Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Drug Metab Dispos ; 52(2): 95-105, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38071533

ABSTRACT

To facilitate the design of drugs readily able to cross the blood brain barrier (BBB), a Madin-Darby canine kidney (MDCK) cell line was established that over expresses both P-glycoprotein (Pgp) and breast cancer resistance protein (BCRP), the main human efflux transporters of the BBB. Proteomics analyses indicate BCRP is expressed at a higher level than Pgp in this cell line. This cell line shows good activity for both transporters [BCRP substrate dantrolene efflux ratio (ER) 16.3 ± 0.9, Pgp substrate quinidine ER 27.5 ± 1.2], and use of selective transporter inhibitors enables an assessment of the relative contributions to overall ERs. The MDCKII-MDR1-BCRP ER negatively correlates with rat unbound brain/unbound plasma ratio, Kpuu Highly brain penetrant compounds with rat Kpuu ≥ 0.3 show ERs ≤ 2 in the MDCKII-MDR1-BCRP assay while compounds predominantly excluded from the brain, Kpuu ≤ 0.05, demonstrate ERs ≥ 20. A subset of compounds with MDCKII-MDR1-BCRP ER < 2 and rat Kpuu < 0.3 were shown to be substrates of rat Pgp using a rat transfected cell line, MDCKII-rMdr1a. These compounds also showed ERs > 2 in the human National Institutes of Health (NIH) MDCKI-MDR1 (high Pgp expression) cell line, which suggests that they are weak human Pgp substrates. Characterization of 37 drugs targeting the central nervous system in the MDCKII-MDR1-BCRP efflux assay show 36 have ERs < 2. In drug discovery, use of the MDCKII-MDR1-BCRP in parallel with the NIH MDCKI-MDR1 cell line is useful for identification of compounds with high brain penetration. SIGNIFICANCE STATEMENT: A single cell line that includes both the major human efflux transporters of the blood brain barrier (MDCKII-MDR1-BCRP) has been established facilitating the rapid identification of efflux substrates and enabling the design of brain penetrant molecules. Efflux ratios using this cell line demonstrate a clear relationship with brain penetration as defined by rat brain Kpuu.


Subject(s)
Blood-Brain Barrier , Neoplasm Proteins , Humans , Animals , Dogs , Rats , Blood-Brain Barrier/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Neoplasm Proteins/metabolism , Membrane Transport Proteins/metabolism , Cell Line , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism
2.
AAPS J ; 25(4): 66, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37380821

ABSTRACT

Capturing human equivalent drug exposures preclinically is a key challenge in the translational process. Motivated by the need to recapitulate the pharmacokinetic (PK) profile of the clinical stage Mcl-1 inhibitor AZD5991 in mice, we describe the methodology used to develop a refined mathematical model relating clinically relevant concentration profiles to efficacy. Administration routes were explored to achieve target exposures matching the clinical exposure of AZD5991. Intravenous infusion using vascular access button (VAB) technology was found to best reproduce clinical target exposures of AZD5991 in mice. Exposure-efficacy relationships were evaluated, demonstrating that dissimilar PK profiles result in differences in target engagement and efficacy outcomes. Thus, these data underscore the importance of accurately ascribing key PK metrics in the translational process to enable clinically meaningful predictions of efficacy.


Subject(s)
Macrocyclic Compounds , Humans , Animals , Mice , Disease Models, Animal , Medical Oncology , Technology
3.
PLoS One ; 18(5): e0285798, 2023.
Article in English | MEDLINE | ID: mdl-37195991

ABSTRACT

The exposure of a dendritic nanoparticle and its conjugated active pharmaceutical ingredient (API) was determined in mouse, rat and dog, with the aim of investigating interspecies differences facilitating clinical translation. Plasma area under the curves (AUCs) were found to be dose proportional across species, while dose normalized concentration time course profiles in plasma, liver and spleen were superimposable in mouse, rat and dog. A physiologically based pharmacokinetic (PBPK) model, previously developed for mouse, was evaluated as a suitable framework to prospectively capture concentration dynamics in rat and dog. The PBPK model, parameterized either by considering species-specific physiology or using alternate scaling methods such as allometry, was shown to capture exposure profiles across species. A sensitivity analysis highlighted API systemic clearance as a key parameter influencing released API levels. The PBPK model was utilized to simulate human exposure profiles, which overlaid dose-normalized data from mouse, rat and dog. The consistency in measured interspecies exposures as well as the capability of the PBPK model to simulate observed dynamics support its use as a powerful translational tool.


