1
|
Umehara K, Parrott N, Schindler E, Legras V, Meneses-Lorente G. PBPK Modeling of Entrectinib and Its Active Metabolite to Derive Dose Adjustments in Pediatric Populations Co-Administered with CYP3A4 Inhibitors. Clin Pharmacol Ther 2024; 116:1130-1140. [PMID: 39023380 DOI: 10.1002/cpt.3386] [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: 04/02/2024] [Accepted: 06/29/2024] [Indexed: 07/20/2024]
Abstract
Physiologically based pharmacokinetic (PBPK) models of entrectinib and its equipotent metabolite, M5, were established in healthy adult subjects and extrapolated to pediatric patients to predict increases in steady-state systemic exposure on co-administration of strong and moderate CYP3A4 inhibitors (itraconazole at 5 mg/kg, erythromycin at 7.5-12.5 mg/kg and fluconazole at 3-12 mg/kg, respectively). Adult model establishment involved the optimization of fraction metabolized by CYP3A4 (0.92 for entrectinib and 0.98 for M5) using data from an itraconazole DDI study. This model captured well the exposure changes of entrectinib and M5 seen in adults co-administered with the strong CYP3A4 inducer rifampicin. In pediatrics, reasonable prediction of entrectinib and M5 pharmacokinetics in ≧2 year olds was achieved when using the default models for physiological development and enzyme ontogenies. However, a two to threefold misprediction of entrectinib and M5 exposures was seen in <2 year olds which may be due to missing mechanistic understanding of gut physiology and/or protein binding in very young children. Model predictions for ≧2 year olds showed that entrectinib AUC(0-t) was increased by approximately sevenfold and five to threefold by strong and high-moderate and low-moderate CYP3A4 inhibitors, respectively. Based on these victim DDI predictions, dose adjustments for entrectinib when given concomitantly with strong and moderate CYP3A4 inhibitors in pediatric subjects were recommended. These simulations informed the approved entrectinib label without the need for additional clinical pharmacology studies.
Collapse
Affiliation(s)
- Kenichi Umehara
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Neil Parrott
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Emilie Schindler
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Valentin Legras
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Georgina Meneses-Lorente
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Roche Products Ltd, Welwyn, UK
| |
Collapse
|
2
|
Kollipara S, Chougule M, Boddu R, Bhatia A, Ahmed T. Playing Hide-and-Seek with Tyrosine Kinase Inhibitors: Can We Overcome Administration Challenges? AAPS J 2024; 26:66. [PMID: 38862853 DOI: 10.1208/s12248-024-00939-1] [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: 04/06/2024] [Accepted: 05/29/2024] [Indexed: 06/13/2024] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have demonstrated significant efficacy against various types of cancers through molecular targeting mechanisms. Over the past 22 years, more than 100 TKIs have been approved for the treatment of various types of cancer indicating the significant progress achieved in this research area. Despite having significant efficacy and ability to target multiple pathways, TKIs administration is associated with challenges. There are reported inconsistencies between observed food effect and labeling administration, challenges of concomitant administration with acid-reducing agents (ARA), pill burden and dosing frequency. In this context, the objective of present review is to visit administration challenges of TKIs and effective ways to tackle them. We have gathered data of 94 TKIs approved in between 2000 and 2022 with respect to food effect, ARA impact, administration schemes (food and PPI restrictions), number of pills per day and administration frequency. Further, trend analysis has been performed to identify inconsistencies in the labeling with respect to observed food effect, molecules exhibiting ARA impact, in order to identify solutions to remove these restrictions through novel formulation approaches. Additionally, opportunities to reduce number of pills per day and dosing frequency for better patient compliance were suggested using innovative formulation interventions. Finally, utility of physiologically based pharmacokinetic modeling (PBPK) for rationale formulation development was discussed with literature reported examples. Overall, this review can act as a ready-to-use-guide for the formulation, biopharmaceutics scientists and medical oncologists to identify opportunities for innovation for TKIs.
Collapse
Affiliation(s)
- Sivacharan Kollipara
- Biopharmaceutics Group, Global Clinical Management, Dr. Reddy's Laboratories Ltd, Integrated Product Development Organization (IPDO), Bachupally, Medchal Malkajgiri District, Hyderabad, 500 090, Telangana, India
| | - Mahendra Chougule
- Biopharmaceutics Group, Global Clinical Management, Dr. Reddy's Laboratories Ltd, Integrated Product Development Organization (IPDO), Bachupally, Medchal Malkajgiri District, Hyderabad, 500 090, Telangana, India
| | - Rajkumar Boddu
- Biopharmaceutics Group, Global Clinical Management, Dr. Reddy's Laboratories Ltd, Integrated Product Development Organization (IPDO), Bachupally, Medchal Malkajgiri District, Hyderabad, 500 090, Telangana, India
| | - Ashima Bhatia
- Biopharmaceutics Group, Global Clinical Management, Dr. Reddy's Laboratories Ltd, Integrated Product Development Organization (IPDO), Bachupally, Medchal Malkajgiri District, Hyderabad, 500 090, Telangana, India
| | - Tausif Ahmed
- Biopharmaceutics Group, Global Clinical Management, Dr. Reddy's Laboratories Ltd, Integrated Product Development Organization (IPDO), Bachupally, Medchal Malkajgiri District, Hyderabad, 500 090, Telangana, India.
| |
Collapse
|
3
|
Wu D, Liu J, Paragas EM, Yadav J, Aliwarga T, Heimbach T, Escotet-Espinoza MS. Assessing and mitigating pH-mediated DDI risks in drug development - formulation approaches and clinical considerations. Drug Metab Rev 2024:1-20. [PMID: 38700278 DOI: 10.1080/03602532.2024.2345632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 04/10/2024] [Indexed: 05/05/2024]
Abstract
pH-mediated drug-drug interactions (DDI) is a prevalent DDI in drug development, especially for weak base compounds with highly pH-dependent solubility. FDA has released a guidance on the evaluation of pH-mediated DDI assessments using in vitro testing and clinical studies. Currently, there is no common practice of ways of testing across the academia and industry. The development of biopredictive method and physiologically-based biopharmaceutics modeling (PBBM) approaches to assess acid-reducing agent (ARA)-DDI have been proven with accurate prediction and could decrease drug development burden, inform clinical design and potentially waive clinical studies. Formulation strategies and careful clinical design could help mitigate the pH-mediated DDI to avoid more clinical studies and label restrictions, ultimately benefiting the patient. In this review paper, a detailed introduction on biorelevant dissolution testing, preclinical and clinical study requirement and PBPK modeling approaches to assess ARA-DDI are described. An improved decision tree for pH-mediated DDI is proposed. Potential mitigations including clinical or formulation strategies are discussed.
Collapse
Affiliation(s)
- Di Wu
- Pharmaceutical Sciences & Clinical Supply, Merck & Co., Inc, Rahway, NJ, USA
| | - Jiaying Liu
- Pharmaceutical Sciences & Clinical Supply, Merck & Co., Inc, Rahway, NJ, USA
| | - Erickson M Paragas
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, South San Francisco, CA, USA
| | - Jaydeep Yadav
- Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Inc, Boston, MA, USA
| | - Theresa Aliwarga
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, South San Francisco, CA, USA
| | - Tycho Heimbach
- Pharmaceutical Sciences & Clinical Supply, Merck & Co., Inc, Rahway, NJ, USA
| | | |
Collapse
|
4
|
Nakayama S, Lukacova V, Tanabe S, Watanabe A, Mullin J, Suarez-Sharp S, Shimizu T. Physiologically Based Pharmacokinetic Absorption Model for Pexidartinib to Evaluate the Impact of Meal Contents and Intake Timing on Drug Exposure. Clin Pharmacol Drug Dev 2024; 13:440-448. [PMID: 38396317 DOI: 10.1002/cpdd.1385] [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: 09/08/2023] [Accepted: 01/23/2024] [Indexed: 02/25/2024]
Abstract
Pexidartinib is a systemic treatment for patients with tenosynovial giant cell tumor not amenable to surgery. Oral absorption of pexidartinib is affected by food; administration with a high-fat meal (HFM) or low-fat meal (LFM) increases absorption by approximately 100% and approximately 60%, respectively, compared with the fasted state. Pexidartinib is currently dosed 250 mg orally twice daily with an LFM (approximately 11-14 g of total fat). We developed a physiologically based pharmacokinetic model to determine the impact on drug exposure of dose timing with respect to meals, meal type, and caloric content. A 15%-16% increase in plasma exposure was predicted when consuming an HFM 1 hour after dosing with an LFM, but almost no effect on pharmacokinetics was predicted when an HFM was consumed 3 hours or more before or after pexidartinib dosing with an LFM. Exposure was not significantly affected when pexidartinib was taken with a 500-kcal LFM over the range of fat (approximately 11-14 g of total fat; 20%-25% calories from fat) for an LFM. These findings on timing of pexidartinib dose with respect to meals should be considered by patients and physicians to reduce the potential for side effects.
