1
|
Liang F, Zhang Y, Xue Q, Yao N. Exploring inter-ethnic and inter-patient variability and optimal dosing of osimertinib: a physiologically based pharmacokinetic modeling approach. Front Pharmacol 2024; 15:1363259. [PMID: 38500771 PMCID: PMC10946252 DOI: 10.3389/fphar.2024.1363259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 02/22/2024] [Indexed: 03/20/2024] Open
Abstract
Purpose: This study aimed to develop and validate a physiologically based pharmacokinetic (PBPK) model for osimertinib (OSI) to predict plasma trough concentration (Ctrough) and pulmonary EGFRm+ (T790M and L858R mutants) inhibition in Caucasian, Japanese, and Chinese populations. The PBPK model was also utilized to investigate inter-ethnic and inter-patient differences in OSI pharmacokinetics (PK) and determine optimal dosing regimens. Methods: Population PBPK models of OSI for healthy and disease populations were developed using physicochemical and biochemical properties of OSI and physiological parameters of different groups. And then the PBPK models were validated using the multiple clinical PK and drug-drug interaction (DDI) study data. Results: The model demonstrated good consistency with the observed data, with most of prediction-to-observation ratios of 0.8-1.25 for AUC, Cmax, and Ctrough. The PBPK model revealed that plasma exposure of OSI was approximately 2-fold higher in patients compared to healthy individuals, and higher exposure observed in Caucasians compared to other ethnic groups. This was primarily attributed to a lower CL/F of OSI in patients and Caucasian. The PBPK model displayed that key factors influencing PK and EGFRm+ inhibition differences included genetic polymorphism of CYP3A4, CYP1A2 expression, plasma free concentration (fup), albumin level, and auto-inhibition/induction on CYP3A4. Inter-patient PK variability was most influenced by CYP3A4 variants, fup, and albumin level. The PBPK simulations indicated that the optimal dosing regimen for patients across the three populations of European, Japanese, and Chinese ancestry was OSI 80 mg once daily (OD) to achieve the desired range of plasma Ctrough (328-677 nmol/L), as well as 80 mg and 160 mg OD for desirable pulmonary EGFRm+ inhibition (>80%). Conclusion: In conclusion, this study's PBPK simulations highlighted potential ethnic and inter-patient variability in OSI PK and EGFRm+ inhibition between Caucasian, Japanese, and Chinese populations, while also providing insights into optimal dosing regimens of OSI.
Collapse
Affiliation(s)
| | | | | | - Na Yao
- Bethune International Peace Hospital, Shijiazhuang, China
| |
Collapse
|
2
|
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
|
3
|
Gao Y, Jiang B, Kim H, Berberich MJ, Che J, Donovan KA, Hatcher JM, Huerta F, Kwiatkowski NP, Liu Y, Liuni PP, Metivier RJ, Murali VK, Nowak RP, Zhang T, Fischer ES, Gray NS, Jones LH. Catalytic Degraders Effectively Address Kinase Site Mutations in EML4-ALK Oncogenic Fusions. J Med Chem 2023; 66:5524-5535. [PMID: 37036171 DOI: 10.1021/acs.jmedchem.2c01864] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Heterobifunctional degraders, known as proteolysis targeting chimeras (PROTACs), theoretically possess a catalytic mode-of-action, yet few studies have either confirmed or exploited this potential advantage of event-driven pharmacology. Degraders of oncogenic EML4-ALK fusions were developed by conjugating ALK inhibitors to cereblon ligands. Simultaneous optimization of pharmacology and compound properties using ternary complex modeling and physicochemical considerations yielded multiple catalytic degraders that were more resilient to clinically relevant ATP-binding site mutations than kinase inhibitor drugs. Our strategy culminated in the design of the orally bioavailable derivative CPD-1224 that avoided hemolysis (a feature of detergent-like PROTACs), degraded the otherwise recalcitrant mutant L1196M/G1202R in vivo, and commensurately slowed tumor growth, while the third generation ALK inhibitor drug lorlatinib had no effect. These results validate our original therapeutic hypothesis by exemplifying opportunities for catalytic degraders to proactively address binding site resistant mutations in cancer.
Collapse
Affiliation(s)
- Yang Gao
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Baishan Jiang
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Hellen Kim
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Matthew J Berberich
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Jianwei Che
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Katherine A Donovan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - John M Hatcher
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Fidel Huerta
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Nicholas P Kwiatkowski
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Yingpeng Liu
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Peter P Liuni
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Rebecca J Metivier
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Vineeth K Murali
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Radosław P Nowak
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Tinghu Zhang
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Eric S Fischer
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Nathanael S Gray
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Lyn H Jones
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| |
Collapse
|
4
|
Fogli S, Tabbò F, Capuano A, Re MD, Passiglia F, Cucchiara F, Scavone C, Gori V, Novello S, Schmidinger M, Danesi R. The expanding family of c-Met inhibitors in solid tumors: a comparative analysis of their pharmacologic and clinical differences. Crit Rev Oncol Hematol 2022; 172:103602. [DOI: 10.1016/j.critrevonc.2022.103602] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 12/16/2022] Open
|
5
|
Ohminato N, Nagayasu M, Ozeki K, Saitoh R, Ono N, Shibahara N, Suda A, Yoshinari K. In vivo- in vitro correlation of antitumor activity of heat shock protein 90 (HSP90) inhibitors with a pharmacokinetics/pharmacodynamics analysis using NCI-N87 xenograft mice. Xenobiotica 2021; 51:968-976. [PMID: 34134599 DOI: 10.1080/00498254.2021.1942588] [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: 10/21/2022]
Abstract
The in vitro antitumor activity (e.g. IC50) of anticancer drugs is important for selecting candidate compounds for in vivo drug efficacy study in the early stage of drug discovery. In this study, we investigated the relationship between in vitro IC50 and in vivo EC50 using six heat shock protein 90 (HSP90) inhibitors.IC50 of each compound was calculated from in vitro cell proliferation assays using the NCI-N87 cancer cell line. Each compound was administered to NCI-N87 xenograft mice, and EC50 and the maximum tumour-killing rate constant were calculated from pharmacokinetics/pharmacodynamics analyses using plasma concentrations and tumour volumes.IC50 obtained in vitro was poorly correlated with EC50 obtained in vivo, while a good correlation (r = 0.856) was observed between them when corrected with the unbound fraction ratio.The results of this study using of HSP90 inhibitors as model compounds suggest importance of the consideration of an unbound fraction to evaluate the relationship between IC50 and EC50. These results will contribute to improvement in the prediction accuracy of in vivo drug efficacy from in vitro activity and the efficiency of drug discovery research.
Collapse
Affiliation(s)
- Noriaki Ohminato
- Chugai Research Institute for Medical Science, Inc., Gotemba, Shizuoka, Japan.,Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Miho Nagayasu
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Kazuhisa Ozeki
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Ryoichi Saitoh
- Research Division, Chugai Pharmaceutical Co., Ltd., Fujisawa, Kanagawa, Japan; f Sustainability Department, Chugai Pharmaceutical Co., Ltd., Chuo-ku, Tokyo, Japan
| | - Naomi Ono
- Research Division, Chugai Pharmaceutical Co., Ltd., Fujisawa, Kanagawa, Japan; f Sustainability Department, Chugai Pharmaceutical Co., Ltd., Chuo-ku, Tokyo, Japan
| | - Norihito Shibahara
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | - Atsushi Suda
- Sustainability Department, Chugai Pharmaceutical Co., Ltd., Chuo-ku, Tokyo, Japan
| | - Kouichi Yoshinari
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
6
|
Jones RDO, Grondine M, Borodovsky A, San Martin M, DuPont M, D'Cruz C, Schuller A, Henry R, Barry E, Castriotta L, Anjum R, Petersson K, Sahota T, Ahmed GF. A pharmacokinetic-pharmacodynamic model for the MET tyrosine kinase inhibitor, savolitinib, to explore target inhibition requirements for anti-tumour activity. Br J Pharmacol 2020; 178:600-613. [PMID: 33125717 DOI: 10.1111/bph.15301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 10/05/2020] [Accepted: 10/20/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Savolitinib (AZD6094, HMPL-504, volitinib) is an oral, potent, and highly MET receptor TK inhibitor. This series of studies aimed to develop a pharmacokinetic-pharmacodynamic (PK/PD) model to link inhibition of MET phosphorylation (pMET) by savolitinib with anti-tumour activity. EXPERIMENTAL APPROACH Cell line-derived xenograft (CDX) experiments using human lung cancer (EBC-1) and gastric cancer (MKN-45) cells were conducted in athymic nude mice using a variety of doses and schedules of savolitinib. Tumour pMET changes and growth inhibition were calculated after 28 days. Population PK/PD techniques were used to construct a PK/PD model for savolitinib. KEY RESULTS Savolitinib showed dose- and dose frequency-dependent anti-tumour activity in the CDX models, with more frequent, lower dosing schedules (e.g., twice daily) being more effective than intermittent, higher dosing schedules (e.g., 4 days on/3 days off or 2 days on/5 days off). There was a clear exposure-response relationship, with maximal suppression of pMET of >90%. Data from additional CDX and patient-derived xenograft (PDX) models overlapped, allowing calculation of a single EC50 of 0.38 ng·ml-1 . Tumour growth modelling demonstrated that prolonged, high levels of pMET inhibition (>90%) were required for tumour stasis and regression in the models. CONCLUSION AND IMPLICATIONS High and persistent levels of MET inhibition by savolitinib were needed for optimal monotherapy anti-tumour activity in preclinical models. The modelling framework developed here can be used to translate tumour growth inhibition from the mouse to human and thus guide choice of clinical dose and schedule.