Subject(s)
Models, Biological , Nanoparticles , Rats , Mice , Humans , Animals , Dogs , Tissue Distribution , Area Under Curve , Liver
4.
Drug Metab Dispos ; 51(7): 892-901, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37041083

ABSTRACT

Candidate drugs may exhibit higher unbound intrinsic clearances (CLint,u) in human liver microsomes (HLMs) relative to human hepatocytes (HHs), posing a challenge as to which value is more predictive of in vivo clearance (CL). This work was aimed at better understanding the mechanism(s) underlying this 'HLM:HH disconnect' via examination of previous explanations, including passive permeability limited CL or cofactor exhaustion in hepatocytes. A series of structurally related, passively permeable (Papps > 5 × 10-6 cm/s), 5-azaquinazolines were studied in different liver fractions, and metabolic rates and routes were determined. A subset of these compounds demonstrated a significant HLM:HH (CLint,u ratio 2-26) disconnect. Compounds were metabolized via combinations of liver cytosol aldehyde oxidase (AO), microsomal cytochrome P450 (CYP) and flavin monooxygenase (FMO). For this series, the lack of concordance between CLint,u determined in HLM and HH contrasted with an excellent correlation of AO dependent CLint,u determined in human liver cytosol[Formula: see text], r2 = 0.95, P < 0.0001). The HLM:HH disconnect for both 5-azaquinazolines and midazolam was as a result of significantly higher CYP activity in HLM and lysed HH fortified with exogenous NADPH relative to intact HH. Moreover, for the 5-azaquinazolines, the maintenance of cytosolic AO and NADPH-dependent FMO activity in HH, relative to CYP, supports the conclusion that neither substrate permeability nor intracellular NADPH for hepatocytes were limiting CLint,u Further studies are required to identify the underlying cause of the lower CYP activities in HH relative to HLM and lysed hepatocytes in the presence of exogenous NADPH. SIGNIFICANCE STATEMENT: Candidate drugs may exhibit higher intrinsic clearance in human liver microsomes relative to human hepatocytes, posing a challenge as to which value is predictive of in vivo clearance. This work demonstrates that the difference in activity determined in liver fractions results from divergent cytochrome P450 but not aldehyde oxidase or flavin monooxygenase activity. This is inconsistent with explanations including substrate permeability limitations or cofactor exhaustion and should inform the focus of further studies to understand this cytochrome P450 specific disconnect phenomenon.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Microsomes, Liver , Humans , Microsomes, Liver/metabolism , NADP/metabolism , Hepatocytes/metabolism , Cytochrome P-450 Enzyme System/metabolism , Aryl Hydrocarbon Hydroxylases/metabolism , Metabolic Clearance Rate
5.
Clin Pharmacol Ther ; 112(3): 562-572, 2022 09.
Article in English | MEDLINE | ID: mdl-35598119

ABSTRACT

Transporters contribute to renal elimination of drugs; therefore drug disposition can be impacted if transporters are inhibited by comedicant drugs. Regulatory agencies have provided guidelines to assess potential drug-drug interaction (DDI) risk for renal organic cation transporter 2 (OCT2) and multidrug and toxin extrusion 1 and 2-K (MATE1/2-K) transporters. Despite this, there are challenges with translating in vitro data using currently available tools to obtain a quantitative assessment of DDI risk in the clinic. Given the high number of drugs and new molecular entities showing in vitro inhibition toward OCT2 and/or MATE1/2-K and the lack of translation to clinically significant effects, it is reasonable to question whether the current in vitro assay design and modeling practice has led to unnecessary clinical evaluation. The aim of this review is to assess and discuss available in vitro and clinical data along with prediction models intended to provide clinical context of risk, including static models proposed by regulatory agencies and physiologically-based pharmacokinetic models, in order to identify best practices and areas of future opportunity. This analysis highlights that different in vitro assay designs, including substrate and cell systems used, strongly influence the derived concentration of drug producing 50% inhibition values and contribute to high variability observed across laboratories. Furthermore, the lack of sensitive index substrates coupled with specific inhibitors for individual transporters necessitates the use of complex models to evaluate clinical DDI risk.


