Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
CPT Pharmacometrics Syst Pharmacol ; 13(4): 589-598, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38303579

RESUMO

Lefamulin is being evaluated as a treatment for bacterial exacerbations in cystic fibrosis (CF). Ivacaftor is approved for the treatment of patients with CF. Lefamulin is a moderate CYP3A inhibitor and co-administration with ivacaftor may result in a drug-drug interaction (DDI). A CF population was built based on literature using the Simcyp Simulator. A previously developed and validated physiologically-based pharmacokinetic (PBPK) model for ivacaftor was used. A PBPK model for lefamulin was developed and verified. Predicted concentrations and pharmacokinetic (PK) parameters for both ivacaftor and lefamulin in healthy subjects and patients with CF were in reasonable agreement with observed data (within 1.4-fold, majority within 1.25-fold). The lefamulin model as a CYP3A4 perpetrator was validated using a different Ki value for oral (p.o.) and intravenous (i.v.) routes. The simulated changes in area under the curve of ivacaftor in patients with CF when co-administered with p.o. and i.v. lefamulin were weak-to-moderate. The predicted change in ivacaftor PK when co-administered with oral lefamulin was less than observed between ivacaftor and fluconazole. These results suggest a low liability for a DDI between lefamulin and ivacaftor in patients with CF.


Assuntos
Aminofenóis , Fibrose Cística , Diterpenos , Compostos Policíclicos , Quinolonas , Tioglicolatos , Humanos , Fibrose Cística/tratamento farmacológico , Quinolonas/uso terapêutico , Inibidores do Citocromo P-450 CYP3A/farmacologia , Citocromo P-450 CYP3A , Interações Medicamentosas , Modelos Biológicos
2.
Clin Pharmacol Ther ; 115(2): 299-308, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37971208

RESUMO

A physiologically-based pharmacokinetic (PBPK) model was developed to simulate plasma concentrations of tucatinib (TUKYSA®) after single-dose or multiple-dose administration of 300 mg b.i.d. orally. This PBPK model was subsequently applied to support evaluation of drug-drug interaction (DDI) risk as a perpetrator resulting from tucatinib inhibition of CYP3A4, CYP2C8, CYP2C9, P-gp, or MATE1/2-K. The PBPK model was also applied to support evaluation of DDI risk as a victim resulting from co-administration with CYP3A4 or CYP2C8 inhibitors, or a CYP3A4 inducer. After refinement with clinical DDI data, the final PBPK model was able to recover the clinically observed single and multiple-dose plasma concentrations for tucatinib when tucatinib was administered as a single agent in healthy subjects. In addition, the final model was able to recover clinically observed plasma concentrations of tucatinib when administered in combination with itraconazole, rifampin, or gemfibrozil as well as clinically observed plasma concentrations of probe substrates of CYP3A4, CYP2C8, CYP2C9, P-gp, or MATE1/2-K. The PBPK model was then applied to prospectively predict the potential perpetrator or victim DDIs with other substrates, inducers, or inhibitors. To simulate a potential interaction with a moderate CYP2C8 inhibitor, two novel PBPK models representing a moderate CYP2C8 inhibitor and a sensitive CYP2C8 substrate were developed based on the existing PBPK models for gemfibrozil and rosiglitazone, respectively. The simulated population geometric mean area under the curve ratio of tucatinib with a moderate CYP2C8 inhibitor ranged from 1.98- to 3.08-fold, and based on these results, no dose modifications were proposed for moderate CYP2C8 inhibitors for the tucatinib label.


