-4:4 were enrolled in a pharmacokinetic study and underwent intensive pharmacokinetic sampling on 3 visits: (i) during the intensive and (ii) continuation phases of antituberculosis treatment with LPV/r-4:4 and (iii) 1 month after antituberculosis treatment completion on LPV/r-4:1. Pharmacometric modeling and simulation were used to compare exposures across weight bands with adult target exposures. Eighty-seven children with a median (interquartile range) age and weight of 19 (4 to 64) months and 8.7 (3.9 to 14.9) kg, respectively, were included in the abacavir analysis. Abacavir pharmacokinetics were best described by a two-compartment model with first-order elimination and transit compartment absorption. After allometric scaling adjusted for the effect of body size, maturation could be identified: clearance was predicted to be fully mature at about 2 years of age and to reach half of this mature value at about 2 months of age. Abacavir bioavailability decreased 36% during treatment with rifampin and LPV/r-4:4 but remained within the median adult recommended exposure, except for children in the 3- to 4.9-kg weight band, in which the exposures were higher. The observed predose morning trough concentrations were higher than the evening values. Though abacavir exposure significantly decreased during concomitant administration of rifampin and LPV/r-4:4, it remained within acceptable ranges. (This study is registered in ClinicalTrials.gov under identifier NCT02348177.).


Asunto(s)
Didesoxinucleósidos/farmacocinética , Infecciones por VIH/tratamiento farmacológico , Lopinavir/farmacocinética , Rifampin/farmacocinética , Ritonavir/farmacocinética , Tuberculosis Pulmonar/tratamiento farmacológico , Fármacos Anti-VIH/farmacocinética , Fármacos Anti-VIH/uso terapéutico , Antituberculosos/farmacocinética , Antituberculosos/uso terapéutico , Disponibilidad Biológica , Niño , Preescolar , Didesoxinucleósidos/uso terapéutico , Combinación de Medicamentos , Interacciones Farmacológicas , Humanos , Lopinavir/uso terapéutico , Estudios Prospectivos , Rifampin/uso terapéutico , Ritonavir/uso terapéutico
13.
Artículo en Inglés | MEDLINE | ID: mdl-32015036

RESUMEN

The purpose of this study was to evaluate the pharmacokinetics of ritonavir-boosted fosamprenavir during pregnancy and postpartum. Amprenavir (the active moiety of fosamprenavir) and ritonavir intensive pharmacokinetic evaluations were performed at steady state during the second and third trimesters of pregnancy and postpartum. Plasma concentrations of amprenavir and ritonavir were measured using high-performance liquid chromatography. The target amprenavir area under the concentration-versus-time curve (AUC) was higher than the 10th percentile (27.7 µg · h/ml) of the median area under the curve for ritonavir-boosted fosamprenavir in adults receiving twice-daily fosamprenavir-ritonavir at 700 mg/100 mg. Twenty-nine women were included in the analysis. The amprenavir AUC from time zero to 12 h (AUC0-12) was lower (geometric mean ratio [GMR], 0.60 [confidence interval {CI}, 0.49 to 0.72] [P < 0.001]) while its apparent oral clearance was higher (GMR, 1.68 [CI, 1.38 to 2.03] [P < 0.001]) in the third trimester than postpartum. Similarly, the ritonavir AUC0-12 was lower in the second (GMR, 0.51 [CI, 0.28 to 0.91] [P = 0.09]) and third (GMR, 0.72 [CI, 0.55 to 0.95] [P = 0.005]) trimesters than postpartum, while its apparent oral clearance was higher in the second (GMR, 1.98 [CI, 1.10 to 3.56] [P = 0.06]) and third (GMR, 1.38 [CI, 1.05 to 1.82] [P = 0.009]) trimesters than postpartum. The amprenavir area under the curve exceeded the target for 6/8 (75%) women in the 2nd trimester, 18/28 (64%) in the 3rd trimester, and 19/22 (86.4%) postpartum, and the trough concentrations (Cmin) of amprenavir were 4- to 16-fold above the mean amprenavir-protein-adjusted 50% inhibitory concentration (IC50) of 0.146 µg/ml. Although amprenavir plasma concentrations in women receiving ritonavir-boosted fosamprenavir were lower during pregnancy than postpartum, the reduced amprenavir concentrations were still above the exposures needed for viral suppression.