Subject(s)
Kidney , Organic Cation Transport Proteins , Drug Interactions , HEK293 Cells , Humans , Kidney/metabolism , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2/metabolism , Renal Elimination
6.
Clin Pharmacol Ther ; 112(4): 770-781, 2022 10.
Article in English | MEDLINE | ID: mdl-34862964

ABSTRACT

The International Consortium for Innovation and Quality (IQ) Physiologically Based Pharmacokinetic (PBPK) Modeling Induction Working Group (IWG) conducted a survey across participating companies around general strategies for PBPK modeling of induction, including experience with its utility to address various questions, regulatory interactions, and regulatory acceptance. The results highlight areas where PBPK modeling is used with high confidence and identifies opportunities where confidence is lower and further evaluation is needed. To enhance the survey results, the PBPK-IWG also collected case studies and analyzed recent literature examples where PBPK models were applied to predict CYP3A induction-mediated drug-drug interactions. PBPK modeling of induction has evolved and progressed significantly, proving to have great potential to accelerate drug discovery and development. With the aim of enabling optimal use for new molecular entities that are either substrates and/or inducers of CYP3A, the PBPK-IWG proposes initial workflows for PBPK application, discusses future trends, and identifies gaps that need to be addressed.


Subject(s)
Cytochrome P-450 CYP3A , Models, Biological , Computer Simulation , Cytochrome P-450 Enzyme System , Drug Interactions , Humans , Workflow
7.
Mol Pharm ; 19(1): 172-187, 2022 01 03.
Article in English | MEDLINE | ID: mdl-34890209

ABSTRACT

A physiologically based pharmacokinetic model was developed to describe the tissue distribution kinetics of a dendritic nanoparticle and its conjugated active pharmaceutical ingredient (API) in plasma, liver, spleen, and tumors. Tumor growth data from MV-4-11 tumor-bearing mice were incorporated to investigate the exposure/efficacy relationship. The nanoparticle demonstrated improved antitumor activity compared to the conventional API formulation, owing to the extended released API concentrations at the site of action. Model simulations further enabled the identification of critical parameters that influence API exposure in tumors and downstream efficacy outcomes upon nanoparticle administration. The model was utilized to explore a range of dosing schedules and their effect on tumor growth kinetics, demonstrating the improved antitumor activity of nanoparticles with less frequent dosing compared to the same dose of naked APIs in conventional formulations.


Subject(s)
Antineoplastic Agents/administration & dosage , Dendrimers/pharmacokinetics , Nanoparticles/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Female , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Tissue Distribution , Treatment Outcome
8.
Cancer Chemother Pharmacol ; 88(3): 451-464, 2021 09.
Article in English | MEDLINE | ID: mdl-34080039