Collapse
Affiliation(s)
- Shintaro Nakayama
- Quantitative Clinical Pharmacology Department, Daiichi Sankyo Co., Ltd, Tokyo, Japan
| | | | - Shuichi Tanabe
- Formulation Technology Research Laboratories, Daiichi Sankyo Co., Ltd, Tokyo, Japan
| | - Akiko Watanabe
- Quantitative Clinical Pharmacology Department, Daiichi Sankyo Co., Ltd, Tokyo, Japan
| | - Jim Mullin
- Simulations Plus, Inc., Lancaster, CA, USA
| | | | - Takako Shimizu
- Quantitative Clinical Pharmacology Department, Daiichi Sankyo Co., Ltd, Tokyo, Japan
| |
Collapse
|
5
|
Chen L, Yao N, Yang H, Zhang S, Zhang K. Prediction of ROS1 and TRKA/B/C occupancy in plasma and cerebrospinal fluid for entrectinib alone and in DDIs using physiologically based pharmacokinetic (PBPK) modeling approach. Cancer Chemother Pharmacol 2024; 93:107-119. [PMID: 37838624 DOI: 10.1007/s00280-023-04598-5] [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: 06/04/2023] [Accepted: 09/20/2023] [Indexed: 10/16/2023]
Abstract
PURPOSE Entrectinib (ENT) is a potent c-ros oncogene 1(ROS1) and neurotrophic tyrosine receptor kinase (NTRKA/B/C) inhibitor. To determine the optimum dosage of ENT using ROS1 and NTRKA/B/C occupancy in plasma and cerebrospinal fluid (CSF) in drug-drug interactions (DDIs), physiologically-based pharmacokinetic (PBPK) models for healthy subjects and cancer population were developed for ENT and M5 (active metabolite). METHODS The PBPK models were built using the modeling parameters of ENT and M5 that were mainly derived from the published paper on the ENT PBPK model, and then validated by the observed pharmacokinetics (PK) in plasma and CSF from healthy subjects and patients. RESULTS The PBPK model showed that AUC, Cmax, and Ctrough ratios between predictions and observations are within the range of 0.5-2.0, except that the M5 AUC ratio is slightly above 2.0 (2.34). Based on the efficacy (> 75% occupancy for ROS1 and NTRKA/B/C) and safety (AUC < 160 μM·h and Cmax < 8.9 μM), the appropriate dosing regimens were identified. The appropriate dosage is 600 mg once daily (OD) when administered alone, reduced to 200 mg and 400 mg OD with itraconazole and fluconazole, respectively. ENT is not recommended for co-administration with rifampicin or efavirenz, but is permitted with fluvoxamine or dexamethasone. CONCLUSION The PBPK models can serve as a powerful approach to predict ENT concentration as well as ROS1 and NTRKA/B/C occupancy in plasma and CSF.
Collapse
Affiliation(s)
- Liangang Chen
- 980 (Bethune International Peace) Hospital of PLA Joint Logistics Support Forces, Shijiazhuang, 050051, China
| | - Na Yao
- 980 (Bethune International Peace) Hospital of PLA Joint Logistics Support Forces, Shijiazhuang, 050051, China
| | - Hongjie Yang
- 980 (Bethune International Peace) Hospital of PLA Joint Logistics Support Forces, Shijiazhuang, 050051, China
| | - Shaofeng Zhang
- Shijiazhuang Medical College, Shijiazhuang, 050599, China
| | - Kai Zhang
- Department of Medical Oncology, Shijiazhuang People's Hospital, Shijiazhuang, 050051, China.
| |
Collapse
|
6
|
Kollipara S, Martins FS, Sanghavi M, Santos GML, Saini A, Ahmed T. Role of Physiologically Based Biopharmaceutics Modeling (PBBM) in Fed Bioequivalence Study Waivers: Regulatory Outlook, Case Studies and Future Perspectives. J Pharm Sci 2024; 113:345-358. [PMID: 38043684 DOI: 10.1016/j.xphs.2023.11.030] [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: 09/14/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
Over the past few decades, physiologically based biopharmaceutics modeling (PBBM) has demonstrated its utility in both new drug and generic product development. Applications of PBBM for fed bioequivalence study waivers is an upcoming area. Recently Innovation & Quality (IQ) consortium demonstrated utility of PBBM to avoid repeat food effect studies for new drugs. In the similar lines, the current manuscript aims to discuss role of PBBM in generic fed bioequivalence study waivers. Generic industry practices related to PBBM model development to predict fed bioequivalence was portrayed with special emphasis on fed bio-predictive media. Media that can simulate fed bioequivalence study outcome were discussed from practical perspective. In-depth analysis, collating the data from 36 products was performed to understand predictability of PBBM for fed bioequivalence. Cases where PBBM was successful to predict fed bioequivalence was correlated with BCS class, formulation category and type of food effect. Further, two case studies were presented wherein fed bioequivalence study waiver obtained with PBBM approach. Lastly, future direction in terms of fed bioequivalence study waivers, regulatory perspectives and best practices for PBBM were portrayed. Overall, this article paves a way to utilize PBBM for generic fed bioequivalence study waivers.
Collapse
Affiliation(s)
- Sivacharan Kollipara
- Biopharmaceutics Group, Global Clinical Management, Dr. Reddy's Laboratories Ltd., Integrated Product Development Organization (IPDO), Bachupally, Medchal Malkajgiri District, Hyderabad, Telangana 500 090, India
| | | | - Maitri Sanghavi
- Biopharmaceutics & Clinical Development, Pharmaceutical Technology Center (PTC), Zydus Lifesciences Ltd., NH-8A, Sarkhej-Bavla Highway, Moraiya, Ahmedabad-382210, Gujrat, India
| | | | - Anuj Saini
- Biopharmaceutics & Clinical Development, Pharmaceutical Technology Center (PTC), Zydus Lifesciences Ltd., NH-8A, Sarkhej-Bavla Highway, Moraiya, Ahmedabad-382210, Gujrat, India
| | - Tausif Ahmed
- Biopharmaceutics Group, Global Clinical Management, Dr. Reddy's Laboratories Ltd., Integrated Product Development Organization (IPDO), Bachupally, Medchal Malkajgiri District, Hyderabad, Telangana 500 090, India.
| |
Collapse
|
7
|
Hu L, Sun Q, Tang L, Cai M, Qian W, Dou T, Wang H, Wu Y, Liu Y. Food Effect on the Pharmacokinetics of VC004, a Tropomyosin Receptor Kinase Inhibitor: A Randomized Crossover Trial in Healthy Chinese Subjects. Clin Drug Investig 2024; 44:79-85. [PMID: 38112942 DOI: 10.1007/s40261-023-01334-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND AND OBJECTIVE VC004 is a novel next-generation tropomyosin receptor kinase (TRK) inhibitor that is approved for the treatment of advanced or metastatic NTRK fusion-positive solid tumors and abrogated the drug resistance of the first-generation TRK inhibitors. The objective of the present study was to evaluate the effect of food on the pharmacokinetics and safety of VC004. METHODS The study was a randomized, open-label, two-period crossover, single-dose, phase I clinical trial. A total of 16 healthy subjects participated the trial. Subjects fasted for 10 h before drug administration in both fasting and fed states. Subjects received VC004 50 mg orally in the fasting state and after a high caloric food in the fed state. Blood samples at the designated time points were collected to determine the plasma concentration of VC004. Safety evaluation in both the fasted and fed periods were assessed via vital sign monitoring and clinical laboratory tests. RESULTS The maximum plasma concentration (Cmax) of VC004 in fed group decreased by 32.8%, corresponding with the slower absorption rate (time to Cmax (Tmax) delayed by almost 3 h) compared with the fasting group. Ratios of geometric means (GMRs) and 90% confidence intervals (90% CIs) of Cmax, the area under the curve of plasma concentration-time from zero to the last measurable concentration (AUC0-t), and AUC from zero to infinity (AUC0-∞) for VC004 between the two states were 67.18 (58.16-77.60), 103.59 (95.04-112.92) and 103.55 (95.63-112.11), respectively. No serious adverse events (AEs) occurred; only three grade 1 or grade 2 adverse events occurred in the fasted group, who recovered by the end of the study. CONCLUSIONS The intake of high calorie food decreased the absorption rate and increased the Tmax of VC004, while the AUC values were similar in both groups. No serious adverse event was reported. In conclusion, food does not alter the pharmacokinetics and safety profile of VC004 in a clinically meaningful manner. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT055528120.
Collapse
Affiliation(s)
- Linlin Hu
- Department of Pharmacy, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
- Department of Phase I Clinical Trial Unit, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
| | - Qiuyue Sun
- Department of Phase I Clinical Trial Unit, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Lu Tang
- Department of Phase I Clinical Trial Unit, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Mingmin Cai
- Department of Phase I Clinical Trial Unit, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Wei Qian
- Department of Phase I Clinical Trial Unit, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Ting Dou
- Department of Phase I Clinical Trial Unit, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Huiping Wang
- Department of Phase I Clinical Trial Unit, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yong Wu
- Jiangsu Vcare PharmaTech Co., Ltd., Nanjing, China
| | | |
Collapse
|
8
|
Moens F, Larsson A, De Blaiser A, Vandevijver G, Spreafico F, Nicolas JM, Lacombe L, Segregur D, Flanagan T, Berben P. Contribution of the Dynamic Intestinal Absorption Model (Diamod) to the Development of a Patient-Centric Drug Formulation. Mol Pharm 2023; 20:6197-6212. [PMID: 37955627 DOI: 10.1021/acs.molpharmaceut.3c00607] [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] [Indexed: 11/14/2023]
Abstract
Compound X is a weak basic drug targeting the early stages of Parkinson's disease, for which a theoretical risk assessment has indicated that elevated gastric pH conditions could potentially result in reduced plasma concentrations. Different in vitro dissolution methodologies varying in level of complexity and a physiologically based pharmacokinetic (PBPK) absorption model demonstrated that the dissolution, solubility, and intestinal absorption of compound X was indeed reduced under elevated gastric pH conditions. These observations were confirmed in a crossover pharmacokinetic study in Beagle dogs. As a result, the development of a formulation resulting in robust performance that is not sensitive to the exposed gastric pH levels is of crucial importance. The dynamic intestinal absorption MODel (Diamod), an advanced in vitro gastrointestinal transfer tool that allows to study the gastrointestinal dissolution and interconnected permeation of drugs, was selected as an in vitro tool for the formulation optimization activities given its promising predictive capacity and its capability to generate insights into the mechanisms driving formulation performance. Different pH-modifiers were screened for their potential to mitigate the pH-effect by decreasing the microenvironmental pH at the dissolution surface. Finally, an optimized formulation containing a clinically relevant dose of the drug and a functional amount of the selected pH-modifier was evaluated for its performance in the Diamod. This monolayer tablet formulation resulted in rapid gastric dissolution and supersaturation, inducing adequate intestinal supersaturation and permeation of compound X, irrespective of the gastric acidity level in the stomach. In conclusion, this study describes the holistic biopharmaceutics approach driving the development of a patient-centric formulation of compound X.