Collapse
Affiliation(s)
- Rhys D O Jones
- Oncology R&D, Research and Early Development, AstraZeneca, Cambridge, UK
| | - Mike Grondine
- Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Alexandra Borodovsky
- Formerly Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Maryann San Martin
- Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Michelle DuPont
- Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Celina D'Cruz
- Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Alwin Schuller
- Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Ryan Henry
- Formerly Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Evan Barry
- Formerly Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Lillian Castriotta
- Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Rana Anjum
- Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | | | - Tarjinder Sahota
- BioPharmaceuticals R&D, Clinical Pharmacology and Safety Sciences, AstraZeneca, Cambridge, UK
| | - Ghada F Ahmed
- Formerly BioPharmaceuticals R&D, Clinical Pharmacology and Safety Sciences, AstraZeneca, Cambridge, UK
| |
Collapse
|
7
|
Zou H, Banerjee P, Leung SSY, Yan X. Application of Pharmacokinetic-Pharmacodynamic Modeling in Drug Delivery: Development and Challenges. Front Pharmacol 2020; 11:997. [PMID: 32719604 PMCID: PMC7348046 DOI: 10.3389/fphar.2020.00997] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/19/2020] [Indexed: 12/19/2022] Open
Abstract
With the advancement of technology, drug delivery systems and molecules with more complex architecture are developed. As a result, the drug absorption and disposition processes after administration of these drug delivery systems and engineered molecules become exceedingly complex. As the pharmacokinetic and pharmacodynamic (PK-PD) modeling allows for the separation of the drug-, carrier- and pharmacological system-specific parameters, it has been widely used to improve understanding of the in vivo behavior of these complex delivery systems and help their development. In this review, we summarized the basic PK-PD modeling theory in drug delivery and demonstrated how it had been applied to help the development of new delivery systems and modified large molecules. The linkage between PK and PD was highlighted. In particular, we exemplified the application of PK-PD modeling in the development of extended-release formulations, liposomal drugs, modified proteins, and antibody-drug conjugates. Furthermore, the model-based simulation using primary PD models for direct and indirect PD responses was conducted to explain the assertion of hypothetical minimal effective concentration or threshold in the exposure-response relationship of many drugs and its misconception. The limitations and challenges of the mechanism-based PK-PD model were also discussed.
Collapse
Affiliation(s)
- Huixi Zou
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Parikshit Banerjee
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Sharon Shui Yee Leung
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Xiaoyu Yan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
8
|
Bland AR, Shrestha N, Rosengren RJ, Ashton JC. Does Crizotinib Auto-Inhibit CYP3A in vivo? Pharmacology 2020; 105:715-718. [PMID: 32460299 DOI: 10.1159/000506996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 03/04/2020] [Indexed: 11/19/2022]
Abstract
Crizotinib is a tyrosine kinase inhibitor used to treat anaplastic lymphoma kinase-positive lung cancer. There is in vitro evidence that crizotinib may auto-inhibit cytochrome P450 3A (CYP3A) activity, with important implications for crizotinib pharmacokinetics. In order to test whether crizotinib treatment alters CYP3A activity in vivo, mice were treated with 5 and 25 mg/kg crizotinib (p.o.) daily for 14 days. Results showed that crizotinib treatment did not alter CYP3A activity as determined by erythromycin N-demethylation. In addition, CYP3A polypeptide expression as measured by Western blot was unchanged. Therefore, our results do not support CYP3A inhibition by crizotinib in vivo.
Collapse
Affiliation(s)
- Abigail R Bland
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Nensi Shrestha
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Rhonda J Rosengren
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - John C Ashton
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand,
| |
Collapse
|
9
|
Song L, Yao X, Liu Y, Zhong W, Jiang J, Liu H, Zhou H, Shi C, Zong K, Wang C, Ma C, Liu D, Hu P. Translational prediction of first-in-human pharmacokinetics and pharmacodynamics of janagliflozin, a selective SGLT2 inhibitor, using allometric scaling, dedrick and PK/PD modeling methods. Eur J Pharm Sci 2020; 147:105281. [PMID: 32126254 DOI: 10.1016/j.ejps.2020.105281] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/18/2020] [Accepted: 02/25/2020] [Indexed: 12/22/2022]
Abstract
AIM Janagliflozin is an orally selective SGLT2 inhibitor. To predict human pharmacokinetics/pharmacodynamics (PK/PD) characteristics of janagliflozin. To design optimal starting dose and effective dose for janagliflozin first-in-human (FIH) study. METHODS Animal PK/PD properties of janagliflozin were obtained from preclinical in vivo and in vitro study. Pharmacologically effective level of same class SGLT2 inhibitors were assessed through preclinical and clinical efficacy data of dapagliflozin, empagliflozin and canagliflozin. Human PK parameters and profiles of janagliflozin were predicted by various methods such as allometric scaling (AS), dedrick and PK/PD modeling analysis. Mechanistic PK/PD model was developed to describe janagliflozin-mediated impact on urinary glucose excretion (UGE). Human IC50 was scaled from rat model-estimated IC50 by correcting interspecies difference of in vitro IC50 and plasma fu of rat and human. The quantitative PK/PD prediction of janagliflozin was evaluated via observed PK/PD profiles of healthy subjects. Predicted PK/PD characteristics of janagliflozin were applied in FIH dose design. Optimal starting dose was suggested by considering preclinical PD and toxicity data of janagliflozin. Effective dose was suggested by considering pharmacologically effective level of same class drugs. RESULTS PK/PD characteristics of janagliflozin in preclinical species were summarized. Pharmacologically effective level for SGLT2 inhibitors was defined as 25~30% ΔUGE (ΔUGE=--(PG*GFR)within24h) based on efficacy data of three same class drugs. Human predicted CL, Vss and F were 1.04 L/h, 77.5 L and 0.80. Predicted AUC and Cmax of janagliflozin of 10 and 50 mg were within 0.47~2.08 fold of observed values. Predicted human UGE0-24 h and UGE0-144 h of 10 and 50 mg dose range were within 0.66~1.41 fold of observed values. Optimal starting dose and pharmacologically active dose (PAD) were suggested as 10 mg and 50 mg. Dose range for FIH study was designed as 10-450 mg. CONCLUSIONS This study predicted human PK/PD characteristics of janagliflozin based on preclinical data and provide optimal dose design for janagliflozin FIH study based on pharmacologically effective level of same class drugs.
Collapse
Affiliation(s)
- Ling Song
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, 100032, China; Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing,100191, China.
| | - Xueting Yao
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, 100032, China
| | - Yang Liu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, 100032, China
| | - Wen Zhong
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, 100032, China
| | - Ji Jiang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, 100032, China
| | - Hongzhong Liu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, 100032, China
| | - Huimin Zhou
- XuanZhu Pharma Co., Ltd., Jinan, Shandong, 250101, China
| | - Chongtie Shi
- XuanZhu Pharma Co., Ltd., Jinan, Shandong, 250101, China
| | - Kaiqi Zong
- XuanZhu Pharma Co., Ltd., Jinan, Shandong, 250101, China
| | - Chong Wang
- XuanZhu Pharma Co., Ltd., Jinan, Shandong, 250101, China
| | - Chuanxiang Ma
- XuanZhu Pharma Co., Ltd., Jinan, Shandong, 250101, China
| | - Dongyang Liu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, 100032, China; Drug Clinical Trial Center, Peking University Third Hospital, Beijing 100191, China.
| | - Pei Hu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital & Chinese Academy of Medical Sciences, Beijing, 100032, China.
