Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Toxicol Appl Pharmacol ; 440: 115922, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35176293

RESUMEN

Although external concentrations are more readily quantified and often used as the metric for regulating and mitigating exposures to environmental chemicals, the toxicological response to an environmental chemical is more directly related to its internal concentrations than the external concentration. The processes of absorption, distribution, metabolism, and excretion (ADME) determine the quantitative relationship between the external and internal concentrations, and these processes are often susceptible to saturation at high concentrations, which can lead to nonlinear changes in internal concentrations that deviate from proportionality. Using generic physiologically-based pharmacokinetic (PBPK) models, we explored how saturable absorption or clearance influence the shape of the internal to external concentration (IEC) relationship. We used the models for hypothetical chemicals to show how differences in kinetic parameters can impact the shape of an IEC relationship; and models for styrene and caffeine to explore how exposure route, frequency, and duration impact the IEC relationships in rat and human exposures. We also analyzed available plasma concentration data for 2,4-dichlorophenoxyacetic acid to demonstrate how a PBPK modeling approach can be an alternative to common statistical methods for analyzing dose proportionality. A PBPK modeling approach can be a valuable tool used in the early stages of a chemical safety assessment program to optimize the design of longer-term animal toxicity studies or to interpret study results.


Asunto(s)
Modelos Biológicos , Animales , Ratas
2.
Regul Toxicol Pharmacol ; 127: 105070, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34718074

RESUMEN

Top dose selection for repeated dose animal studies has generally focused on identification of apical endpoints, use of the limit dose, or determination of a maximum tolerated dose (MTD). The intent is to optimize the ability of toxicity tests performed in a small number of animals to detect effects for hazard identification. An alternative approach, the kinetically derived maximum dose (KMD), has been proposed as a mechanism to integrate toxicokinetic (TK) data into the dose selection process. The approach refers to the dose above which the systemic exposures depart from being proportional to external doses. This non-linear external-internal dose relationship arises from saturation or limitation of TK process(es), such as absorption or metabolism. The importance of TK information is widely acknowledged when assessing human health risks arising from exposures to environmental chemicals, as TK determines the amount of chemical at potential sites of toxicological responses. However, there have been differing opinions and interpretations within the scientific and regulatory communities related to the validity and application of the KMD concept. A multi-stakeholder working group, led by the Health and Environmental Sciences Institute (HESI), was formed to provide an opportunity for impacted stakeholders to address commonly raised scientific and technical issues related to this topic and, more specifically, a weight of evidence approach is recommended to inform design and dose selection for repeated dose animal studies. Commonly raised challenges related to the use of TK data for dose selection are discussed, recommendations are provided, and illustrative case examples are provided to address these challenges or refute misconceptions.


Asunto(s)
Relación Dosis-Respuesta a Droga , Pruebas de Toxicidad/métodos , Toxicocinética , Animales , Pruebas de Carcinogenicidad/métodos , Pruebas de Carcinogenicidad/normas , Dosis Máxima Tolerada , Medición de Riesgo , Pruebas de Toxicidad/normas
3.
Chem Res Toxicol ; 33(1): 223-238, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31532188

RESUMEN

The hepatic risk matrix (HRM) was developed and used to differentiate lead clinical and back-up drug candidates against competitor/marketed drugs within the same pharmaceutical class for their potential to cause human drug-induced liver injury (DILI). The hybrid HRM scoring system blends physicochemical properties (Rule of Two Model: dose and lipophilicity or Partition Model: dose, ionization state, lipophilicity, and fractional carbon bond saturation) with common toxicity mechanisms (cytotoxicity, mitochondrial dysfunction, and bile salt export pump (BSEP) inhibition) that promote DILI. HRM scores are based on bracketed safety margins (<1, 1-10, 10-100, and >100× clinical Cmax,total). On the basis of well-established clinical safety experience of marketed/withdrawn drug candidates, the background analysis consists of 200 drugs from the Liver Toxicity Knowledge Base annotated as Most-DILI- (79), Less-DILI- (56), No-DILI- (47), and Ambiguous-DILI-concern (18) drugs. Scores were generated for over 21 internal and 7 external drug candidates discontinued for unacceptable incidence/magnitude of liver transaminase elevations during clinical trials or withdrawn for liver injury severity. Both hybrid scoring systems identified 70-80% Most-DILI-concern drugs, but more importantly, stratified successful/unsuccessful drug candidates for liver safety (incidence/severity of transaminase elevations and approved drug labels). Incorporating other mechanisms (reactive metabolite and cytotoxic metabolite generation and hepatic efflux transport inhibition, other than BSEP) to the HRM had minimal beneficial impact in DILI prediction/stratification. As is, the hybrid scoring system was positioned for portfolio assessments to contrast DILI risk potential of small molecule drug candidates in early clinical development. This stratified approach for DILI prediction aided decisions regarding drug candidate progression, follow-up mechanistic work, back-up selection, clinical dose selection, and due diligence assessments in favor of compounds with less implied clinical hepatotoxicity risk.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/antagonistas & inhibidores , Animales , Supervivencia Celular , Desarrollo de Medicamentos/métodos , Células Hep G2 , Humanos , Mitocondrias Hepáticas/efectos de los fármacos , Ratas , Medición de Riesgo/métodos
4.
Pharmacol Res Perspect ; 7(6): e00523, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31624633

RESUMEN

Many compounds that appear promising in preclinical species, fail in human clinical trials due to safety concerns. The FDA has strongly encouraged the application of modeling in drug development to improve product safety. This study illustrates how DILIsym, a computational representation of liver injury, was able to reproduce species differences in liver toxicity due to PF-04895162 (ICA-105665). PF-04895162, a drug in development for the treatment of epilepsy, was terminated after transaminase elevations were observed in healthy volunteers (NCT01691274). Liver safety concerns had not been raised in preclinical safety studies. DILIsym, which integrates in vitro data on mechanisms of hepatotoxicity with predicted in vivo liver exposure, reproduced clinical hepatotoxicity and the absence of hepatotoxicity observed in the rat. Simulated differences were multifactorial. Simulated liver exposure was greater in humans than rats. The simulated human hepatotoxicity was demonstrated to be due to the interaction between mitochondrial toxicity and bile acid transporter inhibition; elimination of either mechanism from the simulations abrogated injury. The bile acid contribution occurred despite the fact that the IC50 for bile salt export pump (BSEP) inhibition by PF-04895162 was higher (311 µmol/L) than that has been generally thought to contribute to hepatotoxicity. Modeling even higher PF-04895162 liver exposures than were measured in the rat safety studies aggravated mitochondrial toxicity but did not result in rat hepatotoxicity due to insufficient accumulation of cytotoxic bile acid species. This investigative study highlights the potential for combined in vitro and computational screening methods to identify latent hepatotoxic risks and paves the way for similar and prospective studies.


Asunto(s)
Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/antagonistas & inhibidores , Anticonvulsivantes/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Modelos Biológicos , Quinazolinas/toxicidad , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/metabolismo , Administración Oral , Adolescente , Adulto , Animales , Anticonvulsivantes/administración & dosificación , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Simulación por Computador , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/normas , Epilepsia/tratamiento farmacológico , Células HEK293 , Voluntarios Sanos , Hepatocitos , Humanos , Concentración 50 Inhibidora , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Persona de Mediana Edad , Mitocondrias/efectos de los fármacos , Quinazolinas/administración & dosificación , Ratas , Especificidad de la Especie , Ácido Taurocólico/metabolismo , Adulto Joven
5.
CPT Pharmacometrics Syst Pharmacol ; 8(10): 738-747, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31464379

RESUMEN

Monoclonal antibody (mAb) pharmacokinetics (PK) have largely been predicted via allometric scaling with little consideration for cross-species differences in neonatal Fc receptor (FcRn) affinity or clearance/distribution mechanisms. To address this, we developed a mAb physiologically-based PK model that describes the intracellular trafficking and FcRn recycling of mAbs in a human FcRn transgenic homozygous mouse and human. This model uses mAb-specific in vitro data together with species-specific FcRn tissue expression, tissue volume, and blood-flow physiology to predict mAb in vivo linear PK a priori. The model accurately predicts the terminal half-life of 90% of the mAbs investigated within a twofold error. The mechanistic nature of this model allows us to not only predict linear PK from in vitro data but also explore the PK and target binding of mAbs engineered to have pH-dependent binding to its target or FcRn and could aid in the selection of mAbs with optimal PK and pharmacodynamic properties.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Receptores Fc/genética , Receptores Fc/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Homocigoto , Humanos , Técnicas In Vitro , Modelos Lineales , Ratones , Ratones Transgénicos , Modelos Biológicos , Especificidad de Órganos , Especificidad de la Especie
6.
Clin Pharmacokinet ; 57(4): 491-503, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28653144

RESUMEN

BACKGROUND: Ethnic variability in the pharmacokinetics of organic anion transporting polypeptide (OATP) 1B1 substrates has been observed, but its basis is unclear. A previous study hypothesizes that, without applying an intrinsic ethnic variability in transporter activity, allele frequencies of transporters cannot explain observed ethnic variability in pharmacokinetics. However, this hypothesis contradicts the data collected from compounds that are OATP1B1 substrates but not breast cancer resistance protein (BCRP) substrates. OBJECTIVE: The objective of this study is to evaluate a hypothesis that is physiologically reasonable and more consistent with clinical observations. METHODS: We evaluated if allele frequencies of two transporters (OATP1B1 and BCRP) are key contributors to ethnic variability. In this hypothesis, the same genotype leads to the same activity independent of ethnicity, in contrast to the previous hypothesis of intrinsic ethnic variability in OATP1B1 activity. As a validation, we perform mechanistic pharmacokinetic modeling for SLCO1B1 (encoding OATP1B1) and ABCG2 (encoding BCRP) genotyped pharmacokinetic data from 18 clinical studies with healthy Caucasian and/or Asian subjects. RESULTS: Simulations based on the current hypothesis reasonably describe SLCO1B1 and ABCG2 genotyped pharmacokinetic time course data for five transporter substrates (atorvastatin, pitavastatin, pravastatin, repaglinide, and rosuvastatin) in Caucasian and Asian populations. CONCLUSION: This hypothesis covers the observations that can (e.g., ethnic differences in rosuvastatin pharmacokinetics) or cannot (e.g., lack of differences for pitavastatin pharmacokinetics) be explained by the previous hypothesis. It helps to characterize sources of ethnic variability and provides a foundation for predicting ethnic variability in transporter substrate pharmacokinetics.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Pueblo Asiatico/genética , Frecuencia de los Genes , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Hipoglucemiantes/farmacocinética , Transportador 1 de Anión Orgánico Específico del Hígado/genética , Modelos Biológicos , Proteínas de Neoplasias/genética , Variantes Farmacogenómicas , Población Blanca/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Atorvastatina/administración & dosificación , Atorvastatina/farmacocinética , Carbamatos/administración & dosificación , Carbamatos/farmacocinética , Simulación por Computador , Genotipo , Voluntarios Sanos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Hipoglucemiantes/administración & dosificación , Transportador 1 de Anión Orgánico Específico del Hígado/metabolismo , Proteínas de Neoplasias/metabolismo , Fenotipo , Piperidinas/administración & dosificación , Piperidinas/farmacocinética , Pravastatina/administración & dosificación , Pravastatina/farmacocinética , Quinolinas/administración & dosificación , Quinolinas/farmacocinética , Rosuvastatina Cálcica/administración & dosificación , Rosuvastatina Cálcica/farmacocinética
8.
AAPS J ; 18(3): 746-56, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26951483

RESUMEN

The physiologically based pharmacokinetic (PBPK) model for liver transporter substrates has been established previously and used for predicting drug-drug interactions (DDI) and for clinical practice guidance. So far, nearly all the published PBPK models for liver transporter substrates have one or more hepatic clearance processes (i.e., active uptake, passive diffusion, metabolism, and biliary excretion) estimated by fitting observed systemic data. The estimated hepatic clearance processes are then used to predict liver concentrations and DDI involving either systemic or liver concentration. However, the accuracy and precision of such predictions are unclear. In this study, we try to address this question by using the PBPK model to generate simulated compounds for which we know both systemic and liver profiles. We then developed an approach to assess the accuracy and precision of predicted liver concentration. With hepatic clearance processes estimated using plasma data, model predictions of liver are typically accurate (i.e., true value is bounded by predicted maximum and minimum); however, only for a few compounds are predictions also precise. The results of the current study indicate that extra attention is required when using the current PBPK approach to predict liver concentration and DDI for transporter substrates dependent upon liver concentrations.


Asunto(s)
Hígado/metabolismo , Tasa de Depuración Metabólica/fisiología , Modelos Biológicos , Preparaciones Farmacéuticas/metabolismo , Animales , Humanos , Hígado/efectos de los fármacos , Tasa de Depuración Metabólica/efectos de los fármacos , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/sangre , Ratas , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
9.
Pharm Res ; 33(4): 1003-17, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26718955

RESUMEN

OBJECTIVES: To assess the ability of a previously developed hybrid physiology-based pharmacokinetic-pharmacodynamic (PBPKPD) model in rats to predict the dopamine D2 receptor occupancy (D2RO) in human striatum following administration of antipsychotic drugs. METHODS: A hybrid PBPKPD model, previously developed using information on plasma concentrations, brain exposure and D2RO in rats, was used as the basis for the prediction of D2RO in human. The rat pharmacokinetic and brain physiology parameters were substituted with human population pharmacokinetic parameters and human physiological information. To predict the passive transport across the human blood-brain barrier, apparent permeability values were scaled based on rat and human brain endothelial surface area. Active efflux clearance in brain was scaled from rat to human using both human brain endothelial surface area and MDR1 expression. Binding constants at the D2 receptor were scaled based on the differences between in vitro and in vivo systems of the same species. The predictive power of this physiology-based approach was determined by comparing the D2RO predictions with the observed human D2RO of six antipsychotics at clinically relevant doses. RESULTS: Predicted human D2RO was in good agreement with clinically observed D2RO for five antipsychotics. Models using in vitro information predicted human D2RO well for most of the compounds evaluated in this analysis. However, human D2RO was under-predicted for haloperidol. CONCLUSIONS: The rat hybrid PBPKPD model structure, integrated with in vitro information and human pharmacokinetic and physiological information, constitutes a scientific basis to predict the time course of D2RO in man.


Asunto(s)
Antipsicóticos/farmacología , Antipsicóticos/farmacocinética , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Receptores de Dopamina D2/metabolismo , Esquizofrenia/tratamiento farmacológico , Animales , Antipsicóticos/administración & dosificación , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Antagonistas de los Receptores de Dopamina D2/administración & dosificación , Antagonistas de los Receptores de Dopamina D2/farmacocinética , Antagonistas de los Receptores de Dopamina D2/farmacología , Humanos , Modelos Biológicos , Ratas , Esquizofrenia/metabolismo
10.
Toxicol Sci ; 147(2): 500-14, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26206150

RESUMEN

Severe drug-induced liver injury (DILI) remains a major safety issue due to its frequency of occurrence, idiosyncratic nature, poor prognosis, and diverse underlying mechanisms. Numerous experimental approaches have been published to improve human DILI prediction with modest success. A retrospective analysis of 125 drugs (70 = most-DILI, 55 = no-DILI) from the Food and Drug Administration Liver Toxicity Knowledge Base was used to investigate DILI prediction based on consideration of human exposure alone or in combination with mechanistic assays of hepatotoxic liabilities (cytotoxicity, bile salt export pump inhibition, or mitochondrial inhibition/uncoupling). Using this dataset, human plasma Cmax,total ≥ 1.1 µM alone distinguished most-DILI from no-DILI compounds with high sensitivity/specificity (80/73%). Accounting for human exposure improved the sensitivity/specificity for each assay and helped to derive predictive safety margins. Compounds with plasma Cmax,total ≥ 1.1 µM and triple liabilities had significantly higher odds ratio for DILI than those with single/dual liabilities. Using this approach, a subset of recent pharmaceuticals with evidence of liver injury during clinical development was recognized as potential hepatotoxicants. In summary, plasma Cmax,total ≥ 1.1 µM along with multiple mechanistic liabilities is a major driver for predictions of human DILI potential. In applying this approach during drug development the challenge will be generating accurate estimates of plasma Cmax,total at efficacious doses in advance of generating true exposure data from clinical studies. In the meantime, drug candidates with multiple hepatotoxic liabilities should be deprioritized, since they have the highest likelihood of causing DILI in case their efficacious plasma Cmax,total in humans is higher than anticipated.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Pruebas de Toxicidad , Humanos , Técnicas In Vitro , Concentración 50 Inhibidora , Concentración Máxima Admisible , Estudios Retrospectivos , Pruebas de Toxicidad/estadística & datos numéricos
11.
Front Pharmacol ; 5: 240, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25426072

RESUMEN

Inhibition of the bile salt export pump (BSEP) has been linked to incidence of drug-induced liver injury (DILI), presumably by the accumulation of toxic bile acids in the liver. We have previously constructed and validated a model of bile acid disposition within DILIsym®, a mechanistic model of DILI. In this paper, we use DILIsym® to simulate the DILI response of the hepatotoxic BSEP inhibitors bosentan and CP-724,714 and the non-hepatotoxic BSEP inhibitor telmisartan in humans in order to explore whether we can predict that hepatotoxic BSEP inhibitors can cause bile acid accumulation to reach toxic levels. We also simulate bosentan in rats in order to illuminate potential reasons behind the lack of toxicity in rats compared to the toxicity observed in humans. DILIsym® predicts that bosentan, but not telmisartan, will cause mild hepatocellular ATP decline and serum ALT elevation in a simulated population of humans. The difference in hepatotoxic potential between bosentan and telmisartan is consistent with clinical observations. However, DILIsym® underpredicts the incidence of bosentan toxicity. DILIsym® also predicts that bosentan will not cause toxicity in a simulated population of rats, and that the difference between the response to bosentan in rats and in humans is primarily due to the less toxic bile acid pool in rats. Our simulations also suggest a potential synergistic role for bile acid accumulation and mitochondrial electron transport chain (ETC) inhibition in producing the observed toxicity in CP-724,714, and suggest that CP-724,714 metabolites may also play a role in the observed toxicity. Our work also compares the impact of competitive and noncompetitive BSEP inhibition for CP-724,714 and demonstrates that noncompetitive inhibition leads to much greater bile acid accumulation and potential toxicity. Our research demonstrates the potential for mechanistic modeling to contribute to the understanding of how bile acid transport inhibitors cause DILI.

12.
Drug Metab Dispos ; 42(10): 1646-55, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25092714

RESUMEN

A previously developed physiologically based pharmacokinetic model for hepatic transporter substrates was extended to an organic anion transporting polypeptide substrate, telmisartan. Predictions used in vitro data from sandwich culture human hepatocyte and human liver microsome assays. We have developed a novel method to calibrate partition coefficients (Kps) between nonliver tissues and plasma on the basis of published human positron emission tomography (PET) data to decrease the uncertainty in tissue distribution introduced by in silico-predicted Kps. With in vitro data-predicted hepatic clearances, published empirical scaling factors, and PET-calibrated Kps, the model could accurately recapitulate telmisartan pharmacokinetic (PK) behavior before 2.5 hours. Reasonable predictions also depend on having a model structure that can adequately describe the drug disposition pathways. We showed that the elimination phase (2.5-12 hours) of telmisartan PK could be more accurately recapitulated when enterohepatic recirculation of parent compound derived from intestinal deconjugation of glucuronide metabolite was incorporated into the model. This study demonstrated the usefulness of the previously proposed physiologically based modeling approach for purely predictive intravenous PK simulation and identified additional biologic processes that can be important in prediction.


Asunto(s)
Bencimidazoles/farmacocinética , Benzoatos/farmacocinética , Hepatocitos/metabolismo , Microsomas Hepáticos/metabolismo , Simulación por Computador , Humanos , Modelos Biológicos , Transportadores de Anión Orgánico/metabolismo , Telmisartán
13.
Clin Pharmacokinet ; 53(8): 659-78, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25056496

RESUMEN

Hepatobiliary transport mechanisms have been identified to play a significant role in determining the systemic clearance for a number of widely prescribed drugs and an increasing number of new molecular entities (NMEs). While determining the pharmacokinetics, drug transporters also regulate the target tissue exposure and play a key role in regulating the pharmacological and/or toxicological responses. Consequently, it is of great relevance in drug discovery and development to assess hepatic transporter activity in regard to pharmacokinetic and dose predictions and to evaluate pharmacokinetic variability associated with drug-drug interactions (DDIs) and genetic variants. Mechanistic predictions utilizing physiological-based pharmacokinetic modeling are increasingly used to evaluate transporter contribution and delineate the transporter-enzyme interplay on the basis of hypothesis-driven functional in vitro findings. Significant strides were made in the development of in vitro techniques to facilitate characterization of hepatobiliary transport. However, challenges exist in the quantitative in vitro-in vivo extrapolation of transporter kinetics due to the lack of information on absolute abundance of the transporter in both in vitro and in vivo situations, and/or differential function in the holistic in vitro reagents such as suspended and plated hepatocytes systems, and lack of complete mechanistic understanding of liver model structure. On the other hand, models to predict transporter-mediated DDIs range from basic models to mechanistic static and dynamic models. While basic models provide conservative estimates and are useful upfront in avoiding false negative predictions, mechanistic models integrate multiple victim and perpetrator drugs parameters and are expected to provide quantitative predictions. The aim of this paper is to review the current state of the model-based approaches to predict clinical pharmacokinetics and DDIs of drugs or NMEs that are substrates of hepatic transporters.


Asunto(s)
Descubrimiento de Drogas/métodos , Hígado/efectos de los fármacos , Proteínas de Transporte de Membrana/metabolismo , Modelos Biológicos , Farmacocinética , Transporte Biológico , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Humanos , Hígado/metabolismo , Tasa de Depuración Metabólica , Preparaciones Farmacéuticas/sangre , Preparaciones Farmacéuticas/metabolismo , Valor Predictivo de las Pruebas , Factores de Tiempo , Distribución Tisular
14.
Pharm Res ; 31(10): 2605-17, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24792824

RESUMEN

OBJECTIVES: Dopamine D2 receptor occupancy (D2RO) is the major determinant of efficacy and safety in schizophrenia drug therapy. Excessive D2RO (>80%) is known to cause catalepsy (CAT) in rats and extrapyramidal side effects (EPS) in human. The objective of this study was to use pharmacokinetic and pharmacodynamic modeling tools to relate CAT with D2RO in rats and to compare that with the relationship between D2RO and EPS in humans. METHODS: Severity of CAT was assessed in rats at hourly intervals over a period of 8 h after antipsychotic drug treatment. An indirect response model with and without Markov elements was used to explain the relationship of D2RO and CAT. RESULTS: Both models explained the CAT data well for olanzapine, paliperidone and risperidone. However, only the model with the Markov elements predicted the CAT severity well for clozapine and haloperidol. The relationship between CAT scores in rat and EPS scores in humans was implemented in a quantitative manner. Risk of EPS not exceeding 10% over placebo correlates with less than 86% D2RO and less than 30% probability of CAT events in rats. CONCLUSION: A quantitative relationship between rat CAT and human EPS was elucidated and may be used in drug discovery to predict the risk of EPS in humans from D2RO and CAT scores measured in rats.


Asunto(s)
Antipsicóticos , Catalepsia/metabolismo , Antagonistas de los Receptores de Dopamina D2 , Modelos Biológicos , Receptores de Dopamina D2/metabolismo , Animales , Antipsicóticos/efectos adversos , Antipsicóticos/farmacocinética , Antipsicóticos/farmacología , Benzodiazepinas/efectos adversos , Benzodiazepinas/farmacocinética , Benzodiazepinas/farmacología , Encéfalo/metabolismo , Catalepsia/etiología , Simulación por Computador , Antagonistas de los Receptores de Dopamina D2/efectos adversos , Antagonistas de los Receptores de Dopamina D2/farmacocinética , Antagonistas de los Receptores de Dopamina D2/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Isoxazoles/efectos adversos , Isoxazoles/farmacocinética , Isoxazoles/farmacología , Cadenas de Markov , Olanzapina , Palmitato de Paliperidona , Pirimidinas/efectos adversos , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Ratas , Risperidona/efectos adversos , Risperidona/farmacocinética , Risperidona/farmacología , Índice de Severidad de la Enfermedad
15.
J Pharmacokinet Pharmacodyn ; 41(3): 197-209, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24718648

RESUMEN

Physiologically based pharmacokinetic (PBPK) models provide a framework useful for generating credible human pharmacokinetic predictions from data available at the earliest, preclinical stages of pharmaceutical research. With this approach, the pharmacokinetic implications of in vitro data are contextualized via scaling according to independent physiological information. However, in many cases these models also require model-based estimation of additional empirical scaling factors (SFs) in order to accurately recapitulate known human pharmacokinetic behavior. While this practice clearly improves data characterization, the introduction of empirically derived SFs may belie the extrapolative power commonly attributed to PBPK. This is particularly true when such SFs are compound dependent and/or when there are issues with regard to identifiability. As such, when empirically-derived SFs are necessary, a critical evaluation of parameter estimation and model structure are prudent. In this study, we applied a global optimization method to support model-based estimation of a single set of empirical SFs from intravenous clinical data on seven OATP substrates within the context of a previously published PBPK model as well as a revised PBPK model. The revised model with experimentally measured unbound fraction in liver, permeability between liver compartments, and permeability limited distribution to selected tissues improved data characterization. We utilized large-sample approximation and resampling approaches to estimate confidence intervals for the revised model in support of forward predictions that reflect the derived uncertainty. This work illustrates an objective approach to estimating empirically-derived SFs, systematically refining PBPK model performance and conveying the associated confidence in subsequent forward predictions.


Asunto(s)
Transportadores de Anión Orgánico/metabolismo , Farmacocinética , Algoritmos , Células Cultivadas , Intervalos de Confianza , Hepatocitos/metabolismo , Humanos , Modelos Estadísticos
16.
J Pharmacokinet Pharmacodyn ; 40(5): 557-71, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23933716

RESUMEN

Objectives of the present investigation were: (1) to compare three literature reported tumor growth inhibition (TGI) pharmacodynamic (PD) models and propose an optimal new model that best describes the xenograft TGI data for antibody drug conjugates (ADC), (2) to translate efficacy of the ADC Trastuzumab-emtansine (T-DM1) from mice to patients using the optimized PD model, and (3) to apply the translational strategy to predict clinically efficacious concentrations of a novel in-house anti-5T4 ADC, A1mcMMAF. First, the performance of all four of the PD models (i.e. 3 literature reported + 1 proposed) was evaluated using TGI data of T-DM1 obtained from four different xenografts. Based on the estimates of the pharmacodynamic/pharmacokinetic (PK/PD) modeling, a secondary parameter representing the efficacy index of the drug was calculated, which is termed as the tumor static concentration (TSC). TSC values derived from all four of the models were compared with each other, and with literature reported values, to assess the performance of these models. Subsequently, using the optimized PK/PD model, PD parameters obtained from different cell lines, human PK, and the proposed translational strategy, clinically efficacious doses of T-DM1 were projected. The accuracy of projected efficacious dose range for T-DM1 was verified by comparison with the clinical doses. Aforementioned strategy was then applied to A1mcMMAF for projecting its efficacious concentrations in clinic. TSC values for A1mcMMAF, obtained by fitting TGI data from 4 different xenografts with the proposed PK/PD model, were estimated to range from 0.6 to 11.5 µg mL⁻¹. Accordingly, the clinically efficacious doses for A1mcMMAF were projected retrospectively. All in all, the improved PD model and proposed translational strategy presented here suggest that appropriate correction for the clinical exposure and employing the TSC criterion can help translate mouse TGI data to predict first in human doses of ADCs.


Asunto(s)
Anticuerpos/farmacología , Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Inmunoconjugados/farmacología , Inmunoconjugados/farmacocinética , Neoplasias Experimentales/tratamiento farmacológico , Ado-Trastuzumab Emtansina , Animales , Anticuerpos Monoclonales Humanizados/farmacocinética , Anticuerpos Monoclonales Humanizados/farmacología , Línea Celular Tumoral , Femenino , Humanos , Maitansina/análogos & derivados , Maitansina/farmacocinética , Maitansina/farmacología , Ratones , Ratones Desnudos , Trastuzumab , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
17.
Regul Toxicol Pharmacol ; 66(1): 116-29, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23535119

RESUMEN

The World Health Organization (WHO) International Programme on Chemical Safety (IPCS) Guidance on Characterization and Application of Physiologically Based Pharmacokinetic Models in Risk Assessment (IPCS, 2010) describes key principles for risk assessors and model developers. In the WHO Guidance, a template for model documentation was developed and a case study included. Here the WHO Guidance, including the template, is summarized and an additional case study is presented to illustrate its application, based upon an existing risk assessment for 2-butoxyethanol (CAS NO. 111-76-2). The goal of the WHO Guidance and the current paper is to increase regulatory acceptance of complex biologically descriptive pharmacokinetic (or toxicokinetic) models, such as PBPK models, by facilitating communication and successful interaction between modelers and risk assessors.


Asunto(s)
Glicoles de Etileno/toxicidad , Modelos Biológicos , Medición de Riesgo/métodos , Animales , Glicoles de Etileno/farmacocinética , Sustancias Peligrosas/farmacocinética , Sustancias Peligrosas/toxicidad , Humanos , Cooperación Internacional , Organización Mundial de la Salud
18.
Expert Opin Drug Metab Toxicol ; 9(4): 459-72, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23331046

RESUMEN

INTRODUCTION: Membrane transporters have been recognized to play a key role in determining the absorption, distribution and elimination processes of drugs. The organic anion-transporting polypeptide (OATP)1B1 and OATP1B3 isoforms are selectively expressed in the human liver and are known to cause significant drug-drug interactions (DDIs), as observed with an increasing number of drugs. It is evident that DDIs involving hepatic transporters are capable of altering systemic, as well as tissue-specific, exposure of drug substrates resulting in marked differences in drug safety and/or efficacy. It is therefore essential to quantitatively predict such interactions early in the drug development to mitigate clinical risks. AREAS COVERED: The role of hepatic uptake transporters in drug disposition and clinical DDIs has been reviewed with an emphasis on the current state of the models applicable for quantitative predictions. The readers will also gain insight into the in vitro experimental tools available to characterize transport kinetics, while appreciating the knowledge gaps in the in vitro-in vivo extrapolation (IVIVE), which warrant further investigation. EXPERT OPINION: Static and dynamic models can be convincingly applied to quantitatively predict drug interactions, early in drug discovery, to mitigate clinical risks as well as to avoid unnecessary clinical studies. Compared to basic models, which focus on individual processes, mechanistic models provide the ability to assess DDI potential for compounds with systemic disposition determined by both transporters and metabolic enzymes. However, complexities in the experimental tools and an apparent disconnect in the IVIVE of transport kinetics have limited the physiologically based pharmacokinetic modeling strategies. Emerging data on the expression of transporter proteins and tissue drug concentrations are expected to help bridge these gaps. In addition, detailed characterization of substrate kinetics can facilitate building comprehensive mechanistic models.


Asunto(s)
Hígado/citología , Hígado/efectos de los fármacos , Proteínas de Transporte de Membrana/efectos de los fármacos , Línea Celular , Fenómenos Químicos , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Fluorobencenos/farmacocinética , Fluorobencenos/uso terapéutico , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Cinética , Hígado/metabolismo , Modelos Teóricos , Pravastatina/farmacocinética , Pravastatina/uso terapéutico , Pirimidinas/farmacocinética , Pirimidinas/uso terapéutico , Rosuvastatina Cálcica , Sulfonamidas/farmacocinética , Sulfonamidas/uso terapéutico
19.
Neurodegener Dis ; 12(1): 36-50, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22922480

RESUMEN

BACKGROUND: Reducing brain ß-amyloid (Aß) via inhibition of ß-secretase, or inhibition/modulation of γ-secretase, has been widely pursued as a potential disease-modifying treatment for Alzheimer's disease. Compounds that act through these mechanisms have been screened and characterized with Aß lowering in the brain and/or cerebrospinal fluid (CSF) as the primary pharmacological end point. Interpretation and translation of the pharmacokinetic (PK)/pharmacodynamic (PD) relationship for these compounds is complicated by the relatively slow Aß turnover process in these compartments. OBJECTIVE: To understand Aß turnover kinetics in preclinical species and humans. METHODS: We collected CSF Aß dynamic data after ß- or γ-secretase inhibitor treatment from in-house experiments and the public domain, and analyzed the data using PK/PD modeling to obtain CSF Aß turnover rates (kout) in the mouse, dog, monkey and human. RESULTS: The kout for CSF Aß40 follows allometry (kout = 0.395 × body weight(-0.351)). The kout for CSF Aß40 is approximately 2-fold higher than the turnover of CSF in rodents, but in higher species, the two are comparable. CONCLUSION: The turnover of CSF Aß40 was systematically examined, for the first time, in multiple species through quantitative modeling of multiple data sets. Our result suggests that the clearance mechanisms for CSF Aß in rodents may be different from those in the higher species. The understanding of Aß turnover has considerable implications for the discovery and development of Aß-lowering therapeutics, as illustrated from the perspectives of preclinical PK/PD characterization and preclinical-to-clinical translation.


Asunto(s)
Péptidos beta-Amiloides/líquido cefalorraquídeo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/sangre , Animales , Perros , Humanos , Macaca fascicularis , Ratones , Oligopéptidos/farmacología , Oxadiazoles/farmacología , Pirimidinas/farmacología , Sulfonamidas/farmacología , Tiazinas/farmacología
20.
PLoS One ; 7(9): e44359, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22970204

RESUMEN

A systems-level mathematical model is presented that describes the effects of inhibiting the enzyme 5α-reductase (5aR) on the ventral prostate of the adult male rat under chronic administration of the 5aR inhibitor, finasteride. 5aR is essential for androgen regulation in males, both in normal conditions and disease states. The hormone kinetics and downstream effects on reproductive organs associated with perturbing androgen regulation are complex and not necessarily intuitive. Inhibition of 5aR decreases the metabolism of testosterone (T) to the potent androgen 5α-dihydrotestosterone (DHT). This results in decreased cell proliferation, fluid production and 5aR expression as well as increased apoptosis in the ventral prostate. These regulatory changes collectively result in decreased prostate size and function, which can be beneficial to men suffering from benign prostatic hyperplasia (BPH) and could play a role in prostate cancer. There are two distinct isoforms of 5aR in male humans and rats, and thus developing a 5aR inhibitor is a challenging pursuit. Several inhibitors are on the market for treatment of BPH, including finasteride and dutasteride. In this effort, comparisons of simulated vs. experimental T and DHT levels and prostate size are depicted, demonstrating the model accurately described an approximate 77% decrease in prostate size and nearly complete depletion of prostatic DHT following 21 days of daily finasteride dosing in rats. This implies T alone is not capable of maintaining a normal prostate size. Further model analysis suggests the possibility of alternative dosing strategies resulting in similar or greater effects on prostate size, due to complex kinetics between T, DHT and gene occupancy. With appropriate scaling and parameterization for humans, this model provides a multiscale modeling platform for drug discovery teams to test and generate hypotheses about drugging strategies for indications like BPH and prostate cancer, such as compound binding properties, dosing regimens, and target validation.


Asunto(s)
Inhibidores de 5-alfa-Reductasa/farmacología , Colestenona 5 alfa-Reductasa/metabolismo , Modelos Biológicos , Próstata/efectos de los fármacos , Próstata/enzimología , Inhibidores de 5-alfa-Reductasa/administración & dosificación , Animales , Calibración , Dihidrotestosterona/metabolismo , Relación Dosis-Respuesta a Droga , Finasterida/administración & dosificación , Finasterida/farmacocinética , Finasterida/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Cinética , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Tamaño de los Órganos/efectos de los fármacos , Tamaño de los Órganos/genética , Próstata/metabolismo , Ratas , Testosterona/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...