Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
CPT Pharmacometrics Syst Pharmacol ; 13(1): 118-131, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37833845

ABSTRACT

Hepatic impairment (HI) moderately (<5-fold) affects the systemic exposure (i.e., area under the plasma concentration-time curve [AUC]) of drugs that are substrates of the hepatic sinusoidal organic anion transporting polypeptide (OATP) transporters and are excreted unchanged in the bile and/or urine. However, the effect of HI on their AUC is much greater (>10-fold) for drugs that are also substrates of cytochrome P450 (CYP) 3A enzymes. Using the extended clearance model, through simulations, we identified the ratio of sinusoidal efflux clearance (CL) over the sum of metabolic and biliary CLs as important in predicting the impact of HI on the AUC of dual OATP/CYP3A substrates. Because HI may reduce hepatic CYP3A-mediated CL to a greater extent than biliary efflux CL, the greater the contribution of the former versus the latter, the greater the impact of HI on drug AUC ratio (AUCRHI ). Using physiologically-based pharmacokinetic modeling and simulation, we predicted relatively well the AUCRHI of OATP substrates that are not significantly metabolized (pitavastatin, rosuvastatin, valsartan, and gadoxetic acid). However, there was a trend toward underprediction of the AUCRHI of the dual OATP/CYP3A4 substrates fimasartan and atorvastatin. These predictions improved when the sinusoidal efflux CL of these two drugs was increased in healthy volunteers (i.e., before incorporating the effect of HI), and by modifying the directionality of its modulation by HI (i.e., increase or decrease). To accurately predict the effect of HI on AUC of hepatobiliary cleared drugs it is important to accurately predict all hepatobiliary pathways, including sinusoidal efflux CL.


Subject(s)
Cytochrome P-450 CYP3A , Organic Anion Transporters , Humans , Cytochrome P-450 CYP3A/metabolism , Liver/metabolism , Biological Transport , Rosuvastatin Calcium , Organic Anion Transporters/metabolism , Drug Interactions
2.
CPT Pharmacometrics Syst Pharmacol ; 12(2): 261-273, 2023 02.
Article in English | MEDLINE | ID: mdl-36540952

ABSTRACT

Physiologically based pharmacokinetic models, populated with drug-metabolizing enzyme and transporter (DMET) abundance, can be used to predict the impact of hepatic impairment (HI) on the pharmacokinetics (PK) of drugs. To increase confidence in the predictive power of such models, they must be validated by comparing the predicted and observed PK of drugs in HI obtained by phenotyping (or probe drug) studies. Therefore, we first predicted the effect of all stages of HI (mild to severe) on the PK of drugs primarily metabolized by cytochrome P450 (CYP) 3A enzymes using the default HI module of Simcyp Version 21, populated with hepatic and intestinal CYP3A abundance data. Then, we validated the predictions using CYP3A probe drug phenotyping studies conducted in HI. Seven CYP3A substrates, metabolized primarily via CYP3A (fraction metabolized, 0.7-0.95), with low to high hepatic availability, were studied. For all stages of HI, the predicted PK parameters of drugs were within twofold of the observed data. This successful validation increases confidence in using the DMET abundance data in HI to predict the changes in the PK of drugs cleared by DMET for which phenotyping studies in HI are not available or cannot be conducted. In addition, using CYP3A drugs as an example, through simulations, we identified the salient PK factors that drive the major changes in exposure (area under the plasma concentration-time profile curve) to drugs in HI. This theoretical framework can be applied to any drug and DMET to quickly determine the likely magnitude of change in drug PK due to HI.


Subject(s)
Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System , Humans , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Computer Simulation , Cytochrome P-450 Enzyme System/metabolism , Liver/metabolism , Models, Biological
3.
Pharmacol Ther ; 238: 108271, 2022 10.
Article in English | MEDLINE | ID: mdl-36002079

ABSTRACT

Predicting transporter-based drug clearance (CL) and tissue concentrations (TC) in humans is important to reduce the risk of failure during drug development. In addition, when transporters are present at the tissue:blood interface (e.g., in the liver, blood-brain barrier), predicting TC is important to predict the drug's efficacy and safety. With the advent of quantitative targeted proteomics, in vitro to in vivo extrapolation (IVIVE) of transporter-based drug CL and TC is now possible using transporter-expressing models (cells lines, membrane vesicles) and the in vivo to in vitro relative expression of transporters (REF) as a scaling factor. Unlike other approaches based on physiological scaling, the REF approach is not dependent on the availability of primary cells. Here, we review the REF approach and compare it with other IVIVE approaches such as the relative activity factor approach and physiological scaling. For each of these scaling approaches, we review their underlying principles, assumptions, methodology, predictive performance, as well as advantages and limitations. Finally, we discuss current gaps in IVIVE of transporter-based CL and TC and propose possible reasons for these gaps as well as areas to investigate to bridge these gaps.