Assuntos
Inibidores do Citocromo P-450 CYP2C8 , Genfibrozila , Oxazóis , Piridinas , Quinazolinas , Humanos , Genfibrozila/farmacocinética , Citocromo P-450 CYP3A , Citocromo P-450 CYP2C8 , Citocromo P-450 CYP2C9 , Interações Medicamentosas , Modelos Biológicos , Inibidores do Citocromo P-450 CYP3A
3.
Drug Metab Dispos ; 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-38123941

RESUMO

The utility of PBPK models in support of drug development has been well documented. During the discovery stage, PBPK has increasingly been applied for early risk assessment, prediction of human dose, toxicokinetic dose projection and early formulation assessment. Previous review articles have proposed model building and application strategies for PBPK-based first in human predictions with comprehensive descriptions of the individual components of PBPK models. This includes the generation of decision trees, based on comprehensive literature reviews, to guide the application of PBPK in the discovery setting. The goal of this mini review is to provide additional guidance on the real-world application of PBPK, in support of the discovery stage of drug development. In this mini review, our goal is to provide guidance on the typical steps involved in the development and application of a PBPK model during drug discovery to assist in decision making. We have illustrated our recommended approach through description of case examples, where PBPK has been successfully applied to aid in human PK projection, candidate selection and prediction of drug interaction liability for parent and metabolite. Through these case studies, we have highlighted fundamental issues, including pre-verification in preclinical species, the application of empirical scalars in the prediction of in vivo clearance from in vitro systems, in silico prediction of permeability and the exploration of aqueous and biorelevant solubility data to predict dissolution. In addition, current knowledge gaps have been highlighted and future directions proposed. Significance Statement Through description of three case studies, we have highlighted the fundamental principles of PBPK application during drug discovery. These include pre-verification of the model in preclinical species, application of empirical scalars where necessary in the prediction of clearance, in silico prediction of permeability, and the exploration of aqueous and biorelevant solubility data to predict dissolution. In addition, current knowledge gaps have been highlighted and future directions proposed.

4.
CPT Pharmacometrics Syst Pharmacol ; 12(9): 1335-1346, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37587640

RESUMO

As part of a collaboration between Medicines for Malaria Venture (MMV), Certara UK and Monash University, physiologically-based pharmacokinetic (PBPK) models were developed for 20 antimalarials, using data obtained from standardized in vitro assays and clinical studies within the literature. The models have been applied within antimalarial drug development at MMV for more than 5 years. During this time, a strategy for their impactful use has evolved. All models are described in the supplementary material and are available to researchers. Case studies are also presented, demonstrating real-world development and clinical applications, including the assessment of the drug-drug interaction liability between combination partners or with co-administered drugs. This work emphasizes the benefit of PBPK modeling for antimalarial drug development and decision making, and presents a strategy to integrate it into the research and development process. It also provides a repository of shared information to benefit the global health research community.


Assuntos
Antimaláricos , Humanos , Desenvolvimento de Medicamentos , Projetos de Pesquisa , Universidades
5.
CPT Pharmacometrics Syst Pharmacol ; 12(10): 1553-1564, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37614073

RESUMO

Pomotrelvir is a new chemical entity and potent direct-acting antiviral inhibitor of the main protease of coronaviruses. Here the cytochrome P450 (CYP)-mediated drug-drug interaction (DDI) potential of pomotrelvir was evaluated for major CYP isoforms, starting with in vitro assays followed by the basic static model assessment. The identified CYP3A4-mediated potential DDIs were evaluated clinically at a supratherapeutic dose of 1050 mg twice daily (b.i.d.) of pomotrelvir, including pomotrelvir coadministration with ritonavir (strong inhibitor of CYP3A4) or midazolam (sensitive substrate of CYP3A4). Furthermore, a physiologically-based pharmacokinetic (PBPK) model was developed within the Simcyp Population-based Simulator using in vitro and in vivo information and validated with available human pharmacokinetic (PK) data. The PBPK model was simulated to assess the DDI potential for CYP isoforms that pomotrelvir has shown a weak to moderate DDI in vitro and for CYP3A4 at the therapeutic dose of 700 mg b.i.d. To support the use of pomotrelvir in women of childbearing potential, the impact of pomotrelvir on the exposure of the representative oral hormonal contraceptive drugs ethinyl estradiol and levonorgestrel was assessed using the PBPK model. The overall assessment suggested weak inhibition of pomotrelvir on CYP3A4 and minimal impact of a strong CYP3A4 inducer or inhibitor on pomotrelvir PK. Therefore, pomotrelvir is not anticipated to have clinically meaningful DDIs at the clinical dose. These comprehensive in vitro, in clinic, and in silico efforts indicate that the DDI potential of pomotrelvir is minimal, so excluding patients on concomitant medicines in clinical studies would not be required.