ABSTRACT

PURPOSE: Limited information is available regarding the drug-drug interaction (DDI) potential of molecular targeted agents and rituximab plus cyclophosphamide, doxorubicin (hydroxydaunorubicin), vincristine (Oncovin), and prednisone (R-CHOP) therapy. The addition of the Bruton tyrosine kinase (BTK) inhibitor ibrutinib to R-CHOP therapy results in increased toxicity versus R-CHOP alone, including higher incidence of peripheral neuropathy. Vincristine is a substrate of P-glycoprotein (P-gp, ABCB1); drugs that inhibit P-gp could potentially cause increased toxicity when co-administered with vincristine through DDI. While the combination of the BTK inhibitor acalabrutinib and R-CHOP is being explored clinically, the DDI potential between these therapies is unknown. METHODS: A human mechanistic physiology-based pharmacokinetic (PBPK) model of vincristine following intravenous dosing was developed to predict potential DDI interactions with combination therapy. In vitro absorption, distribution, metabolism, and excretion and in vivo clinical PK parameters informed PBPK model development, which was verified by comparing simulated vincristine concentrations with observed clinical data. RESULTS: While simulations suggested no DDI between vincristine and ibrutinib or acalabrutinib in plasma, simulated vincristine exposure in muscle tissue was increased in the presence of ibrutinib but not acalabrutinib. Extrapolation of the vincristine mechanistic PBPK model to other P-gp substrates further suggested DDI risk when ibrutinib (area under the concentration-time curve [AUC] ratio: 1.8), but not acalabrutinib (AUC ratio: 0.92), was given orally with venetoclax or digoxin. CONCLUSION: Overall, these data suggest low DDI risk between acalabrutinib and P-gp substrates with negligible increase in the potential risk of vincristine-induced peripheral neuropathy when acalabrutinib is added to R-CHOP therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Models, Biological , Peripheral Nervous System Diseases/chemically induced , Vincristine/administration & dosage , Administration, Intravenous , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Area Under Curve , Caco-2 Cells , Computer Simulation , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Cyclophosphamide/pharmacokinetics , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Doxorubicin/pharmacokinetics , Drug Interactions , Female , Humans , Male , Middle Aged , Molecular Targeted Therapy , Prednisone/administration & dosage , Prednisone/adverse effects , Prednisone/pharmacokinetics , Rituximab/administration & dosage , Rituximab/adverse effects , Rituximab/pharmacokinetics , Tissue Distribution , Vincristine/adverse effects , Vincristine/pharmacokinetics , Young Adult
9.
J Med Chem ; 63(23): 14530-14559, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32910656

ABSTRACT

Herein we report the optimization of a series of tricyclic indazoles as selective estrogen receptor degraders (SERD) and antagonists for the treatment of ER+ breast cancer. Structure based design together with systematic investigation of each region of the molecular architecture led to the identification of N-[1-(3-fluoropropyl)azetidin-3-yl]-6-[(6S,8R)-8-methyl-7-(2,2,2-trifluoroethyl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl]pyridin-3-amine (28). This compound was demonstrated to be a highly potent SERD that showed a pharmacological profile comparable to fulvestrant in its ability to degrade ERα in both MCF-7 and CAMA-1 cell lines. A stringent control of lipophilicity ensured that 28 had favorable physicochemical and preclinical pharmacokinetic properties for oral administration. This, combined with demonstration of potent in vivo activity in mouse xenograft models, resulted in progression of this compound, also known as AZD9833, into clinical trials.


Subject(s)
Antineoplastic Agents/administration & dosage , Selective Estrogen Receptor Modulators/administration & dosage , Administration, Oral , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Biological Availability , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Cyclization , Drug Discovery , Female , Humans , Lipids/chemistry , Molecular Structure , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/pharmacokinetics , Structure-Activity Relationship
10.
Drug Metab Dispos ; 48(11): 1137-1146, 2020 11.
Article in English | MEDLINE | ID: mdl-32847864

ABSTRACT

The use of in vitro in vivo extrapolation (IVIVE) from human hepatocyte (HH) and human liver microsome (HLM) stability assays is a widely accepted predictive methodology for human metabolic clearance (CLmet). However, a systematic underprediction of CLmet from both matrices appears to be universally apparent, which can be corrected for via an empirical regression offset. After physiological scaling, intrinsic clearance (CLint) for compounds metabolized via the same enzymatic pathway should be equivalent for both matrices. Compounds demonstrating significantly higher HLM CLint relative to HH CLint have been encountered, raising questions regarding how to predict CLmet for such compounds. Here, we determined the HLM:HH CLint ratio for 140 marketed drugs/compounds, compared this ratio as a function of physiochemical properties and drug metabolism enzyme dependence, and examined methodologies to predict CLmet from both matrices. The majority (78%) of compounds displaying a high HLM:HH CLint ratio were CYP3A substrates. Using HH CLint for CYP3A substrates, the current IVIVE regression offset approach remains an appropriate strategy to predict CLmet (% compounds overpredicted/correctly predicted/underpredicted 27/62/11, respectively). However, using the same approach for HLM significantly overpredicts CLmet for CYP3A substrates (% compounds overpredicted/correctly predicted/underpredicted 56/33/11, respectively), highlighting that a different IVIVE offset is required for CYP3A substrates using HLM. This work furthers the understanding of compound properties associated with a disproportionately high HLM:HH CLint ratio and outlines a successful IVIVE approach for such compounds. SIGNIFICANCE STATEMENT: Oral drug discovery programs typically strive for low clearance compounds to ensure sufficient target engagement. Human liver microsomes and isolated human hepatocytes are used to optimize and predict human hepatic metabolic clearance. After physiological scaling, intrinsic clearance for compounds of the same metabolic pathway should be equivalent between matrices. However, a disconnect in intrinsic clearance is sometimes apparent. The work described attempts to further understand this phenomenon, and by achieving a mechanistic understanding, improvements in clearance predictions may be realized.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Hepatobiliary Elimination , Hepatocytes/enzymology , Microsomes, Liver/enzymology , Datasets as Topic , Drug Evaluation, Preclinical/methods , Female , Humans , Liver/cytology , Liver/enzymology , Male , Models, Biological , Recombinant Proteins/metabolism
11.
Drug Discov Today ; 25(10): 1793-1800, 2020 10.
Article in English | MEDLINE | ID: mdl-32693163

ABSTRACT

Proteolysis-targeting chimeras (PROTACs) are an emerging therapeutic modality with the potential to open target space not accessible to conventional small molecules via a degradation-based mechanism; however, their bifunctional nature can result in physicochemical properties that breach commonly accepted limits for small-molecule oral drugs. We offer a drug metabolism and pharmacokinetics (DMPK) perspective on the optimisation of oral PROTACs across a diverse set of projects within Oncology R&D at AstraZeneca, highlighting some of the challenges that they have presented to our established screening cascade. Furthermore, we challenge some of the perceptions and dogma surrounding the feasibility of oral PROTACS and demonstrate that acceptable oral PK properties for this modality can be regularly achievable despite the physicochemical property challenges they present.


Subject(s)
Drug Delivery Systems , Pharmaceutical Preparations/administration & dosage , Proteins/metabolism , Administration, Oral , Humans , Pharmaceutical Preparations/metabolism , Proteolysis
12.
Trends Pharmacol Sci ; 41(6): 390-408, 2020 06.
Article in English | MEDLINE | ID: mdl-32359836

ABSTRACT

During drug discovery and prior to the first human dose of a novel candidate drug, the pharmacokinetic (PK) behavior of the drug in humans is predicted from preclinical data. This helps to inform the likelihood of achieving therapeutic exposures in early clinical development. Once clinical data are available, the observed human PK are compared with predictions, providing an opportunity to assess and refine prediction methods. Application of best practice in experimental data generation and predictive methodologies, and a focus on robust mechanistic understanding of the candidate drug disposition properties before nomination to clinical development, have led to maximizing the probability of successful PK predictions so that 83% of AstraZeneca drug development projects progress in the clinic with no PK issues; and 71% of key PK parameter predictions [64% of area under the curve (AUC) predictions; 78% of maximum concentration (Cmax) predictions; and 70% of half-life predictions] are accurate to within twofold. Here, we discuss methods to predict human PK used by AstraZeneca, how these predictions are assessed and what can be learned from evaluating the predictions for 116 candidate drugs.


Subject(s)
Drug Discovery , Pharmacokinetics , Humans
13.
Drug Discov Today ; 24(5): 1067-1073, 2019 05.
Article in English | MEDLINE | ID: mdl-30710641

ABSTRACT

Due to the blood-brain barrier (BBB) limiting the exposure of therapeutics to the central nervous system (CNS), patients with brain malignancies are challenging to treat, typically have poor prognoses, and represent a significant unmet medical need. Preclinical data report osimertinib to have significant BBB penetration and emerging clinical data demonstrate encouraging activity against CNS malignancies. Here, we discuss the oncology drug candidates AZD3759 and AZD1390 as case examples of discovery projects designing in BBB penetrance. We demonstrate how these innovative kinase inhibitors were recognized as brain penetrant and outline our view of experimental approaches and strategies that can facilitate the discovery of new brain-penetrant therapies for the treatment of primary and secondary CNS malignancies as well as other CNS disorders.


Subject(s)
Acrylamides/pharmacokinetics , Aniline Compounds/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Brain Neoplasms/metabolism , Brain/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Brain/diagnostic imaging , Brain Neoplasms/drug therapy , Drug Discovery , Humans , Piperazines/pharmacokinetics , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Quinazolines/pharmacokinetics , Quinazolines/therapeutic use
14.
Mol Pharm ; 10(4): 1191-206, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23253040

ABSTRACT

In the present age of pharmaceutical research and development, focused delivery of decision making data is more imperative than ever before. Resulting from several years' success, failure and consequential learning, this article also proffers advice and guidance on which in vitro and in vivo experiments to perform to facilitate efficient and cost-effective pursuit of candidate drugs with acceptable human pharmacokinetic profiles. Predictive in silico models are important in directing design toward compounds with the highest probability of having suitable DMPK properties rather than in predicting human pharmacokinetics per se, and the value and utility of such approaches are reviewed with the intention of providing direction to DMPK scientists. Relating to absorption, distribution, elimination and effective half-life, strategies are described to provide direction in commonly encountered scenarios.


Subject(s)
Drug Evaluation, Preclinical/methods , Metabolic Clearance Rate , Pharmaceutical Preparations/administration & dosage , Pharmacokinetics , Absorption , Algorithms , Animals , Computer Simulation , Drug Design , Humans , Models, Theoretical , Technology, Pharmaceutical/methods
15.
Drug Metab Dispos ; 39(6): 1008-13, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21357702

ABSTRACT

Identifying any extrahepatic excretion phenomenon in preclinical species is crucial for an accurate prediction of the pharmacokinetics in man. This understanding is particularly key for drugs with a small volume of distribution, because they require an especially low total clearance to be suitable for a once-a-day dosing regimen in man. In this study, three animal scaling techniques were applied for the prediction of the human renal clearance of 36 diverse drugs that show active secretion or net reabsorption: 1) direct correlations between renal clearance in man and each of the two main preclinical species (rat and dog); 2) simple allometry; and 3) Mahmood's renal clearance scaling method. The results show clearly that the predictions to man for the methods are improved significantly when corrections are made for species differences in plasma protein binding. Overall, the most accurate predictions were obtained by using a direct correlation with the dog renal clearance after correcting for differences in plasma protein binding and kidney blood flow (r² = 0.84), where predictions, on average, were within 2-fold of the observed renal clearance values in human.


Subject(s)
Drug Evaluation, Preclinical , Kidney/metabolism , Pharmaceutical Preparations/blood , Absorption , Animals , Blood Proteins/metabolism , Dogs , Drug Evaluation, Preclinical/statistics & numerical data , Drugs, Investigational/pharmacokinetics , Humans , Inactivation, Metabolic , Male , Mathematical Computing , Metabolic Clearance Rate , Predictive Value of Tests , Protein Binding , Rats , Species Specificity
16.
Drug Metab Dispos ; 37(6): 1259-68, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19307295

ABSTRACT

Prototypic CYP3A4 inducers were tested in a pregnane X receptor (PXR) reporter gene assay, Fa2N-4 cells, HepaRG cells, and primary human hepatocytes, along with negative controls, using CYP3A4 mRNA and activity endpoints, where appropriate. Over half of the compounds tested (14 of 24) were identified as time-dependent inhibitors of CYP3A4 and high mRNA/activity ratios (>10) were consistent with CYP3A4 time-dependent inhibition for compounds such as troleandomycin, ritonavir, and verapamil. Induction response was compared between two human donors; there was an excellent correlation in the EC(50) estimates (r(2) = 0.89, p < 0.001), and a weak but statistically significant correlation was noted for maximum observed induction at an optimum concentration (E(max)) (r(2) = 0.38, p = 0.001). E(max) and EC(50) estimates determined from the PXR reporter gene assay and Fa2N-4 and HepaRG cells were compared with those from hepatocytes. Overall, EC(50) values generated using hepatocytes agreed with those generated in the PXR reporter gene assay (r(2) = 0.85, p < 0.001) and Fa2N-4 (r(2) = 0.65, p < 0.001) and HepaRG (r(2) = 0.99, p < 0.001) cells. However, E(max) values generated in hepatocytes were only significantly correlated to those determined in Fa2N-4 (r(2) = 0.33, p = 0.005) and HepaRG cells (r(2) = 0.79, p < 0.001). "Gold standard" cytochrome P450 induction data can be generated using primary human hepatocytes, but a restricted, erratic supply and interdonor variability somewhat restrict routine application within a drug discovery setting. HepaRG cells are a valuable recent addition to the armory of in vitro tools for assessing CYP3A4 induction and seem to be an excellent surrogate of primary cells.


Subject(s)
Cytochrome P-450 CYP3A/biosynthesis , Drug Evaluation, Preclinical , Drug Interactions , Cells, Cultured , Drug Design , Enzyme Induction/drug effects , Hepatocytes/metabolism , Humans , Pharmaceutical Preparations , Pregnane X Receptor , Receptors, Steroid/metabolism , Reverse Transcriptase Polymerase Chain Reaction
17.
Drug Metab Dispos ; 36(6): 1126-34, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18356267

ABSTRACT

Unbound IC(50) (IC(50,u)) values of 15 drugs were determined in eight recombinantly expressed human cytochromes P450 (P450s) and human hepatocytes, and the data were used to simulate clinical area under the plasma concentration-time curve changes (deltaAUC) on coadministration with prototypic CYP2D6 substrates. Significant differences in IC(50,u) values between enzyme sources were observed for quinidine (0.02 microM in recombinant CYP2D6 versus 0.5 microM in hepatocytes) and propafenone (0.02 versus 4.1 microM). The relative contribution of individual P450s toward the oxidative metabolism of clinical probes desipramine, imipramine, tolterodine, propranolol, and metoprolol was estimated via determinations of intrinsic clearance using recombinant P450s (rP450s). Simulated deltaAUC were compared with those observed in vivo via the ratios of unbound inhibitor concentration at the entrance to the liver to inhibition constants determined against rP450s ([I](in,u)/K(i)) and incorporating parallel substrate elimination pathways. For this dataset, there were 20% false negatives (observed deltaAUC >or= 2, predicted deltaAUC < 2), 77% correct predictions, and 3% false positives. Thus, the [I](in,u)/K(i) approach appears relatively successful at estimating the degree of clinical interactions and can be incorporated into drug discovery strategies. Using a Simcyp ADME (absorption, metabolism, distribution, elimination) simulator (Simcyp Ltd., Sheffield, UK), there were 3% false negatives, 94% correct simulations, and 3% false positives. False-negative predictions were rationalized as a result of mechanism-based inhibition, production of inhibitory metabolites, and/or hepatic uptake. Integrating inhibition and reaction phenotyping data from automated rP450 screens have shown applicability to predict the occurrence and degree of in vivo drug-drug interactions, and such data may identify the clinical consequences for candidate drugs as both "perpetrators" and "victims" of P450-mediated interactions.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Hepatocytes/metabolism , Recombinant Proteins/metabolism , Area Under Curve , Cells, Cultured , Computer Simulation , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Forecasting , Humans , Pharmaceutical Preparations/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics
18.
Bioorg Med Chem Lett ; 17(21): 6013-8, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17827008

ABSTRACT

Starting from adenosine triphosphate (ATP), the identification of a novel series of P2Y(12) receptor antagonists and exploitation of their SAR is described. Modifications of the acidic side chain and the purine core and investigation of hydrophobic substituents led to a series of neutral molecules. The leading compound, 17 (AZD6140), is currently in a large phase III clinical trial for the treatment of acute coronary syndromes and prevention of thromboembolic clinical sequelae.