| |
Collapse
|
10
|
Iida H, Fujikawa R, Kozaki R, Harada R, Hosokawa Y, Ogawara KI, Ohno T. Pharmacokinetic-Pharmacodynamic-Efficacy Modeling of ONO-7579, a Novel Pan-Tropomyosin Receptor Kinase Inhibitor, in a Murine Xenograft Tumor Model. J Pharmacol Exp Ther 2020; 373:361-369. [PMID: 32217770 DOI: 10.1124/jpet.119.264499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/23/2020] [Indexed: 12/26/2022] Open
Abstract
The orally available and novel small molecule ONO-7579 (N-{2-[4-(2-amino-5-chloropyridin-3-yl)phenoxy]pyrimidin-5-yl}-N'-[2-(methanesulfonyl)-5-(trifluoromethyl)phenyl]urea) is a highly potent and selective pan-tropomyosin receptor kinase (TRK) inhibitor. The objective of the present study was to characterize the pharmacokinetic (PK), pharmacodynamic (PD), and antitumor efficacy relationships of ONO-7579 in mice xenografted with a human colorectal cancer cell line, KM12 (harboring the tropomyosin 3 (TPM3) -neurotrophic tyrosine receptor kinase 1 fusion gene), via a PK/PD modeling approach. Plasma and tumor concentrations of ONO-7579, tumor levels of phosphorylated TPM3-TRKA (pTRKA), and tumor volumes in the murine model were measured with a single or multiple dose of ONO-7579 (0.06-0.60 mg/kg) administered once daily. The PK/PD/efficacy models were developed in a sequential manner. Changes in plasma concentrations of ONO-7579 were described with an oral one-compartment model. Tumor concentrations of ONO-7579 were higher than plasma concentrations, and changes in ONO-7579 tumor concentrations were described with an additional tumor compartment that had no influence on plasma concentrations. pTRKA in tumors was described with a direct Emax model, and the tumor ONO-7579 concentration causing 50% of the maximum effect was estimated to be 17.6 ng/g. In addition, a pTRKA-driven tumor growth inhibition model indicated that ONO-7579 started to sharply increase the antitumor effect at pTRKA inhibition rates >60% and required >91.5% to reduce tumors. In conclusion, the developed PK/PD/efficacy models revealed a "switch-like" relationship between pTRKA inhibition rate and antitumor effect in a murine KM12 xenograft model, demonstrating that pTRKA in tumors could serve as an effective biomarker for scheduling the dose regimen in early-stage clinical studies. SIGNIFICANCE STATEMENT: In recent years, clinical development of TRK inhibitors in patients with neurotrophic tyrosine receptor kinase fusion-positive solid tumors has been accelerated. This research found that phosphorylated TRKA was a useful biomarker for explaining the antitumor efficacy of TRK inhibitors using a pharmacokinetic/pharmacodynamic modeling approach in xenograft mice. This finding suggests a rational dosing regimen in early-stage clinical studies for ONO-7579 (N-{2-[4-(2-amino-5-chloropyridin-3-yl)phenoxy]pyrimidin-5-yl}-N'-[2-(methanesulfonyl)-5-(trifluoromethyl)phenyl]urea), a novel pan-TRK inhibitor.
Collapse
Affiliation(s)
- Hiroyuki Iida
- Clinical Pharmacology (H.I., T.O.), Research Center of Oncology (R.F., R.K.), and Pharmacokinetic Research Laboratories (R.H., Y.H.), Ono Pharmaceutical Company Limited, Osaka, Japan; and Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.)
| | - Ryu Fujikawa
- Clinical Pharmacology (H.I., T.O.), Research Center of Oncology (R.F., R.K.), and Pharmacokinetic Research Laboratories (R.H., Y.H.), Ono Pharmaceutical Company Limited, Osaka, Japan; and Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.)
| | - Ryohei Kozaki
- Clinical Pharmacology (H.I., T.O.), Research Center of Oncology (R.F., R.K.), and Pharmacokinetic Research Laboratories (R.H., Y.H.), Ono Pharmaceutical Company Limited, Osaka, Japan; and Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.)
| | - Ryuichi Harada
- Clinical Pharmacology (H.I., T.O.), Research Center of Oncology (R.F., R.K.), and Pharmacokinetic Research Laboratories (R.H., Y.H.), Ono Pharmaceutical Company Limited, Osaka, Japan; and Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.)
| | - Yuya Hosokawa
- Clinical Pharmacology (H.I., T.O.), Research Center of Oncology (R.F., R.K.), and Pharmacokinetic Research Laboratories (R.H., Y.H.), Ono Pharmaceutical Company Limited, Osaka, Japan; and Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.)
| | - Ken-Ichi Ogawara
- Clinical Pharmacology (H.I., T.O.), Research Center of Oncology (R.F., R.K.), and Pharmacokinetic Research Laboratories (R.H., Y.H.), Ono Pharmaceutical Company Limited, Osaka, Japan; and Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.)
| | - Tomoya Ohno
- Clinical Pharmacology (H.I., T.O.), Research Center of Oncology (R.F., R.K.), and Pharmacokinetic Research Laboratories (R.H., Y.H.), Ono Pharmaceutical Company Limited, Osaka, Japan; and Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.)
| |
Collapse
|
11
|
Guerrero YA, Desai D, Sullivan C, Kindt E, Spilker ME, Maurer TS, Solomon DE, Bartlett DW. A Microfluidic Perfusion Platform for In Vitro Analysis of Drug Pharmacokinetic-Pharmacodynamic (PK-PD) Relationships. AAPS JOURNAL 2020; 22:53. [PMID: 32124093 DOI: 10.1208/s12248-020-0430-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/08/2020] [Indexed: 12/14/2022]
Abstract
Static in vitro cell culture studies cannot capture the dynamic concentration profiles of drugs, nutrients, and other factors that cells experience in physiological systems. This limits the confidence in the translational relevance of in vitro experiments and increases the reliance on empirical testing of exposure-response relationships and dose optimization in animal models during preclinical drug development, introducing additional challenges owing to species-specific differences in drug pharmacokinetics (PK) and pharmacodynamics (PD). Here, we describe the development of a microfluidic cell culture device that enables perfusion of cells under 2D or 3D culture conditions with temporally programmable concentration profiles. Proof-of-concept studies using doxorubicin and gemcitabine demonstrated the ability of the microfluidic PK-PD device to examine dose- and time-dependent effects of doxorubicin as well as schedule-dependent effects of doxorubicin and gemcitabine combination therapy on cell viability using both step-wise drug concentration profiles and species-specific (i.e., mouse, human) drug PK profiles. The results demonstrate the importance of including physiologically relevant dynamic drug exposure profiles during in vitro drug testing to more accurately mimic in vivo drug effects, thereby improving translatability across nonclinical studies and reducing the reliance on animal models during drug development.
Collapse
Affiliation(s)
- Yadir A Guerrero
- Neofluidics, 6650 Lusk Blvd, Suite 101, San Diego, California, 92121, USA
| | - Diti Desai
- Neofluidics, 6650 Lusk Blvd, Suite 101, San Diego, California, 92121, USA
| | - Connor Sullivan
- Neofluidics, 6650 Lusk Blvd, Suite 101, San Diego, California, 92121, USA
| | - Erick Kindt
- Pharmacokinetics, Dynamics, & Metabolism, Pfizer Worldwide Research and Development, Pfizer Inc., 10646 Science Center Drive, San Diego, California, 92121, USA
| | - Mary E Spilker
- Pharmacokinetics, Dynamics, & Metabolism, Pfizer Worldwide Research and Development, Pfizer Inc., 10646 Science Center Drive, San Diego, California, 92121, USA
| | - Tristan S Maurer
- Pharmacokinetics, Dynamics, & Metabolism, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, USA
| | - Deepak E Solomon
- Neofluidics, 6650 Lusk Blvd, Suite 101, San Diego, California, 92121, USA.
| | - Derek W Bartlett
- Pharmacokinetics, Dynamics, & Metabolism, Pfizer Worldwide Research and Development, Pfizer Inc., 10646 Science Center Drive, San Diego, California, 92121, USA.
| |
Collapse
|
12
|
Yamazaki S, Gukasyan HJ, Wang H, Uryu S, Sharma S. Translational Pharmacokinetic-Pharmacodynamic Modeling for an Orally Available Novel Inhibitor of Epigenetic Regulator Enhancer of Zeste Homolog 2. J Pharmacol Exp Ther 2020; 373:220-229. [PMID: 32094296 DOI: 10.1124/jpet.119.263491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/12/2020] [Indexed: 11/22/2022] Open
Abstract
PF06821497 has been identified as an orally available small-molecule enhancer of zeste homolog 2 inhibitor. The objectives of the present study were to characterize pharmacokinetic-pharmacodynamic-disease relationships of PF06821497 in xenograft mouse models with diffuse large B-cell lymphoma (Karpas422). An indirect-response model reasonably fit dose-dependent pharmacodynamic responses [histone H3 on lysine 27 (H3K27) me3 inhibition] with an unbound EC 50 of 76 nM, whereas a signal-transduction model sufficiently fit dose-dependent disease responses (tumor growth inhibition) with an unbound tumor stasis concentration (T sc ) of 168 nM. Thus, effective concentration for 70% of maximal effect (EC70) for H3K27me3 inhibition was roughly comparable to T sc , suggesting that 70% H3K27me3 inhibition could be required for tumor stasis. Consistently, an integrated pharmacokinetic-pharmacodynamic-disease model adequately describing tumor growth inhibition also suggested that ∼70% H3K27me3 inhibition was associated with tumor stasis. Based on these results, we would propose that an EC70 estimate for H3K27me3 inhibition corresponding to tumor stasis could be considered a minimum target efficacious concentration of PF06821497 in cancer patients. SIGNIFICANCE STATEMENT: Using a mathematical modeling approach, the quantitative relationships of an orally available anticancer small-molecule enhancer of zeste homolog 2 inhibitor, PF06821497, were characterized among pharmacokinetics, pharmacodynamic biomarker inhibition, and disease responses in nonclinical xenograft models with diffuse large B-cell lymphoma. The modeling results suggest that >70% histone H3 on lysine 27 (H3K27) me3 inhibition would be required for tumor stasis (i.e., 100% tumor growth inhibition). Accordingly, we would propose that an effective concentration for 70% of maximal effect estimate for H3K27me3 inhibition could be considered a minimum target efficacious concentration of PF06821497 in cancer patients.
Collapse
Affiliation(s)
- Shinji Yamazaki
- Pharmacokinetics, Dynamics and Metabolism (S.Y.), Pharmaceutical Science (H.J.G.), and Oncology Research Unit (H.W., S.U., S.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Hovhannes J Gukasyan
- Pharmacokinetics, Dynamics and Metabolism (S.Y.), Pharmaceutical Science (H.J.G.), and Oncology Research Unit (H.W., S.U., S.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Hui Wang
- Pharmacokinetics, Dynamics and Metabolism (S.Y.), Pharmaceutical Science (H.J.G.), and Oncology Research Unit (H.W., S.U., S.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Sean Uryu
- Pharmacokinetics, Dynamics and Metabolism (S.Y.), Pharmaceutical Science (H.J.G.), and Oncology Research Unit (H.W., S.U., S.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Shikhar Sharma
- Pharmacokinetics, Dynamics and Metabolism (S.Y.), Pharmaceutical Science (H.J.G.), and Oncology Research Unit (H.W., S.U., S.S.), Pfizer Worldwide Research & Development, San Diego, California
| |
Collapse
|
13
|
Gu Y, Sai Y, Wang J, Yu M, Wang G, Zhang L, Ren H, Fan S, Ren Y, Qing W, Su W. Preclinical pharmacokinetics, disposition, and translational pharmacokinetic/pharmacodynamic modeling of savolitinib, a novel selective cMet inhibitor. Eur J Pharm Sci 2019; 136:104938. [PMID: 31132401 DOI: 10.1016/j.ejps.2019.05.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/14/2019] [Accepted: 05/23/2019] [Indexed: 01/16/2023]
Abstract
Savolitinib is a novel small-molecule selective cMet inhibitor. This work characterized its pharmacokinetics in preclinical phase, established the preclinical relationships between PK, cMet modulation and anti-tumor efficacy. In vitro and in vivo animal studies were performed for PK characterization. Savolitinib showed good absorption, moderate tissue distribution, low to intermediate clearance, and low accumulation. Hepatic oxidative metabolism followed by urinary and biliary excretions was the major elimination pathway. Based on preclinical PK data, human PK profiles were predicted using empirical methods. Pharmacodynamic studies for evaluating cMet inhibition and anti-tumor efficacy were conducted in nude mice bearing Hs746t xenograft. PK/PD models were built to link the PD measurements to nude mouse PK. The established integrated preclinical PK/PD model contained a two-compartment non-linear PK model, a biomarker link model and a tumor growth transit model. The IC50 of cMet inhibition and the concentration achieving half of the maximal Hs746t tumor reduction by savolitinib were equal to 12.5 and 3.7 nM (free drug), respectively. Based on the predicted human PK data, as well as the established PK/PD model in nude mouse, the human PD (cMet inhibition) profiles were also simulated. This research supported clinical development of savolitinib. Understanding the preclinical PK/PD relationship of savolitinib provides translational insights into the cMet-targeted drug development.
Collapse
Affiliation(s)
- Yi Gu
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Yang Sai
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Jian Wang
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Meijing Yu
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Guanglin Wang
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Li Zhang
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Hongcan Ren
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Shiming Fan
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Yongxin Ren
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Weiguo Qing
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Weiguo Su
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| |
Collapse
|
14
|
Vizirianakis IS, Miliotou AN, Mystridis GA, Andriotis EG, Andreadis II, Papadopoulou LC, Fatouros DG. Tackling pharmacological response heterogeneity by PBPK modeling to advance precision medicine productivity of nanotechnology and genomics therapeutics. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1605828] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Ioannis S. Vizirianakis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Androulla N. Miliotou
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - George A. Mystridis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleftherios G. Andriotis
- Laboratory of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis I. Andreadis
- Laboratory of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Lefkothea C. Papadopoulou
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios G. Fatouros
- Laboratory of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
15
|
Goto A, Abe S, Koshiba S, Yamaguchi K, Sato N, Kurahashi Y. Current status and future perspective on preclinical pharmacokinetic and pharmacodynamic (PK/PD) analysis: Survey in Japan pharmaceutical manufacturers association (JPMA). Drug Metab Pharmacokinet 2019; 34:148-154. [PMID: 30827921 DOI: 10.1016/j.dmpk.2019.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/24/2018] [Accepted: 01/15/2019] [Indexed: 11/29/2022]
Abstract
Preclinical pharmacokinetic/pharmacodynamic (PK/PD) analysis is an efficient tool for the translational research and proof of mechanism/concept in animals. The questionnaire survey on the practice of preclinical PK/PD analysis was conducted in the member companies of the Japan Pharmaceutical Manufacturers Association (JPMA). According to the survey, 60% of companies conducted preclinical PK/PD analysis and its impact for drug development was different between each of the companies. The frequently analyzed therapeutic areas of preclinical PK/PD analysis were neurology, inflammation and metabolic disease, and those are different from the therapeutic area (infectious disease and oncology) in which PK/PD analysis was considered as effective by the present survey. Many companies which have used preclinical PK/PD analysis for the translation to human PK/PD and for the prediction of dose/regimen had good communication with other research & development (R&D) departments (e.g. pharmacology/clinical pharmacology). The increase in resources for preclinical PK/PD analysis including education was highly demanded. As a future perspective, the closer collaboration between pharmacokinetics scientists, pharmacologists, toxicologists and clinical pharmacologists and the increase in resources including upskilling and the comprehension of preclinical PK/PD analysis by the project team are considered to lead to efficient contributions to improve the success ratio of drug discovery and development.
Collapse
Affiliation(s)
- Akihiko Goto
- Non-Clinical Evaluation Expert Committee, Drug Evaluation Committee, Japan Pharmaceutical Manufacturers Association (JPMA), 2-3-11, Nihombashi-honcho, Chuo-ku, Tokyo, 103-0023, Japan; Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, 2-26-1, Muraoka-Higashi, Fujisawa, Kanagawa, 251-8555, Japan.
| | - Sadahiro Abe
- Non-Clinical Evaluation Expert Committee, Drug Evaluation Committee, Japan Pharmaceutical Manufacturers Association (JPMA), 2-3-11, Nihombashi-honcho, Chuo-ku, Tokyo, 103-0023, Japan; Clinical Pharmacology, Clinical Research, Pfizer R&D Japan, Inc., Shinjuku Bunka Quint Building, 3-22-7, Yoyogi, Shibuya-ku, Tokyo, 151-8589, Japan
| | - Shoko Koshiba
- Non-Clinical Evaluation Expert Committee, Drug Evaluation Committee, Japan Pharmaceutical Manufacturers Association (JPMA), 2-3-11, Nihombashi-honcho, Chuo-ku, Tokyo, 103-0023, Japan; Pharmacokinetic Research Laboratories, Translational Research Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., 1188 Shimotogari, Nagaizumi-cho, Sunto-gun, Shizuoka, 411-8731, Japan
| | - Koji Yamaguchi
- Non-Clinical Evaluation Expert Committee, Drug Evaluation Committee, Japan Pharmaceutical Manufacturers Association (JPMA), 2-3-11, Nihombashi-honcho, Chuo-ku, Tokyo, 103-0023, Japan
| | - Nobuo Sato
- Non-Clinical Evaluation Expert Committee, Drug Evaluation Committee, Japan Pharmaceutical Manufacturers Association (JPMA), 2-3-11, Nihombashi-honcho, Chuo-ku, Tokyo, 103-0023, Japan; Pharmacokinetics and Analysis Laboratory, Pharmaceutical Research Center, Meiji Seika Pharma Co., Ltd., 760 Morooka-cho, Kohoku-ku, Yokohama, 222-8567, Japan
| | - Yoshikazu Kurahashi
- Drug Metabolism & Pharmacokinetics Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho Takatsuki, Osaka, 569-1125, Japan
| |
Collapse
|
16
|
Srivastava AK, Hollingshead MG, Govindharajulu JP, Covey JM, Liston D, Simpson MA, Peggins JO, Bottaro DP, Wright JJ, Kinders RJ, Doroshow JH, Parchment RE. Molecular Pharmacodynamics-Guided Scheduling of Biologically Effective Doses: A Drug Development Paradigm Applied to MET Tyrosine Kinase Inhibitors. Mol Cancer Ther 2018; 17:698-709. [PMID: 29444985 PMCID: PMC5935559 DOI: 10.1158/1535-7163.mct-17-0552] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 12/11/2017] [Accepted: 12/29/2017] [Indexed: 11/16/2022]
Abstract
The development of molecularly targeted agents has benefited from use of pharmacodynamic markers to identify "biologically effective doses" (BED) below MTDs, yet this knowledge remains underutilized in selecting dosage regimens and in comparing the effectiveness of targeted agents within a class. We sought to establish preclinical proof-of-concept for such pharmacodynamics-based BED regimens and effectiveness comparisons using MET kinase small-molecule inhibitors. Utilizing pharmacodynamic biomarker measurements of MET signaling (tumor pY1234/1235MET/total MET ratio) in a phase 0-like preclinical setting, we developed optimal dosage regimens for several MET kinase inhibitors and compared their antitumor efficacy in a MET-amplified gastric cancer xenograft model (SNU-5). Reductions in tumor pY1234/1235MET/total MET of 95%-99% were achievable with tolerable doses of EMD1214063/MSC2156119J (tepotinib), XL184 (cabozantinib), and XL880/GSK1363089 (foretinib), but not ARQ197 (tivantinib), which did not alter the pharmacodynamic biomarker. Duration of kinase suppression and rate of kinase recovery were specific to each agent, emphasizing the importance of developing customized dosage regimens to achieve continuous suppression of the pharmacodynamic biomarker at the required level (here, ≥90% MET kinase suppression). The customized dosage regimen of each inhibitor yielded substantial and sustained tumor regression; the equivalent effectiveness of customized dosage regimens that achieve the same level of continuous molecular target control represents preclinical proof-of-concept and illustrates the importance of proper scheduling of targeted agent BEDs. Pharmacodynamics-guided biologically effective dosage regimens (PD-BEDR) potentially offer a superior alternative to pharmacokinetic guidance (e.g., drug concentrations in surrogate tissues) for developing and making head-to-head comparisons of targeted agents. Mol Cancer Ther; 17(3); 698-709. ©2018 AACR.