Collapse
Affiliation(s)
| | - Adam Larsson
- ProDigest BV, Technologiepark 82, 9052 Ghent, Belgium
| | | | | | | | - Jean-Marie Nicolas
- UCB Pharma SA, Early Solutions, Chemin du Foriest 1, 1420 Braine l'Alleud, Belgium
| | - Lucie Lacombe
- UCB Pharma SA, Product Design & Performance, Chemin du Foriest 1, 1420 Braine l'Alleud, Belgium
| | - Domagoj Segregur
- UCB Pharma SA, Product Design & Performance, Chemin du Foriest 1, 1420 Braine l'Alleud, Belgium
| | - Talia Flanagan
- UCB Pharma SA, Product Design & Performance, Chemin du Foriest 1, 1420 Braine l'Alleud, Belgium
| | - Philippe Berben
- UCB Pharma SA, Product Design & Performance, Chemin du Foriest 1, 1420 Braine l'Alleud, Belgium
| |
Collapse
|
9
|
Tsakalozou E, Mohamed MEF, Polak S, Heimbach T. Applications of Modeling and Simulation Approaches in Support of Drug Product Development of Oral Dosage Forms and Locally Acting Drug Products: a Symposium Summary. AAPS J 2023; 25:96. [PMID: 37783902 DOI: 10.1208/s12248-023-00862-x] [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: 07/25/2023] [Accepted: 09/16/2023] [Indexed: 10/04/2023] Open
Abstract
The number of modeling and simulation applications, including physiologically based pharmacokinetic (PBPK) models, physiologically based biopharmaceutics modeling (PBBM), and empirical models, has been constantly increasing along with the regulatory acceptance of these methodologies. While aiming at minimizing unnecessary human testing, these methodologies are used today to support the development and approval of novel drug products and generics. Modeling approaches are leveraged today for assessing drug-drug interaction, informing dose adjustments in renally or hepatically impaired patients, perform dose selection in pediatrics and pregnant women and diseased populations, and conduct biopharmaceutics-related assessments such as establish clinically relevant specifications for drug products and achieve quality assurance throughout the product life cycle. In the generics space, PBPK analyses are utilized toward virtual bioequivalence assessments within the scope of alternative bioequivalence approaches, product-specific guidance development, and food effect assessments among others. Case studies highlighting the evolving and expanding role of modeling and simulation approaches within the biopharmaceutics space were presented at the symposium titled "Model Informed Drug Development (MIDD): Role in Dose Selection, Vulnerable Populations, and Biowaivers - Chemical Entities" and Prologue "PBPK/PBBM to inform the Bioequivalence Safe Space, Food Effects, and pH-mediated DDIs" at the American Association of Pharmaceutical Scientists (AAPS) PharmSci 360 Annual Meeting in Boston, MA, on October 16-19, 2022, and are summarized here.
Collapse
Affiliation(s)
- Eleftheria Tsakalozou
- Division of Quantitative Methods and Modeling, Office of Research and Standards (ORS), Office of Generic Drugs (OGD), Center for Drug Evaluation and Research (CDER), US Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, Maryland, USA.
| | | | - Sebastian Polak
- Certara UK, Simcyp Division, Sheffield, UK
- Jagiellonian University Medical College, Krakow, Poland
| | - Tycho Heimbach
- Pharmaceutical Sciences, MRL, Merck & Co., Inc, Rahway, New Jersey, 07065, USA
| |
Collapse
|
10
|
Wang X, Chen F, Guo N, Gu Z, Lin H, Xiang X, Shi Y, Han B. Application of physiologically based pharmacokinetics modeling in the research of small-molecule targeted anti-cancer drugs. Cancer Chemother Pharmacol 2023; 92:253-270. [PMID: 37466731 DOI: 10.1007/s00280-023-04566-z] [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: 04/14/2023] [Accepted: 07/05/2023] [Indexed: 07/20/2023]
Abstract
INTRODUCTION Physiologically based pharmacokinetics (PBPK) models are increasingly used in the drug research and development, especially in anti-cancer drugs. Between 2001 and 2020, a total of 89 small-molecule targeted antitumor drugs were approved in China and the United States, some of which already included PBPK modeling in their application or approval packages. This article intended to review the prevalence and application of PBPK model in these drugs. METHOD Article search was performed in the PubMed to collect English research articles on small-molecule targeted anti-cancer drugs using PBPK modeling. The selected articles were classified into nine categorizes according to the application areas and further analyzed. RESULT From 2001 to 2020, more than 60% of small-molecule targeted anti-cancer drugs (54/89) were studied using PBPK model with a wide range of application. Ninety research articles were included, of which 48 involved enzyme-mediated drug-drug interaction (DDI). Of these retrieved articles, Simcyp, GastroPlus, and PK-Sim were the most widely model building platforms, which account for 63.8%, 15.2%, and 8.6%, respectively. CONCLUSION PBPK modeling is commonly and widely used to research small-molecule targeted anti-cancer drugs.
Collapse
Affiliation(s)
- Xiaowen Wang
- Department of Pharmacy, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, China
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, China
| | - Fang Chen
- Department of Pharmacy, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Nan Guo
- Department of Pharmacy, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, China
| | - Zhichun Gu
- Department of Pharmacy, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Houwen Lin
- Department of Pharmacy, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoqiang Xiang
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, China
| | - Yufei Shi
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, China.
| | - Bing Han
- Department of Pharmacy, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, China.
| |
Collapse
|
11
|
Kesisoglou F, Basu S, Belubbi T, Bransford P, Chung J, Dodd S, Dolton M, Heimbach T, Kulkarni P, Lin W, Moir A, Parrott N, Pepin X, Ren X, Sharma P, Stamatopoulos K, Tistaert C, Vaidhyanathan S, Wagner C, Riedmaier AE. Streamlining Food Effect Assessment - Are Repeated Food Effect Studies Needed? An IQ Analysis. AAPS J 2023; 25:60. [PMID: 37322223 DOI: 10.1208/s12248-023-00822-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/16/2023] [Indexed: 06/17/2023] Open
Abstract
Current regulatory guidelines on drug-food interactions recommend an early assessment of food effect to inform clinical dosing instructions, as well as a pivotal food effect study on the to-be-marketed formulation if different from that used in earlier trials. Study waivers are currently only granted for BCS class 1 drugs. Thus, repeated food effect studies are prevalent in clinical development, with the initial evaluation conducted as early as the first-in-human studies. Information on repeated food effect studies is not common in the public domain. The goal of the work presented in this manuscript from the Food Effect PBPK IQ Working Group was to compile a dataset on these studies across pharmaceutical companies and provide recommendations on their conduct. Based on 54 studies collected, we report that most of the repeat food effect studies do not result in meaningful differences in the assessment of the food effect. Seldom changes observed were more than twofold. There was no clear relationship between the change in food effect and the formulation change, indicating that in most cases, once a compound is formulated appropriately within a specific formulation technology, the food effect is primarily driven by inherent compound properties. Representative examples of PBPK models demonstrate that following appropriate validation of the model with the initial food effect study, the models can be applied to future formulations. We recommend that repeat food effect studies should be approached on a case-by-case basis taking into account the totality of the evidence including the use of PBPK modeling.
