1
|
Foti RS. Utility of physiologically based pharmacokinetic modeling in predicting and characterizing clinical drug interactions. Drug Metab Dispos 2025; 53:100021. [PMID: 39884811 DOI: 10.1124/dmd.123.001384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/09/2023] [Accepted: 01/02/2024] [Indexed: 02/01/2024] Open
Abstract
Physiologically based pharmacokinetic (PBPK) modeling is a mechanistic dynamic modeling approach that can be used to predict or retrospectively describe changes in drug exposure due to drug-drug interactions (DDIs). With advancements in commercially available PBPK software, PBPK DDI modeling has become a mainstream approach from early drug discovery through to late-stage drug development and is often used to support regulatory packages for new drug applications. This Minireview will briefly describe the approaches to predicting DDI using PBPK and static modeling approaches, the basic model structures and features inherent to PBPK DDI models, and key examples where PBPK DDI models have been used to describe complex DDI mechanisms. Future directions aimed at using PBPK models to characterize transporter-mediated DDI, predict DDI in special populations, and assess the DDI potential of protein therapeutics will be discussed. A summary of the 209 PBPK DDI examples published to date in 2023 will be provided. Overall, current data and trends suggest a continued role for PBPK models in the characterization and prediction of DDI for therapeutic molecules. SIGNIFICANCE STATEMENT: Physiologically based pharmacokinetic (PBPK) models have been a key tool in the characterization of various pharmacokinetic phenomena, including drug-drug interactions. This Minireview will highlight recent advancements and publications around physiologically based pharmacokinetic drug-drug interaction modeling, an important area of drug discovery and development research in light of the increasing prevalence of polypharmacology in clinical settings.
Collapse
Affiliation(s)
- Robert S Foti
- Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co, Inc, Boston, Massachusetts.
| |
Collapse
|
2
|
Isoherranen N. Physiologically based pharmacokinetic modeling of small molecules: How much progress have we made? Drug Metab Dispos 2025; 53:100013. [PMID: 39884807 DOI: 10.1124/dmd.123.000960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/10/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024] Open
Abstract
Physiologically based pharmacokinetic (PBPK) models of small molecules have become mainstream in drug development and in academic research. The use of PBPK models is continuously expanding, with the majority of work now focusing on predictions of drug-drug interactions, drug-disease interactions, and changes in drug disposition across lifespan. Recently, publications that use PBPK modeling to predict drug disposition during pregnancy and in organ impairment have increased reflecting the advances in incorporating diverse physiologic changes into the models. Because of the expanding computational power and diversity of modeling platforms available, the complexity of PBPK models has also increased. Academic efforts have provided clear advances in better capturing human physiology in PBPK models and incorporating more complex mathematical concepts into PBPK models. Examples of such advances include the segregated gut model with a series of gut compartments allowing modeling of physiologic blood flow distribution within an organ and zonation of metabolic enzymes and series compartment liver models allowing simulations of hepatic clearance for high extraction drugs. Despite these advances in academic research, the progress in assessing model quality and defining model acceptance criteria based on the intended use of the models has not kept pace. This Minireview suggests that awareness of the need for predefined criteria for model acceptance has increased, but many manuscripts still lack description of scientific justification and/or rationale for chosen acceptance criteria. As artificial intelligence and machine learning approaches become more broadly accepted, these tools offer promise for development of comprehensive assessment for existing observed data and analysis of model performance. SIGNIFICANCE STATEMENT: Physiologically based pharmacokinetic (PBPK) modeling has become a mainstream application in academic literature and is broadly used for predictions, analysis, and evaluation of pharmacokinetic data. Significant progress has been made in developing advanced PBPK models that better capture human physiology, but oftentimes sufficient justification for the chosen model acceptance criterion and model structure is still missing. This Minireview provides a summary of the current landscape of PBPK applications used and highlights the need for advancing PBPK modeling science and training in academia.
Collapse
Affiliation(s)
- Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington.
| |
Collapse
|
3
|
Prieto Garcia L, Vildhede A, Nordell P, Ahlström C, Montaser AB, Terasaki T, Lennernäs H, Sjögren E. Physiologically based pharmacokinetics modeling and transporter proteomics to predict systemic and local liver and muscle disposition of statins. CPT Pharmacometrics Syst Pharmacol 2024; 13:1029-1043. [PMID: 38576225 PMCID: PMC11179708 DOI: 10.1002/psp4.13139] [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: 11/22/2023] [Revised: 02/22/2024] [Accepted: 03/25/2024] [Indexed: 04/06/2024] Open
Abstract
Statins are used to reduce liver cholesterol levels but also carry a dose-related risk of skeletal muscle toxicity. Concentrations of statins in plasma are often used to assess efficacy and safety, but because statins are substrates of membrane transporters that are present in diverse tissues, local differences in intracellular tissue concentrations cannot be ruled out. Thus, plasma concentration may not be an adequate indicator of efficacy and toxicity. To bridge this gap, we used physiologically based pharmacokinetic (PBPK) modeling to predict intracellular concentrations of statins. Quantitative data on transporter clearance were scaled from in vitro to in vivo conditions by integrating targeted proteomics and transporter kinetics data. The developed PBPK models, informed by proteomics, suggested that organic anion-transporting polypeptide 2B1 (OATP2B1) and multidrug resistance-associated protein 1 (MRP1) play a pivotal role in the distribution of statins in muscle. Using these PBPK models, we were able to predict the impact of alterations in transporter function due to genotype or drug-drug interactions on statin systemic concentrations and exposure in liver and muscle. These results underscore the potential of proteomics-guided PBPK modeling to scale transporter clearance from in vitro data to real-world implications. It is important to evaluate the role of drug transporters when predicting tissue exposure associated with on- and off-target effects.
Collapse
Affiliation(s)
- Luna Prieto Garcia
- Department of Pharmaceutical Bioscience, Translational Drug Discovery and DevelopmentUppsala UniversityUppsalaSweden
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Anna Vildhede
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Pär Nordell
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Christine Ahlström
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Ahmed B. Montaser
- School of Pharmacy, Faculty of Health SciencesUniversity of Eastern FinlandKuopioFinland
| | - Tetsuya Terasaki
- School of Pharmacy, Faculty of Health SciencesUniversity of Eastern FinlandKuopioFinland
| | - Hans Lennernäs
- Department of Pharmaceutical Bioscience, Translational Drug Discovery and DevelopmentUppsala UniversityUppsalaSweden
| | - Erik Sjögren
- Department of Pharmaceutical Bioscience, Translational Drug Discovery and DevelopmentUppsala UniversityUppsalaSweden
| |
Collapse
|
4
|
Small BG, Hatley O, Jamei M, Gardner I, Johnson TN. Incorporation and Performance Verification of Hepatic Portal Blood Flow Shunting in Minimal and Full PBPK Models of Liver Cirrhosis. Clin Pharmacol Ther 2023; 114:1264-1273. [PMID: 37620290 DOI: 10.1002/cpt.3032] [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: 06/01/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Patho-physiological changes in liver cirrhosis create portacaval shunts that allow blood flow to bypass the hepatic portal vein into the systemic circulation affecting drug pharmacokinetics (PKs). The objectives of this work were to implement a physiologically-based pharmacokinetic (PBPK) framework describing shunted blood flows in virtual patients with differing degrees of liver cirrhosis; and to assess the minimal and full PBPK model's performance using drugs with intermediate to high hepatic extraction. Single dose concentration-time profiles and PK parameters for oral ibrutinib, midazolam, propranolol, and buspirone were simulated in healthy volunteers (HVs) and subjects with cirrhosis (Child-Pugh severity score (CP-A, CP-B, or CP-C)). Model performance was verified by comparing predicted to observed fold-changes in PK parameters between HVs and cirrhotic subjects. The verified model was used to simulate the PK changes for simvastatin in patients with cirrhosis. The predicted area under the curve ratios (AUCCirr :AUCHV ) for ibrutinib were 3.38, 6.87, and 11.46 using the minimal PBPK model with shunt and 1.61, 2.58, and 4.33 without the shunt, these compared with observed values of 4.33, 8.14, and 9.04, respectively. For ibrutinib, propranolol, and buspirone, including a shunt in the PBPK model improved the prediction of the AUCCirr :AUCHV and maximum plasma concentration ratios (CmaxCirr :CmaxHV ). For midazolam, an intermediate extraction drug, the differences were less clear. Simulated simvastatin dose adjustments in cirrhosis suggested that 20 mg in CP-A and 10 mg in CP-B could be used clinically. A mechanistic model-informed understanding of the anatomic and pathophysiology of cirrhosis will facilitate improved dose prediction and adjustment in this vulnerable population.
Collapse
Affiliation(s)
- Ben G Small
- Certara UK Limited (Simcyp Division), Sheffield, UK
| | | | - Masoud Jamei
- Certara UK Limited (Simcyp Division), Sheffield, UK
| | - Iain Gardner
- Certara UK Limited (Simcyp Division), Sheffield, UK
| | | |
Collapse
|
5
|
Dong J, Prieto Garcia L, Huang Y, Tang W, Lundahl A, Elebring M, Ahlström C, Vildhede A, Sjögren E, Någård M. Understanding Statin-Roxadustat Drug-Drug-Disease Interaction Using Physiologically-Based Pharmacokinetic Modeling. Clin Pharmacol Ther 2023; 114:825-835. [PMID: 37376792 DOI: 10.1002/cpt.2980] [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: 02/27/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023]
Abstract
A different drug-drug interaction (DDI) scenario may exist in patients with chronic kidney disease (CKD) compared with healthy volunteers (HVs), depending on the interplay between drug-drug and disease (drug-drug-disease interaction (DDDI)). Physiologically-based pharmacokinetic (PBPK) modeling, in lieu of a clinical trial, is a promising tool for evaluating these complex DDDIs in patients. However, the prediction confidence of PBPK modeling in the severe CKD population is still low when nonrenal pathways are involved. More mechanistic virtual disease population and robust validation cases are needed. To this end, we aimed to: (i) understand the implications of severe CKD on statins (atorvastatin, simvastatin, and rosuvastatin) pharmacokinetics (PK) and DDI; and (ii) predict untested clinical scenarios of statin-roxadustat DDI risks in patients to guide suitable dose regimens. A novel virtual severe CKD population was developed incorporating the disease effect on both renal and nonrenal pathways. Drug and disease PBPK models underwent a four-way validation. The verified PBPK models successfully predicted the altered PKs in patients for substrates and inhibitors and recovered the observed statin-rifampicin DDIs in patients and the statin-roxadustat DDIs in HVs within 1.25- and 2-fold error. Further sensitivity analysis revealed that the severe CKD effect on statins PK is mainly mediated by hepatic BCRP for rosuvastatin and OATP1B1/3 for atorvastatin. The magnitude of statin-roxadustat DDI in patients with severe CKD was predicted to be similar to that in HVs. PBPK-guided suitable dose regimens were identified to minimize the risk of side effects or therapeutic failure of statins when co-administered with roxadustat.
Collapse
Affiliation(s)
- Jin Dong
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Luna Prieto Garcia
- Drug Metabolism and Pharmacokinetics, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Gothenburg, Sweden
- Department of Pharmaceutical Biosciences, Translational Drug Discovery and Development, Uppsala University, Uppsala, Sweden
| | - Yingbo Huang
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Weifeng Tang
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Anna Lundahl
- Drug Metabolism and Pharmacokinetics, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Gothenburg, Sweden
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Marie Elebring
- Drug Metabolism and Pharmacokinetics, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Gothenburg, Sweden
| | - Christine Ahlström
- Drug Metabolism and Pharmacokinetics, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Vildhede
- Drug Metabolism and Pharmacokinetics, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Gothenburg, Sweden
| | - Erik Sjögren
- Department of Pharmaceutical Biosciences, Translational Drug Discovery and Development, Uppsala University, Uppsala, Sweden
| | - Mats Någård
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| |
Collapse
|
6
|
Clewell H, Campbell J, Linakis M. Recent Applications Of Physiologically Based Pharmacokinetic Modeling To Assess The Toxicity Of Mixtures: A Review. CURRENT OPINION IN TOXICOLOGY 2023. [DOI: 10.1016/j.cotox.2023.100390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
7
|
A Physiologically-Based Pharmacokinetic Model of Ruxolitinib and Posaconazole to Predict CYP3A4-Mediated Drug-Drug Interaction Frequently Observed in Graft versus Host Disease Patients. Pharmaceutics 2022; 14:pharmaceutics14122556. [PMID: 36559050 PMCID: PMC9785192 DOI: 10.3390/pharmaceutics14122556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/13/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Ruxolitinib (RUX) is approved for the treatment of steroid-refractory acute and chronic graft versus host disease (GvHD). It is predominantly metabolized via cytochrome P450 (CYP) 3A4. As patients with GvHD have an increased risk of invasive fungal infections, RUX is frequently combined with posaconazole (POS), a strong CYP3A4 inhibitor. Knowledge of RUX exposure under concomitant POS treatment is scarce and recommendations on dose modifications are inconsistent. A physiologically based pharmacokinetic (PBPK) model was developed to investigate the drug-drug interaction (DDI) between POS and RUX. The predicted RUX exposure was compared to observed concentrations in patients with GvHD in the clinical routine. PBPK models for RUX and POS were independently set up using PK-Sim® Version 11. Plasma concentration-time profiles were described successfully and all predicted area under the curve (AUC) values were within 2-fold of the observed values. The increase in RUX exposure was predicted with a DDI ratio of 1.21 (Cmax) and 1.59 (AUC). Standard dosing in patients with GvHD led to higher RUX exposure than expected, suggesting further dose reduction if combined with POS. The developed model can serve as a starting point for further simulations of the implemented DDI and can be extended to further perpetrators of CYP-mediated PK-DDIs or disease-specific physiological changes.
Collapse
|