Subject(s)
Adenosine Triphosphate/therapeutic use , Adenosine/analogs & derivatives , Membrane Proteins/antagonists & inhibitors , Purinergic P2 Receptor Antagonists , Thrombosis/prevention & control , Adenosine/therapeutic use , Administration, Oral , Animals , Humans , Receptors, Purinergic P2Y12 , Ticagrelor
19.
Chem Biol Interact ; 168(1): 2-15, 2007 May 20.
Article in English | MEDLINE | ID: mdl-17208208

ABSTRACT

This review promotes the value of isolated hepatocytes in modern Drug Discovery programmes and outlines how increased understanding, particularly in the area of in vitro-in vivo extrapolation (IVIVE), has led to more widespread use. The importance of in vitro metabolic intrinsic clearance data for predicting in vivo clearance has been acknowledged for several years and the greater utility of hepatocytes, compared with hepatic microsomes and liver slices, for this application is discussed. The application of hepatocytes in predicting drug-drug interactions (DDIs) resulting from reversible and irreversible (time-dependent) inhibition is relatively novel but affords the potential to study both phase I and phase II processes together with any impact of drug efflux and/or uptake (cellular accumulation). Progress in this area is reviewed along with current opinions on the comparative use of primary hepatocytes and higher throughput reporter gene-based systems for studying cytochrome P450 (CYP) induction. The appreciation of the role of transporter proteins in drug disposition continues to evolve. The study of hepatic uptake using isolated hepatocytes and the interplay between drug transport and metabolism with respect to both clearance and DDIs and subsequent IVIVE is also considered.


Subject(s)
Drug Design , Hepatocytes/metabolism , Pharmaceutical Preparations/metabolism , Animals , Cells, Cultured , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Hepatocytes/enzymology , Humans , Metabolic Clearance Rate , Metabolic Networks and Pathways
20.
Drug Metab Dispos ; 34(8): 1291-300, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16679385

ABSTRACT

Primary human hepatocytes in culture are commonly used to evaluate cytochrome P450 (P450) induction via an enzyme activity endpoint. However, other processes can confound data interpretation. To this end, the impact of time-dependent P450 inhibition in this system was evaluated. Using a substrate-cassette approach, P450 activities were determined after incubation with the prototypic inhibitors tienilic acid (CYP2C9), erythromycin, troleandomycin, and fluoxetine (CYP3A4). Kinetic analysis of enzyme inactivation in hepatocytes was used to describe the effect of these time-dependent inhibitors and derive the inhibition parameters kinact and KI) which generally were in good agreement with the values derived using recombinant P450s and human liver microsomes (HLMs). Tienilic acid selectively inhibited CYP2C9-dependent diclofenac 4'-hydroxylation activity, and erythromycin, troleandomycin, and fluoxetine inhibited CYP3A4-dependent midazolam 1'-hydroxylation in a time- and concentration-dependent manner. Fluoxetine also inhibited CYP2C19-dependent S-mephenytoin 4'-hydroxylation in a time- and concentration-dependent manner in hepatocytes, HLMs, and recombinant CYP2C19 (KI 0.4 microM and kinact 0.5 min(-1)). As expected, the effect of fluoxetine on CYP2D6 in hepatocytes was consistent with potent yet reversible inhibition. A very weak time-dependent CYP2C9 inhibitor (AZ1, a proprietary AstraZeneca compound; KI 30 microM and kinact 0.02 min(-1)) effectively abolished CYP2C9 activity over 24 h at low (micromolar) concentrations in primary cultured human hepatocytes. This work demonstrates that caution is warranted in the interpretation of enzyme induction studies with metabolically stable, weak time-dependent inhibitors, which may have dramatic inhibitory effects on P450 activity in this system. Therefore, in addition to enzyme activity, mRNA and/or protein levels should be measured to fully evaluate the P450 induction potential of a drug candidate.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Hepatocytes/enzymology , Aged , Aged, 80 and over , Cells, Cultured , Cytochrome P-450 Enzyme System/metabolism , Erythromycin/pharmacology , Female , Fluoxetine/pharmacology , Hepatocytes/drug effects , Humans , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Middle Aged , Ticrynafen/pharmacology , Troleandomycin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...