Collapse
Affiliation(s)
| | - Sumit Basu
- Clinical Pharmacology - Oncology, Novartis Institutes of Biomedical Research, East Hanover, New Jersey, USA
| | - Tejashree Belubbi
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Philip Bransford
- Data & Computational Sciences, Vertex Pharmaceuticals, Boston, Massachusetts, USA
| | - John Chung
- Drug Product Technologies, Amgen Inc., Thousand Oaks, California, USA
| | - Stephanie Dodd
- Chemical & Pharmaceutical Profiling, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | | | - Tycho Heimbach
- Pharmaceutical Sciences, Merck & Co., Inc., Rahway, NJ, USA
| | | | - Wen Lin
- Pharmacokinetics and Drug Metabolism, Sanofi, Bridgewater, New Jersey, USA
| | - Andrea Moir
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| | - Neil Parrott
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Xavier Pepin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Charter Way, Macclesfield, SK10 2NA, UK
- Regulatory Affairs, Simulations Plus, Lancaster, CA, USA
| | - Xiaojun Ren
- Modeling & Simulation, PK Sciences, Novartis Institutes of Biomedical Research, East Hanover, New Jersey, USA
| | - Pradeep Sharma
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | | | | | - Shruthi Vaidhyanathan
- Drug Product Science and Technology, Bristol-Myers Squibb, New Brunswick, New Jersey, USA
| | - Christian Wagner
- Global Drug Product Development, Global CMC Development, the Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | |
Collapse
|
12
|
de Waal T, Brouwers J, Rayyan M, Stillhart C, Vinarova L, Vinarov Z, Augustijns P. Characterization of neonatal and infant enterostomy fluids - Part II: Drug solubility. Int J Pharm 2023:123141. [PMID: 37321462 DOI: 10.1016/j.ijpharm.2023.123141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
Previous research revealed marked differences in the composition of intestinal fluids between infants and adults. To explore the impact on the solubilization of orally administered drugs, the present study assessed the solubility of five poorly water-soluble, lipophilic drugs in intestinal fluid pools from 19 infant enterostomy patients (infant HIF). For some but not all drugs, the average solubilizing capacity of infant HIF was similar to that of HIF obtained from adults (adult HIF) in fed conditions. Commonly used fed state simulated intestinal fluids (FeSSIF(-V2)) predicted fairly well drug solubility in the aqueous fraction of infant HIF, but did not account for the substantial solubilization by the lipid phase of infant HIF. Despite similarities in the average solubilities of some drugs in infant HIF and adult HIF or SIF, the underlying solubilization mechanisms likely differ, considering important compositional differences (e.g., low bile salt levels). Finally, the huge variability in composition of infant HIF pools resulted in a highly variable solubilizing capacity, potentially causing variations in drug bioavailability. The current study warrants future research focusing on (i) understanding the mechanisms underlying drug solubilization in infant HIF and (ii) evaluating the sensitivity of oral drug products to interpatient variations in drug solubilization.
Collapse
Affiliation(s)
- Tom de Waal
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Joachim Brouwers
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Maissa Rayyan
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | | | - Liliya Vinarova
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, Sofia University, Sofia, Bulgaria
| | - Zahari Vinarov
- Department of Chemical and Pharmaceutical Engineering, Faculty of Chemistry and Pharmacy, Sofia University, Sofia, Bulgaria
| | - Patrick Augustijns
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Yang E, Yu K, Lee S. Prediction of gastric pH-mediated drug exposure using physiologically-based pharmacokinetic modeling: A case study of itraconazole. CPT Pharmacometrics Syst Pharmacol 2023; 12:865-877. [PMID: 36967484 PMCID: PMC10272297 DOI: 10.1002/psp4.12959] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/01/2023] [Accepted: 02/14/2023] [Indexed: 05/24/2024] Open
Abstract
Abnormal gastric acidity, including achlorhydria, can act as a significant source of variability in orally administered drugs especially with pH-sensitive solubility profiles, such as weak bases, potentially resulting in an undesirable therapeutic response. This study aimed to evaluate the utility of physiologically-based pharmacokinetic (PBPK) modeling in the prediction of gastric pH-mediated drug exposure by using itraconazole, a weak base, as a case. An itraconazole PBPK model was developed on the mechanistic basis of its absorption kinetics in a middle-out manner from a stepwise in vitro-in vivo extrapolation to in vivo refinement. Afterward, an independent prospective clinical study evaluating gastric pH and itraconazole pharmacokinetics (PKs) under normal gastric acidity and esomeprazole-induced gastric hypoacidity was conducted for model validation. Validation was performed by comparing the predicted data with the clinical observations, and the valid model was subsequently applied to predict PK changes under achlorhydria. The developed itraconazole PBPK model showed reasonable reproducibility for gastric pH-mediated exposure observed in the clinical investigation. Based on the model-based simulations, itraconazole exposure was expected to be decreased up to 65% under achlorhydria, and furthermore, gastric pH-mediated exposure could be mechanistically interpreted according to sequential variation in total solubility, dissolution, and absorption. This study suggested the utility of PBPK modeling in the prediction of gastric pH-mediated exposure, especially for drugs whose absorption is susceptible to gastric pH. Our findings will serve as a leading model for further mechanistic assessment of exposure depending on gastric pH for various drugs, ultimately contributing to personalized pharmacotherapy.
Collapse
Affiliation(s)
- Eunsol Yang
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and Hospital101 Daehak‐ro, Jongno‐guSeoul03080Republic of Korea
- Kidney Research InstituteSeoul National University Medical Research Center103 Daehak‐ro, Jongno‐guSeoul03080Republic of Korea
| | - Kyung‐Sang Yu
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and Hospital101 Daehak‐ro, Jongno‐guSeoul03080Republic of Korea
| | - SeungHwan Lee
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and Hospital101 Daehak‐ro, Jongno‐guSeoul03080Republic of Korea
| |
Collapse
|
14
|
de Waal T, Brouwers J, Berben P, Flanagan T, Tack J, Vandenberghe W, Vanuytsel T, Augustijns P. Characterization of Aspirated Duodenal Fluids from Parkinson's Disease Patients. Pharmaceutics 2023; 15:pharmaceutics15041243. [PMID: 37111729 PMCID: PMC10145225 DOI: 10.3390/pharmaceutics15041243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/16/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Parkinson's disease, one of the most common neurodegenerative diseases, may not only affect the motor system, but also the physiology of the gastrointestinal tract. Delayed gastric emptying, impaired motility and altered intestinal bacteria are well-established consequences of the disease, which can have a pronounced effect on the absorption of orally administered drugs. In contrast, no studies have been performed into the composition of intestinal fluids. It is not unlikely that Parkinson's disease also affects the composition of intestinal fluids, a critical factor in the in vitro and in silico simulation of drug dissolution, solubilization and absorption. In the current study, duodenal fluids were aspirated from Parkinson's disease (PD) patients and age-matched healthy controls (healthy controls, HC) consecutively in fasted and fed conditions. The fluids were then characterized for pH, buffer capacity, osmolality, total protein, phospholipids, bile salts, cholesterol and lipids. In a fasted state, the intestinal fluid composition was highly similar in PD patients and healthy controls. In general, the same was true for fed-state fluids, apart from a slightly slower and less pronounced initial change in factors directly affected by the meal (i.e., buffer capacity, osmolality, total protein and lipids) in PD patients. The absence of a fast initial increase for these factors immediately after meal intake, as was observed in healthy controls, might result from slower gastric emptying in PD patients. Irrespective of the prandial state, a higher relative amount of secondary bile salts was observed in PD patients, potentially indicating altered intestinal bacterial metabolism. Overall, the data from this study indicate that only minor disease-specific adjustments in small intestinal fluid composition should be considered when simulating intestinal drug absorption in PD patients.
Collapse
Affiliation(s)
- Tom de Waal
- Drug Delivery and Disposition, KU Leuven, 3000 Leuven, Belgium
| | | | - Philippe Berben
- Pharmaceutical Sciences, UCB Pharma SA, 1420 Braine-l'Alleud, Belgium
| | - Talia Flanagan
- Pharmaceutical Sciences, UCB Pharma SA, 1420 Braine-l'Alleud, Belgium
| | - Jan Tack
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
- Translational Research Center for Gastrointestinal Disorders, TARGID, KU Leuven, 3000 Leuven, Belgium
| | - Wim Vandenberghe
- Department of Neurology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Tim Vanuytsel
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
- Translational Research Center for Gastrointestinal Disorders, TARGID, KU Leuven, 3000 Leuven, Belgium
| | | |
Collapse
|
15
|
de Waal T, Brouwers J, Mols R, Hoffman I, Rayyan M, Augustijns P. Characterization of neonatal and infant enterostomy fluids. Int J Pharm 2023; 639:122943. [PMID: 37059240 DOI: 10.1016/j.ijpharm.2023.122943] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/16/2023]
Abstract
The composition of gastrointestinal (GI) fluids is crucial for the dissolution, solubilization, and absorption of orally administered drugs. Disease- or age-related changes in GI fluid composition could significantly affect the pharmacokinetics of oral drugs. However, limited studies have been conducted on the characteristics of GI fluids in neonates and infants due to practical and ethical challenges. The current study collected enterostomy fluids from 21 neonate and infant patients over an extended period of time and from different regions of the small intestine and colon. The fluids were characterized for pH, buffer capacity, osmolality, total protein, bile salts, phospholipids, cholesterol, and lipid digestion products. The study found a large variability in the fluid characteristics among the different patients, in line with the highly heterogeneous study population. Compared to adult intestinal fluids, the enterostomy fluids from neonates and infants had low bile salt concentrations, with an increasing trend as a function of age; no secondary bile salts were detected. In contrast, total protein and lipid concentrations were relatively high, even in the distal small intestine. These findings suggest marked differences in intestinal fluid composition between neonates and infants versus adults, which may affect the absorption of certain drugs.
Collapse
Affiliation(s)
- Tom de Waal
- Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | | | - Raf Mols
- Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Ilse Hoffman
- Paediatric Gastroenterology. Hepatology and Nutrition, University Hospitals Leuven, Leuven, Belgium
| | - Maissa Rayyan
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | | |
Collapse
|
16
|
Pepin X, McAlpine V, Moir A, Mann J. Acalabrutinib Maleate Tablets: The Physiologically Based Biopharmaceutics Model behind the Drug Product Dissolution Specification. Mol Pharm 2023; 20:2181-2193. [PMID: 36859819 DOI: 10.1021/acs.molpharmaceut.3c00005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Acalabrutinib maleate tablets correspond to an improved formulation compared to acalabrutinib capsules as they can be dosed with and without acid reducing agents and therefore benefit more cancer patients. The dissolution specification for the drug product was determined using all the information available on the drug safety, efficacy, and in vitro performance. In addition, a physiologically based biopharmaceutics model was developed for acalabrutinib maleate tablets on the back of a previously published model for acalabrutinib capsules to establish that the proposed drug product dissolution specification would ensure safe and effective products for all patients including those under acid reducing agent treatment. The model was built, validated, and used to predict the exposure of virtual batches where the dissolution was slower than that of the clinical target. A combination of exposure prediction and the use of a PK-PD model allowed it to be demonstrated that the proposed drug product dissolution specification was acceptable. This combination of models enabled a larger safe space than would have been granted by consideration of bioequivalence only.
Collapse
Affiliation(s)
- Xavier Pepin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Charter Way, SK10 2NA Macclesfield, United Kingdom
| | - Vivien McAlpine
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, SK10 2NA Macclesfield, United Kingdom
| | - Andrea Moir
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, SK10 2NA Macclesfield, United Kingdom
| | - James Mann
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, SK10 2NA Macclesfield, United Kingdom
| |
Collapse
|
17
|
Meneses-Lorente G, Guerini E, Mercier F, Parrott N, Kowalski K, Chow-Maneval E, Buchheit V, Bergthold G, Fox E, Phipps A, Djebli N. Entrectinib dose confirmation in pediatric oncology patients: pharmacokinetic considerations. Cancer Chemother Pharmacol 2023; 91:239-246. [PMID: 36884068 PMCID: PMC10033473 DOI: 10.1007/s00280-023-04510-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 02/08/2023] [Indexed: 03/09/2023]
Abstract
PURPOSE Entrectinib is a central nervous system-active potent inhibitor of tropomyosin receptor kinase (TRK), with anti-tumor activity against neurotrophic NTRK gene fusion-positive tumors. This study investigates the pharmacokinetics of entrectinib and its active metabolite (M5) in pediatric patients and aims to understand whether the pediatric dose of 300 mg/m2 once daily (QD) provides an exposure that is consistent with the approved adult dose (600 mg QD). METHODS Forty-three patients aged from birth to 22 years were administered entrectinib (250-750 mg/m2 QD) orally with food in 4-week cycles. Entrectinib formulations included capsules without acidulant (F1) and capsules with acidulant (F2B and F06). RESULTS Although there was interpatient variability with F1, entrectinib and M5 exposures increased dose dependently. Lower systemic exposures were observed in pediatric patients receiving 400 mg/m2 QD entrectinib (F1) versus adults receiving either the same dose/formulation or the recommended flat dose of 600 mg QD (~ 300 mg/m2 for a 70 kg adult) due to suboptimal F1 performance in the pediatric study. The observed pediatric exposures following 300 mg/m2 QD entrectinib (F06) were comparable to those in adults receiving 600 mg QD. CONCLUSIONS Overall, the F1 formulation of entrectinib was associated with lower systemic exposure in pediatric patients compared with the commercial acidulant formulation (F06). Systemic exposures achieved in pediatric patients with the F06 recommended dose (300 mg/m2) were within the known efficacious range in adults, confirming the adequacy of the recommended dose regimen with the commercial formulation.
Collapse
Affiliation(s)
| | - Elena Guerini
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Francois Mercier
- Biostatistics, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Neil Parrott
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Karey Kowalski
- PharmD Clinical Pharmacology, Ignyta, Inc., San Diego, CA, USA
| | | | - Vincent Buchheit
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Guillaume Bergthold
- Product Development Oncology and Hematology Department, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Elizabeth Fox
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Alex Phipps
- Department of Clinical Pharmacology, Roche Innovation Center Welwyn, Roche Products Ltd., Welwyn Garden City, UK
| | - Nassim Djebli
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
18
|
Zhou D, Chen B, Sharma S, Tang W, Pepin X. Physiologically Based Absorption Modelling to Explore the Formulation and Gastric pH Changes on the Pharmacokinetics of Acalabrutinib. Pharm Res 2023; 40:375-386. [PMID: 35478298 DOI: 10.1007/s11095-022-03268-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/20/2022] [Indexed: 10/18/2022]
Abstract
Acalabrutinib, a selective Bruton's tyrosine kinase inhibitor, is a biopharmaceutics classification system class II drug. The aim of this study was to develop a physiologically based pharmacokinetic (PBPK) model to mechanistically describe absorption of immediate release capsule formulation of acalabrutinib in humans. Integration of in vitro biorelevant measurements, dissolution studies and in silico modelling provided clinically relevant inputs for the mechanistic absorption PBPK model. The batch specific dissolution data were integrated in two ways, by fitting a diffusion layer model scalar to the drug product dissolution with integration of drug substance laser diffraction particle size data, or by fitting a product particle size distribution to the dissolution data. The latter method proved more robust and biopredictive. In both cases, the drug surface solubility was well predicted by the Simcyp simulator. The model using the product particle size distribution (P-PSD) for each clinical batch adequately captured the PK profiles of acalabrutinib and its active metabolite. Average fold errors were 0.89 for both Cmax and AUC, suggesting good agreement between predicted and observed PK values. The model also accurately predicted pH-dependent drug-drug interactions between omeprazole and acalabrutinib, which was similar across all clinical formulations. The model predicted acalabrutinib geometric mean AUC ratios (with omeprazole vs acalabrutinib alone) were 0.51 and 0.68 for 2 batches of formulations, which are close to observed values of 0.43 and 0.51~0.63, respectively. The mechanistic absorption PBPK model could be potentially used for future applications such as optimizing formulations or predicting the PK for different batches of the drug product.
Collapse
Affiliation(s)
- Diansong Zhou
- Clinical Pharmacology & Quantitative Pharmacology, AstraZeneca, BioPharmaceuticals R&D, Boston, Massachusetts, USA. .,AstraZeneca, 35 Gatehouse Dr., Waltham, Massachusett, 02451, USA.
| | - Buyun Chen
- Clinical Pharmacology & Quantitative Pharmacology, AstraZeneca, BioPharmaceuticals R&D, South San Francisco, California, USA
| | - Shringi Sharma
- Clinical Pharmacology & Quantitative Pharmacology, AstraZeneca, BioPharmaceuticals R&D, South San Francisco, California, USA
| | - Weifeng Tang
- Clinical Pharmacology & Quantitative Pharmacology, AstraZeneca, BioPharmaceuticals R&D, Gaithersburg, Maryland, USA
| | - Xavier Pepin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| |
Collapse
|
19
|
Pepin XJH, Hammarberg M, Mattinson A, Moir A. Physiologically Based Biopharmaceutics Model for Selumetinib Food Effect Investigation and Capsule Dissolution Safe Space - Part I: Adults. Pharm Res 2023; 40:387-403. [PMID: 36002614 DOI: 10.1007/s11095-022-03339-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/09/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE A physiologically based biopharmaceutics model (PBBM) was developed to mechanistically investigate the effect of formulation and food on selumetinib pharmacokinetics. METHODS Selumetinib is presented as a hydrogen sulfate salt, and in vitro and in vivo data were used to verify the precipitation rate to apply to simulations. Dissolution profiles observed for capsules and granules were used to derive product-particle size distributions for model input. The PBBM incorporated gut efflux and first-pass gut metabolism, based on intravenous and oral pharmacokinetic data, alongside in vitro data for the main enzyme isoform and P-glycoprotein efflux. The PBBM was validated across eight clinical scenarios. RESULTS The quality-control dissolution method for selumetinib capsules was found to be clinically relevant through PBBM validation. A safe space for capsule dissolution was established using a virtual batch. The effect of food (low fat vs high fat) on capsules and granules was elucidated by the PBBM. For capsules, a lower amount was dissolved in the fed state due to a pH increase in the stomach followed by higher precipitation in the small intestine. First-pass gut extraction is higher for capsules in the fed state due to drug dilution in the stomach chyme and reduced concentration in the lumen. The enteric-coated granules dissolve more slowly than capsules after stomach emptying, attenuating the difference in first-pass gut extraction between prandial states. CONCLUSIONS The PBBM was instrumental in understanding and explaining the different behaviors of the selumetinib formulations. The model can be used to predict the impact of food in humans.
Collapse
Affiliation(s)
- Xavier J H Pepin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| | - Maria Hammarberg
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden. .,AstraZeneca, Pepparedsleden, SE-431 83, Mölndal, Sweden.
| | - Alexandra Mattinson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| | - Andrea Moir
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| |
Collapse
|
20
|
Physiologically Based Biopharmaceutics Modeling of Food Effect for Basmisanil: A Retrospective Case Study of the Utility for Formulation Bridging. Pharmaceutics 2023; 15:pharmaceutics15010191. [PMID: 36678820 PMCID: PMC9862143 DOI: 10.3390/pharmaceutics15010191] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/21/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Basmisanil, is a lipophilic drug substance, exhibiting poor solubility and good permeability (BCS class 2). A validated physiologically based biopharmaceutics model (PBBM) has been previously described for tablets dosed in the fed state. The PBBM captured the less than proportional increases in exposure at higher doses well and indicated that absorption was dissolution rate-limited below 200 mg while solubility was limiting for higher doses. In this study, a model for dosing in the fasted state is described and is verified for simulation of the food effect where exposures were ~1.5 fold higher when a 660 mg tablet was given with food. The model is then applied to simulate the food effect for a granules formulation given at a lower dose (120 mg). The food effect at the lower dose was reasonably simulated with a ratio of simulated/observed food effect of 1.35 for Cmax and 0.83 for AUC. Sensitivity analysis was carried out for uncertain model parameters to confirm that the model could predict the magnitude of the positive food effect with moderate to high confidence. This study suggests that a verified PBBM can provide a useful alternative to a repeat food effect study when formulation changes are minor. However, there is need for further evaluation of the approach and a definition of what formulation changes are minor in this context. In addition, this work highlights some uncertainties in the handling of solubility in PBBM, in particular around temperature dependency of solubility and the parameterization of bile salt solubilization using measurements in biorelevant media.
Collapse
|
21
|
Kiyota T, Ando Y, Kambayashi A. Dynamic Changes in Gastrointestinal Fluid Characteristics after Food Ingestion Are Important for Quantitatively Predicting the In Vivo Performance of Oral Solid Dosage Forms in Humans in the Fed State. Mol Pharm 2023; 20:357-369. [PMID: 36373973 DOI: 10.1021/acs.molpharmaceut.2c00666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The aim of this study was to develop a simulation model to predict the in vivo performance of solid oral dosage forms in humans in the fed state. We focused on investigating the effect of dynamic changes in gastrointestinal (GI) fluid characteristics in the fed state on the in vivo performance of solid dosage forms. We used six solid dosage forms containing weak base drugs as model formulations, two with positive food effects in humans, two with negative food effects, and two which are not affected by food ingestion. These model drug formulations were used to perform biorelevant dissolution tests in the stomach and small intestine under both prandial states. The in vitro properties of the drug products obtained from these tests were then coupled with in silico models (fasted or fed) to predict food effects in humans. We successfully incorporated the dynamic changes in GI fluid characteristics and their effects on the in vivo dissolution of drugs into the prediction model for the fed state. This newly designed physiologically based biopharmaceutics modeling approach provided the precise and quantitative prediction of food effects (i.e., changes in Cmax and AUC after food ingestion) in humans while considering the dynamic changes in fluid characteristics in the fed state.
Collapse
Affiliation(s)
- Tsuyoshi Kiyota
- Pharmaceutical Research and Technology Laboratories, Astellas Pharma Inc., 180 Ozumi, Yaizu, Shizuoka425-0072, Japan
| | - Yuki Ando
- Pharmaceutical Research and Technology Laboratories, Astellas Pharma Inc., 180 Ozumi, Yaizu, Shizuoka425-0072, Japan
| | - Atsushi Kambayashi
- Pharmaceutical Research and Technology Laboratories, Astellas Pharma Inc., 180 Ozumi, Yaizu, Shizuoka425-0072, Japan.,School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka422-8526, Japan
| |
Collapse
|
22
|
Desai AV, Robinson GW, Gauvain K, Basu EM, Macy ME, Maese L, Whipple NS, Sabnis AJ, Foster JH, Shusterman S, Yoon J, Weiss BD, Abdelbaki MS, Armstrong AE, Cash T, Pratilas CA, Corradini N, Marshall LV, Farid-Kapadia M, Chohan S, Devlin C, Meneses-Lorente G, Cardenas A, Hutchinson KE, Bergthold G, Caron H, Chow Maneval E, Gajjar A, Fox E. Entrectinib in children and young adults with solid or primary CNS tumors harboring NTRK, ROS1, or ALK aberrations (STARTRK-NG). Neuro Oncol 2022; 24:1776-1789. [PMID: 35395680 PMCID: PMC9527518 DOI: 10.1093/neuonc/noac087] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Entrectinib is a TRKA/B/C, ROS1, ALK tyrosine kinase inhibitor approved for the treatment of adults and children aged ≥12 years with NTRK fusion-positive solid tumors and adults with ROS1 fusion-positive non-small-cell lung cancer. We report an analysis of the STARTRK-NG trial, investigating the recommended phase 2 dose (RP2D) and activity of entrectinib in pediatric patients with solid tumors including primary central nervous system tumors. METHODS STARTRK-NG (NCT02650401) is a phase 1/2 trial. Phase 1, dose-escalation of oral, once-daily entrectinib, enrolled patients aged <22 years with solid tumors with/without target NTRK1/2/3, ROS1, or ALK fusions. Phase 2, basket trial at the RP2D, enrolled patients with intracranial or extracranial solid tumors harboring target fusions or neuroblastoma. Primary endpoints: phase 1, RP2D based on toxicity; phase 2, objective response rate (ORR) in patients harboring target fusions. Safety-evaluable patients: ≥1 dose of entrectinib; response-evaluable patients: measurable/evaluable baseline disease and ≥1 dose at RP2D. RESULTS At data cutoff, 43 patients, median age of 7 years, were response-evaluable. In phase 1, 4 patients experienced dose-limiting toxicities. The most common treatment-related adverse event was weight gain (48.8%). Nine patients experienced bone fractures (20.9%). In patients with fusion-positive tumors, ORR was 57.7% (95% CI 36.9-76.7), median duration of response was not reached, and median (interquartile range) duration of treatment was 10.6 months (4.2-18.4). CONCLUSIONS Entrectinib resulted in rapid and durable responses in pediatric patients with solid tumors harboring NTRK1/2/3 or ROS1 fusions.
Collapse
Affiliation(s)
- Ami V Desai
- Department of Pediatrics, Section of Hematology/Oncology/Stem Cell Transplantation, University of Chicago Medical Center, Chicago, Illinois, USA
| | - Giles W Robinson
- Division of Neuro-Oncology, Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Karen Gauvain
- Pediatric Neuro-Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ellen M Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Margaret E Macy
- Pediatric Hematology-Oncology, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Luke Maese
- Department of Pediatrics, Division of Hematology/Oncology, University of Utah/Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Nicholas S Whipple
- Pediatric Hematology-Oncology, University of Utah, Salt Lake City, Utah, USA
| | - Amit J Sabnis
- Division of Pediatric Oncology, Department of Pediatrics, University of California, San Francisco, California, USA
| | - Jennifer H Foster
- Department of Pediatrics, Hematology-Oncology, Texas Children’s Hospital, Houston, Texas, USA
| | - Suzanne Shusterman
- Pediatric Hematology and Oncology, Dana Farber Cancer Institute/Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| | - Janet Yoon
- Department of Pediatrics, University of California San Diego, San Diego, California, USA
| | - Brian D Weiss
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Mohamed S Abdelbaki
- Division of Hematology & Oncology, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Amy E Armstrong
- Division of Pediatric Hematology/Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Thomas Cash
- Pediatric Hematology/Oncology, Aflac Cancer & Blood Disorders Center, Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Christine A Pratilas
- Department of Oncology, Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nadège Corradini
- Department of Pediatric Hematology and Oncology, Institute of Pediatric Hematology and Oncology (IHOPe), Léon Bérard Cancer Centre, Lyon, France
| | - Lynley V Marshall
- Children and Young People’s Unit, The Royal Marsden Hospital and The Institute of Cancer Research, London, UK
| | | | - Saibah Chohan
- PDD Data & Statistical Sciences, F. Hoffmann-La Roche Ltd., Mississauga, Ontario, Canada
| | - Clare Devlin
- Pharma Development Oncology and Hematology, Roche Products Ltd., Welwyn Garden City, UK
| | | | - Alison Cardenas
- Clinical Safety, Genentech, Inc., South San Francisco, California, USA
| | | | | | - Hubert Caron
- Product Development Oncology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Amar Gajjar
- Division of Neuro-Oncology, Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Elizabeth Fox
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
23
|
Jiang Q, Li M, Li H, Chen L. Entrectinib, a new multi-target inhibitor for cancer therapy. Biomed Pharmacother 2022; 150:112974. [PMID: 35447552 DOI: 10.1016/j.biopha.2022.112974] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/29/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022] Open
Abstract
Clinical practice shows that when single-target drugs treat multi-factor diseases such as tumors, cardiovascular system and endocrine system diseases, it is often difficult to achieve good therapeutic effects, and even serious adverse reactions may occur. Multi-target drugs can simultaneously regulate multiple links of disease, improve efficacy, reduce adverse reactions, and improve drug resistance. They are ideal drugs for treating complex diseases, and therefore have become the main direction of drug development. At present, some multi-target drugs have been successfully used in many major diseases. Entrectinib is an oral small molecule inhibitor that targets TRK, ROS1, and ALK. It is used to treat locally advanced or metastatic solid tumors with NTRK1/2/3, ROS1 and ALK gene fusion mutations. It can pass through the blood-brain barrier and is the only TRK inhibitor clinically proven to be effective against primary and metastatic brain diseases. In 2019, entrectinib was approved by the FDA to treat adult patients with ROS1-positive metastatic non-small cell lung cancer. Case reports showed that continuous administration of entrectinib was effective and tolerable. In this review, we give a brief introduction to TKK, ROS1 and ALK, and on this basis, we give a detailed and comprehensive introduction to the mechanism of action, pharmacokinetics, pharmacodynamics, clinical efficacy, tolerability and drug interactions of entrectinib.
Collapse
Affiliation(s)
- Qinghua Jiang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Mingxue Li
- Wuya College of Innovation, School of Pharmacy, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hua Li
- Wuya College of Innovation, School of Pharmacy, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Lixia Chen
- Wuya College of Innovation, School of Pharmacy, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
24
|
K Y, Kollipara S, Ahmed T, Chachad S. Applications of PBPK/PBBM modeling in generic product development: An industry perspective. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103152] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
25
|
Wilson CG, Aarons L, Augustijns P, Brouwers J, Darwich AS, De Waal T, Garbacz G, Hansmann S, Hoc D, Ivanova A, Koziolek M, Reppas C, Schick P, Vertzoni M, García-Horsman JA. Integration of advanced methods and models to study drug absorption and related processes: An UNGAP perspective. Eur J Pharm Sci 2021; 172:106100. [PMID: 34936937 DOI: 10.1016/j.ejps.2021.106100] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 01/09/2023]
Abstract
This collection of contributions from the European Network on Understanding Gastrointestinal Absorption-related Processes (UNGAP) community assembly aims to provide information on some of the current and newer methods employed to study the behaviour of medicines. It is the product of interactions in the immediate pre-Covid period when UNGAP members were able to meet and set up workshops and to discuss progress across the disciplines. UNGAP activities are divided into work packages that cover special treatment populations, absorption processes in different regions of the gut, the development of advanced formulations and the integration of food and pharmaceutical scientists in the food-drug interface. This involves both new and established technical approaches in which we have attempted to define best practice and highlight areas where further research is needed. Over the last months we have been able to reflect on some of the key innovative approaches which we were tasked with mapping, including theoretical, in silico, in vitro, in vivo and ex vivo, preclinical and clinical approaches. This is the product of some of us in a snapshot of where UNGAP has travelled and what aspects of innovative technologies are important. It is not a comprehensive review of all methods used in research to study drug dissolution and absorption, but provides an ample panorama of current and advanced methods generally and potentially useful in this area. This collection starts from a consideration of advances in a priori approaches: an understanding of the molecular properties of the compound to predict biological characteristics relevant to absorption. The next four sections discuss a major activity in the UNGAP initiative, the pursuit of more representative conditions to study lumenal dissolution of drug formulations developed independently by academic teams. They are important because they illustrate examples of in vitro simulation systems that have begun to provide a useful understanding of formulation behaviour in the upper GI tract for industry. The Leuven team highlights the importance of the physiology of the digestive tract, as they describe the relevance of gastric and intestinal fluids on the behaviour of drugs along the tract. This provides the introduction to microdosing as an early tool to study drug disposition. Microdosing in oncology is starting to use gamma-emitting tracers, which provides a link through SPECT to the next section on nuclear medicine. The last two papers link the modelling approaches used by the pharmaceutical industry, in silico to Pop-PK linking to Darwich and Aarons, who provide discussion on pharmacometric modelling, completing the loop of molecule to man.
Collapse
Affiliation(s)
- Clive G Wilson
- Strathclyde Institute of Pharmacy & Biomedical Sciences, Glasgow, U.K.
| | | | | | | | | | | | | | | | | | | | - Mirko Koziolek
- NCE Formulation Sciences, Abbvie Deutschland GmbH & Co. KG, Germany
| | | | - Philipp Schick
- Department of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, University of Greifswald, Germany
| | | | | |
Collapse
|
26
|
Development and application of a physiologically based pharmacokinetic model for entrectinib in rats and scale-up to humans: Route-dependent gut wall metabolism. Biomed Pharmacother 2021; 146:112520. [PMID: 34902744 DOI: 10.1016/j.biopha.2021.112520] [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: 10/27/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 11/21/2022] Open
Abstract
Entrectinib (Rozlytrek®) is an oral antineoplastic agent approved by the U.S. Food and Drug Administration in 2019 for the treatment of c-ros oncogene 1 (ROS1)-positive non-small cell lung cancer and neurotrophic tyrosine receptor kinase (NTRK) fusion-positive solid tumors. Although there have been a few studies on the pharmacokinetics of entrectinib, the relative contributions of several kinetic factors determining the oral bioavailability and systemic exposure of entrectinib are still worthy of investigation. Experimental data on the intestinal absorption and disposition of entrectinib in rats were acquired from studies on in vitro protein binding/tissue S9 metabolism, in situ intestinal perfusion, and in vivo dose-escalation/hepatic extraction. Using these datasets, an in-house whole-body physiologically based pharmacokinetic (PBPK) model incorporating the QGut model concepts and segregated blood flow in the gut was constructed and optimized with respect to drug-specific parameters. The established rat PBPK model was further extrapolated to humans through relevant physiological scale-up and parameter optimization processes. The optimized rat and human PBPK models adequately captured the impact of route-dependent gut metabolism on the systemic exposure to entrectinib and closely mirrored various preclinical and clinical observations. Our proposed PBPK model could be useful in optimizing dosage regimens and predicting drug interaction potential in various clinical conditions, after partial modification and validation.
Collapse
|
27
|
Osman HM, Tuncbilek M. Entrectinib: A new Selective Tyrosine Kinase Inhibitor Approved for the Treatment of Pediatric and Adult Patients with NTRK Fusion-positive, Recurrent or Advanced Solid Tumors. Curr Med Chem 2021; 29:2602-2616. [PMID: 34521321 DOI: 10.2174/0929867328666210914121324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/13/2021] [Accepted: 07/23/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Entrectinib is a highly potent ATP-competitive and selective inhibitor of tyrosine kinases - Trk A B C, ALK, and ROS1. It was developed by Roche and initially approved in Japan in 2019 for the treatment of pediatric and adult patients with NTRK fusion-positive, recurrent, or advanced solid tumors. In August 2019, entrectinib received accelerated approval by the U.S FDA for this indication. It is also the first FDA-approved drug designed to target both NTRK and ROS1. OBJECTIVE We aim to summarize recent studies related to the synthesis, mechanism of action, and clinical trials of the newly approved selective tyrosine kinase inhibitor entrectinib. METHOD We conduct a literature review of the research studies on the new highly-potent small-molecule entrectinib. CONCLUSION Entrectinib, based on three clinical studies (ALKA, STARTRK-1, and STARTRK-2), was well tolerated, with a manageable safety profile. It induced clinically meaningful responses in recurrent or advanced solid tumors associated with NTRK fusion-positive or ROS1+ NSCLC. It demonstrated substantial efficacy in patients with CNS metastases.
Collapse
Affiliation(s)
- Hind M Osman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, 06100 Ankara. Turkey
| | - Meral Tuncbilek
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, 06100 Ankara. Turkey
| |
Collapse
|
28
|
Djebli N, Buchheit V, Parrott N, Guerini E, Cleary Y, Fowler S, Frey N, Yu L, Mercier F, Phipps A, Meneses-Lorente G. Physiologically-Based Pharmacokinetic Modelling of Entrectinib Parent and Active Metabolite to Support Regulatory Decision-Making. Eur J Drug Metab Pharmacokinet 2021; 46:779-791. [PMID: 34495458 DOI: 10.1007/s13318-021-00714-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2021] [Indexed: 01/11/2023]
Abstract
BACKGROUND AND OBJECTIVE Entrectinib is a selective inhibitor of ROS1/TRK/ALK kinases, recently approved for oncology indications. Entrectinib is predominantly cleared by cytochrome P450 (CYP) 3A4, and modulation of CYP3A enzyme activity profoundly alters the pharmacokinetics of both entrectinib and its active metabolite M5. We describe development of a combined physiologically based pharmacokinetic (PBPK) model for entrectinib and M5 to support dosing recommendations when entrectinib is co-administered with CYP3A4 inhibitors or inducers. METHODS A PBPK model was established in Simcyp® Simulator. The initial model based on in vitro-in vivo extrapolation was refined using sensitivity analysis and non-linear mixed effects modeling to optimize parameter estimates and to improve model fit to data from a clinical drug-drug interaction study with the strong CYP3A4 inhibitor, itraconazole. The model was subsequently qualified against clinical data, and the final qualified model used to simulate the effects of moderate to strong CYP3A4 inhibitors and inducers on entrectinib and M5 pharmacokinetics. RESULTS The final model showed good predictive performance for entrectinib and M5, meeting commonly used predictive performance acceptance criteria in each case. The model predicted that co-administration of various moderate CYP3A4 inhibitors (verapamil, erythromycin, clarithromycin, fluconazole, and diltiazem) would result in an average increase in entrectinib exposure between 2.2- and 3.1-fold, with corresponding average increases for M5 of approximately 2-fold. Co-administration of moderate CYP3A4 inducers (efavirenz, carbamazepine, phenytoin) was predicted to result in an average decrease in entrectinib exposure between 45 and 79%, with corresponding average decreases for M5 of approximately 50%. CONCLUSIONS The model simulations were used to derive dosing recommendations for co-administering entrectinib with CYP3A4 inhibitors or inducers. PBPK modeling has been used in lieu of clinical studies to enable regulatory decision-making.
Collapse
Affiliation(s)
- Nassim Djebli
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | - Vincent Buchheit
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Neil Parrott
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Elena Guerini
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Yumi Cleary
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Stephen Fowler
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Nicolas Frey
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Li Yu
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Jersey City, NJ, USA
| | - François Mercier
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Alex Phipps
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Roche Products Ltd, Welwyn, UK
| | - Georgina Meneses-Lorente
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Roche Products Ltd, Welwyn, UK
| |
Collapse
|
29
|
Meneses-Lorente G, Fowler S, Guerini E, Kowalski K, Chow-Maneval E, Yu L, Mercier F, Ullah M, Umehara K, Brink A, Buchheit V, Zwanziger E, Phipps A, Djebli N. In vitro and clinical investigations to determine the drug-drug interaction potential of entrectinib, a small molecule inhibitor of neurotrophic tyrosine receptor kinase (NTRK). Invest New Drugs 2021; 40:68-80. [PMID: 34417912 PMCID: PMC8763936 DOI: 10.1007/s10637-021-01156-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/26/2021] [Indexed: 12/15/2022]
Abstract
Background Entrectinib is a CNS-active, potent inhibitor of tyrosine receptor kinases A/B/C, ROS1 and anaplastic lymphoma kinase approved for use in patients with solid tumors. We describe the in vitro and clinical studies investigating potential entrectinib drug-drug interactions. Methods In vitro studies with human biomaterials assessed the enzymes involved in entrectinib metabolism, and whether entrectinib modulates the activity of the major cytochrome P450 (CYP) enzymes or drug transporter P-glycoprotein. Clinical studies investigated the effect of a strong CYP3A4 inhibitor (itraconazole) and inducer (rifampin) on single-dose entrectinib pharmacokinetics. The effect of entrectinib on sensitive probe substrates for CYP3A4 (midazolam) and P-glycoprotein (digoxin) were also investigated. Results Entrectinib is primarily metabolized by CYP3A4. In vitro, entrectinib is a CYP3A4/5 inhibitor (IC50 2 μM) and a weak CYP3A4 inducer. Entrectinib inhibited P-glycoprotein (IC50 1.33 μM) but is a poor substrate. In healthy subjects, itraconazole increased entrectinib Cmax and AUC by 73% and 504%, respectively, and rifampin decreased entrectinib Cmax and AUC by 56% and 77%, respectively. Single dose entrectinib did not affect midazolam AUC, although Cmax decreased by 34%. Multiple dose entrectinib increased midazolam AUC by 50% and decreased Cmax by 21%. Single dose entrectinib increased digoxin AUC and Cmax by 18% and 28%, respectively, but did not affect digoxin renal clearance. Conclusions Entrectinib is a CYP3A4 substrate and is sensitive to the effects of coadministered moderate/strong CYP3A4 inhibitors and strong inducers, and requires dose adjustment. Entrectinib is a weak inhibitor of CYP3A4 and P-glycoprotein and no dose adjustments are required with CYP3A4/P- glycoprotein substrates. Registration Number (Study 2) NCT03330990 (first posted online November 6, 2017) As studies 1 and 3 are phase 1 trials in healthy subjects, they are not required to be registered.
Collapse
Affiliation(s)
| | - Stephen Fowler
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Elena Guerini
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | | | - Li Yu
- Roche Innovation Center, Little Falls, NJ, USA
| | - Francois Mercier
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Mohammed Ullah
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Kenichi Umehara
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Andreas Brink
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Vincent Buchheit
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Elke Zwanziger
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Nassim Djebli
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
30
|
Wu F, Shah H, Li M, Duan P, Zhao P, Suarez S, Raines K, Zhao Y, Wang M, Lin HP, Duan J, Yu L, Seo P. Biopharmaceutics Applications of Physiologically Based Pharmacokinetic Absorption Modeling and Simulation in Regulatory Submissions to the U.S. Food and Drug Administration for New Drugs. AAPS JOURNAL 2021; 23:31. [DOI: 10.1208/s12248-021-00564-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/01/2021] [Indexed: 12/19/2022]
|
31
|
Meneses-Lorente G, Bentley D, Guerini E, Kowalski K, Chow-Maneval E, Yu L, Brink A, Djebli N, Mercier F, Buchheit V, Phipps A. Characterization of the pharmacokinetics of entrectinib and its active M5 metabolite in healthy volunteers and patients with solid tumors. Invest New Drugs 2021; 39:803-811. [PMID: 33462752 PMCID: PMC8068699 DOI: 10.1007/s10637-020-01047-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/09/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Entrectinib is an oral, CNS-active, potent inhibitor of tyrosine receptor kinases A/B/C, tyrosine kinase ROS proto-oncogene 1, and anaplastic lymphoma kinase approved for use in patients with solid tumors. We describe 3 clinical studies, including one investigating the single/multiple dose pharmacokinetics of entrectinib in patients and two studies in healthy volunteers investigating the absorption/distribution/metabolism/excretion (ADME) of entrectinib, its relative bioavailability, and effect of food on pharmacokinetics. METHODS The patient study is open-label with dose-escalation and expansion phases. Volunteers received entrectinib (100-400 mg/m2, and 600-800 mg) once daily with food in continuous 28-day cycles. In the ADME study, volunteers received a single oral dose of [14C]entrectinib 600 mg. In the third study, volunteers received single doses of entrectinib 600 mg as the research and marketed formulations in the fasted state (Part 1), and the marketed formulation in the fed and fasted states (Part 2). Entrectinib and its major active metabolite M5 were assessed in all studies. RESULTS Entrectinib was absorbed in a dose-dependent manner with maximum concentrations at ~4 h postdose and an elimination half-life of ~20 h. Entrectinib was cleared mainly through metabolism and both entrectinib and metabolites were eliminated mainly in feces (minimal renal excretion). At steady-state, the M5-to-entrectinib AUC ratio was 0.5 (with 600 mg entrectinib research formulation in patients). The research and marketed formulations were bioequivalent and food had no relevant effect on pharmacokinetics. CONCLUSIONS Entrectinib is well absorbed, with linear PK that is suitable for once-daily dosing, and can be taken with or without food.
Collapse
Affiliation(s)
| | | | - Elena Guerini
- Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | | | | | - Li Yu
- Roche Innovation Center, Little Falls, NJ, USA
| | - Andreas Brink
- Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Nassim Djebli
- Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Francois Mercier
- Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Vincent Buchheit
- Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Alex Phipps
- Roche Innovation Centre Welwyn, Roche Products Ltd, Welwyn Garden City, UK
| |
Collapse
|
32
|
de Waal T, Rubbens J, Grimm M, Vandecaveye V, Tack J, Weitschies W, Brouwers J, Augustijns P. Exploring the Effect of Esomeprazole on Gastric and Duodenal Fluid Volumes and Absorption of Ritonavir. Pharmaceutics 2020; 12:pharmaceutics12070670. [PMID: 32708859 PMCID: PMC7408179 DOI: 10.3390/pharmaceutics12070670] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
Proton-pump inhibitors (PPIs), frequently prescribed to lower gastric acid secretion, often exert an effect on the absorption of co-medicated drug products. A previous study showed decreased plasma levels of the lipophilic drug ritonavir after co-administration with the PPI Nexium (40 mg esomeprazole), even though duodenal concentrations were not affected. The present study explored if a PPI-induced decrease in gastrointestinal (GI) fluid volume might contribute to the reduced absorption of ritonavir. In an exploratory cross-over study, five volunteers were given a Norvir tablet (100 mg ritonavir) orally, once without PPI pre-treatment and once after a three-day pre-treatment with the PPI esomeprazole. Blood samples were collected for eight hours to assess ritonavir absorption and magnetic resonance imaging (MRI) was used to determine the gastric and duodenal fluid volumes during the first three hours after administration of the tablet. The results confirmed that PPI intake reduced ritonavir plasma concentrations by 40%. The gastric residual volume and gastric fluid volume decreased by 41% and 44% respectively, while the duodenal fluid volume was reduced by 33%. These data suggest that the PPI esomeprazole lowers the available fluid volume for dissolution, which may limit the amount of ritonavir that can be absorbed. Although additional factors may play a role, the effect of PPI intake on the GI fluid volume should be considered when simulating the absorption of poorly soluble drugs like ritonavir in real-life conditions.
Collapse
Affiliation(s)
- Tom de Waal
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49—Box 921, 3000 Leuven, Belgium; (T.d.W.); (J.R.); (J.B.)
| | - Jari Rubbens
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49—Box 921, 3000 Leuven, Belgium; (T.d.W.); (J.R.); (J.B.)
| | - Michael Grimm
- Center of Drug Absorption and Transport, Institute of Pharmacy, University of Greifswald, 17489 Greifswald, Germany; (M.G.); (W.W.)
| | | | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders, TARGID, KU Leuven, 3000 Leuven, Belgium;
| | - Werner Weitschies
- Center of Drug Absorption and Transport, Institute of Pharmacy, University of Greifswald, 17489 Greifswald, Germany; (M.G.); (W.W.)
| | - Joachim Brouwers
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49—Box 921, 3000 Leuven, Belgium; (T.d.W.); (J.R.); (J.B.)
| | - Patrick Augustijns
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49—Box 921, 3000 Leuven, Belgium; (T.d.W.); (J.R.); (J.B.)
- Correspondence:
| |
Collapse
|
33
|
Mackie A, Mulet-Cabero AI, Torcello-Gómez A. Simulating human digestion: developing our knowledge to create healthier and more sustainable foods. Food Funct 2020; 11:9397-9431. [DOI: 10.1039/d0fo01981j] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The gold standard for nutrition studies is clinical trials but they are expensive and variable, and do not always provide the mechanistic information required, hence the increased use ofin vitroand increasinglyin silicosimulations of digestion.
Collapse
Affiliation(s)
- Alan Mackie
- The School of Food Science and Nutrition
- University of Leeds
- Leeds
- UK
| | | | | |
Collapse
|