Asunto(s)
Carbamatos/farmacocinética , Furanos/farmacocinética , Infecciones por VIH/tratamiento farmacológico , Inhibidores de la Proteasa del VIH/farmacocinética , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Ritonavir/farmacocinética , Sulfonamidas/farmacocinética , Adulto , Área Bajo la Curva , Carbamatos/efectos adversos , Femenino , Furanos/efectos adversos , Inhibidores de la Proteasa del VIH/efectos adversos , Humanos , Edad Materna , Embarazo , Trimestres del Embarazo , ARN Viral/sangre , Ritonavir/efectos adversos , Sulfonamidas/efectos adversos , Carga Viral
14.
Artículo en Inglés | MEDLINE | ID: mdl-32071050

RESUMEN

Treating malaria in HIV-coinfected individuals should consider potential drug-drug interactions. Artemether-lumefantrine is the most widely recommended treatment for uncomplicated malaria globally. Lumefantrine is metabolized by CYP3A4, an enzyme that commonly used antiretrovirals often induce or inhibit. A population pharmacokinetic meta-analysis was conducted using individual participant data from 10 studies with 6,100 lumefantrine concentrations from 793 nonpregnant adult participants (41% HIV-malaria-coinfected, 36% malaria-infected, 20% HIV-infected, and 3% healthy volunteers). Lumefantrine exposure increased 3.4-fold with coadministration of lopinavir-ritonavir-based antiretroviral therapy (ART), while it decreased by 47% with efavirenz-based ART and by 59% in the patients with rifampin-based antituberculosis treatment. Nevirapine- or dolutegravir-based ART and malaria or HIV infection were not associated with significant effects. Monte Carlo simulations showed that those on concomitant efavirenz or rifampin have 49% and 80% probability of day 7 concentrations <200 ng/ml, respectively, a threshold associated with an increased risk of treatment failure. The risk of achieving subtherapeutic concentrations increases with larger body weight. An extended 5-day and 6-day artemether-lumefantrine regimen is predicted to overcome these drug-drug interactions with efavirenz and rifampin, respectively.


Asunto(s)
Fármacos Anti-VIH/farmacocinética , Antimaláricos/farmacocinética , Terapia Antirretroviral Altamente Activa , Lumefantrina/farmacocinética , Adolescente , Adulto , Anciano , Fármacos Anti-VIH/uso terapéutico , Antimaláricos/uso terapéutico , Combinación Arteméter y Lumefantrina/farmacocinética , Combinación Arteméter y Lumefantrina/uso terapéutico , Peso Corporal , Simulación por Computador , Interacciones Farmacológicas , Femenino , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Humanos , Lopinavir/farmacocinética , Lopinavir/uso terapéutico , Lumefantrina/uso terapéutico , Malaria/complicaciones , Malaria/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Método de Montecarlo , Ritonavir/farmacocinética , Ritonavir/uso terapéutico , Adulto Joven
15.
Artículo en Inglés | MEDLINE | ID: mdl-32641296

RESUMEN

Coronavirus disease 2019 (COVID-19) leads to inflammatory cytokine release, which can downregulate the expression of metabolizing enzymes. This cascade affects drug concentrations in the plasma. We investigated the association between lopinavir (LPV) and hydroxychloroquine (HCQ) plasma concentrations and the levels of the acute-phase inflammation marker C-reactive protein (CRP). LPV plasma concentrations in 92 patients hospitalized at our institution were prospectively collected. Lopinavir-ritonavir was administered every 12 hours, 800/200 mg on day 1 and 400/100 mg on day 2 until day 5 or 7. HCQ was given at 800 mg, followed by 400 mg after 6, 24, and 48 h. Hematological, liver, kidney, and inflammation laboratory values were analyzed on the day of drug level determination. The median age of study participants was 59 (range, 24 to 85) years, and 71% were male. The median durations from symptom onset to hospitalization and treatment initiation were 7 days (interquartile range [IQR], 4 to 10) and 8 days (IQR, 5 to 10), respectively. The median LPV trough concentration on day 3 of treatment was 26.5 µg/ml (IQR, 18.9 to 31.5). LPV plasma concentrations positively correlated with CRP values (r = 0.37, P < 0.001) and were significantly lower when tocilizumab was preadministered. No correlation was found between HCQ concentrations and CRP values. High LPV plasma concentrations were observed in COVID-19 patients. The ratio of calculated unbound drug fraction to published SARS-CoV-2 50% effective concentrations (EC50) indicated insufficient LPV concentrations in the lung. CRP values significantly correlated with LPV but not HCQ plasma concentrations, implying inhibition of cytochrome P450 3A4 (CYP3A4) metabolism by inflammation.


Asunto(s)
Antivirales/farmacocinética , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Hidroxicloroquina/farmacocinética , Lopinavir/farmacocinética , Neumonía Viral/tratamiento farmacológico , Ritonavir/farmacocinética , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antivirales/sangre , Antivirales/farmacología , Betacoronavirus/inmunología , Betacoronavirus/patogenicidad , Biomarcadores/sangre , Proteína C-Reactiva/metabolismo , COVID-19 , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/mortalidad , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/mortalidad , Síndrome de Liberación de Citoquinas/virología , Esquema de Medicación , Combinación de Medicamentos , Femenino , Hospitales Universitarios , Humanos , Hidroxicloroquina/sangre , Hidroxicloroquina/farmacología , Tiempo de Internación/estadística & datos numéricos , Lopinavir/sangre , Lopinavir/farmacología , Masculino , Persona de Mediana Edad , Pandemias , Neumonía Viral/inmunología , Neumonía Viral/mortalidad , Neumonía Viral/virología , Estudios Retrospectivos , Ritonavir/sangre , Ritonavir/farmacología , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Análisis de Supervivencia
16.
Invest New Drugs ; 38(5): 1526-1532, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32306204

RESUMEN

Introduction Oral formulations of docetaxel have successfully been developed as an alternative for intravenous administration. Co-administration with the enzyme inhibitor ritonavir boosts the docetaxel plasma exposure. In dose-escalation trials, the maximum tolerated doses for two different dosing regimens were established and dose-limiting toxicities (DLTs) were recorded. The aim of current analysis was to develop a pharmacokinetic (PK)-toxicodynamic (TOX) model to quantify the relationship between docetaxel plasma exposure and DLTs. Methods A total of 85 patients was included in the current analysis, 18 patients showed a DLT in the four-week observation period. A PK-TOX model was developed and simulations based on the PK-TOX model were performed. Results The final PK-TOX model was characterized by an effect compartment representing the toxic effect of docetaxel, which was linked to the probability of developing a DLT. Simulations of once-weekly, once-daily 60 mg and once-weekly, twice-daily 30 mg followed by 20 mg of oral docetaxel suggested that 14% and 34% of patients, respectively, would have a probability >25% to develop a DLT in a four-week period. Conclusions A PK-TOX model was successfully developed. This model can be used to evaluate the probability of developing a DLT following treatment with oral docetaxel and ritonavir in different dosing regimens.


Asunto(s)
Antineoplásicos , Protocolos de Quimioterapia Combinada Antineoplásica , Docetaxel , Modelos Biológicos , Ritonavir , Administración Oral , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Simulación por Computador , Docetaxel/administración & dosificación , Docetaxel/efectos adversos , Docetaxel/farmacocinética , Docetaxel/toxicidad , Humanos , Ritonavir/administración & dosificación , Ritonavir/efectos adversos , Ritonavir/farmacocinética , Ritonavir/toxicidad
17.
Mol Pharm ; 17(12): 4564-4571, 2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33151697

RESUMEN

Seeded growth rates of ritonavir in copovidone at 75% relative humidity (RH) and 50 °C were evaluated by single-particle tracking second harmonic generation (SHG) microscopy and found to be ∼3-fold slower for crystallites at the surface compared to the bulk. The shelf lives of final dosage forms containing amorphous solid dispersions (ASDs) are often dictated by the rates of active pharmaceutical ingredient crystallization. Upon exposure to elevated RH, the higher anticipated water content near the surfaces of ASDs has the potential to substantially impact nucleation and growth kinetics relative to the bulk. However, quantitative assessment of these differences in growth rates is complicated by challenges associated with discrimination of the two contributions (supersaturation and molecular mobility) in ensemble-averaged measurements. In the present study, "sandwich" materials were prepared, in which sparse populations of ritonavir single-crystalline seeds were pressed between two similar ASD films to assess bulk crystallization rates. These sandwich materials were compared and contrasted with analogously prepared "open-faced" samples, without the capping film, to assess the surface crystallization rates. Single-particle analysis by SHG microscopy time-series during in situ crystallization produced average growth rates of 3.8 µm/h for bulk columnar crystals with a particle-to-particle standard deviation of 0.9 µm/h. In addition, columnar crystal growth rates for surface particles were measured to be 1.3 µm/h and radiating crystal growth rates for surface particles were measured to be 1.0 µm/h, both with a particle-to-particle deviation of 0.4 µm/h. The observed appearance of radiating crystals upon surface seeding is attributed to reduced ritonavir solubility upon water adsorption at the interface, leading to higher defect densities in crystal growth. Despite substantial differences in crystal habit, correction of the surface growth rates by a factor of 4 from geometric effects resulted in relatively minor but statistically significant differences in the growth kinetics for the two local environments. These results are consistent, with viscosity being a relatively weak function of water absorption coupled with primarily diffusion-limited growth kinetics.


Asunto(s)
Excipientes/química , Ritonavir/química , Disponibilidad Biológica , Química Farmacéutica , Cristalización , Liberación de Fármacos , Estabilidad de Medicamentos , Almacenaje de Medicamentos , Ritonavir/farmacocinética , Solubilidad
18.
Mol Pharm ; 17(7): 2329-2344, 2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32427480

RESUMEN

Ritonavir is a well-known CYP3A4 and CYP2D6 enzyme inhibitor, frequently used to assess the drug-drug interaction (DDI) liability of susceptible drugs. It is also used as a pharmacokinetic booster to increase exposure to CYP3A4 substrates. This study aimed to develop a mechanistic absorption and disposition model to describe exposure to ritonavir following oral dosing of the commercial amorphous solid dispersion tablet, Norvir, under fasted and fed conditions. A mechanistic description of ritonavir absorption from Norvir tablets may help to improve the design of DDI studies. Key parameters of amorphous ritonavir including free base solubility (solubility of the unbound, un-ionized species), bile micelle partition coefficients, formulation wetting/disintegration, and in vivo precipitation parameters were either obtained from the literature or estimated by modeling in vitro biopharmaceutic experiments. Based on variety of in vitro evidence, a main assumption of the model is that ritonavir does not form a crystalline precipitate while resident in the gastrointestinal tract. In the model, if simulated luminal concentration exceeds the amorphous solubility limit, then precipitation to an amorphous form is immediate. Simulated and observed Cmax and AUC0-t parameters were well captured (within 1.5-fold) for both fasted and fed states in healthy volunteers. By accounting for luminal fluid viscosity differences in the different prandial states (affecting drug diffusivity) as well as the effect of drug free fraction on gut wall permeation rates, it was possible to explain the negative food effect observed for Norvir tablets in humans. In summary, a biopharmaceutic in vitro in vivo extrapolation approach provides confidence in (verification of) key input parameters of the physiologically-based pharmacokinetic ritonavir model which resulted in successful simulation of observed plasma profiles.


Asunto(s)
Productos Biológicos/farmacocinética , Ingestión de Alimentos , Ayuno , Absorción Intestinal/efectos de los fármacos , Ritonavir/farmacocinética , Administración Oral , Productos Biológicos/administración & dosificación , Productos Biológicos/química , Biofarmacia , Simulación por Computador , Dieta Alta en Grasa , Interacciones Farmacológicas , Voluntarios Sanos , Humanos , Concentración de Iones de Hidrógeno , Modelos Biológicos , Permeabilidad , Ritonavir/administración & dosificación , Ritonavir/química , Solubilidad , Comprimidos , Viscosidad , Agua/química
19.
Ann Clin Microbiol Antimicrob ; 19(1): 3, 2020 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-31969147