Collapse
Affiliation(s)
- Apurva K Srivastava
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Melinda G Hollingshead
- Biological Testing Branch, Developmental Therapeutics Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jeevan Prasaad Govindharajulu
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Joseph M Covey
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Dane Liston
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Melanie A Simpson
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - James O Peggins
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Donald P Bottaro
- Urologic Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - John J Wright
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Robert J Kinders
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Ralph E Parchment
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland.
| |
Collapse
|
17
|
C-547, a 6-methyluracil derivative with long-lasting binding and rebinding on acetylcholinesterase: Pharmacokinetic and pharmacodynamic studies. Neuropharmacology 2018; 131:304-315. [DOI: 10.1016/j.neuropharm.2017.12.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 12/07/2017] [Accepted: 12/19/2017] [Indexed: 01/09/2023]
|
18
|
Yang F, Wang B, Liu Z, Xia X, Wang W, Yin D, Sheng L, Li Y. Prediction of a Therapeutic Dose for Buagafuran, a Potent Anxiolytic Agent by Physiologically Based Pharmacokinetic/Pharmacodynamic Modeling Starting from Pharmacokinetics in Rats and Human. Front Pharmacol 2017; 8:683. [PMID: 29066968 PMCID: PMC5641330 DOI: 10.3389/fphar.2017.00683] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/13/2017] [Indexed: 01/29/2023] Open
Abstract
Physiologically based pharmacokinetic (PBPK)/pharmacodynamic (PD) models can contribute to animal-to-human extrapolation and therapeutic dose predictions. Buagafuran is a novel anxiolytic agent and phase I clinical trials of buagafuran have been completed. In this paper, a potentially effective dose for buagafuran of 30 mg t.i.d. in human was estimated based on the human brain concentration predicted by a PBPK/PD modeling. The software GastroPlusTM was used to build the PBPK/PD model for buagafuran in rat which related the brain tissue concentrations of buagafuran and the times of animals entering the open arms in the pharmacological model of elevated plus-maze. Buagafuran concentrations in human plasma were fitted and brain tissue concentrations were predicted by using a human PBPK model in which the predicted plasma profiles were in good agreement with observations. The results provided supportive data for the rational use of buagafuran in clinic.
Collapse
Affiliation(s)
- Fen Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Center of Drug Clinical Trial, Peking University Cancer Hospital and Institute, Beijing, China.,Clinical Pharmacology Research Center, Peking Union Medical College Hospital and Chinese Academy of Medical Sciences, Beijing, China
| | - Baolian Wang
- Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihao Liu
- Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuejun Xia
- Department of Drug Delivery System, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weijun Wang
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dali Yin
- Department of Synthetic Medicinal Chemistry, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Sheng
- Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Li
- Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
19
|
Verheijen RB, Yu H, Schellens JHM, Beijnen JH, Steeghs N, Huitema ADR. Practical Recommendations for Therapeutic Drug Monitoring of Kinase Inhibitors in Oncology. Clin Pharmacol Ther 2017; 102:765-776. [PMID: 28699160 PMCID: PMC5656880 DOI: 10.1002/cpt.787] [Citation(s) in RCA: 205] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/27/2017] [Accepted: 06/29/2017] [Indexed: 01/16/2023]
Abstract
Despite the fact that pharmacokinetic exposure of kinase inhibitors (KIs) is highly variable and clear relationships exist between exposure and treatment outcomes, fixed dosing is still standard practice. This review aims to summarize the available clinical pharmacokinetic and pharmacodynamic data into practical guidelines for individualized dosing of KIs through therapeutic drug monitoring (TDM). Additionally, we provide an overview of prospective TDM trials and discuss the future steps needed for further implementation of TDM of KIs.
Collapse
Affiliation(s)
- Remy B Verheijen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Huixin Yu
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Jan H M Schellens
- Department of Medical Oncology and Clinical Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology and Clinical Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Department of Clinical Pharmacy, Utrecht University Medical Center, Utrecht, The Netherlands
| |
Collapse
|
20
|
Carrara L, Lavezzi SM, Borella E, De Nicolao G, Magni P, Poggesi I. Current mathematical models for cancer drug discovery. Expert Opin Drug Discov 2017; 12:785-799. [PMID: 28595492 DOI: 10.1080/17460441.2017.1340271] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Pharmacometric models represent the most comprehensive approaches for extracting, summarizing and integrating information obtained in the often sparse, limited, and less-than-optimally designed experiments performed in the early phases of oncology drug discovery. Whilst empirical methodologies may be enough for screening and ranking candidate drugs, modeling approaches are needed for optimizing and making economically viable the learn-confirm cycles within an oncology research program and anticipating the dose regimens to be investigated in the subsequent clinical development. Areas covered: Papers appearing in the literature of approximately the last decade reporting modeling approaches applicable to anticancer drug discovery have been listed and commented. Papers were selected based on the interest in the proposed methodology or in its application. Expert opinion: The number of modeling approaches used in the discovery of anticancer drugs is consistently increasing and new models are developed based on the current directions of research of new candidate drugs. These approaches have contributed to a better understanding of new oncological targets and have allowed for the exploitation of the relatively sparse information generated by preclinical experiments. In addition, they are used in translational approaches for guiding and supporting the choice of dosing regimens in early clinical development.
Collapse
Affiliation(s)
- Letizia Carrara
- a Dipartimento di Ingegneria Industriale e dell'Informazione , Università degli Studi di Pavia , Pavia , Italy
| | - Silvia Maria Lavezzi
- a Dipartimento di Ingegneria Industriale e dell'Informazione , Università degli Studi di Pavia , Pavia , Italy
| | - Elisa Borella
- a Dipartimento di Ingegneria Industriale e dell'Informazione , Università degli Studi di Pavia , Pavia , Italy
| | - Giuseppe De Nicolao
- a Dipartimento di Ingegneria Industriale e dell'Informazione , Università degli Studi di Pavia , Pavia , Italy
| | - Paolo Magni
- a Dipartimento di Ingegneria Industriale e dell'Informazione , Università degli Studi di Pavia , Pavia , Italy
| | - Italo Poggesi
- b Global Clinical Pharmacology , Janssen Research and Development , Cologno Monzese , Italy
| |
Collapse
|
21
|
Venkatakrishnan K, Ecsedy JA. Enhancing value of clinical pharmacodynamics in oncology drug development: An alliance between quantitative pharmacology and translational science. Clin Pharmacol Ther 2016; 101:99-113. [PMID: 27804123 DOI: 10.1002/cpt.544] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/23/2016] [Accepted: 10/23/2016] [Indexed: 01/08/2023]
Abstract
Clinical pharmacodynamic evaluation is a key component of the "pharmacologic audit trail" in oncology drug development. We posit that its value can and should be greatly enhanced via application of a robust quantitative pharmacology framework informed by biologically mechanistic considerations. Herein, we illustrate examples of intersectional blindspots across the disciplines of quantitative pharmacology and translational science and offer a roadmap aimed at enhancing the caliber of clinical pharmacodynamic research in the development of oncology therapeutics.
Collapse
Affiliation(s)
- K Venkatakrishnan
- Quantitative Clinical Pharmacology, Takeda Pharmaceuticals International Co, Cambridge, Massachusetts, USA
| | - J A Ecsedy
- Translational and Biomarker Research, Takeda Pharmaceuticals International Co, Cambridge, Massachusetts, USA
| |
Collapse
|
22
|
Yamazaki S, Spilker ME, Vicini P. Translational modeling and simulation approaches for molecularly targeted small molecule anticancer agents from bench to bedside. Expert Opin Drug Metab Toxicol 2016; 12:253-65. [PMID: 26799750 DOI: 10.1517/17425255.2016.1141895] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Recent advances in molecular biology have enabled personalized cancer therapies with molecularly targeted agents (MTAs), which offer a promising future for cancer therapy. Dynamic modeling and simulation (M&S) is a powerful mathematical approach linking drug exposures to pharmacological responses, providing a quantitative assessment of in vivo drug potency. Accordingly, a growing emphasis is being placed upon M&S to quantitatively understand therapeutic exposure-response relationships of MTAs in nonclinical models. AREAS COVERED An overview of M&S approaches for MTAs in nonclinical models is presented with discussion about mechanistic extrapolation of antitumor efficacy from bench to bedside. Emphasis is placed upon recent advances in M&S approaches linking drug exposures, biomarker responses (e.g. target modulation) and pharmacological outcomes (e.g. antitumor efficacy). EXPERT OPINION For successful personalized cancer therapies with MTAs, it is critical to mechanistically and quantitatively understand their exposure-response relationships in nonclinical models, and to logically and properly apply such knowledge to the clinic. Particularly, M&S approaches to predict pharmacologically active concentrations of MTAs in patients based upon nonclinical data would be highly valuable in guiding the design and execution of clinical trials. Proactive approaches to understand their exposure-response relationships could substantially increase probability of achieving a positive proof-of-concept in the clinic.
Collapse
Affiliation(s)
- Shinji Yamazaki
- a Pharmacokinetics, Dynamics & Metabolism , Pfizer Worldwide Research & Development , San Diego , CA , USA
| | - Mary E Spilker
- a Pharmacokinetics, Dynamics & Metabolism , Pfizer Worldwide Research & Development , San Diego , CA , USA
| | - Paolo Vicini
- a Pharmacokinetics, Dynamics & Metabolism , Pfizer Worldwide Research & Development , San Diego , CA , USA
| |
Collapse
|
23
|
Capistrano R, Vangestel C, Wouters A, Dockx Y, Pauwels P, Stroobants S, Apers S, Lardon F, Pieters L, Staelens S. Efficacy Screening of Gloriosa Superba Extracts in a Murine Pancreatic Cancer Model Using (18)F-FDG PET/CT for Monitoring Treatment Response. Cancer Biother Radiopharm 2016; 31:99-109. [PMID: 27093343 DOI: 10.1089/cbr.2015.1954] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE In vivo efficacy of two herbal extracts of Gloriosa superba L. (Colchicaceae) was investigated in a murine pancreatic tumor model by tumor volume measurements and Positron Emission Tomography (PET) imaging using 2-deoxy-2-[(18)F]fluoro-d-glucose ((18)F-FDG). MATERIALS AND METHODS A crude extract of G. superba (GS) seeds rich in colchicine and a colchicine-poor extract (GS2B) containing mostly colchicoside as a putative prodrug were prepared. PANC02-bearing C57BL/6 mice were treated with either placebo, gemcitabine, or one of the extracts (three different doses) for 10 days. Tumor volume measurements were performed daily during treatment and additionally (18)F-FDG Positron emission tomography/computed tomography was acquired at baseline and after 7 days of treatment. Ki-67 and cleaved caspase-3 immunostaining was performed on the resected tumors. RESULTS After 7 days of treatment, a dose-dependent tumor growth inhibition of both extracts was observed with the highest in vivo response at the highest dose of GS and GS2B and gemcitabine. A positive significant correlation was found between Ki-67 scores and relative tumor volumes (RTV), and a negative significant correlation between caspase-3 staining scores and RTV. A decrease in (18)F-FDG uptake was clearly observed in all treatment groups. CONCLUSIONS The therapeutic efficacy of the two different herbal extracts was demonstrated in an in vivo pancreatic tumor model. (18)F-FDG PET was able to detect an early response as overall lower (18)F-FDG uptake was measured in the treated groups.
Collapse
Affiliation(s)
- Rica Capistrano
- 1 Department of Pharmaceutical Sciences, Natural Products & Food Research and Analysis (NatuRA), University of Antwerp , Antwerp, Belgium
| | - Christel Vangestel
- 2 Department of Nuclear Medicine, Antwerp University Hospital , Edegem, Belgium .,3 Molecular Imaging Center Antwerp (MICA), University of Antwerp , Antwerp, Belgium
| | - An Wouters
- 4 Center for Oncological Research (CORE), University of Antwerp , Antwerp, Belgium
| | - Yanina Dockx
- 3 Molecular Imaging Center Antwerp (MICA), University of Antwerp , Antwerp, Belgium
| | - Patrick Pauwels
- 4 Center for Oncological Research (CORE), University of Antwerp , Antwerp, Belgium
| | - Sigrid Stroobants
- 2 Department of Nuclear Medicine, Antwerp University Hospital , Edegem, Belgium .,3 Molecular Imaging Center Antwerp (MICA), University of Antwerp , Antwerp, Belgium
| | - Sandra Apers
- 1 Department of Pharmaceutical Sciences, Natural Products & Food Research and Analysis (NatuRA), University of Antwerp , Antwerp, Belgium
| | - Filip Lardon
- 4 Center for Oncological Research (CORE), University of Antwerp , Antwerp, Belgium
| | - Luc Pieters
- 1 Department of Pharmaceutical Sciences, Natural Products & Food Research and Analysis (NatuRA), University of Antwerp , Antwerp, Belgium
| | - Steven Staelens
- 3 Molecular Imaging Center Antwerp (MICA), University of Antwerp , Antwerp, Belgium
| |
Collapse
|
24
|
Gabrielsson J, Gibbons FD, Peletier LA. Mixture dynamics: Combination therapy in oncology. Eur J Pharm Sci 2016; 88:132-46. [PMID: 27050307 DOI: 10.1016/j.ejps.2016.02.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 01/02/2016] [Accepted: 02/29/2016] [Indexed: 01/05/2023]
Abstract
In recent years combination therapies have become increasingly popular in most therapeutic areas. We present a qualitative and quantitative approach and elucidate some of the challenges and solutions to a more optimal therapy. For tumor growth this involves the study of semi-mechanistic cell-growth/kill models with multiple sites of action. We introduce such models and analyze their dynamic properties using simulations and mathematical analysis. This is done for two specific case studies, one involving a single compound and one a combination of two compounds. We generalize the notion of Tumor Static Concentration to cases when two compounds are involved and develop a graphical method for determining the optimal combination of the two compounds, using ideas akin to those used in studies employing isobolograms. In studying the dynamics of the second case study we focus, not only on the different concentrations, but also on the different dosing regimens and pharmacokinetics of the two compounds.
Collapse
Affiliation(s)
- Johan Gabrielsson
- Swedish University of Agricultural Sciences, Department of Biomedical Sciences and Veterinary Public Health, Division of Pharmacology and Toxicology, Box 7028, SE-750 07 Uppsala, Sweden.
| | - Francis D Gibbons
- DMPK Modeling & Simulation, Oncology IMED, AstraZeneca, Waltham, MA 02151, USA.
| | - Lambertus A Peletier
- Mathematical Institute, Leiden University, PB 9512, 2300 RA Leiden, The Netherlands.
| |
Collapse
|
25
|
Reichel A, Lienau P. Pharmacokinetics in Drug Discovery: An Exposure-Centred Approach to Optimising and Predicting Drug Efficacy and Safety. Handb Exp Pharmacol 2016; 232:235-260. [PMID: 26330260 DOI: 10.1007/164_2015_26] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The role of pharmacokinetics (PK) in drug discovery is to support the optimisation of the absorption, distribution, metabolism and excretion (ADME) properties of lead compounds with the ultimate goal to attain a clinical candidate which achieves a concentration-time profile in the body that is adequate for the desired efficacy and safety profile. A thorough characterisation of the lead compounds aiming at the identification of the inherent PK liabilities also includes an early generation of PK/PD relationships linking in vitro potency and target exposure/engagement with expression of pharmacological activity (mode-of-action) and efficacy in animal studies. The chapter describes an exposure-centred approach to lead generation, lead optimisation and candidate selection and profiling that focuses on a stepwise generation of an understanding between PK/exposure and PD/efficacy relationships by capturing target exposure or surrogates thereof and cellular mode-of-action readouts in vivo. Once robust PK/PD relationship in animal PD models has been constructed, it is translated to anticipate the pharmacologically active plasma concentrations in patients and the human therapeutic dose and dosing schedule which is also based on the prediction of the PK behaviour in human as described herein. The chapter outlines how the level of confidence in the predictions increases with the level of understanding of both the PK and the PK/PD of the new chemical entities (NCE) in relation to the disease hypothesis and the ability to propose safe and efficacious doses and dosing schedules in responsive patient populations. A sound identification of potential drug metabolism and pharmacokinetics (DMPK)-related development risks allows proposing of an effective de-risking strategy for the progression of the project that is able to reduce uncertainties and to increase the probability of success during preclinical and clinical development.
Collapse
Affiliation(s)
- Andreas Reichel
- Research Pharmacokinetics, Global Drug Discovery, Bayer Pharma, Berlin, Germany.
| | - Philip Lienau
- Research Pharmacokinetics, Global Drug Discovery, Bayer Pharma, Berlin, Germany.
| |
Collapse
|
26
|
Deciphering the In Vivo Performance of a Monoclonal Antibody to Neutralize Its Soluble Target at the Site of Action in a Mouse Collagen-Induced Arthritis Model. Pharm Res 2015; 33:1040-9. [PMID: 26718954 DOI: 10.1007/s11095-015-1850-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/22/2015] [Indexed: 12/25/2022]
Abstract
PURPOSE Study the disposition and target-neutralization capability of an anti-interleukin-6 (IL-6) monoclonal antibody (mAb) at the joint in a mouse collagen-induced arthritis (CIA) model. METHODS A mechanistic pharmacokinetic/pharmacodynamic study was conducted in a mouse CIA model using CNTO 345, a rat anti-mouse IL-6 mAb, as model compound. The drug, total/free IL-6 concentrations in both serum and joint lavage fluid were quantitatively assessed and compared to those in the normal control mice. RESULTS CNTO 345 exhibited higher clearance and significantly higher joint lavage/serum ratio in the CIA mice than in the normal control mice. The mAb concentrations in the joint lavage are approximately proportional to the serum concentrations at all the time points being examined. Dosing of CNTO 345 led to sustained free IL-6 suppression in both serum and joint lavage in a dose-dependent manner. A dose-dependent increase in total IL-6 was observed in serum, but not in the joint lavage fluid. Though no change in disease activity was observed following a single dose of anti-IL-6 mAb at peak of the disease, a dose-dependent decrease in serum amyloid A, a downstream biomarker of IL-6, was observed. CONCLUSIONS This study provided quantitative assessments of the distribution and target-neutralization capability of an anti-IL-6 mAb at the site of action in an animal disease model.
Collapse
|
27
|
Optimization of human dose prediction by using quantitative and translational pharmacology in drug discovery. Future Med Chem 2015; 7:2351-69. [PMID: 26599348 DOI: 10.4155/fmc.15.143] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In this perspective article, we explain how quantitative and translational pharmacology, when well-implemented, is believed to lead to improved clinical candidates and drug targets that are differentiated from current treatment options. Quantitative and translational pharmacology aims to build and continuously improve the quantitative relationship between drug exposure, target engagement, efficacy, safety and its interspecies relationship at every phase of drug discovery. Drug hunters should consider and apply these concepts to develop compounds with a higher probability of interrogating the clinical biological hypothesis. We offer different approaches to set an initial effective concentration or pharmacokinetic-pharmacodynamic target in man and to predict human pharmacokinetics that determine together the predicted human dose and dose schedule. All concepts are illustrated with ample literature examples.
Collapse
|
28
|
Sachs JR, Mayawala K, Gadamsetty S, Kang SP, de Alwis DP. Optimal Dosing for Targeted Therapies in Oncology: Drug Development Cases Leading by Example. Clin Cancer Res 2015; 22:1318-24. [DOI: 10.1158/1078-0432.ccr-15-1295] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/05/2015] [Indexed: 11/16/2022]
|
29
|
Kirouac DC, Lahdenranta J, Du J, Yarar D, Onsum MD, Nielsen UB, McDonagh CF. Model-Based Design of a Decision Tree for Treating HER2+ Cancers Based on Genetic and Protein Biomarkers. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2015. [PMID: 26225238 PMCID: PMC4394616 DOI: 10.1002/psp4.19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human cancers are incredibly diverse with regard to molecular aberrations, dependence on oncogenic signaling pathways, and responses to pharmacological intervention. We wished to assess how cellular dependence on the canonical PI3K vs. MAPK pathways within HER2+ cancers affects responses to combinations of targeted therapies, and biomarkers predictive of their activity. Through an integrative analysis of mechanistic model simulations and in vitro cell line profiling, we designed a six-arm decision tree to stratify treatment of HER2+ cancers using combinations of targeted agents. Activating mutations in the PI3K and MAPK pathways (PIK3CA and KRAS), and expression of the HER3 ligand heregulin determined sensitivity to combinations of inhibitors against HER2 (lapatinib), HER3 (MM-111), AKT (MK-2206), and MEK (GSK-1120212; trametinib), in addition to the standard of care trastuzumab (Herceptin). The strategy used to identify effective combinations and predictive biomarkers in HER2-expressing tumors may be more broadly extendable to other human cancers.
Collapse
Affiliation(s)
- D C Kirouac
- Merrimack Pharmaceuticals Cambridge, Massachusetts, USA
| | - J Lahdenranta
- Merrimack Pharmaceuticals Cambridge, Massachusetts, USA
| | - J Du
- Merrimack Pharmaceuticals Cambridge, Massachusetts, USA
| | - D Yarar
- Merrimack Pharmaceuticals Cambridge, Massachusetts, USA
| | - M D Onsum
- Merrimack Pharmaceuticals Cambridge, Massachusetts, USA
| | - U B Nielsen
- Merrimack Pharmaceuticals Cambridge, Massachusetts, USA
| | - C F McDonagh
- Merrimack Pharmaceuticals Cambridge, Massachusetts, USA
| |
Collapse
|
30
|
Hamilton G, Rath B, Burghuber O. Pharmacokinetics of crizotinib in NSCLC patients. Expert Opin Drug Metab Toxicol 2015; 11:835-42. [PMID: 25732197 DOI: 10.1517/17425255.2015.1021685] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION For a subpopulation of NSCLC patients genetic rearrangement of the anaplastic lymphoma kinase (ALK) was found as driver mutation, which can be targeted by the selective inhibitor crizotinib. AREAS COVERED This article presents an overview of the clinical studies that provided the characterization of the pharmacokinetic parameters for the administration of crizotinib to cancer patients and the factors influencing the clinical profiles of this drug. EXPERT OPINION Crizotinib is administered orally as a capsule and clinical studies indicated 250 mg crizotinib BID continuously as the maximal tolerated dose in cancer patients. Bioavailability is ∼ 40% and pharmacokinetic parameters are influenced by food only to a minor degree. This dose of the drug corresponds to a significant inhibition of the mutated ALK, retards tumor growth and achieves clinical responses in the majority of patients. Crizotinib lactam is the single metabolite with minor inhibitory activity for the ALK fusion protein. Metabolization is executed mainly by CYP3A4/5 and is modulated by other drugs interacting with this cytochrome oxidase. Despite the one-fits-all approach in administration of crizotinib at a fixed dose the pharmacokinetic parameters indicate a stable steady state upon continuous administration, which achieves sufficient inhibition of the ALK drug target.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Ludwig Boltzmann Cluster of Translational Oncology , 1090 Vienna , Austria +43 1 40400 66270 ; +43 1 40400 66270 ;
| | | | | |
Collapse
|
31
|
Yamazaki S, Lam JL, Zou HY, Wang H, Smeal T, Vicini P. Mechanistic understanding of translational pharmacokinetic-pharmacodynamic relationships in nonclinical tumor models: a case study of orally available novel inhibitors of anaplastic lymphoma kinase. Drug Metab Dispos 2014; 43:54-62. [PMID: 25349124 DOI: 10.1124/dmd.114.061143] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The orally available novel small molecules PF06463922 [(10R)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-h][2,5,11]benzoxadiazacyclotetradecine-3-carbonitrile] and PF06471402 [(10R)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(azeno)pyrazolo[4,3-h][2,5,11]benzoxadiazacyclo-tetradecine-3-carbonitrile] are second-generation anaplastic lymphoma kinase (ALK) inhibitors targeted to both naïve and resistant patients with non-small cell lung cancer (NSCLC) to the first-generation ALK inhibitor crizotinib. The objectives of the present study were to characterize and compare the pharmacokinetic-pharmacodynamic (PKPD) relationships of PF06463922 and PF06471402 for target modulation in tumor and antitumor efficacy in athymic mice implanted with H3122 NSCLC cells expressing a crizotinib-resistant echinoderm microtubule-associated protein-like 4 (EML4)-ALK mutation, EML4-ALK(L1196M). Furthermore, the PKPD relationships for these ALK inhibitors were evaluated and compared between oral administration and subcutaneous constant infusion (i.e., between different pharmacokinetic [PK] profiles). Oral and subcutaneous PK profiles of these ALK inhibitors were adequately described by a one-compartment PK model. An indirect response model extended with a modulator fit the time courses of PF06463922- and PF06471402-mediated target modulation (i.e., ALK phosphorylation) with an estimated unbound EC50,in vivo of 36 and 20 nM, respectively, for oral administration, and 100 and 69 nM, respectively, for subcutaneous infusion. A drug-disease model based on the turnover concept fit tumor growth curves inhibited by PF06463922 and PF06471402 with estimated unbound tumor stasis concentrations of 51 and 27 nM, respectively, for oral administration, and 116 and 70 nM, respectively, for subcutaneous infusion. Thus, the EC50,in vivo to EC60,in vivo estimates for ALK inhibition corresponded to the concentrations required tumor stasis in all cases, suggesting that the pharmacodynamic relationships of target modulation to antitumor efficacy were consistent among the ALK inhibitors, even when the PK profiles with different administration routes were considerably different.
Collapse
Affiliation(s)
- Shinji Yamazaki
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Justine L Lam
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Helen Y Zou
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Hui Wang
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Tod Smeal
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Paolo Vicini
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| |
Collapse
|
32
|
Stroh M, Duda DG, Takimoto CH, Yamazaki S, Vicini P. Translation of anticancer efficacy from nonclinical models to the clinic. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e128. [PMID: 25098530 PMCID: PMC4150926 DOI: 10.1038/psp.2014.28] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/28/2014] [Indexed: 01/06/2023]
Abstract
Mouse cancer models have provided critical insights into tumor biology; however, clinical translation of these findings has been challenging. This perspective posits that factors impacting on successful translation start with limitations in capturing human cancer pathophysiology and end with challenges in generating robust translatable preclinical end points. A comprehensive approach that considers clinically relevant mouse models with both an integrated biomarker strategy and a complementary modeling and simulation effort will strengthen the current oncology drug development paradigm.
Collapse
Affiliation(s)
- M Stroh
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | - D G Duda
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - C H Takimoto
- Translational Medicine Early Development, Janssen Research & Development, Radnor, Pennsylvania, USA
| | - S Yamazaki
- Pharmacokinetics, Dynamics and Metabolism-New Biological Entities, Pfizer, San Diego, California, USA
| | - P Vicini
- Pharmacokinetics, Dynamics and Metabolism-New Biological Entities, Pfizer, San Diego, California, USA
| |
Collapse
|
33
|
Yamazaki S, Lam JL, Zou HY, Wang H, Smeal T, Vicini P. Translational pharmacokinetic-pharmacodynamic modeling for an orally available novel inhibitor of anaplastic lymphoma kinase and c-Ros oncogene 1. J Pharmacol Exp Ther 2014; 351:67-76. [PMID: 25073473 DOI: 10.1124/jpet.114.217141] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
An orally available macrocyclic small molecule, PF06463922 [(10R)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-h][2,5,11]benzoxadiazacyclotetradecine-3-carbonitrile], is a selective inhibitor of anaplastic lymphoma kinase (ALK) and c-Ros oncogene 1 (ROS1). The objectives of the present study were to characterize the pharmacokinetic-pharmacodynamic relationships of PF06463922 between its systemic exposures, pharmacodynamic biomarker (target modulation), and pharmacologic response (antitumor efficacy) in athymic mice implanted with H3122 non-small cell lung carcinomas expressing echinoderm microtubule-associated protein-like 4 (EML4)-ALK mutation (EML4-ALK(L1196M)) and with NIH3T3 cells expressing CD74-ROS1. In these nonclinical tumor models, PF06463922 was orally administered to animals with EML4-ALK(L1196M) and CD74-ROS1 at twice daily doses of 0.3-20 and 0.01-3 mg/kg per dose, respectively. Plasma concentration-time profiles of PF06463922 were adequately described by a one-compartment pharmacokinetic model. Using the model-simulated plasma concentrations, a pharmacodynamic indirect response model with a modulator sufficiently fit the time courses of target modulation (i.e., ALK phosphorylation) in tumors of EML4-ALK(L1196M)-driven models with EC50,in vivo of 36 nM free. A drug-disease model based on an indirect response model reasonably fit individual tumor growth curves in both EML4-ALK(L1196M)- and CD74-ROS1-driven models with the estimated tumor stasis concentrations of 51 and 6.2 nM free, respectively. Thus, the EC60,in vivo (52 nM free) for ALK inhibition roughly corresponded to the tumor stasis concentration in an EML4-ALK(L1196M)-driven model, suggesting that 60% ALK inhibition would be required for tumor stasis. Accordingly, we proposed that the EC60,in vivo for ALK inhibition corresponding to the tumor stasis could be considered a minimum target efficacious concentration of PF06463922 for cancer patients in a phase I trial.
Collapse
Affiliation(s)
- Shinji Yamazaki
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Justine L Lam
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Helen Y Zou
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Hui Wang
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Tod Smeal
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Paolo Vicini
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| |
Collapse
|
34
|
Practical Guidelines for Therapeutic Drug Monitoring of Anticancer Tyrosine Kinase Inhibitors: Focus on the Pharmacokinetic Targets. Clin Pharmacokinet 2014; 53:305-25. [DOI: 10.1007/s40262-014-0137-2] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
35
|
Huang Q, Johnson TW, Bailey S, Brooun A, Bunker KD, Burke BJ, Collins MR, Cook AS, Cui JJ, Dack KN, Deal JG, Deng YL, Dinh D, Engstrom LD, He M, Hoffman J, Hoffman RL, Johnson PS, Kania RS, Lam H, Lam JL, Le PT, Li Q, Lingardo L, Liu W, Lu MW, McTigue M, Palmer CL, Richardson PF, Sach NW, Shen H, Smeal T, Smith GL, Stewart AE, Timofeevski S, Tsaparikos K, Wang H, Zhu H, Zhu J, Zou HY, Edwards MP. Design of Potent and Selective Inhibitors to Overcome Clinical Anaplastic Lymphoma Kinase Mutations Resistant to Crizotinib. J Med Chem 2014; 57:1170-87. [DOI: 10.1021/jm401805h] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Qinhua Huang
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Ted W. Johnson
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Simon Bailey
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Alexei Brooun
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Kevin D. Bunker
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Benjamin J. Burke
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Michael R. Collins
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Andrew S. Cook
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - J. Jean Cui
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Kevin N. Dack
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Judith G. Deal
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Ya-Li Deng
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Dac Dinh
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Lars D. Engstrom
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Mingying He
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Jacqui Hoffman
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Robert L. Hoffman
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Patrick S. Johnson
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Robert S. Kania
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Hieu Lam
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Justine L. Lam
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Phuong T. Le
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Qiuhua Li
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Laura Lingardo
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Wei Liu
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Melissa West Lu
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Michele McTigue
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Cynthia L. Palmer
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Paul F. Richardson
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Neal W. Sach
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Hong Shen
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Tod Smeal
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Graham L. Smith
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Albert E. Stewart
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Sergei Timofeevski
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Konstantinos Tsaparikos
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Hui Wang
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Huichun Zhu
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Jinjiang Zhu
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Helen Y. Zou
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Martin P. Edwards
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
| |
Collapse
|
36
|
Wu B, Sun YN. Pharmacokinetics of Peptide-Fc fusion proteins. J Pharm Sci 2013; 103:53-64. [PMID: 24285510 DOI: 10.1002/jps.23783] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 10/15/2013] [Accepted: 10/22/2013] [Indexed: 01/11/2023]
Abstract
Peptide-Fc fusion proteins (or peptibodies) are chimeric proteins generated by fusing a biologically active peptide with the Fc-domain of immunoglobulin G. In this review, we describe recent studies that have evaluated the absorption, distribution, metabolism, and excretion characteristics of peptibodies. Key features of the pharmacokinetics of peptibodies include their extended half-life due to recycling by the neonatal Fc receptor (FcRn), a substantial contribution by renal excretion to total clearance and, for certain peptibodies, target-mediated drug disposition. The prolonged half-life of peptibodies permits less-frequent dose administration compared with small therapeutic peptides, thereby supporting patient convenience and compliance. Hence, a considerable number of peptibodies are currently in preclinical and clinical development. Investigation of the metabolism (biotransformation) of biologics is an evolving area of research: ligand-binding mass spectrometry techniques have been employed for the characterization of the peptibody romiplostim, providing a new approach to evaluation of the degradation products of biologics. Pharmacokinetic/pharmacodynamic modeling and simulation techniques have been used to predict the pharmacokinetics of peptibodies which can inform clinical decision-making, particularly selection of dosing regimens. This integrated review highlights the distinct pharmacokinetic characteristics of peptibodies and their influence on the drug development process for this emerging family of therapeutics.
Collapse
Affiliation(s)
- Benjamin Wu
- Department of Pharmacokinetics and Drug Metabolism, Quantitative Pharmacology Group, Amgen Inc, Thousand Oaks, California, 91320
| | | |
Collapse
|