Assuntos
Citocromo P-450 CYP3A , Hepatite C Crônica , Humanos , Feminino , Antivirais/farmacologia , Sistema Enzimático do Citocromo P-450 , Interações Medicamentosas , Isoformas de Proteínas , Modelos Biológicos , Inibidores do Citocromo P-450 CYP3A/farmacologia , Simulação por Computador
6.
Pharmaceutics ; 15(5)2023 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-37242718

RESUMO

The bioequivalence of bempedoic acid oral suspension and commercial immediate release (IR) tablet formulations were assessed using a physiologically based pharmacokinetic (PBPK) model. The mechanistic model, developed from clinical mass balance results and in vitro intrinsic solubility, permeability, and dissolution data, was verified against observed clinical pharmacokinetics (PK) results. Model inputs included a fraction of a dose in solution (0.01%), viscosity (118.8 cps), and median particle diameter (50 µm) for the suspension and particle diameter (36.4 µm) for IR tablets. Dissolution was determined in the relevant media (pH 1.2-6.8) in vitro. Model simulations of bioequivalence predicted oral suspension (test) to IR tablet (reference) geometric mean ratio estimates of 96.9% (90% confidence interval [CI]: 92.6-101) for maximum concentration and 98.2% (90% CI: 87.3-111) for the area under the concentration-time curve. Sensitivity analyses showed gastric transit time had a minor impact on model predictions. Oral suspension biopharmaceutical safe space was defined by extremes of particle size and the percent of bempedoic acid in solution. PBPK model simulations predicted that the rate and extent of bempedoic acid absorption are unlikely to exhibit clinically meaningful differences when dosed as an oral suspension compared with an IR tablet without requiring a clinical bioequivalence study in adults.

7.
CPT Pharmacometrics Syst Pharmacol ; 12(4): 532-544, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36896795

RESUMO

Vonoprazan is metabolized extensively through CYP3A and is an in vitro time-dependent inhibitor of CYP3A. A tiered approach was applied to understand the CYP3A victim and perpetrator drug-drug interaction (DDI) potential for vonoprazan. Mechanistic static modeling suggested vonoprazan is a potential clinically relevant CYP3A inhibitor. Thus, a clinical study was conducted to evaluate the impact of vonoprazan on the exposure of oral midazolam, an index substrate for CYP3A. A physiologically-based pharmacokinetic (PBPK) model for vonoprazan was also developed using in vitro data, drug- and system-specific parameters, and clinical data and observations from a [14 C] human absorption, distribution, metabolism, and excretion study. The PBPK model was refined and verified using data from a clinical DDI study with the strong CYP3A inhibitor, clarithromycin, to confirm the fraction metabolized by CYP3A, and the oral midazolam clinical DDI data assessing vonoprazan as a time-dependent inhibitor of CYP3A. The verified PBPK model was applied to simulate the anticipated changes in vonoprazan exposure due to moderate and strong CYP3A inducers (efavirenz and rifampin, respectively). The clinical midazolam DDI study indicated weak inhibition of CYP3A, with a less than twofold increase in midazolam exposure. PBPK simulations projected a 50% to 80% reduction in vonoprazan exposure when administered concomitantly with moderate or strong CYP3A inducers. Based on these results, the vonoprazan label was revised and states that lower doses of sensitive CYP3A substrates with a narrow therapeutic index should be used when administered concomitantly with vonoprazan, and co-administration with moderate and strong CYP3A inducers should be avoided.


Assuntos
Indutores do Citocromo P-450 CYP3A , Inibidores do Citocromo P-450 CYP3A , Humanos , Inibidores do Citocromo P-450 CYP3A/farmacocinética , Indutores do Citocromo P-450 CYP3A/farmacocinética , Midazolam/farmacocinética , Citocromo P-450 CYP3A/metabolismo , Interações Medicamentosas , Modelos Biológicos
8.
Front Pharmacol ; 13: 974423, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36225583

RESUMO

Background: Atezolizumab has been studied in multiple indications for both pediatric and adult patient populations. Generally, clinical studies enrolling pediatric patients may not collect sufficient pharmacokinetic data to characterize the drug exposure and disposition because of operational, ethical, and logistical challenges including burden to children and blood sample volume limitations. Therefore, mechanistic modeling and simulation may serve as a tool to predict and understand the drug exposure in pediatric patients. Objective: To use mechanistic physiologically-based pharmacokinetic (PBPK) modeling to predict atezolizumab exposure at a dose of 15 mg/kg (max 1,200 mg) in pediatric patients to support dose rationalization and label recommendations. Methods: A minimal mechanistic PBPK model was used which incorporated age-dependent changes in physiology and biochemistry that are related to atezolizumab disposition such as endogenous IgG concentration and lymph flow. The PBPK model was developed using both in vitro data and clinically observed data in adults and was verified across dose levels obtained from a phase I and multiple phase III studies in both pediatric patients and adults. The verified model was then used to generate PK predictions for pediatric and adult subjects ranging from 2- to 29-year-old. Results: Individualized verification in children and in adults showed that the simulated concentrations of atezolizumab were comparable (76% within two-fold and 90% within three-fold, respectively) to the observed data with no bias for either over- or under-prediction. Applying the verified model, the predicted exposure metrics including Cmin, Cmax, and AUCtau were consistent between pediatric and adult patients with a geometric mean of pediatric exposure metrics between 0.8- to 1.25-fold of the values in adults. Conclusion: The results show that a 15 mg/kg (max 1,200 mg) atezolizumab dose administered intravenously in pediatric patients provides comparable atezolizumab exposure to a dose of 1,200 mg in adults. This suggests that a dose of 15 mg/kg will provide adequate and effective atezolizumab exposure in pediatric patients from 2- to 18-year-old.

9.
JCO Glob Oncol ; 8: e2200199, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36198134

RESUMO

PURPOSE: Pediatric CNS tumors are increasingly a priority, particularly with the WHO designation of low-grade glioma (LGG) as one of six index childhood cancers. There are currently limited data on outcomes of pediatric patients with LGGs in low- and middle-income countries (LMICs). METHODS: To better understand the outcomes of LGGs in LMICs, this systematic review interrogated nine literature databases. RESULTS: The search identified 14,977 publications. Sixteen studies from 19 countries met the selection criteria and were included for data abstraction and analysis. Eleven studies (69%) were retrospective reviews from single institutions, and one (6%) captured institutional data prospectively. The studies captured a total of 957 patients with a median of 49 patients per study. Seven (44%) of the studies described the treatment modalities used. Of 373 patients for whom there was information, 173 (46%) had a gross total or near total resection, 109 (29%) had a subtotal resection, and 91 (24%) had only a biopsy performed. Seven studies, with a total of 476 patients, described the frequency of use of radiotherapy and/or chemotherapy in the cohorts: 83 of these patients received radiotherapy and 76 received chemotherapy. The 5-year overall survival ranged from 69.2% to 93.5%, although lower survival rates were reported at earlier time points. We identified limitations in the published studies with respect to the cohort sizes and methodologies. CONCLUSION: The included studies reported survival rates frequently exceeding 80%, although the ultimate number of studies was limited, pointing to the paucity of studies describing the outcomes of children with LGGs in LMICs. This study underscores the need for more robust data on outcomes in pediatric LGG.


Assuntos
Neoplasias Encefálicas , Neoplasias do Sistema Nervoso Central , Glioma , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Criança , Países em Desenvolvimento , Glioma/patologia , Glioma/cirurgia , Humanos , Estudos Retrospectivos
10.
Pediatr Blood Cancer ; 69(12): e30007, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36129384

RESUMO

Cure4Kids is a free web-based knowledge platform for professionals providing care for children with cancer and hematologic diseases, offering its users a comprehensive suite of learning opportunities. It has been a resource for the pediatric oncology community across the world for the past two decades, with 60,107 users having logged in 1,412,514 times with 22,045,553 content hits. A transformation of Cure4Kids is being planned and will include an improved user interface, increased interactivity, and more content.


Assuntos
Aprendizagem , Neoplasias , Criança , Humanos , Neoplasias/terapia , Internet
11.
Drug Metab Dispos ; 50(10): 1322-1331, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36100353

RESUMO

Drugs that modulate cytokine levels are often used for the treatment of cancer as well as inflammatory or immunologic disorders. Pharmacokinetic drug-biologic interactions (DBIs) may arise from suppression or elevation of cytochrome P450 (P450) enzymes caused by the increase or decrease in cytokine levels after administration of these therapies. There is in vitro and in vivo evidence that demonstrates a clear link between raised interleukin (IL)-6 levels and P450 suppression, in particular CYP3A4. However, despite this, the changes in IL-6 levels in vivo rarely lead to significant drug interactions (area under the curve and Cmax ratios < 2-fold). The clinical significance of such interactions therefore remains questionable and is dependent on the therapeutic index of the small molecule therapy. Physiologically based pharmacokinetic (PBPK) modeling has been used successfully to predict the impact of raised IL-6 on P450 activities. Beyond IL-6, published data show little evidence that IL-8, IL-10, and IL-17 suppress P450 enzymes. In vitro data suggest that IL-1ß, IL-2, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ can cause suppression of P450 enzymes. Despite in vivo there being a link between IL-6 levels and P450 suppression, the evidence to support a direct effect of IL-2, IL-8, IL-10, IL-17, IFN-γ, TNF-α, or vascular endothelial growth factor on P450 activity is inconclusive. This commentary will discuss the relevance of such drug-biologic interactions and whether current PBPK models considering only IL-6 are sufficient. SIGNIFICANCE STATEMENT: This commentary summarizes the current in vitro and in vivo literature regarding cytokine-mediated cytochrome P450 suppression and compares the relative suppressive potential of different cytokines in reference to interleukin (IL)-6. It also discusses the relevance of drug-biologic interactions to therapeutic use of small molecule drugs and whether current physiologically based pharmacokinetic models considering only IL-6 are sufficient to predict the extent of drug-biologic interactions.


Assuntos
Produtos Biológicos , Interleucina-6 , Sistema Enzimático do Citocromo P-450/metabolismo , Citocinas , Interações Medicamentosas , Interleucina-10 , Interleucina-17 , Interleucina-2 , Interleucina-6/metabolismo , Interleucina-8 , Preparações Farmacêuticas/metabolismo , Fator de Necrose Tumoral alfa , Fator A de Crescimento do Endotélio Vascular
13.
AAPS J ; 24(4): 72, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35650328

RESUMO

New drugs may in some cases need to be tested in paediatric and pregnant patients. However, it is difficult to recruit such patients and there are many ethical issues around their inclusion in clinical trials. Modelling and simulation can help to plan well-designed clinical trials with a reduced number of participants and to bridge gaps where recruitment is difficult. Physiologically based pharmacokinetic (PBPK) models for small molecule drugs have been used to aid study design and dose adjustments in paediatrics and pregnancy, with several publications in the literature. However, published PBPK models for monoclonal antibodies (mAb) in these populations are scarce. Here, the current status of mAb PBPK models in paediatrics and pregnancy is discussed. Seven mAb PBPK models published for paediatrics were found, which report good prediction accuracy across a wide age range. No mAb PBPK models for pregnant women have been published to date. Current challenges to the development of such PBPK models are discussed, including gaps in our knowledge of relevant physiological processes and availability of clinical data to verify models. As the availability of such data increases, it will help to improve our confidence in the PBPK model predictive ability. Advantages for using PBPK models to predict mAb PK in paediatrics and pregnancy are discussed. For example, the ability to incorporate ontogeny and gestational changes in physiology, prediction of maternal, placental and foetal exposure and the ability to make predictions from in vitro and preclinical data prior to clinical data being available.


Assuntos
Modelos Biológicos , Pediatria , Anticorpos Monoclonais , Criança , Simulação por Computador , Feminino , Humanos , Placenta , Gravidez
14.
Clin Transl Sci ; 15(6): 1417-1429, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35233913

RESUMO

Trazodone is approved for the treatment of major depressive disorders, marketed as immediate release (IR), prolonged release, and once a day (OAD) formulation. The different formulations allow different administration schedules and may be useful to facilitate patients' compliance to the antidepressant treatment. A previously verified physiologically-based pharmacokinetic model based on in vitro and in vivo information on trazodone pharmacokinetics was applied, aiming at predicting brain receptor occupancy (RO) after single and repeated dosing of the IR formulation and repeated dosing of the OAD formulation in healthy subjects. Receptors included in the simulations were selected using static calculations of RO based on the maximum unbound brain concentration (Cmax,brain,u ) of trazodone for each formulation and dosing scheme, resulting in 16 receptors being simulated. Seven receptors were simulated for the IR low dose formulation (30 mg), with similar tonset and duration of coverage (range: 0.09-0.25 h and 2.1->24 h, respectively) as well as RO (range: 0.64-0.92) predicted between day 1 and day 7 of dosing. The 16 receptors evaluated for the OAD formulation (300 mg) showed high RO (range: 0.97-0.84 for the receptors also covered by the IR formulation and 0.73-0.48 for the remaining) correlating with affinity and similar duration of time above the target threshold to the IR formulation (range: 2->24 h). The dose-dependent receptor coverage supports the multimodal activity of trazodone, which may further contribute to its fast antidepressant action and effectiveness in controlling different symptoms in depressed patients.


Assuntos
Transtorno Depressivo Maior , Trazodona , Antidepressivos , Encéfalo , Preparações de Ação Retardada , Transtorno Depressivo Maior/tratamento farmacológico , Humanos
15.
Diabetes Care ; 2021 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-34362816

RESUMO

OBJECTIVE: To evaluate glucose control using fast-acting insulin aspart (faster aspart) compared with insulin aspart (IAsp) delivered by the MiniMed Advanced Hybrid Closed-Loop (AHCL) system in adults with type 1 diabetes. RESEARCH DESIGN AND METHODS: In this randomized, open-label, crossover study, participants were assigned to receive faster aspart or IAsp in random order. Stages 1 and 2 comprised of 6 weeks in closed loop, preceded by 2 weeks in open loop. This was followed by stage 3, whereby participants changed directly back to the insulin formulation used in stage 1 for 1 week in closed loop. Participants chose their own meals except for two standardized meal tests, a missed meal bolus and late meal bolus. The primary outcome was the percentage of time sensor glucose values were from 70 to 180 mg/dL (time in range; [TIR]). RESULTS: Twenty-five adults (52% male) were recruited; the median (interquartile range) age was 48 (37, 57) years, and the median HbA1c was 7.0% (6.6, 7.2) (53 [49, 55] mmol/mol). Faster aspart demonstrated greater overall TIR compared with IAsp (82.3% [78.5, 83.7] vs. 79.6% [77.0, 83.4], respectively; mean difference 1.9% [0.5, 3.3]; P = 0.007). Four-hour postprandial glucose TIR was higher using faster aspart compared with IAsp for all meals combined (73.6% [69.4, 80.2] vs. 72.1% [64.5, 78.5], respectively; median difference 3.5% [1.0, 7.3]; P = 0.003). There was no ketoacidosis or severe hypoglycemia. CONCLUSIONS: Faster aspart safely improved glucose control compared with IAsp in a group of adults with well-controlled type 1 diabetes using AHCL. The modest improvement was mainly related to mealtime glycemia. While the primary outcome demonstrated statistical significance, the clinical impact may be small, given an overall difference in TIR of 1.9%.

16.
Eur J Drug Metab Pharmacokinet ; 46(4): 513-525, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34143391

RESUMO

BACKGROUND AND OBJECTIVES: Lenabasum is a synthetic agonist of the cannabinoid receptor type 2 (CB2) with anti-inflammatory and antifibrotic properties. Utilizing Simcyp, we developed a physiologically based pharmacokinetic (PBPK) model based on physicochemical properties, cell culture data, and cytochrome P450 (CYP) phenotyping, inhibition, and induction data. METHODS: Clinical data from healthy volunteers treated with 20 mg of lenabasum in a single ascending dose (SAD) study were used for model development. The model was verified using lenabasum SAD (10 and 40 mg) data as well as multiple dose (20 mg three times per day) data. Lenabasum is a CYP substrate, and the model predicted lenabasum clearance of 51% by CYP2C9, 37% by CYP2C8, and 12% by CYP3A4. Lenabasum is also an inhibitor of these isozymes. RESULTS: The model accurately described the area under the plasma concentration-time curve (AUC) and maximum plasma concentration (Cmax) for lenabasum within 1.19-fold and 1.25-fold accuracy, respectively, of the observed clinical values. The simulations of CYP inducers predicted that the strongest interaction would occur with rifampin, with the AUC decreasing to 0.36 of the control value, whereas the simulations of CYP inhibitors predicted that the greatest effect would occur with fluconazole, with a 1.43-fold increase in AUC. CONCLUSIONS: Our model is a useful tool for predicting the pharmacokinetics of lenabasum and adjustments to its dosing in possible drug-drug interaction scenarios.


Assuntos
Agonistas de Receptores de Canabinoides/farmacocinética , Dronabinol/análogos & derivados , Modelos Biológicos , Adulto , Área Sob a Curva , Células CACO-2 , Agonistas de Receptores de Canabinoides/administração & dosagem , Simulação por Computador , Indutores das Enzimas do Citocromo P-450/farmacologia , Inibidores das Enzimas do Citocromo P-450/farmacologia , Relação Dose-Resposta a Droga , Dronabinol/administração & dosagem , Dronabinol/farmacocinética , Interações Medicamentosas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
17.
Front Bioinform ; 1: 731340, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-36303796

RESUMO

Quantitative modeling is increasingly utilized in the drug discovery and development process, from the initial stages of target selection, through clinical studies. The modeling can provide guidance on three major questions-is this the right target, what are the right compound properties, and what is the right dose for moving the best possible candidate forward. In this manuscript, we present a site-of-action modeling framework which we apply to monoclonal antibodies against soluble targets. We give a comprehensive overview of how we construct the model and how we parametrize it and include several examples of how to apply this framework for answering the questions postulated above. The utilities and limitations of this approach are discussed.

18.
CPT Pharmacometrics Syst Pharmacol ; 9(9): 534-541, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32697437

RESUMO

Monoclonal antibodies (mAbs) can be engineered to have "extended half-life" and "catch and release" properties to improve target coverage. We have developed a mAb physiologically-based pharmacokinetic model that describes intracellular trafficking, neonatal Fc receptor (FcRn) recycling, and nonspecific clearance of mAbs. We extended this model to capture target binding as a function of target affinity, expression, and turnover. For mAbs engineered to have an extended half-life, the model was able to accurately predict the terminal half-life (82% within 2-fold error of the observed value) in the human FcRn transgenic (Tg32) homozygous mouse and human. The model also accurately captures the trend in pharmacokinetic and target coverage data for a set of mAbs with differing catch and release properties in the Tg32 mouse. The mechanistic nature of this model allows us to explore different engineering techniques early in drug discovery, potentially expanding the number of "druggable" targets.


Assuntos
Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais/farmacocinética , Antígenos de Histocompatibilidade Classe I/genética , Receptores Fc/genética , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/metabolismo , Anticorpos Amplamente Neutralizantes/imunologia , Anticorpos Amplamente Neutralizantes/metabolismo , Simulação por Computador , Desenvolvimento de Medicamentos , Descoberta de Drogas , Anticorpos Anti-HIV/imunologia , Anticorpos Anti-HIV/metabolismo , Meia-Vida , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Homozigoto , Humanos , Camundongos , Camundongos Transgênicos , Modelos Imunológicos , Mutação , Ligação Proteica/imunologia , Engenharia de Proteínas/métodos , Receptores Fc/efeitos dos fármacos
19.
Clin Pharmacol Ther ; 108(5): 976-984, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32531808

RESUMO

We use a mechanistic lung model to demonstrate that accumulation of chloroquine (CQ), hydroxychloroquine (HCQ), and azithromycin (AZ) in the lungs is sensitive to changes in lung pH, a parameter that can be affected in patients with coronavirus disease 2019 (COVID-19). A reduction in pH from 6.7 to 6 in the lungs, as observed in respiratory disease, led to 20-fold, 4.0-fold, and 2.7-fold increases in lung exposure of CQ, HCQ, and AZ, respectively. Simulations indicated that the relatively high concentrations of CQ and HCQ in lung tissue were sustained long after administration of the drugs had stopped. Patients with COVID-19 often present with kidney failure. Our simulations indicate that renal impairment (plus lung pH reduction) caused 30-fold, 8.0-fold, and 3.4-fold increases in lung exposures for CQ, HCQ, and AZ, respectively, with relatively small accompanying increases (20 to 30%) in systemic exposure. Although a number of different dosage regimens were assessed, the purpose of our study was not to provide recommendations for a dosing strategy, but to demonstrate the utility of a physiologically-based pharmacokinetic modeling approach to estimate lung concentrations. This, used in conjunction with robust in vitro and clinical data, can help in the assessment of COVID-19 therapeutics going forward.


Assuntos
Azitromicina/farmacocinética , Infecções por Coronavirus , Hidroxicloroquina/farmacocinética , Pulmão , Pandemias , Pneumonia Viral , Anti-Infecciosos/farmacocinética , Antivirais/farmacocinética , Betacoronavirus/fisiologia , Disponibilidade Biológica , COVID-19 , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/fisiopatologia , Relação Dose-Resposta a Droga , Desenho de Fármacos , Humanos , Concentração de Íons de Hidrogênio , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Taxa de Depuração Metabólica , Pneumonia Viral/tratamento farmacológico , Pneumonia Viral/metabolismo , Pneumonia Viral/fisiopatologia , Pneumonia Viral/virologia , SARS-CoV-2 , Tratamento Farmacológico da COVID-19
20.
Behav Ther ; 51(2): 253-267, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32138936

RESUMO

Standardized developmentally based assessment systems have transformed the capacity to identify transdiagnostic behavioral markers of mental disorder risk in early childhood, notably, clinically significant irritability and externalizing behaviors. However, behavior-based instruments that both differentiate risk for persistent psychopathology from normative misbehavior, and are feasible for community clinicians to implement, are in nascent phases of development. Young children's facial expressions during frustration challenges may form the basis for novel assessments tools that are flexible, quick, and easy to implement as markers of psychopathology to complement validated questionnaires. However, the accuracy of facial expressions to correctly classify young children falling above and below clinical cut-offs is unknown. Our goal was to test how facial expressions during frustration, defined by different facial muscular movements, related to individual differences in irritability and externalizing behaviors and discriminated children with clinically significant levels from peers. Participants were 79 children (ages 3-7) who completed a short, moderately frustrating computer task while facial expressions were recorded. Only negative facial expressions that included eye constriction related to irritability and externalizing behaviors and were clinically discriminating. Moreover, these expressions significantly discriminated children with and without clinically significant irritability and externalizing symptoms with high Area Under the Curve (AUC) values (> .75) indicating good clinical utility. In contrast, expressions without eye constriction showed no clinical utility. The presence of negative expressions with eye constriction in response to a short frustration prompt may serve as an indicator of early psychopathology, raising the potential for novel assessment tools that may enhance precision of early identification.


Assuntos
Expressão Facial , Músculos Faciais/fisiopatologia , Humor Irritável/fisiologia , Transtornos Mentais/fisiopatologia , Criança , Pré-Escolar , Feminino , Frustração , Humanos , Masculino , Comportamento Problema
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...