RESUMEN

BACKGROUND: To evaluate the pharmacokinetic of plasma lopinavir (LPV) and ritonavir (RTV) when co-administered with three times weekly (TPW) rifabutin (RBT) at a dose of either 150 or 300 mg in African tuberculosis (TB) and HIV co-infected adult patients. METHODS: This is a pharmacokinetic study conducted in Ouagadougou among patients treated with a standard dosage of LPV/RTV 400/100 mg twice daily and RBT 150 mg TPW (arm A = 9 patients) or rifabutin 300 mg TPW (arm B = 7 patients) based regimens. Patients were recruited from the Bogodogo and Kossodo district hospitals in Ouagadougou from May 2013 to December 2015. Study inclusion criteria were that the patients were between 18 and 60 years of age, HIV-1 infected with pulmonary tuberculosis confirmed or suspected. Subsequent blood samples for pharmacokinetic monitoring were collected at 1, 2, 3, 4, 6, 8 and 12 h after combined drug ingestion for plasma drug monitoring using HPLC/MS assays. RESULTS: The medians LPV Cmax and Tmax were respectively, 20 µg/mL and 4 h for the RBT 150 mg group (arm A) and 7.7 µg/mL and 3 h for the RBT 300 mg group (arm B). The AUC0-12 of LPV was 111.8 µg h/mL in patients belonging to arm A versus 69.9 µg/mL for those in arm B (p = 0.313). The C0 of LPV was lower than 4 µg/mL in three patients receiving RBT 300 mg. Of note, the RTV plasma concentrations were nearly halved among patients on RBT 300 mg compared to those on lower RBT doses. The AUC0-12 of RTV in arm A was 12.7 µg h/mL versus 6.6 µg h/ml in arm B (p = 0.313). CONCLUSION: In our study, the pharmacokinetic of LPV and RTV was found to be highly variable when coadministrated with RBT 150 mg or 300 mg three times per week. There is a need for specific large study to verify clinical and virological effects of this variation, especially when coadministrated with RBT of 300 mg TPW, and to prevent viral resistance in response to under-dosing of LPV. Trial registration PACTR201310000629390. Registered 28 October 2013, http://www.pactr.org/.


Asunto(s)
Coinfección/tratamiento farmacológico , Infecciones por VIH/tratamiento farmacológico , Lopinavir/farmacocinética , Ritonavir/farmacocinética , Tuberculosis/tratamiento farmacológico , Adolescente , Adulto , Fármacos Anti-VIH/uso terapéutico , Burkina Faso , Femenino , VIH-1 , Humanos , Lopinavir/sangre , Masculino , Persona de Mediana Edad , Rifabutina/administración & dosificación , Rifabutina/uso terapéutico , Ritonavir/sangre , Adulto Joven
20.
Artículo en Inglés | MEDLINE | ID: mdl-30783000

RESUMEN

Identifying and understanding potential drug-drug interactions (DDIs) are vital for the treatment of human immunodeficiency virus type 1 (HIV-1) infection. This article discusses DDIs between doravirine, a nonnucleoside reverse transcriptase inhibitor (NNRTI), and cytochrome P450 3A (CYP3A) substrates and drugs that modulate CYP3A activity. Consistent with previously published in vitro data and DDI trials with the CYP3A substrates midazolam and atorvastatin, doravirine did not have any meaningful impact on the pharmacokinetics of the CYP3A substrates ethinyl estradiol and levonorgestrel. Coadministration of doravirine with CYP3A inhibitors (ritonavir or ketoconazole) increased doravirine exposure approximately 3-fold. However, these increases were not considered clinically meaningful. Conversely, previously published trials showed that coadministered CYP3A inducers (rifampin and rifabutin) decreased doravirine exposure by 88% and 50%, respectively (K. L. Yee, S. G. Khalilieh, R. I. Sanchez, R. Liu, et al., Clin Drug Investig 37:659-667, 2017 [https://doi.org/10.1007/s40261-017-0513-4]; S. G. Khalilieh, K. L. Yee, R. I. Sanchez, R. Liu, et al., J Clin Pharmacol 58:1044-1052, 2018 [https://doi.org/10.1002/jcph.1103]), while doravirine exposure following prior efavirenz administration led to an initial reduction in doravirine exposure of 62%, but the reduction became less pronounced with time (K. L. Yee, R. I. Sanchez, P. Auger, R. Liu, et al., Antimicrob Agents Chemother 61:e01757-16, 2017 [https://doi.org/10.1128/AAC.01757-16]). Overall, the coadministration of doravirine with CYP3A inhibitors and substrates is, therefore, supported by these data together with efficacy and safety data from clinical trials, while coadministration with strong CYP3A inducers, such as rifampin, cannot be recommended. Concomitant dosing with rifabutin (a CYP3A inducer less potent than rifampin) is acceptable if doravirine dosing is adjusted from once to twice daily; however, the effect of other moderate inducers on doravirine pharmacokinetics is unknown.


Asunto(s)
Inhibidores del Citocromo P-450 CYP3A/farmacocinética , Citocromo P-450 CYP3A/metabolismo , Piridonas/farmacocinética , Triazoles/farmacocinética , Adolescente , Adulto , Anciano , Alquinos , Benzoxazinas/farmacocinética , Ciclopropanos , Interacciones Farmacológicas , Femenino , Humanos , Cetoconazol/farmacocinética , Masculino , Persona de Mediana Edad , Ritonavir/farmacocinética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA