1
|
Sundheimer JK, Benzel J, Longuespée R, Burhenne J, Pfister SM, Maaß KK, Sauter M, Pajtler KW. Experimental Insights and Recommendations for Successfully Performing Cerebral Microdialysis With Hydrophobic Drug Candidates. Clin Transl Sci 2025; 18:e70226. [PMID: 40286321 PMCID: PMC12033007 DOI: 10.1111/cts.70226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 03/14/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025] Open
Abstract
Cerebral microdialysis in rodents represents a robust and versatile technique for quantifying the pharmacologically relevant unbound fraction of drugs in the brain. When this unbound fraction is simultaneously determined in plasma, it facilitates the calculation of the corresponding unbound plasma-to-brain partition coefficient (Kp,uu) for a given compound in vivo. This coefficient is critical for understanding the penetration and distribution of drugs across the blood-brain barrier (BBB). However, obtaining valid and accurate microdialysis data can be particularly challenging for hydrophobic drugs due to their pronounced non-specific interactions with the components of the microdialysis system. The present study reports the outcomes of comprehensive microdialysis investigations in rodents, focusing on three hydrophobic compounds: actinomycin D, selinexor, and ulixertinib. These compounds exhibited varying degrees of non-specific binding to the surfaces of the microdialysis apparatus, leading to low recovery rates and substantial carry-over effects. To diminish these limitations, strategies such as surface coating and the use of optimized materials were employed to enhance the reliability of the microdialysis system. To ensure the robustness and reproducibility of microdialysis-related research outcomes, our experimental findings were supplemented with a narrative literature review. This review encompassed keyword-driven PubMed-indexed publications on microdialysis from 1970 to 2024, providing a broader context for the challenges and solutions associated with the technique. By integrating empirical results with practical recommendations, this study offers a comprehensive resource aimed at advancing the application of cerebral microdialysis in preclinical drug development, particularly for compounds with challenging physicochemical properties.
Collapse
Affiliation(s)
- Julia K. Sundheimer
- Hopp Children's Cancer Center at the NCT Heidelberg (KiTZ)HeidelbergGermany
- Division of Pediatric Neurooncology, German Cancer Consortium (DKTK)German Cancer Research Center (DKFZ)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Julia Benzel
- Hopp Children's Cancer Center at the NCT Heidelberg (KiTZ)HeidelbergGermany
- Division of Pediatric Neurooncology, German Cancer Consortium (DKTK)German Cancer Research Center (DKFZ)HeidelbergGermany
- Department of Pediatric Hematology, Oncology and ImmunologyHeidelberg University HospitalHeidelbergGermany
| | - Rémi Longuespée
- Medical Faculty Heidelberg/Heidelberg University Hospital, Internal Medicine IX—Department of Clinical Pharmacology and PharmacoepidemiologyHeidelberg UniversityHeidelbergGermany
- Metabolic Crosstalk in CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Jürgen Burhenne
- Medical Faculty Heidelberg/Heidelberg University Hospital, Internal Medicine IX—Department of Clinical Pharmacology and PharmacoepidemiologyHeidelberg UniversityHeidelbergGermany
| | - Stefan M. Pfister
- Hopp Children's Cancer Center at the NCT Heidelberg (KiTZ)HeidelbergGermany
- Division of Pediatric Neurooncology, German Cancer Consortium (DKTK)German Cancer Research Center (DKFZ)HeidelbergGermany
- Department of Pediatric Hematology, Oncology and ImmunologyHeidelberg University HospitalHeidelbergGermany
| | - Kendra K. Maaß
- Hopp Children's Cancer Center at the NCT Heidelberg (KiTZ)HeidelbergGermany
- Division of Pediatric Neurooncology, German Cancer Consortium (DKTK)German Cancer Research Center (DKFZ)HeidelbergGermany
- Department of Pediatric Hematology, Oncology and ImmunologyHeidelberg University HospitalHeidelbergGermany
| | - Max Sauter
- Medical Faculty Heidelberg/Heidelberg University Hospital, Internal Medicine IX—Department of Clinical Pharmacology and PharmacoepidemiologyHeidelberg UniversityHeidelbergGermany
| | - Kristian W. Pajtler
- Hopp Children's Cancer Center at the NCT Heidelberg (KiTZ)HeidelbergGermany
- Division of Pediatric Neurooncology, German Cancer Consortium (DKTK)German Cancer Research Center (DKFZ)HeidelbergGermany
- Department of Pediatric Hematology, Oncology and ImmunologyHeidelberg University HospitalHeidelbergGermany
| |
Collapse
|
2
|
Moreno V, Manuel Sepúlveda J, Reardon DA, Pérez-Núñez Á, González León P, Hanna B, Filvaroff E, Aronchik I, Chang H, Amoroso B, Zuraek M, Sanchez-Perez T, Mendez C, Stephens D, Nikolova Z, Vogelbaum MA. Trotabresib, an oral potent bromodomain and extraterminal inhibitor, in patients with high-grade gliomas: A phase I, "window-of-opportunity" study. Neuro Oncol 2023; 25:1113-1122. [PMID: 36455228 PMCID: PMC10237409 DOI: 10.1093/neuonc/noac263] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND The bromodomain and extraterminal protein (BET) inhibitor trotabresib has demonstrated antitumor activity in patients with advanced solid tumors, including high-grade gliomas. CC-90010-GBM-001 (NCT04047303) is a phase I study investigating the pharmacokinetics, pharmacodynamics, and CNS penetration of trotabresib in patients with recurrent high-grade gliomas scheduled for salvage resection. METHODS Patients received trotabresib 30 mg/day on days 1-4 before surgery, followed by maintenance trotabresib 45 mg/day 4 days on/24 days off after surgery. Primary endpoints were plasma pharmacokinetics and trotabresib concentrations in resected tissue. Secondary and exploratory endpoints included safety, pharmacodynamics, and antitumor activity. RESULTS Twenty patients received preoperative trotabresib and underwent resection with no delays or cancelations of surgery; 16 patients received maintenance trotabresib after recovery from surgery. Trotabresib plasma pharmacokinetics were consistent with previous data. Mean trotabresib brain tumor tissue:plasma ratio was 0.84 (estimated unbound partition coefficient [KPUU] 0.37), and modulation of pharmacodynamic markers was observed in blood and brain tumor tissue. Trotabresib was well tolerated; the most frequent grade 3/4 treatment-related adverse event during maintenance treatment was thrombocytopenia (5/16 patients). Six-month progression-free survival was 12%. Two patients remain on treatment with stable disease at cycles 25 and 30. CONCLUSIONS Trotabresib penetrates the blood-brain-tumor barrier in patients with recurrent high-grade glioma and demonstrates target engagement in resected tumor tissue. Plasma pharmacokinetics, blood pharmacodynamics, and safety were comparable with previous results for trotabresib in patients with advanced solid tumors. Investigation of adjuvant trotabresib + temozolomide and concomitant trotabresib + temozolomide + radiotherapy in patients with newly diagnosed glioblastoma is ongoing (NCT04324840).
Collapse
Affiliation(s)
- Victor Moreno
- START Madrid-FJD, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | | | - David A Reardon
- Department of Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Ángel Pérez-Núñez
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Pedro González León
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Bishoy Hanna
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Ida Aronchik
- Bristol Myers Squibb, San Francisco, California, USA
| | - Henry Chang
- Bristol Myers Squibb, San Francisco, California, USA
| | - Barbara Amoroso
- Centre for Innovation and Translational Research Europe, A Bristol Myers Squibb Company, Seville, Spain
| | | | - Tania Sanchez-Perez
- Centre for Innovation and Translational Research Europe, A Bristol Myers Squibb Company, Seville, Spain
| | - Cristina Mendez
- Centre for Innovation and Translational Research Europe, A Bristol Myers Squibb Company, Seville, Spain
| | | | - Zariana Nikolova
- Centre for Innovation and Translational Research Europe, A Bristol Myers Squibb Company, Seville, Spain
| | | |
Collapse
|
3
|
Leow JWH, Gu Y, Chan ECY. Investigating the relevance of CYP2J2 inhibition for drugs known to cause intermediate to high risk torsades de pointes. Eur J Pharm Sci 2023; 187:106475. [PMID: 37225005 DOI: 10.1016/j.ejps.2023.106475] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/10/2023] [Accepted: 05/21/2023] [Indexed: 05/26/2023]
Abstract
Cardiac cytochrome P450 2J2 (CYP2J2) metabolizes endogenous polyunsaturated fatty acid, arachidonic acid (AA), to bioactive regioisomeric epoxyeicosatrienoic acid (EET) metabolites. This endogenous metabolic pathway has been postulated to play a homeostatic role in cardiac electrophysiology. However, it is unknown if drugs that cause intermediate to high risk torsades de pointes (TdP) exhibit inhibitory effects against CYP2J2 metabolism of AA to EETs. In this study, we demonstrated that 11 out of 16 drugs screened with intermediate to high risk of TdP as defined by the Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative are concurrently reversible inhibitors of CYP2J2 metabolism of AA, with unbound inhibitory constant (Ki,AA,u) values ranging widely from 0.132 to 19.9 µM. To understand the physiological relevancy of Ki,AA,u, the in vivo unbound drug concentration within human heart tissue (Cu,heart) was calculated via experimental determination of in vitro unbound partition coefficient (Kpuu) for 10 CYP2J2 inhibitors using AC16 human ventricular cardiomyocytes as well as literature-derived values of fraction unbound in plasma (fu,p) and plasma drug concentrations in clinical scenarios leading to TdP. Notably, all CYP2J2 inhibitors screened belonging to the high TdP risk category, namely vandetanib and bepridil, exhibited highest Kpuu values of 18.2 ± 1.39 and 7.48 ± 1.16 respectively although no clear relationship between Cu,heart and risk of TdP could eventually be determined. R values based on basic models of reversible inhibition as per FDA guidelines were calculated using unbound plasma drug concentrations (Cu,plasma) and adapted using Cu,heart which suggested that 4 out of 10 CYP2J2 inhibitors with intermediate to high risk of TdP demonstrate greatest potential for clinically relevant in vivo cardiac drug-AA interactions. Our results shed novel insights on the relevance of CYP2J2 inhibition in drugs with risk of TdP. Further studies ascertaining the role of CYP2J2 metabolism of AA in cardiac electrophysiology, characterizing inherent cardiac ion channel activities of drugs with risk of TdP as well as in vivo evidence of drug-AA interactions will be required prior to determining if CYP2J2 inhibition could be an alternative mechanism contributing to drug-induced TdP.
Collapse
Affiliation(s)
- Jacqueline Wen Hui Leow
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543
| | - Yuxiang Gu
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543.
| |
Collapse
|
4
|
Poller B, Werner S, Domange N, Mettler L, Stein RR, Loretan J, Wartmann M, Faller B, Huth F. Time Matters - In vitro Cellular Disposition Kinetics Help Rationalizing Cellular Potency Disconnects. Xenobiotica 2022; 52:878-889. [PMID: 36189672 DOI: 10.1080/00498254.2022.2130837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2022]
Abstract
Loss in potency is commonly observed in early drug discovery when moving from biochemical to more complex cellular systems. Among other factors, low permeability is often considered to cause such potency disconnects.We developed a novel cellular disposition assay in MDCK cells to determine passive uptake clearance (PSinf), cell-to-medium ratios at steady-state (Kp) and the time to reach 90% steady-state (TTSS90) from a single experiment in a high-throughput format.The assay was validated using 40 marketed drugs, showing a wide distribution of PSinf and Kp values. The parameters generally correlated with transcellular permeability and lipophilicity, while PSinf data revealed better resolution in the high and low permeability ranges compared to traditional permeability data. A linear relationship between the Kp/PSinf ratio and TTSS90 was mathematically derived and experimentally validated, demonstrating the dependency of TTSS90 on the rate and extent of cellular accumulation.Cellular disposition parameters could explain potency (IC50) disconnects noted for seven Bruton's tyrosine kinase degrader compounds in a cellular potency assay. In contrast to transcellular permeability, PSinf data enabled identification of the compounds with IC50 disconnects based on their time to reach equilibrium. Overall, the novel assay offers the possibility to address potency disconnects in early drug discovery.
Collapse
Affiliation(s)
- Birk Poller
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Sophie Werner
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Norbert Domange
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Lina Mettler
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Richard R Stein
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Jacqueline Loretan
- Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Markus Wartmann
- Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Bernard Faller
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Felix Huth
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| |
Collapse
|
5
|
Wang Z, Yong Chan EC. Inhibition of Cytochrome P450 2J2-Mediated Metabolism of Rivaroxaban and Arachidonic Acid by Ibrutinib and Osimertinib. Drug Metab Dispos 2022; 50:1332-1341. [PMID: 35817438 DOI: 10.1124/dmd.122.000928] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/29/2022] [Indexed: 11/22/2022] Open
Abstract
Covalent tyrosine kinase inhibitors (TKIs) ibrutinib and osimertinib are associated with cardiac arrhythmia. The interactions between these TKIs with CYP2J2 that is highly expressed in the human heart are unknown. In vitro metabolism experiments were performed to characterize CYP2J2-mediated metabolism of ibrutinib and osimertinib. Unbound distribution coefficient (Kpuu) for both TKIs was determined in AC16 cardiomyocytes. In vitro reversible and time-dependent CYP2J2 inhibition experiments were conducted with exogenous and endogenous substrates, namely rivaroxaban and arachidonic acid (AA), respectively, where kinetic parameters were estimated via one-site and multisite kinetic modeling. Ibrutinib was efficiently metabolized by CYP2J2 to a hydroxylated metabolite, M35, following substrate inhibition kinetics. Osimertinib is not a substrate of CYP2J2. Both TKIs depicted Kpuu values above 1 and equipotently inhibited CYP2J2-mediated hydroxylation of rivaroxaban in a concentration-dependent manner without time-dependency. The mode of reversible inhibition of CYP2J2-mediated metabolism of rivaroxaban and AA by osimertinib was described by Michaelis-Menten kinetics, whereas a two-site kinetic model recapitulated the atypical inhibitory kinetics of ibrutinib, assuming multiple substrate-binding domains within the CYP2J2 active site. The inhibition of ibrutinib and osimertinib on cardiac AA metabolism could be clinically significant considering the preferable distribution of both TKIs to cardiomyocytes with R cut-off values of 1.160 and 1.026, respectively. The dysregulation of CYP2J2-mediated metabolism of AA to cardioprotective epoxyeicosatrienoic acids by ibrutinib and osimertinib serves as a novel mechanism for TKI-induced cardiac arrhythmia. Mechanistic characterization of CYP2J2-mediated typical and atypical enzyme kinetics further illuminates the unique catalytic properties of CYP2J2. SIGNIFICANCE STATEMENT: We reported for the first time that ibrutinib is efficiently metabolized by CYP2J2. By using rivaroxaban and arachidonic acid (AA) as substrates, we characterized the typical and atypical inhibition kinetics of CYP2J2 by ibrutinib and osimertinib. The inhibition of both drugs on cardiac AA metabolism could be clinically significant considering their preferable distribution to cardiomyocytes. Our findings serve as a novel mechanism for drug-induced cardiac arrhythmia and shed insights into the multisite interactions between CYP2J2 and ligands.
Collapse
Affiliation(s)
- Ziteng Wang
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| |
Collapse
|
6
|
Weng Y, Fonseca KR, Bi YA, Mathialagan S, Riccardi K, Tseng E, Bessire AJ, Cerny MA, Tess DA, Rodrigues AD, Kalgutkar AS, Litchfield JE, Di L, Varma MVS. Transporter-Enzyme Interplay in the Pharmacokinetics of PF-06835919, A First-in-class Ketohexokinase Inhibitor for Metabolic Disorders and Non-alcoholic Fatty Liver Disease. Drug Metab Dispos 2022; 50:DMD-AR-2022-000953. [PMID: 35779864 DOI: 10.1124/dmd.122.000953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 06/27/2022] [Indexed: 11/22/2022] Open
Abstract
Excess dietary fructose consumption promotes metabolic dysfunction thereby increasing the risk of obesity, type 2 diabetes, non-alcoholic steatohepatitis (NASH), and related comorbidities. PF-06835919, a first-in-class ketohexokinase (KHK) inhibitor, showed reversal of such metabolic disorders in preclinical models and clinical studies, and is under clinical development for the potential treatment of NASH. In this study, we evaluated the transport and metabolic pathways of PF-06835919 disposition and assessed pharmacokinetics in preclinical models. PF-06835919 showed active uptake in cultured primary human hepatocytes, and substrate activity to organic anion transporter (OAT)2 and organic anion transporting-polypeptide (OATP)1B1 in transfected cells. "SLC-phenotyping" studies in human hepatocytes suggested contribution of passive uptake, OAT2- and OATP1B-mediated transport to the overall uptake to be about 15%, 60% and 25%, respectively. PF-06835919 showed low intrinsic metabolic clearance in vitro, and was found to be metabolized via both oxidative pathways (58%) and acyl glucuronidation (42%) by CYP3A, CYP2C8, CYP2C9 and UGT2B7. Following intravenous dosing, PF-06835919 showed low clearance (0.4-1.3 mL/min/kg) and volume of distribution (0.17-0.38 L/kg) in rat, dog and monkey. Human oral pharmacokinetics are predicted within 20% error when considering transporter-enzyme interplay in a PBPK model. Finally, unbound liver-to-plasma ratio (Kpuu) measured in vitro using rat, NHP and human hepatocytes was found to be approximately 4, 25 and 10, respectively. Similarly, liver Kpuu in rat and monkey following intravenous dosing of PF-06835919 was found to be 2.5 and 15, respectively, and notably higher than the muscle and brain Kpuu, consistent with the active uptake mechanisms observed in vitro. Significance Statement This work characterizes the transport/metabolic pathways in the hepatic disposition of PF-06835919, a first-in-class KHK inhibitor for the treatment of metabolic disorders and NASH. Phenotyping studies using transfected systems, human hepatocytes and liver microsomes signifies the role of OAT2 and OATP1B1 in the hepatic uptake and multiple enzymes in the metabolism of PF-06835919. Data presented suggest hepatic transporter-enzyme interplay in determining its systemic concentrations and potential enrichment in liver, a target site for KHK inhibition.
Collapse
Affiliation(s)
- Yan Weng
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., United States
| | | | | | - Sumathy Mathialagan
- Pharmacokinetics, Pharmacodynamics, and Metabolism, Medicine Design, Pfizer Inc, United States
| | | | - Elaine Tseng
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Global Research and Development, United States
| | | | | | | | | | - Amit S Kalgutkar
- Medicine Design, Pfizer Worldwide Research and Development, United States
| | | | - Li Di
- Pharmacokintics Dynamics and Metabolism, Pfizer Inc., United States
| | | |
Collapse
|
7
|
Ramsden D, Perloff ES, Whitcher-Johnstone A, Ho T, Patel R, Kozminski KD, Fullenwider CL, Zhang JG. Predictive In Vitro-In Vivo Extrapolation for Time Dependent Inhibition of CYP1A2, CYP2C8, CYP2C9, CYP2C19, and CYP2D6 Using Pooled Human Hepatocytes, Human Liver Microsomes, and a Simple Mechanistic Static Model. Drug Metab Dispos 2022; 50:114-127. [PMID: 34789487 DOI: 10.1124/dmd.121.000718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/12/2021] [Indexed: 11/22/2022] Open
Abstract
Inactivation of Cytochrome P450 (CYP450) enzymes can lead to significant increases in exposure of comedicants. The majority of reported in vitro to in vivo extrapolation (IVIVE) data have historically focused on CYP3A, leaving the assessment of other CYP isoforms insubstantial. To this end, the utility of human hepatocytes (HHEP) and human liver microsomes (HLM) to predict clinically relevant drug-drug interactions was investigated with a focus on CYP1A2, CYP2C8, CYP2C9, CYP2C19, and CYP2D6. Evaluation of IVIVE for CYP2B6 was limited to only weak inhibition. A search of the University of Washington Drug-Drug Interaction Database was conducted to identify a clinically relevant weak, moderate, and strong inhibitor for selective substrates of CYP1A2, CYP2C8, CYP2C9, CYP2C19, and CYP2D6, resulting in 18 inhibitors for in vitro characterization against 119 clinical interaction studies. Pooled human hepatocytes and HLM were preincubated with increasing concentrations of inhibitors for designated timepoints. Time dependent inhibition was detected in HLM for four moderate/strong inhibitors, suggesting that some optimization of incubation conditions (i.e., lower protein concentrations) is needed to capture weak inhibition. Clinical risk assessment was conducted by incorporating the in vitro derived kinetic parameters maximal rate of enzyme inactivation (min-1) (kinact) and concentration of inhibitor resulting in 50% of the maximum enzyme inactivation (KI) into static equations recommended by regulatory authorities. Significant overprediction was observed when applying the basic models recommended by regulatory agencies. Mechanistic static models, which consider the fraction of metabolism through the impacted enzyme, using the unbound hepatic inlet concentration lead to the best overall prediction accuracy with 92% and 85% of data from HHEPs and HLM, respectively, within twofold of the observed value. SIGNIFICANCE STATEMENT: Coupling time-dependent inactivation parameters derived from pooled human hepatocytes and human liver microsomes (HLM) with a mechanistic static model provides an easy and quantitatively accurate means to determine clinical drug-drug interaction risk from in vitro data. Optimization is needed to evaluate time-dependent inhibition (TDI) for weak and moderate inhibitors using HLM. Recommendations are made with respect to input parameters for in vitro to in vivo extrapolation (IVIVE) of TDI with non-CYP3A enzymes using available data from HLM and human hepatocytes.
Collapse
Affiliation(s)
- Diane Ramsden
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| | - Elke S Perloff
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| | - Andrea Whitcher-Johnstone
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| | - Thuy Ho
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| | - Reena Patel
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| | - Kirk D Kozminski
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| | - Cody L Fullenwider
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| | - J George Zhang
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| |
Collapse
|
8
|
Di L. An update on the importance of plasma protein binding in drug discovery and development. Expert Opin Drug Discov 2021; 16:1453-1465. [PMID: 34403271 DOI: 10.1080/17460441.2021.1961741] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: Plasma protein binding (PPB) remains a controversial topic in drug discovery and development. Fraction unbound (fu) is a critical parameter that needs to be measured accurately, because it has significant impacts on the predictions of drug-drug interactions (DDI), estimations of therapeutic indices (TI), and developments of PK/PD relationships. However, it is generally not advisable to change PPB through structural modifications, because PPB on its own has little relevance for in vivo efficacy.Areas covered: PPB fundamentals are discussed including the three main classes of drug binding proteins (i.e., albumin, alpha1-acid glycoprotein, and lipoproteins) and their physicochemical properties, in vivo half-life, and synthesis rate. State-of-the-art methodologies for PPB are highlighted. Applications of PPB in drug discovery and development are presented.Expert opinion: PPB is an old topic in pharmacokinetics, but there are still many misconceptions. Improving the accuracy of PPB for highly bound compounds is an ongoing effort in the field with high priority. As the field continues to generate high quality data, the regulatory agencies will increase their confidence in our ability to accurately measure PPB of highly bound compounds, and experimental fu values below 0.01 will more likely be used for DDI predictions in the future.
Collapse
Affiliation(s)
- Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT, US
| |
Collapse
|
9
|
Di L, Riccardi K, Tess D. Evolving approaches on measurements and applications of intracellular free drug concentration and Kp uu in drug discovery. Expert Opin Drug Metab Toxicol 2021; 17:733-746. [PMID: 34058926 DOI: 10.1080/17425255.2021.1935866] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Introduction: Intracellular-free drug concentration (Cu,cell) and unbound partition coefficient (Kpuu) are two important parameters to develop pharmacokinetic and pharmacodynamic relationships, predict drug-drug interaction potentials and estimate therapeutic indices.Area covered: Methods on measurements of Cu,cell, Kpuu, partition coefficient (Kp) and fraction unbound of cells (fuc) are discussed. Advantages and limitations of several fuc methods are reviewed. Applications highlighted here are bridging the potency gaps between biochemical and cell-based assays, in vitro hepatocyte assay to predict in vivo liver-to-plasma Kpuu, the role of Kpuu in prediction of hepatic clearance for enzyme- and transporter-mediated mechanisms using extended clearance equation, and structural attributes governing tissue Kpuu.Expert opinion: Cu,cell and Kpuu are of growing applications in drug discovery. Methods for measurements of these properties continue to evolve in order to achieve higher precision/accuracy and obtain more detailed information at the subcellular levels. Future directions of the field include the development of in vitro and in silico models to predict tissue Kpuu, direct measurement of free drug concentration in subcellular organelles, and further investigations into the critical elements governing cell and tissue Kpuu. Significant innovation is needed to advance this complex, but highly impactful and exciting area of science.
Collapse
Affiliation(s)
- Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT, USA
| | - Keith Riccardi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT, USA.,Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Cambridge, MA
| | - David Tess
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT, USA.,Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Cambridge, MA
| |
Collapse
|
10
|
Abstract
The study of enzyme kinetics in drug metabolism involves assessment of rates of metabolism and inhibitory potencies over a suitable concentration range. In all but the very simplest in vitro system, these drug concentrations can be influenced by a variety of nonspecific binding reservoirs that can reduce the available concentration to the enzyme system(s) under investigation. As a consequence, the apparent kinetic parameters, such as Km or Ki, that are derived can deviate from the true values. There are a number of sources of these nonspecific binding depots or barriers, including membrane permeation and partitioning, plasma or serum protein binding, and incubational binding. In the latter case, this includes binding to the assay apparatus as well as biological depots, depending on the characteristics of the in vitro matrix being used. Given the wide array of subcellular, cellular, and recombinant enzyme systems utilized in drug metabolism, each of these has different components which can influence the free drug concentration. The physicochemical properties of the test compound are also paramount in determining the influential factors in any deviation between true and apparent kinetic behavior. This chapter describes the underlying mechanisms determining the free drug concentration in vitro and how these factors can be accounted for in drug metabolism studies, illustrated with case studies from the literature.
Collapse
Affiliation(s)
- Nigel J Waters
- Preclinical Development, Black Diamond Therapeutics, Cambridge, MA, USA
| | - R Scott Obach
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc, Groton, CT, USA
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc, Groton, CT, USA
| |
Collapse
|
11
|
Determination of Fraction Unbound and Unbound Partition Coefficient to Estimate Intracellular Free Drug Concentration. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2021. [DOI: 10.1007/978-1-0716-1250-7_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
12
|
Keefer C, Chang G, Carlo A, Novak JJ, Banker M, Carey J, Cianfrogna J, Eng H, Jagla C, Johnson N, Jones R, Jordan S, Lazzaro S, Liu J, Scott Obach R, Riccardi K, Tess D, Umland J, Racich J, Varma M, Visswanathan R, Di L. Mechanistic insights on clearance and inhibition discordance between liver microsomes and hepatocytes when clearance in liver microsomes is higher than in hepatocytes. Eur J Pharm Sci 2020; 155:105541. [DOI: 10.1016/j.ejps.2020.105541] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/18/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023]
|
13
|
Van Brantegem P, Chatterjee S, De Bruyn T, Annaert P, Deferm N. Drug-induced cholestasis assay in primary hepatocytes. MethodsX 2020; 7:101080. [PMID: 33088729 PMCID: PMC7559231 DOI: 10.1016/j.mex.2020.101080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/23/2020] [Indexed: 01/14/2023] Open
Abstract
Drug-induced cholestasis (DIC) is a major cause of clinical failure of drug candidates. Numerous patients worldwide are affected when exposed to marketed drugs exhibiting a DIC signature. Prospective identification of DIC during early compound development remains challenging. Here we describe the optimized in vitro procedure for early assessment and prediction of an increased DIC risk. Our method is based on three principles:•Exposure of primary human hepatocyte cultures to test compounds in the absence and presence of a physiologically relevant mixture of endogenous bile salts.•Rapid and quantitative assessment of the influence of concomitant bile salt exposure on hepatocyte functionality and integrity after 24 h or 48 h of incubation.•Translation of the in vitro result, expressed as a DIC index (DICI) value, into an in vivo safety margin.Using our historical control data, a new (data driven) DICI cut-off value of 0.78 was established for discerning cholestatic and non-cholestatic compounds. Our DIC assay protocol was further improved by now relying on the principle of the no observable adverse effect level (NOAEL) for determining the highest test compound concentration corresponding to a DICI ≥ 0.78. Predicted safety margin values were subsequently calculated for compounds displaying hepatotoxic and/or cholestatic effects in patients, thus enabling evaluation of the performance of our DIC assay. Of note, this assay can be extended to explore the role of drug metabolites in precipitating DIC.
Collapse
Affiliation(s)
- Pieter Van Brantegem
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Sagnik Chatterjee
- Pharmaceutical Candidate Optimization, Biocon, Bristol-Myers Squibb R& D Center (BBRC), Syngene International Ltd., Bangalore, India
| | - Tom De Bruyn
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc, South San Francisco, CA, USA
| | - Pieter Annaert
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Neel Deferm
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| |
Collapse
|
14
|
Riccardi K, Ryu S, Tess D, Li R, Luo L, Johnson N, Jordan S, Patel R, Di L. Comparison of Fraction Unbound Between Liver Homogenate and Hepatocytes at 4°C. AAPS JOURNAL 2020; 22:91. [DOI: 10.1208/s12248-020-00476-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/26/2020] [Indexed: 01/18/2023]
|
15
|
Ryu S, Riccardi K, Patel R, Zueva L, Burchett W, Di L. Applying Two Orthogonal Methods to Assess Accuracy of Plasma Protein Binding Measurements for Highly Bound Compounds. J Pharm Sci 2019; 108:3745-3749. [DOI: 10.1016/j.xphs.2019.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/15/2019] [Accepted: 08/06/2019] [Indexed: 12/17/2022]
|
16
|
Zhang D, Hop CECA, Patilea-Vrana G, Gampa G, Seneviratne HK, Unadkat JD, Kenny JR, Nagapudi K, Di L, Zhou L, Zak M, Wright MR, Bumpus NN, Zang R, Liu X, Lai Y, Khojasteh SC. Drug Concentration Asymmetry in Tissues and Plasma for Small Molecule-Related Therapeutic Modalities. Drug Metab Dispos 2019; 47:1122-1135. [PMID: 31266753 PMCID: PMC6756291 DOI: 10.1124/dmd.119.086744] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/10/2019] [Indexed: 02/06/2023] Open
Abstract
The well accepted "free drug hypothesis" for small-molecule drugs assumes that only the free (unbound) drug concentration at the therapeutic target can elicit a pharmacologic effect. Unbound (free) drug concentrations in plasma are readily measurable and are often used as surrogates for the drug concentrations at the site of pharmacologic action in pharmacokinetic-pharmacodynamic analysis and clinical dose projection in drug discovery. Furthermore, for permeable compounds at pharmacokinetic steady state, the free drug concentration in tissue is likely a close approximation of that in plasma; however, several factors can create and maintain disequilibrium between the free drug concentration in plasma and tissue, leading to free drug concentration asymmetry. These factors include drug uptake and extrusion mechanisms involving the uptake and efflux drug transporters, intracellular biotransformation of prodrugs, membrane receptor-mediated uptake of antibody-drug conjugates, pH gradients, unique distribution properties (covalent binders, nanoparticles), and local drug delivery (e.g., inhalation). The impact of these factors on the free drug concentrations in tissues can be represented by K p,uu, the ratio of free drug concentration between tissue and plasma at steady state. This review focuses on situations in which free drug concentrations in tissues may differ from those in plasma (e.g., K p,uu > or <1) and discusses the limitations of the surrogate approach of using plasma-free drug concentration to predict free drug concentrations in tissue. This is an important consideration for novel therapeutic modalities since systemic exposure as a driver of pharmacologic effects may provide limited value in guiding compound optimization, selection, and advancement. Ultimately, a deeper understanding of the relationship between free drug concentrations in plasma and tissues is needed.
Collapse
Affiliation(s)
- Donglu Zhang
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Cornelis E C A Hop
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Gabriela Patilea-Vrana
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Gautham Gampa
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Herana Kamal Seneviratne
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Jashvant D Unadkat
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Jane R Kenny
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Karthik Nagapudi
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Li Di
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Lian Zhou
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Mark Zak
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Matthew R Wright
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Namandjé N Bumpus
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Richard Zang
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Xingrong Liu
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - Yurong Lai
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| | - S Cyrus Khojasteh
- Genentech, South San Francisco, California (D.Z., C.E.C.A.H., J.R.K., K.N., M.Z., M.R.W., R.Z., S.C.K.); Department of Medicine, Division of Clinical Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, Maryland (H.K.S., N.N.B.); Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (G.G.); Department of Pharmaceutics, University of Washington, Seattle, Washington (G.P.-V., J.D.U.); Biogen, Cambridge, Massachusetts (X.L.); Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, Connecticut (L.D.); Drug Disposition, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (L.Z.); and Drug Metabolism, Gilead Sciences, Foster City, California (Y.L.)
| |
Collapse
|
17
|
Treyer A, Ullah M, Parrott N, Molitor B, Fowler S, Artursson P. Impact of Intracellular Concentrations on Metabolic Drug-Drug Interaction Studies. AAPS JOURNAL 2019; 21:77. [PMID: 31214810 PMCID: PMC6581936 DOI: 10.1208/s12248-019-0344-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 05/23/2019] [Indexed: 12/16/2022]
Abstract
Accurate prediction of drug-drug interactions (DDI) is a challenging task in drug discovery and development. It requires determination of enzyme inhibition in vitro which is highly system-dependent for many compounds. The aim of this study was to investigate whether the determination of intracellular unbound concentrations in primary human hepatocytes can be used to bridge discrepancies between results obtained using human liver microsomes and hepatocytes. Specifically, we investigated if Kpuu could reconcile differences in CYP enzyme inhibition values (Ki or IC50). Firstly, our methodology for determination of Kpuu was optimized for human hepatocytes, using four well-studied reference compounds. Secondly, the methodology was applied to a series of structurally related CYP2C9 inhibitors from a Roche discovery project. Lastly, the Kpuu values of three commonly used CYP3A4 inhibitors—ketoconazole, itraconazole, and posaconazole—were determined and compared to compound-specific hepatic enrichment factors obtained from physiologically based modeling of clinical DDI studies with these three compounds. Kpuu obtained in suspended human hepatocytes gave good predictions of system-dependent differences in vitro. The Kpuu was also in fair agreement with the compound-specific hepatic enrichment factors in DDI models and can therefore be used to improve estimations of enrichment factors in physiologically based pharmacokinetic modeling.
Collapse
Affiliation(s)
- Andrea Treyer
- Department of Pharmacy, Uppsala University, Box 580, SE-751 23, Uppsala, Sweden
| | - Mohammed Ullah
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Neil Parrott
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Birgit Molitor
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Stephen Fowler
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Box 580, SE-751 23, Uppsala, Sweden. .,Science for Life Laboratory Drug Discovery and Development platform (SciLifelab DDD-P), Uppsala, Sweden. .,Uppsala University Drug Optimization and Pharmaceutical Profiling Platform (UDOPP), Uppsala University, Uppsala, Sweden.
| |
Collapse
|
18
|
Riccardi KA, Tess DA, Lin J, Patel R, Ryu S, Atkinson K, Di L, Li R. A Novel Unified Approach to Predict Human Hepatic Clearance for Both Enzyme- and Transporter-Mediated Mechanisms Using Suspended Human Hepatocytes. Drug Metab Dispos 2019; 47:484-492. [PMID: 30787098 DOI: 10.1124/dmd.118.085639] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 02/05/2019] [Indexed: 02/13/2025] Open
Abstract
The accurate prediction of human pharmacokinetics is critically important in modern drug discovery since it drives both pharmacological and toxicological effects. Although significant progress has been made in predicting drug disposition by hepatic drug-metabolizing enzymes, predicting transporter-mediated clearance is still highly uncertain. Furthermore, different approaches are often used to predict clearance with and without transporter involvement, hence the major clearance pathway for a compound must first be determined to know which approach to use. As a result of these challenges, a novel unified method has been developed using cryopreserved suspended human hepatocytes to predict human hepatic clearance for both enzyme- and transporter-mediated mechanisms. This method hypothesizes that, once in vitro metabolic stability is scaled by partition coefficients between hepatocytes and buffer with 4% bovine serum albumin, in vivo clearance can be better predicted. With this method, good in vitro-in vivo correlation of human hepatic clearance has been obtained for a set of 32 structurally diverse compounds, including such transporters as organic anion-transporting polypeptide substrates. The clearance predictions for most compounds are within 3-fold of observed values. This is the first time that multiple compounds result in good in vitro-in vivo extrapolation using an entirely "bottom-up" approach without any empirical scaling factor when transporter-mediated clearance is involved. Potential exceptions are compounds with significant biliary and/or extra-hepatic clearance. The method offers an alternative approach to more accurately predict human hepatic clearance when multiple complex mechanisms are involved.
Collapse
Affiliation(s)
- Keith A Riccardi
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (K.A.R., J.L., R.P., S.R., K.A., L.D.) and Systems Modeling and Simulation, Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts (D.A.T., R.L.)
| | - David A Tess
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (K.A.R., J.L., R.P., S.R., K.A., L.D.) and Systems Modeling and Simulation, Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts (D.A.T., R.L.)
| | - Jian Lin
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (K.A.R., J.L., R.P., S.R., K.A., L.D.) and Systems Modeling and Simulation, Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts (D.A.T., R.L.)
| | - Roshan Patel
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (K.A.R., J.L., R.P., S.R., K.A., L.D.) and Systems Modeling and Simulation, Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts (D.A.T., R.L.)
| | - Sangwoo Ryu
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (K.A.R., J.L., R.P., S.R., K.A., L.D.) and Systems Modeling and Simulation, Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts (D.A.T., R.L.)
| | - Karen Atkinson
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (K.A.R., J.L., R.P., S.R., K.A., L.D.) and Systems Modeling and Simulation, Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts (D.A.T., R.L.)
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (K.A.R., J.L., R.P., S.R., K.A., L.D.) and Systems Modeling and Simulation, Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts (D.A.T., R.L.)
| | - Rui Li
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut (K.A.R., J.L., R.P., S.R., K.A., L.D.) and Systems Modeling and Simulation, Medicine Design, Pfizer Worldwide R&D, Cambridge, Massachusetts (D.A.T., R.L.)
| |
Collapse
|
19
|
Li Z, Di L, Maurer TS. Theoretical Considerations for Direct Translation of Unbound Liver-to-Plasma Partition Coefficient from In Vitro to In Vivo. AAPS JOURNAL 2019; 21:43. [DOI: 10.1208/s12248-019-0314-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/26/2019] [Indexed: 12/25/2022]
|
20
|
Willmes DM, Kurzbach A, Henke C, Schumann T, Zahn G, Heifetz A, Jordan J, Helfand SL, Birkenfeld AL. The longevity gene INDY ( I 'm N ot D ead Y et) in metabolic control: Potential as pharmacological target. Pharmacol Ther 2018; 185:1-11. [DOI: 10.1016/j.pharmthera.2017.10.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
21
|
Treyer A, Mateus A, Wiśniewski JR, Boriss H, Matsson P, Artursson P. Intracellular Drug Bioavailability: Effect of Neutral Lipids and Phospholipids. Mol Pharm 2018; 15:2224-2233. [DOI: 10.1021/acs.molpharmaceut.8b00064] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Andrea Treyer
- Department of Pharmacy, Uppsala University, Uppsala 75123, Sweden
| | - André Mateus
- Department of Pharmacy, Uppsala University, Uppsala 75123, Sweden
| | - Jacek R Wiśniewski
- Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | | | - Pär Matsson
- Department of Pharmacy, Uppsala University, Uppsala 75123, Sweden
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Uppsala 75123, Sweden
- Science for Life Laboratory Drug Discovery and Development Platform (SciLifelab DDD-P), Uppsala 75123, Sweden
- Uppsala University Drug Optimization and Pharmaceutical Profiling Platform (UDOPP), Uppsala University, Uppsala 75123, Sweden
| |
Collapse
|
22
|
Riccardi K, Ryu S, Lin J, Yates P, Tess D, Li R, Singh D, Holder BR, Kapinos B, Chang G, Di L. Comparison of Species and Cell-Type Differences in Fraction Unbound of Liver Tissues, Hepatocytes, and Cell Lines. Drug Metab Dispos 2018; 46:415-421. [PMID: 29437874 DOI: 10.1124/dmd.117.079152] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 01/24/2018] [Indexed: 01/02/2023] Open
Abstract
Fraction unbound (fu) of liver tissue, hepatocytes, and other cell types is an essential parameter used to estimate unbound liver drug concentration and intracellular free drug concentration. fu,liver and fu,cell are frequently measured in multiple species and cell types in drug discovery and development for various applications. A comparison study of 12 matrices for fu,liver and fu,cell of hepatocytes in five different species (mouse, rat, dog, monkey, and human), as well as fu,cell of Huh7 and human embryonic kidney 293 cell lines, was conducted for 22 structurally diverse compounds with the equilibrium dialysis method. Using an average bioequivalence approach, our results show that the average difference in binding to liver tissue, hepatocytes, or different cell types was within 2-fold of that of the rat fu,liver Therefore, we recommend using rat fu,liver as a surrogate for liver binding in other species and cell types in drug discovery. This strategy offers the potential to simplify binding studies and reduce cost, thereby enabling a more effective and practical determination of fu for liver tissues, hepatocytes, and other cell types. In addition, fu under hepatocyte stability incubation conditions should not be confused with fu,cell, as one is a diluted fu and the other is an undiluted fu Cell density also plays a critical role in the accurate measurement of fu,cell.
Collapse
Affiliation(s)
- Keith Riccardi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., S.R., J.L., D.S., B.R.H., B.K., G.C., L.D.); and Early Clinical Development (P.Y.), and Pharmacokinetics, Dynamics and Metabolism (D.T., R.L.), Pfizer Inc., Cambridge, Massachusetts
| | - Sangwoo Ryu
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., S.R., J.L., D.S., B.R.H., B.K., G.C., L.D.); and Early Clinical Development (P.Y.), and Pharmacokinetics, Dynamics and Metabolism (D.T., R.L.), Pfizer Inc., Cambridge, Massachusetts
| | - Jian Lin
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., S.R., J.L., D.S., B.R.H., B.K., G.C., L.D.); and Early Clinical Development (P.Y.), and Pharmacokinetics, Dynamics and Metabolism (D.T., R.L.), Pfizer Inc., Cambridge, Massachusetts
| | - Phillip Yates
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., S.R., J.L., D.S., B.R.H., B.K., G.C., L.D.); and Early Clinical Development (P.Y.), and Pharmacokinetics, Dynamics and Metabolism (D.T., R.L.), Pfizer Inc., Cambridge, Massachusetts
| | - David Tess
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., S.R., J.L., D.S., B.R.H., B.K., G.C., L.D.); and Early Clinical Development (P.Y.), and Pharmacokinetics, Dynamics and Metabolism (D.T., R.L.), Pfizer Inc., Cambridge, Massachusetts
| | - Rui Li
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., S.R., J.L., D.S., B.R.H., B.K., G.C., L.D.); and Early Clinical Development (P.Y.), and Pharmacokinetics, Dynamics and Metabolism (D.T., R.L.), Pfizer Inc., Cambridge, Massachusetts
| | - Dhirender Singh
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., S.R., J.L., D.S., B.R.H., B.K., G.C., L.D.); and Early Clinical Development (P.Y.), and Pharmacokinetics, Dynamics and Metabolism (D.T., R.L.), Pfizer Inc., Cambridge, Massachusetts
| | - Brian R Holder
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., S.R., J.L., D.S., B.R.H., B.K., G.C., L.D.); and Early Clinical Development (P.Y.), and Pharmacokinetics, Dynamics and Metabolism (D.T., R.L.), Pfizer Inc., Cambridge, Massachusetts
| | - Brendon Kapinos
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., S.R., J.L., D.S., B.R.H., B.K., G.C., L.D.); and Early Clinical Development (P.Y.), and Pharmacokinetics, Dynamics and Metabolism (D.T., R.L.), Pfizer Inc., Cambridge, Massachusetts
| | - George Chang
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., S.R., J.L., D.S., B.R.H., B.K., G.C., L.D.); and Early Clinical Development (P.Y.), and Pharmacokinetics, Dynamics and Metabolism (D.T., R.L.), Pfizer Inc., Cambridge, Massachusetts
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., S.R., J.L., D.S., B.R.H., B.K., G.C., L.D.); and Early Clinical Development (P.Y.), and Pharmacokinetics, Dynamics and Metabolism (D.T., R.L.), Pfizer Inc., Cambridge, Massachusetts
| |
Collapse
|
23
|
Industry Perspective on Contemporary Protein-Binding Methodologies: Considerations for Regulatory Drug-Drug Interaction and Related Guidelines on Highly Bound Drugs. J Pharm Sci 2017; 106:3442-3452. [DOI: 10.1016/j.xphs.2017.09.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/07/2017] [Indexed: 11/21/2022]
|
24
|
Riede J, Camenisch G, Huwyler J, Poller B. Current In Vitro Methods to Determine Hepatic Kp uu : A Comparison of Their Usefulness and Limitations. J Pharm Sci 2017; 106:2805-2814. [DOI: 10.1016/j.xphs.2017.03.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 12/20/2022]
|
25
|
Sun Y, Chothe PP, Sager JE, Tsao H, Moore A, Laitinen L, Hariparsad N. Quantitative Prediction of CYP3A4 Induction: Impact of Measured, Free, and Intracellular Perpetrator Concentrations from Human Hepatocyte Induction Studies on Drug-Drug Interaction Predictions. Drug Metab Dispos 2017; 45:692-705. [PMID: 28336578 DOI: 10.1124/dmd.117.075481] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 03/21/2017] [Indexed: 02/13/2025] Open
Abstract
Typically, concentration-response curves are based upon nominal inducer concentrations for in-vitro-to-in-vivo extrapolation of CYP3A4 induction. The limitation of this practice is that it assumes the hepatocyte culture model is a static system. We assessed whether correcting for: 1) changes in perpetrator concentration in the induction medium during the incubation period, 2) perpetrator binding to proteins in the induction medium, and 3) nonspecific binding of perpetrator can improve the accuracy of CYP3A4 induction predictions. Of the seven compounds used in this evaluation, significant parent loss and nonspecific binding were observed for rifampicin (29.3-38.3%), pioglitazone (64.3-78.6%), and rosiglitazone (57.1-75.5%). As a result, the free measured EC50 values (EC50u) of pioglitazone, rosiglitazone, and rifampicin were significantly lower than the nominal EC50 values. In general, the accuracy of the induction predictions, using multiple static models, improved when corrections were made for measured medium concentrations, medium protein binding, and nonspecific binding of the perpetrator, as evidenced by 18-29% reductions in the root mean square error. The relative induction score model performed better than the basic static and mechanistic static models, resulting in lower prediction error and no false-positive or false-negative predictions. However, even when the EC50u value was used, the induction prediction for bosentan, which is a substrate of organic anion transporter proteins, was overpredicted by approximately 2-fold. Accounting for the ratio of unbound intracellular concentrations to unbound medium concentrations (Kpuu,in vitro) (0.5-7.5) and the predicted multiple-dose Kpuu,in vivo (0.6) for bosentan resulted in induction predictions within 35% of the observed interaction.
Collapse
Affiliation(s)
- Yongkai Sun
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Paresh P Chothe
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Jennifer E Sager
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Hong Tsao
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Amanda Moore
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Leena Laitinen
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Niresh Hariparsad
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Inc., Boston, Massachusetts
| |
Collapse
|
26
|
Riccardi K, Lin J, Li Z, Niosi M, Ryu S, Hua W, Atkinson K, Kosa RE, Litchfield J, Di L. Novel Method to Predict In Vivo Liver-to-Plasma K puu for OATP Substrates Using Suspension Hepatocytes. Drug Metab Dispos 2017; 45:576-580. [PMID: 28258068 DOI: 10.1124/dmd.116.074575] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/01/2017] [Indexed: 02/13/2025] Open
Abstract
The ability to predict human liver-to-plasma unbound partition coefficient (Kpuu) is of great importance to estimate unbound liver concentration, develop PK/PD relationships, predict efficacy and toxicity in the liver, and model the drug-drug interaction potential for drugs that are asymmetrically distributed into the liver. A novel in vitro method has been developed to predict in vivo Kpuu with good accuracy using cryopreserved suspension hepatocytes in InVitroGRO HI media with 4% BSA. Validation was performed using six OATP substrates with rat in vivo Kpuu data from i.v. infusion studies where a steady state was achieved. Good in vitro-in vivo correlation (IVIVE) was observed as the in vitro Kpuu values were mostly within 2-fold of in vivo Kpuu Good Kpuu IVIVE in human was also observed with in vivo Kpuu data of dehydropravastatin from positron emission tomography and in vivo Kpuu data from PK/PD modeling for pravastatin and rosuvastatin. Under the specific Kpuu assay conditions, the drug-metabolizing enzymes and influx/efflux transporters appear to function at physiologic levels. No scaling factors are necessary to predict in vivo Kpuu from in vitro data. The novel in vitro Kpuu method provides a useful tool in drug discovery to project in vivo Kpuu.
Collapse
Affiliation(s)
- Keith Riccardi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Jian Lin
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Zhenhong Li
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Mark Niosi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Sangwoo Ryu
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Wenyi Hua
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Karen Atkinson
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Rachel E Kosa
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - John Litchfield
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| |
Collapse
|