Subject(s)
Membrane Transport Proteins , Models, Biological , Biological Transport , Drug Interactions , Humans , Liver/metabolism , Membrane Transport Proteins/metabolism , Metabolic Clearance Rate
4.
Clin Pharmacol Ther ; 110(1): 108-122, 2021 07.
Article in English | MEDLINE | ID: mdl-33759449

ABSTRACT

Numerous drugs are currently under accelerated clinical investigation for the treatment of coronavirus disease 2019 (COVID-19); however, well-established safety and efficacy data for these drugs are limited. The goal of this study was to predict the potential of 25 small molecule drugs in clinical trials for COVID-19 to cause clinically relevant drug-drug interactions (DDIs), which could lead to potential adverse drug reactions (ADRs) with the use of concomitant medications. We focused on 11 transporters, which are targets for DDIs. In vitro potency studies in membrane vesicles or HEK293 cells expressing the transporters coupled with DDI risk assessment methods revealed that 20 of the 25 drugs met the criteria from regulatory authorities to trigger consideration of a DDI clinical trial. Analyses of real-world data from electronic health records, including a database representing nearly 120,000 patients with COVID-19, were consistent with several of the drugs causing transporter-mediated DDIs (e.g., sildenafil, chloroquine, and hydroxychloroquine). This study suggests that patients with COVID-19, who are often older and on various concomitant medications, should be carefully monitored for ADRs. Future clinical studies are needed to determine whether the drugs that are predicted to inhibit transporters at clinically relevant concentrations, actually result in DDIs.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , COVID-19 , Drug Interactions , Drug-Related Side Effects and Adverse Reactions , Membrane Transport Proteins/metabolism , Virus Internalization/drug effects , Virus Replication/drug effects , Antiviral Agents/pharmacokinetics , COVID-19/virology , Clinical Trials as Topic , Drug Monitoring/methods , Drug Monitoring/standards , Drug-Related Side Effects and Adverse Reactions/etiology , Drug-Related Side Effects and Adverse Reactions/metabolism , Drug-Related Side Effects and Adverse Reactions/prevention & control , Electronic Health Records/statistics & numerical data , HEK293 Cells , Humans , Hydroxychloroquine/pharmacokinetics , Risk Assessment/methods , SARS-CoV-2/drug effects , SARS-CoV-2/physiology
5.
AAPS J ; 23(2): 35, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33649974

ABSTRACT

A rare cause of megaloblastic anemia (MA) is thiamine-responsive megaloblastic anemia (TRMA), a genetic disorder caused by mutations in SLC19A2 (encoding THTR1), a thiamine transporter. The study objectives were to (1) functionally characterize selected TRMA-associated SLC19A2 variants and (2) determine whether current prescription drugs associated with drug-induced MA (DIMA) may act via inhibition of SLC19A2. Functional characterization of selected SLC19A2 variants was performed by confocal microscopy and isotopic uptake studies of [3H]-thiamine in HEK293 cells. Sixty-three drugs associated with DIMA were screened for SLC19A2 inhibition in isotopic uptake studies. Three previously uncharacterized SLC19A2 variants identified in TRMA patients exhibited disrupted localization to the plasma membrane along with near-complete loss-of-function. Ten of 63 drugs inhibited SLC19A2-mediated thiamine transport ≥ 50% at screening concentrations; however, with the exception of erythromycin, none was predicted to inhibit SLC19A2 at clinically relevant unbound plasma concentrations. Data from electronic health records revealed reduced levels of thiamine pyrophosphate (TPP) in patients prescribed erythromycin, consistent with inhibition of SLC19A2-mediated thiamine transport. Here, we confirmed the role of three SLC19A2 variants in TRMA pathology. Additionally, we report that inhibition of SLC19A2 is a potential, but uncommon mechanism for DIMA.


Subject(s)
Anemia, Megaloblastic/genetics , Diabetes Mellitus/genetics , Erythromycin/adverse effects , Hearing Loss, Sensorineural/genetics , Membrane Transport Proteins/genetics , Thiamine Deficiency/congenital , Thiamine Pyrophosphate/antagonists & inhibitors , Adult , Anemia, Megaloblastic/blood , Anemia, Megaloblastic/chemically induced , Cell Membrane/metabolism , Diabetes Mellitus/blood , Diabetes Mellitus/chemically induced , Drug Interactions , Erythromycin/pharmacokinetics , Female , Genetic Variation , HEK293 Cells , Hearing Loss, Sensorineural/blood , Hearing Loss, Sensorineural/chemically induced , Humans , Loss of Function Mutation , Male , Membrane Transport Proteins/metabolism , Thiamine Deficiency/blood , Thiamine Deficiency/chemically induced , Thiamine Deficiency/genetics , Thiamine Pyrophosphate/blood , Thiamine Pyrophosphate/metabolism
6.
Hum Mutat ; 40(7): 983-995, 2019 07.
Article in English | MEDLINE | ID: mdl-30950137

ABSTRACT

Deleterious variants in SLC2A2 cause Fanconi-Bickel Syndrome (FBS), a glycogen storage disorder, whereas less common variants in SLC2A2 associate with numerous metabolic diseases. Phenotypic heterogeneity in FBS has been observed, but its causes remain unknown. Our goal was to functionally characterize rare SLC2A2 variants found in FBS and metabolic disease-associated variants to understand the impact of these variants on GLUT2 activity and expression and establish genotype-phenotype correlations. Complementary RNA-injected Xenopus laevis oocytes were used to study mutant transporter activity and membrane expression. GLUT2 homology models were constructed for mutation analysis using GLUT1, GLUT3, and XylE as templates. Seventeen FBS variants were characterized. Only c.457_462delCTTATA (p.Leu153_Ile154del) exhibited residual glucose uptake. Functional characterization revealed that only half of the variants were expressed on the plasma membrane. Most less common variants (except c.593 C>A (p.Thr198Lys) and c.1087 G>T (p.Ala363Ser)) exhibited similar GLUT2 transport activity as the wild type. Structural analysis of GLUT2 revealed that variants affect substrate-binding, steric hindrance, or overall transporter structure. The mutant transporter that is associated with a milder FBS phenotype, p.Leu153_Ile154del, retained transport activity. These results improve our overall understanding of the underlying causes of FBS and impact of GLUT2 function on various clinical phenotypes ranging from rare to common disease.


Subject(s)
Fanconi Syndrome/genetics , Glucose Transporter Type 2/chemistry , Glucose Transporter Type 2/metabolism , Mutation , Animals , Binding Sites , Cell Membrane/metabolism , Fanconi Syndrome/metabolism , Female , Genetic Association Studies , Glucose/metabolism , Glucose Transporter Type 2/genetics , Humans , Models, Molecular , Oocytes/metabolism , Xenopus
7.
Clin Pharmacol Ther ; 106(3): 623-631, 2019 09.
Article in English | MEDLINE | ID: mdl-30924126

ABSTRACT

Allopurinol, which lowers uric acid (UA) concentration, is increasingly being recognized for its benefits in cardiovascular and renal disease. However, response to allopurinol is variable. We gathered samples from 4,446 multiethnic subjects for a genome-wide association study of allopurinol response. Consistent with previous studies, we observed that the Q141K variant in ABCG2 (rs2231142), which encodes the efflux pump breast cancer resistance protein (BCRP), associated with worse response to allopurinol. However, for the first time this association reached genome-wide level significance (P = 8.06 × 10-11 ). Additionally, we identified a novel association with a variant in GREM2 (rs1934341, P = 3.22 × 10-6 ). In vitro studies identified oxypurinol, the active metabolite of allopurinol, as an inhibitor of the UA transporter GLUT9, suggesting that oxypurinol may modulate UA reabsorption. These results provide strong evidence for a role of BCRP Q141K in allopurinol response, and suggest that allopurinol may have additional hypouricemic effects beyond xanthine oxidase inhibition.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Allopurinol/pharmacology , Neoplasm Proteins/genetics , Uric Acid/metabolism , Aged , Aged, 80 and over , Cytokines/genetics , Ethnicity , Female , Genome-Wide Association Study , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Humans , Male , Middle Aged , Oxypurinol/pharmacology , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL