1
|
Sharifian A, Varshosaz J, Aliomrani M, Kazemi M. Nose to brain delivery of ibudilast micelles for treatment of multiple sclerosis in an experimental autoimmune encephalomyelitis animal model. Int J Pharm 2023; 638:122936. [PMID: 37030640 DOI: 10.1016/j.ijpharm.2023.122936] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
Multiple sclerosis is a chronic inflammatory disease of the central nervous system ultimate to neurodegeneration and demyelination. Ibudilast is a phosphodiesterase inhibitor, effective on the function of glial cells and lymphocytes, and inhibits the release of TNF-α by inflammatory cells. Dysregulation of glia is one of the most important pathological causes of MS. Therefore, ibudilast as a glial attenuator can be a useful treatment. The objective of the present study was to investigate the effect of nasal spray of polydopamine coated micelles of surfactin, a biosurfactant, loaded with ibudilast on its brain targeted delivery and effectiveness in remylination and neuroprotection in animal model of MS. In animal studies the micelles were administrated intranasally in different doses of 10, 25 and 50 mg/kg/day in C57/BL6 mice immunized by experimental autoimmune encephalomyelitis (EAE) model. The results of Luxol fast blue staining indicated increment in myelin fiber percent more significantly (p<0.05) in the groups treated with the polydopamine coated micelles (PDAM) compared to nasal spray of free drug or oral administration. These formulations also increased expression of Mbp, Olig2 and Mog genes in the corpus callosum. These results suggest a positive outcome of polydopamine coated micelles loaded with ibudilast in active MS as an anti-inflammatory and neuroprotective agent.
Collapse
Affiliation(s)
- Akram Sharifian
- Department of Pharmaceutics, Novel Drug Delivery Systems Research Centre, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Jaleh Varshosaz
- Department of Pharmaceutics, Novel Drug Delivery Systems Research Centre, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mehdi Aliomrani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohammad Kazemi
- Department of Genetics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
2
|
Fischer H, Senn C, Ullah M, Cantrill C, Schuler F, Yu L. Calculation of an Apical Efflux Ratio from P-Glycoprotein (P-gp) In Vitro Transport Experiments Shows an Improved Correlation with In Vivo Cerebrospinal Fluid Measurements in Rats: Impact on P-gp Screening and Compound Optimization. J Pharmacol Exp Ther 2020; 376:322-329. [PMID: 33288523 DOI: 10.1124/jpet.120.000158] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/02/2020] [Indexed: 11/22/2022] Open
Abstract
P-glycoprotein (P-gp) is a major blood-brain barrier (BBB) efflux transporter. In vitro approaches, including bidirectional efflux ratio (ER), are used to measure P-gp-mediated transport, but findings can be inconsistent across models. We propose a novel, more physiologically relevant, in vitro model: unidirectional apical efflux ratio (AP-ER)-a ratio of permeability rates at the apical side of the BBB with and without P-gp inhibitor. To test our approach, ER and AP-ER were calculated for 3227 structurally diverse compounds in porcine kidney epithelial cells (LLC-PK1) overexpressing human or mouse P-gp and classified based on their passive transcellular P-gp permeability or charged properties. In vivo rat infusion studies were performed for selected compounds with high ER but low AP-ER. One-third of the 3227 compounds had bidirectional ER that was much higher than AP-ER; very few had AP-ER higher than ER. Compounds with a large difference between AP-ER and ER were typically basic compounds with low-to-medium passive permeability and high lipophilicity and/or amphiphilicity, leading to strong membrane binding. Outcomes in the human model were similar to those in mice, suggesting AP-ER/ER ratios may be conserved for at least two species. AP-ER predicted measured cerebrospinal fluid (CSF) concentration better than ER for the five compounds tested in our in vivo rat infusion studies. We report superior estimations of the CSF concentrations of the compounds when based on less resource-intensive AP-ER versus classic ER. Better understanding of the properties leading to high P-gp-mediated efflux in vivo could support more efficient brain-penetrant compound screening and optimization. SIGNIFICANCE STATEMENT: To address inconsistencies associated with the historical, bidirectional efflux ratio (ER) calculation of P-glycoprotein-mediated transport, we propose to use the novel, more physiologically relevant, unidirectional apical efflux ratio (AP-ER) model. In vitro experiments suggested that compounds with strong membrane binding showed the largest difference between AP-ER and ER, and in vivo infusion studies showed that AP-ER predicted cerebrospinal fluid concentrations of compounds better than ER; outcomes in the human model were similar to those in mice.
Collapse
Affiliation(s)
- Holger Fischer
- Roche Pharmaceutical Research and Early Development, DMPK/PD project leader (H.F.), Comparative Pharmacology (C.S.), Investigative Safety, Pharmaceutical Sciences (M.U., C.C.), and Immunology, Infectious Disease and Ophthalmology (F.S.), Roche Innovation Center, Basel, Switzerland; and Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Translational and Clinical Research Center, Inc., and LIYU Pharmaceutical Consulting LCC, New Jersey, USA (L.Y.)
| | - Claudia Senn
- Roche Pharmaceutical Research and Early Development, DMPK/PD project leader (H.F.), Comparative Pharmacology (C.S.), Investigative Safety, Pharmaceutical Sciences (M.U., C.C.), and Immunology, Infectious Disease and Ophthalmology (F.S.), Roche Innovation Center, Basel, Switzerland; and Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Translational and Clinical Research Center, Inc., and LIYU Pharmaceutical Consulting LCC, New Jersey, USA (L.Y.)
| | - Mohammed Ullah
- Roche Pharmaceutical Research and Early Development, DMPK/PD project leader (H.F.), Comparative Pharmacology (C.S.), Investigative Safety, Pharmaceutical Sciences (M.U., C.C.), and Immunology, Infectious Disease and Ophthalmology (F.S.), Roche Innovation Center, Basel, Switzerland; and Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Translational and Clinical Research Center, Inc., and LIYU Pharmaceutical Consulting LCC, New Jersey, USA (L.Y.)
| | - Carina Cantrill
- Roche Pharmaceutical Research and Early Development, DMPK/PD project leader (H.F.), Comparative Pharmacology (C.S.), Investigative Safety, Pharmaceutical Sciences (M.U., C.C.), and Immunology, Infectious Disease and Ophthalmology (F.S.), Roche Innovation Center, Basel, Switzerland; and Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Translational and Clinical Research Center, Inc., and LIYU Pharmaceutical Consulting LCC, New Jersey, USA (L.Y.)
| | - Franz Schuler
- Roche Pharmaceutical Research and Early Development, DMPK/PD project leader (H.F.), Comparative Pharmacology (C.S.), Investigative Safety, Pharmaceutical Sciences (M.U., C.C.), and Immunology, Infectious Disease and Ophthalmology (F.S.), Roche Innovation Center, Basel, Switzerland; and Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Translational and Clinical Research Center, Inc., and LIYU Pharmaceutical Consulting LCC, New Jersey, USA (L.Y.)
| | - Li Yu
- Roche Pharmaceutical Research and Early Development, DMPK/PD project leader (H.F.), Comparative Pharmacology (C.S.), Investigative Safety, Pharmaceutical Sciences (M.U., C.C.), and Immunology, Infectious Disease and Ophthalmology (F.S.), Roche Innovation Center, Basel, Switzerland; and Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Translational and Clinical Research Center, Inc., and LIYU Pharmaceutical Consulting LCC, New Jersey, USA (L.Y.)
| |
Collapse
|
3
|
Shaw RC, Tamagnan GD, Tavares AAS. Rapidly (and Successfully) Translating Novel Brain Radiotracers From Animal Research Into Clinical Use. Front Neurosci 2020; 14:871. [PMID: 33117115 PMCID: PMC7559529 DOI: 10.3389/fnins.2020.00871] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 07/27/2020] [Indexed: 12/26/2022] Open
Abstract
The advent of preclinical research scanners for in vivo imaging of small animals has added confidence into the multi-step decision-making process of radiotracer discovery and development. Furthermore, it has expanded the utility of imaging techniques available to dissect clinical questions, fostering a cyclic interaction between the clinical and the preclinical worlds. Significant efforts from medicinal chemistry have also made available several high-affinity and selective compounds amenable for radiolabeling, that target different receptors, transporters and enzymes in vivo. This substantially increased the range of applications of molecular imaging using positron emission tomography (PET) or single photon emission computed tomography (SPECT). However, the process of developing novel radiotracers for in vivo imaging of the human brain is a multi-step process that has several inherent pitfalls and technical difficulties, which often hampers the successful translation of novel imaging agents from preclinical research into clinical use. In this paper, the process of radiotracer development and its relevance in brain research is discussed; as well as, its pitfalls, technical challenges and future promises. Examples of successful and unsuccessful translation of brain radiotracers will be presented.
Collapse
Affiliation(s)
- Robert C. Shaw
- BHF Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Adriana Alexandre S. Tavares
- BHF Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
4
|
Huang L, Wells MC, Zhao Z. A Practical Perspective on the Evaluation of Small Molecule CNS Penetration in Drug Discovery. Drug Metab Lett 2020; 13:78-94. [PMID: 30854983 DOI: 10.2174/1872312813666190311125652] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 02/20/2019] [Accepted: 03/01/2019] [Indexed: 01/16/2023]
Abstract
The separation of the brain from blood by the blood-brain barrier and the bloodcerebrospinal fluid (CSF) barrier poses unique challenges for the discovery and development of drugs targeting the central nervous system (CNS). This review will describe the role of transporters in CNS penetration and examine the relationship between unbound brain (Cu-brain) and unbound plasma (Cu-plasma) or CSF (CCSF) concentration. Published data demonstrate that the relationship between Cu-brain and Cu-plasma or CCSF can be affected by transporter status and passive permeability of a drug and CCSF may not be a reliable surrogate for CNS penetration. Indeed, CCSF usually over-estimates Cu-brain for efflux substrates and it provides no additional value over Cu-plasma as the surrogate of Cu-brain for highly permeable non-efflux substrates. A strategy described here for the evaluation of CNS penetration is to use in vitro permeability, P-glycoprotein (Pgp) and breast cancer resistance protein efflux assays and Cu-brain/Cu-plasma in preclinical species. Cu-plasma should be used as the surrogate of Cu-brain for highly permeable non-efflux substrates with no evidence of impaired distribution into the brain. When drug penetration into the brain is impaired, we recommend using (total brain concentration * unbound fraction in the brain) as Cu-brain in preclinical species or Cu-plasma/in vitro Pgp efflux ratio if Pgp is the major limiting mechanism for brain penetration.
Collapse
Affiliation(s)
- Liyue Huang
- Epizyme Inc, 400 Technology Square, Cambridge, MA-02139, United States
| | - Mary C Wells
- Vertex Pharmaceuticals, 50 Northern Ave, Boston, MA-02210, United States
| | - Zhiyang Zhao
- Alliance Pharma, Inc. 17 Lee Blvd. Malvern, PA-19355, United States
| |
Collapse
|
5
|
Schubert JW, Harrison ST, Mulhearn J, Gomez R, Tynebor R, Jones K, Bunda J, Hanney B, Wai JM, Cox C, McCauley JA, Sanders JM, Magliaro B, O'Brien J, Pajkovic N, Huszar Agrapides SL, Taylor A, Gotter A, Smith SM, Uslaner J, Browne S, Risso S, Egbertson M. Discovery, Optimization, and Biological Characterization of 2,3,6‐Trisubstituted Pyridine‐Containing M
4
Positive Allosteric Modulators. ChemMedChem 2019; 14:943-951. [PMID: 30920765 DOI: 10.1002/cmdc.201900088] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Indexed: 11/08/2022]
Affiliation(s)
| | - Scott T. Harrison
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - James Mulhearn
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - Robert Gomez
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - Robert Tynebor
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - Kristen Jones
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - Jaime Bunda
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - Barbara Hanney
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | | | - Chris Cox
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - John A. McCauley
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - John M. Sanders
- Department of Computational and Structural ChemistryMerck & Co., Inc. West Point PA USA
| | - Brian Magliaro
- Department of In Vitro PharmacologyMerck & Co., Inc. West Point PA USA
| | - Julie O'Brien
- Department of In Vitro PharmacologyMerck & Co., Inc. West Point PA USA
| | - Natasa Pajkovic
- Department of Pharmacokinetics, Pharmacodynamics, and Drug MetabolismMerck & Co., Inc West Point PA USA
| | | | - Anne Taylor
- Department of In Vivo PharmacologyMerck & Co., Inc. West Point PA USA
| | - Anthony Gotter
- Department of Neuroscience ResearchMerck & Co., Inc. West Point PA USA
| | - Sean M. Smith
- Department of Neuroscience ResearchMerck & Co., Inc. West Point PA USA
| | - Jason Uslaner
- Department of Neuroscience ResearchMerck & Co., Inc. West Point PA USA
| | - Susan Browne
- Department of In Vivo PharmacologyMerck & Co., Inc. West Point PA USA
| | - Stefania Risso
- Department of Neuroscience ResearchMerck & Co., Inc. West Point PA USA
| | - Melissa Egbertson
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| |
Collapse
|
6
|
The Impact of Endogenous Breast Cancer Resistance Protein on Human P-Glycoprotein-Mediated Transport Assays Using LLC-PK1 Cells Transfected With Human P-Glycoprotein. J Pharm Sci 2019; 108:1085-1089. [DOI: 10.1016/j.xphs.2018.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/19/2018] [Accepted: 10/08/2018] [Indexed: 01/13/2023]
|
7
|
Miyamoto R, Nozawa T, Kimura M, Shiozuka K, Tabata K. Development and Validation of Semiautomated 96-Well Transport Assay Using LLC-PK1 Cells Transfected with Human P-Glycoprotein for High-Throughput Screening. Assay Drug Dev Technol 2015; 13:79-87. [DOI: 10.1089/adt.2014.621] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Rei Miyamoto
- Drug Metabolism and Pharmacokinetics Research Division, Astellas Research Technologies Co., Ltd., Ibaraki, Japan
| | - Takashi Nozawa
- Analysis and Pharmacokinetics Research Labs, Astellas Pharma, Inc., Ibaraki, Japan
| | - Mayuko Kimura
- Drug Metabolism and Pharmacokinetics Research Division, Astellas Research Technologies Co., Ltd., Ibaraki, Japan
| | - Koichi Shiozuka
- Drug Metabolism and Pharmacokinetics Research Division, Astellas Research Technologies Co., Ltd., Ibaraki, Japan
| | - Kenji Tabata
- Analysis and Pharmacokinetics Research Labs, Astellas Pharma, Inc., Ibaraki, Japan
| |
Collapse
|
8
|
Johnson TW, Richardson PF, Bailey S, Brooun A, Burke BJ, Collins MR, Cui JJ, Deal JG, Deng YL, Dinh D, Engstrom LD, He M, Hoffman J, Hoffman RL, Huang Q, Kania RS, Kath JC, Lam H, Lam JL, Le PT, Lingardo L, Liu W, McTigue M, Palmer CL, Sach NW, Smeal T, Smith GL, Stewart AE, Timofeevski S, Zhu H, Zhu J, Zou HY, Edwards MP. Discovery of (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 (PF-06463922), a Macrocyclic Inhibitor of Anaplastic Lymphoma Kinase (ALK) and c-ros Oncogene 1 (ROS1) with Preclinical Brain Exposure and Broad-Spectrum Potency against ALK-Resistant Mutations. J Med Chem 2014; 57:4720-44. [DOI: 10.1021/jm500261q] [Citation(s) in RCA: 350] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Ted W. Johnson
- 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
| | - 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
| | - 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
| | - J. Jean Cui
- 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
| | - Qinhua Huang
- 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
| | - John C. Kath
- 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
| | - 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
| | - 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
| | - Neal W. Sach
- 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
| | - 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
|
9
|
Wilhelm I, Krizbai IA. In vitro models of the blood-brain barrier for the study of drug delivery to the brain. Mol Pharm 2014; 11:1949-63. [PMID: 24641309 DOI: 10.1021/mp500046f] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The most important obstacle to the drug delivery into the brain is the presence of the blood-brain barrier, which limits the traffic of substances between the blood and the nervous tissue. Therefore, adequate in vitro models need to be developed in order to characterize the penetration properties of drug candidates into the central nervous system. This review article summarizes the presently used and the most promising in vitro BBB models based on the culture of brain endothelial cells. Robust models can be obtained using primary porcine brain endothelial cells and rodent coculture models, which have low paracellular permeability and express functional efflux transporters, showing good correlation of drug penetration data with in vivo results. Models mimicking the in vivo anatomophysiological complexity of the BBB are also available, including triple coculture (culture of brain endothelial cells in the presence of pericytes and astrocytes), dynamic, and microfluidic models; however, these are not suitable for rapid, high throughput studies. Potent human cell lines would be needed for easily available and reproducible models which avoid interspecies differences.
Collapse
Affiliation(s)
- Imola Wilhelm
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences , Temesvári krt. 62, 6726 Szeged, Hungary
| | | |
Collapse
|
10
|
Caruso A, Alvarez-Sánchez R, Hillebrecht A, Poirier A, Schuler F, Lavé T, Funk C, Belli S. PK/PD assessment in CNS drug discovery: Prediction of CSF concentration in rodents for P-glycoprotein substrates and application to in vivo potency estimation. Biochem Pharmacol 2013; 85:1684-99. [DOI: 10.1016/j.bcp.2013.02.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 02/13/2013] [Accepted: 02/14/2013] [Indexed: 12/22/2022]
|
11
|
Deo AK, Theil FP, Nicolas JM. Confounding Parameters in Preclinical Assessment of Blood–Brain Barrier Permeation: An Overview With Emphasis on Species Differences and Effect of Disease States. Mol Pharm 2013; 10:1581-95. [DOI: 10.1021/mp300570z] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Anand K. Deo
- UCB Pharma S.A., Chemin du Foriest, B-1420 Braine-l’Alleud,
Belgium
| | | | | |
Collapse
|
12
|
Hamill TG, Eng W, Jennings A, Lewis R, Thomas S, Wood S, Street L, Wisnoski D, Wolkenberg S, Lindsley C, Sanabria-Bohórquez SM, Patel S, Riffel K, Ryan C, Cook J, Sur C, Burns HD, Hargreaves R. The synthesis and preclinical evaluation in rhesus monkey of [¹⁸F]MK-6577 and [¹¹C]CMPyPB glycine transporter 1 positron emission tomography radiotracers. Synapse 2011; 65:261-70. [PMID: 20687108 DOI: 10.1002/syn.20842] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Two positron emission tomography radiotracers for the glycine transporter 1 (GlyT1) are reported here. Each radiotracer is a propylsulfonamide-containing benzamide and was labeled with either carbon-11 or fluorine-18. [¹¹C]CMPyPB was synthesized by the alkylation of a 3-hydroxypyridine precursor using [¹¹C]MeI, and [¹⁸F]MK-6577 was synthesized by a nucleophilic aromatic substitution reaction using a 2-chloropyridine precursor. Each tracer shows good uptake into rhesus monkey brain with the expected distribution of highest uptake in the pons, thalamus, and cerebellum and lower uptake in the striatum and gray matter of the frontal cortex. In vivo blockade and chase studies of [¹⁸F]MK-6577 showed a large specific signal and reversible binding. In vitro autoradiographic studies with [¹⁸F]MK-6577 showed a large specific signal in both rhesus monkey and human brain slices and a distribution consistent with the in vivo results and those reported in the literature. In vivo metabolism studies in rhesus monkeys demonstrated that only more-polar metabolites are formed for each tracer. Of these two tracers, [¹⁸F]MK-6577 was more extensively characterized and is a promising clinical positron emission tomography tracer for imaging GlyT1 and for measuring GlyT1 occupancy of therapeutic compounds.
Collapse
Affiliation(s)
- Terence G Hamill
- Discovery Imaging, Merck Research Laboratories, West Point, Pennsylvania 19486, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Wager TT, Villalobos A, Verhoest PR, Hou X, Shaffer CL. Strategies to optimize the brain availability of central nervous system drug candidates. Expert Opin Drug Discov 2011; 6:371-81. [DOI: 10.1517/17460441.2011.564158] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
14
|
Hostetler ED, Eng W, Joshi AD, Sanabria-Bohórquez S, Kawamoto H, Ito S, O'Malley S, Krause S, Ryan C, Patel S, Williams M, Riffel K, Suzuki G, Ozaki S, Ohta H, Cook J, Burns HD, Hargreaves R. Synthesis, characterization, and monkey PET studies of [18F]MK-1312, a PET tracer for quantification of mGluR1 receptor occupancy by MK-5435. Synapse 2010; 65:125-35. [DOI: 10.1002/syn.20826] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
15
|
Hostetler ED, Sanabria-Bohórquez S, Fan H, Zeng Z, Gantert L, Williams M, Miller P, O'Malley S, Kameda M, Ando M, Sato N, Ozaki S, Tokita S, Ohta H, Williams D, Sur C, Cook JJ, Burns HD, Hargreaves R. Synthesis, characterization, and monkey positron emission tomography (PET) studies of [18F]Y1-973, a PET tracer for the neuropeptide Y Y1 receptor. Neuroimage 2010; 54:2635-42. [PMID: 21078401 DOI: 10.1016/j.neuroimage.2010.11.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 10/25/2010] [Accepted: 11/02/2010] [Indexed: 11/28/2022] Open
Abstract
Neuropeptide Y receptor subtype 1 (NPY Y1) has been implicated in appetite regulation, and antagonists of NPY Y1 are being explored as potential therapeutics for obesity. An NPY Y1 PET tracer is useful for determining the level of target engagement by NPY Y1 antagonists in preclinical and clinical studies. Here we report the synthesis and evaluation of [(18)F]Y1-973, a novel PET tracer for NPY Y1. [(18)F]Y1-973 was radiolabeled by reaction of a primary chloride with [(18)F]KF/K2.2.2 followed by deprotection with HCl. [(18)F]Y1-973 was produced with high radiochemical purity (>98%) and high specific activity (>1000 Ci/mmol). PET studies in rhesus monkey brain showed that the distribution of [(18)F]Y1-973 was consistent with the known NPY Y1 distribution; uptake was highest in the striatum and cortical regions and lowest in the pons, cerebellum nuclei, and brain stem. Blockade of [(18)F]Y1-973 uptake with NPY Y1 antagonist Y1-718 revealed a specific signal that was dose-dependently reduced in all regions of grey matter to a similarly low level of tracer uptake, indicative of an NPY Y1 specific signal. In vitro autoradiographic studies with [(18)F]Y1-973 in rhesus monkey and human brain tissue slices revealed an uptake distribution consistent with the in vivo PET studies. Highest binding density was observed in the dentate gyrus, caudate-putamen, and cortical regions; moderate binding density in the hypothalamus and thalamus; and lowest binding density in the globus pallidus and cerebellum. In vitro saturation binding studies in rhesus monkey and human caudate-putamen homogenates confirmed a similarly high B(max)/K(d) ratio for [(18)F]Y1-973, suggesting the tracer may provide a specific signal in human brain of similar magnitude to that observed in rhesus monkey. [(18)F]Y1-973 is a suitable PET tracer for imaging NPY Y1 in rhesus monkey with potential for translation to human PET studies.
Collapse
Affiliation(s)
- Eric D Hostetler
- Imaging, Merck Research Laboratories, West Point, PA 19486, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Sanftner LM, Gibbons JA, Gross MI, Suzuki BM, Gaeta FCA, Johnson KW. Cross-species comparisons of the pharmacokinetics of ibudilast. Xenobiotica 2010; 39:964-77. [PMID: 19925385 DOI: 10.3109/00498250903254340] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
To enable clinical development of ibudilast for new indications, its pharmacokinetics were characterized in mice, rats, rabbits, dogs, cynomolgus monkeys, and minipigs. Animal pharmacokinetics were compared with a separate study in healthy volunteers. Following oral dosing, the dose-normalized area under the curve (AUC) (DN-AUC(24h)) in humans is 896 ((ng*h ml(-1))/(mg kg(-1))), and in animals ranges from 0.3 to 87. The variability among species cannot be explained by intrinsic clearance, which in intravenous dosing experiments shows only moderate interspecies variation (13-41 l h(-1) m(-2)). A portal vein rat pharmacokinetics model suggested that differences in first-pass gut clearance may explain some of the interspecies variation in oral bioavailability. Ibudilast shows auto-induction of metabolism in some animals, but not in humans. Plasma protein binding in humans and some animals is greater than or equal to 95%. The primary metabolite 6,7-dihyrdodiol-ibudilast is measurable and has been quantitated in plasma from animals and humans. Finally, biodistribution studies show that ibudilast distributes rapidly, extensively, and reversibly to the central nervous system.
Collapse
Affiliation(s)
- L M Sanftner
- Research and Development, Avigen Inc., 1301 Harbor Bay Parkway, Alameda, CA 94502, USA
| | | | | | | | | | | |
Collapse
|
17
|
Hamill TG, Sato N, Jitsuoka M, Tokita S, Sanabria S, Eng W, Ryan C, Krause S, Takenaga N, Patel S, Zeng Z, Williams D, Sur C, Hargreaves R, Burns HD. Inverse agonist histamine H3 receptor PET tracers labelled with carbon-11 or fluorine-18. Synapse 2009; 63:1122-32. [DOI: 10.1002/syn.20689] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
18
|
Zheng M, Wang J, Lubinski J, Flint OP, Krishna R, Yao M, Pursley JM, Thakur A, Boulton DW, Santone KS, Barten DM, Anderson JJ, Felsenstein KM, Hansel SB. Studies on the pharmacokinetics and metabolism of a gamma-secretase inhibitor BMS-299897, and exploratory investigation of CYP enzyme induction. Xenobiotica 2009; 39:544-55. [PMID: 19480557 DOI: 10.1080/00498250902928555] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BMS-299897 is a gamma-secretase inhibitor that was effective in reducing amyloid beta-peptide (A beta) in transgenic mice and guinea pigs. Therefore, pharmacokinetic and drug metabolism studies were conducted in animals to support its clinical development. The compound appeared to have low to intermediate total body clearance and was orally bioavailable (24-100%). The oral absorption of BMS-299897 from solid dosage forms appeared to be dissolution rate-limited. BMS-299897 was distributed into extravascular space (V(ss) >or= 1.3 l kg(-1)), including brain (brain-to-plasma ratio = 0.13-0.50). BMS-299897 appeared to be a P-glycoprotein (P-gp) substrate as the brain-to-plasma ratio was two-fold higher in the mdr1a knockout mouse as compared with the wild-type. Apparent autoinduction by BMS-299897 was observed in murine and rat efficacy and toxicity studies. In vitro, BMS-299897 was a weaker inducer of cytochrome P450 3A4 (CYP3A4) and a weaker transactivator of human pregnane X receptor (hPXR) as compared with rifampicin. Induction of human UGT1A and UGT2B was evaluated in primary human hepatocytes, but the results were inconclusive. A low potential for autoinduction in humans was predicted at a clinical dose of 250 mg and the prediction was consistent with the findings from a clinical multiple-dose study with BMS-299897 in probable Alzheimer's patients.
Collapse
Affiliation(s)
- M Zheng
- Metabolism and Pharmacokinetics, Bristol-Myers Squibb Research and Development, Wallingford, CT, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Tang C, Kuo Y, Pudvah NT, Ellis JD, Michener MS, Egbertson M, Graham SL, Cook JJ, Hochman JH, Prueksaritanont T. Effect of P-glycoprotein-mediated efflux on cerebrospinal fluid concentrations in rhesus monkeys. Biochem Pharmacol 2009; 78:642-7. [DOI: 10.1016/j.bcp.2009.05.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Revised: 05/18/2009] [Accepted: 05/19/2009] [Indexed: 10/20/2022]
|
20
|
Satoh A, Sagara T, Sakoh H, Hashimoto M, Nakashima H, Kato T, Goto Y, Mizutani S, Azuma-Kanoh T, Tani T, Okuda S, Okamoto O, Ozaki S, Iwasawa Y, Ohta H, Kawamoto H. Identification of an orally active opioid receptor-like 1 (ORL1) receptor antagonist 4-{3-[(2R)-2,3-dihydroxypropyl]-2-oxo-2,3-dihydro-1H-benzimidazol-1-yl}-1-[(1S,3S,4R)-spiro[bicyclo[2.2.1]heptane-2,1'-cyclopropan]-3-ylmethyl]piperidine as clinical candidate. J Med Chem 2009; 52:4091-4. [PMID: 19537798 DOI: 10.1021/jm900581g] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Our efforts to optimize prototype opioid receptor-like 1 (ORL1) antagonist 1 led to the discovery of 4-{3-[(2R)-2,3-dihydroxypropyl]-2-oxo-2,3-dihydro-1H-benzimidazol-1-yl}-1-[(1S,3S,4R)-spiro[bicyclo[2.2.1]heptane-2,1'-cyclopropan]-3-ylmethyl]piperidine 10. 10 showed potent ORL1 antagonistic activity, excellent selectivity over other opioid receptors, and in vivo efficacy after oral dosing. Currently clinical trials of 10 are underway.
Collapse
Affiliation(s)
- Atsushi Satoh
- Tsukuba Research Institute, Banyu Pharmaceutical Co, Ltd, Tsukuba 300-2611, Ibaraki, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Suzuki T, Kameda M, Ando M, Miyazoe H, Sekino E, Ito S, Masutani K, Kamijo K, Takezawa A, Moriya M, Ito M, Ito J, Nakase K, Matsushita H, Ishihara A, Takenaga N, Tokita S, Kanatani A, Sato N, Fukami T. Discovery of novel diarylketoxime derivatives as selective and orally active melanin-concentrating hormone 1 receptor antagonists. Bioorg Med Chem Lett 2009; 19:5339-45. [PMID: 19683441 DOI: 10.1016/j.bmcl.2009.07.132] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Revised: 07/28/2009] [Accepted: 07/29/2009] [Indexed: 12/11/2022]
Abstract
Optimization of the lead 2a led to the identification of a novel diarylketoxime class of melanin-concentrating hormone 1 receptor (MCH-1R) antagonists. Our focus was directed toward improvement of hERG activity and metabolic stability. The representative derivative 4b showed potent and dose-dependent body weight reduction in diet-induced obese (DIO) C57BL/6J mice after oral administration. The synthesis and structure-activity relationships of the novel diarylketoxime MCH-1R antagonists are described.
Collapse
Affiliation(s)
- Takao Suzuki
- Department of Medicinal Chemistry, Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ando M, Sato N, Nagase T, Nagai K, Ishikawa S, Takahashi H, Ohtake N, Ito J, Hirayama M, Mitobe Y, Iwaasa H, Gomori A, Matsushita H, Tadano K, Fujino N, Tanaka S, Ohe T, Ishihara A, Kanatani A, Fukami T. Discovery of pyridone-containing imidazolines as potent and selective inhibitors of neuropeptide Y Y5 receptor. Bioorg Med Chem 2009; 17:6106-22. [DOI: 10.1016/j.bmc.2009.05.069] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 05/27/2009] [Accepted: 05/28/2009] [Indexed: 11/16/2022]
|
23
|
Sasaki T, Takahashi T, Nagase T, Mizutani T, Ito S, Mitobe Y, Miyamoto Y, Kanesaka M, Yoshimoto R, Tanaka T, Takenaga N, Tokita S, Sato N. Synthesis, structure–activity relationships, and biological profiles of a dihydrobenzoxathiin class of histamine H3 receptor inverse agonists. Bioorg Med Chem Lett 2009; 19:4232-6. [DOI: 10.1016/j.bmcl.2009.05.101] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 05/21/2009] [Accepted: 05/25/2009] [Indexed: 11/25/2022]
|
24
|
He H, Lyons K, Shen X, Yao Z, Bleasby K, Chan G, Hafey M, Li X, Xu S, Salituro GM, Cohen LH, Tang W. Utility of unbound plasma drug levels and P-glycoprotein transport data in prediction of central nervous system exposure. Xenobiotica 2009; 39:687-93. [DOI: 10.1080/00498250903015402] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
25
|
Suzuki T, Moriya M, Sakamoto T, Suga T, Kishino H, Takahashi H, Ishikawa M, Nagai K, Imai Y, Sekino E, Ito M, Iwaasa H, Ishihara A, Tokita S, Kanatani A, Sato N, Fukami T. Discovery of novel spiro-piperidine derivatives as highly potent and selective melanin-concentrating hormone 1 receptor antagonists. Bioorg Med Chem Lett 2009; 19:3072-7. [DOI: 10.1016/j.bmcl.2009.04.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Revised: 04/01/2009] [Accepted: 04/03/2009] [Indexed: 11/25/2022]
|
26
|
Syvänen S, Lindhe O, Palner M, Kornum BR, Rahman O, Långström B, Knudsen GM, Hammarlund-Udenaes M. Species differences in blood-brain barrier transport of three positron emission tomography radioligands with emphasis on P-glycoprotein transport. Drug Metab Dispos 2008; 37:635-43. [PMID: 19047468 DOI: 10.1124/dmd.108.024745] [Citation(s) in RCA: 251] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Species differences occur in the brain concentrations of drugs, but the reasons for these differences are not yet apparent. This study was designed to compare brain uptake of three radiolabeled P-glycoprotein (P-gp) substrates across species using positron emission tomography. Brain concentrations and brain-to-plasma ratios were compared; [(11)C]verapamil in rats, guinea pigs, and monkeys; [(11)C](S)-(2-methoxy-5-(5-trifluoromethyltetrazol-1-yl)-phenylmethylamino)-2(S)-phenylpiperidine (GR205171) in rats, guinea pigs, monkeys, and humans; and [(18)F]altanserin in rats, minipigs, and humans. The fraction of the unbound radioligand in plasma was studied along with its metabolism. The effect of P-gp inhibition was investigated by administering cyclosporin A (CsA). Pronounced species differences were found in the brain and brain-to-plasma concentrations of [(11)C]verapamil, [(11)C]GR205171, and [(18)F]altanserin with higher brain distribution in humans, monkeys, and minipigs than in rats and guinea pigs. For example, the brain-to-plasma ratio of [(11)C]GR205171 was almost 9-fold higher in humans compared with rats. The species differences were still present after P-gp inhibition, although the increase in brain concentrations after P-gp inhibition was somewhat greater in rats than in the other species. Differences in plasma protein binding and metabolism did not explain the species-related differences. The findings are important for interpretation of brain drug delivery when extrapolating preclinical data to humans. Compounds found to be P-gp substrates in rodents are likely to also be substrates in higher species, but sufficient blood-brain barrier permeability may be retained in humans to allow the compound to act at intracerebral targets.
Collapse
Affiliation(s)
- Stina Syvänen
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Nagase T, Mizutani T, Sekino E, Ishikawa S, Ito S, Mitobe Y, Miyamoto Y, Yoshimoto R, Tanaka T, Ishihara A, Takenaga N, Tokita S, Sato N. Synthesis and Evaluation of Structurally Constrained Quinazolinone Derivatives as Potent and Selective Histamine H3 Receptor Inverse Agonists. J Med Chem 2008; 51:6889-901. [DOI: 10.1021/jm800569w] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Tsuyoshi Nagase
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba 300-2611, Japan
| | - Takashi Mizutani
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba 300-2611, Japan
| | - Etsuko Sekino
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba 300-2611, Japan
| | - Shiho Ishikawa
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba 300-2611, Japan
| | - Sayaka Ito
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba 300-2611, Japan
| | - Yuko Mitobe
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba 300-2611, Japan
| | - Yasuhisa Miyamoto
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba 300-2611, Japan
| | - Ryo Yoshimoto
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba 300-2611, Japan
| | - Takeshi Tanaka
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba 300-2611, Japan
| | - Akane Ishihara
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba 300-2611, Japan
| | - Norihiro Takenaga
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba 300-2611, Japan
| | - Shigeru Tokita
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba 300-2611, Japan
| | - Nagaaki Sato
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd., Okubo 3, Tsukuba 300-2611, Japan
| |
Collapse
|
28
|
Synthesis and evaluation of a spiro-isobenzofuranone class of histamine H3 receptor inverse agonists. Bioorg Med Chem Lett 2008; 18:5101-6. [DOI: 10.1016/j.bmcl.2008.07.125] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2008] [Revised: 07/28/2008] [Accepted: 07/30/2008] [Indexed: 11/23/2022]
|
29
|
Sato N, Jitsuoka M, Shibata T, Hirohashi T, Nonoshita K, Moriya M, Haga Y, Sakuraba A, Ando M, Ohe T, Iwaasa H, Gomori A, Ishihara A, Kanatani A, Fukami T. (9S)-9-(2-hydroxy-4,4-dimethyl-6-oxo-1-cyclohexen-1-yl)-3,3-dimethyl-2,3,4,9-tetrahydro-1H-xanthen-1-one, a selective and orally active neuropeptide Y Y5 receptor antagonist. J Med Chem 2008; 51:4765-70. [PMID: 18637668 DOI: 10.1021/jm8003587] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
(9S)-9-(2-Hydroxy-4,4-dimethyl-6-oxo-1-cyclohexen-1-yl)-3,3-dimethyl-2,3,4,9-tetrahydro-1H-xanthen-1-one ((S)-1) was identified as a selective and orally active neuropeptide Y Y5 receptor antagonist. The structure-activity relationship for this structural class was investigated and showed that limited substitution on the phenyl ring was tolerated and that modification of the 4,4-dimethyl group of the cyclohexenone and the 3,3-dimethyl group of the xanthenone parts slightly improved potency. The plasma concentration-time profile after oral administration of (S)-1 in Sprague-Dawley (SD) rats showed significant in vivo racemization of (S)-1 and that (S)-1 is cleared much more quickly than (R)-1. The duration of (S)-1 in SD rats after oral administration of (RS)-1 racemate was twice as long as that following oral administration of (S)-1. The C max values of (S)-1 after administration of (S)-1 and (RS)-1 were comparable, and the brain to plasma ratio for (S)-1 was 0.34 in SD rats. In our acute D-Trp (34)NPY-induced food intake model, both (S)-1 and (RS)-1 showed potent and dose-dependent efficacy. Therefore, the use of (RS)-1 is suitable for studies that require sustained plasma exposure of (S)-1.
Collapse
Affiliation(s)
- Nagaaki Sato
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co, Ltd, Okubo 3, Tsukuba 300-2611, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Nagase T, Mizutani T, Ishikawa S, Sekino E, Sasaki T, Fujimura T, Ito S, Mitobe Y, Miyamoto Y, Yoshimoto R, Tanaka T, Ishihara A, Takenaga N, Tokita S, Fukami T, Sato N. Synthesis, Structure−Activity Relationships, and Biological Profiles of a Quinazolinone Class of Histamine H3 Receptor Inverse Agonists. J Med Chem 2008; 51:4780-9. [DOI: 10.1021/jm8003834] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Tsuyoshi Nagase
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Takashi Mizutani
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Shiho Ishikawa
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Etsuko Sekino
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Takahide Sasaki
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Takashi Fujimura
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Sayaka Ito
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Yuko Mitobe
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Yasuhisa Miyamoto
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Ryo Yoshimoto
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Takeshi Tanaka
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Akane Ishihara
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Norihiro Takenaga
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Shigeru Tokita
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Takehiro Fukami
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| | - Nagaaki Sato
- Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co., Ltd, Okubo 3, Tsukuba, Ibaraki 300-2611, Japan
| |
Collapse
|
31
|
Storch CH, Nikendei C, Schild S, Haefeli WE, Weiss J, Herzog W. Expression and activity of P-glycoprotein (MDR1/ABCB1) in peripheral blood mononuclear cells from patients with anorexia nervosa compared with healthy controls. Int J Eat Disord 2008; 41:432-8. [PMID: 18348284 DOI: 10.1002/eat.20519] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Pharmacotherapeutic strategies for treatment of anorexia nervosa (AN) are characterized by limited success. Some drugs used (antipsychotics, selective serotonin reuptake inhibitors) are transported by P-glycoprotein (P-gp), a transporter with major impact on pharmacokinetics of substrate drugs. Biochemical alterations seen in AN patients could lead to increased expression and/or activity of P-gp and therefore to diminished access of drugs to the brain. The aim of our study was to investigate expression and activity levels of P-gp in peripheral blood mononuclear cells (PBMCs) in AN patients. METHOD PBMCs of 16 AN patients and 16 controls were isolated. Activity of P-gp was determined by flow cytometry and expression was quantified by reverse-transcriptase-real-time-polymerase-chain-reaction. RESULTS Neither a significant difference in P-gp expression (AN: 0.00154 +/- 0.00088 [MDR1/beta2 mg], control: 0.00244 +/- 0.0013 [MDR1/beta2 mg], p = .138) nor a difference in P-gp activity (rhodamine 123 ratio AN: 1.79 +/- 0.73, control: 2.03 +/- 0.42, p = .20) between AN patients and healthy controls could be detected. In contrast to previous studies, expression and activity of P-gp correlated significantly (p = .0031). CONCLUSION Failure in pharmacotherapy with P-gp substrates in AN patients are probably neither caused by different P-gp expression nor activity levels.
Collapse
Affiliation(s)
- Caroline Henrike Storch
- Department of Internal Medicine VI, Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
32
|
Jeffrey P, Summerfield SG. Challenges for blood-brain barrier (BBB) screening. Xenobiotica 2008; 37:1135-51. [PMID: 17968740 DOI: 10.1080/00498250701570285] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Whilst blood-brain barrier permeability is an important determinant in achieving efficacious central nervous system drug concentrations, it should not be viewed or measured in isolation. Recent studies have highlighted the need for an integrated approach where optimal central nervous system penetration is achieved through the correct balance of permeability, a low potential for active efflux, and the appropriate physicochemical properties that allow for drug partitioning and distribution into brain tissue. Integrating data from permeability studies performed incorporating an assessment of active efflux by P-glycoprotein in combination with drug-free fraction measurements in blood and brain has furthered the understanding of the impact of the blood-brain barrier on central nervous system uptake and the underlying physicochemical properties that contribute to central nervous system drug disposition. This approach moves away from screening and ranking compounds in assays designed to measure or predict central nervous system penetration in the somewhat arbitrary units of brain-blood (or plasma) ratios.
Collapse
Affiliation(s)
- P Jeffrey
- Neurology & GI Centre of Excellence for Drug Discovery, GlaxoSmithKline, Harlow, UK
| | | |
Collapse
|
33
|
Venkatakrishnan K, Tseng E, Nelson FR, Rollema H, French JL, Kaplan IV, Horner WE, Gibbs MA. Central nervous system pharmacokinetics of the Mdr1 P-glycoprotein substrate CP-615,003: intersite differences and implications for human receptor occupancy projections from cerebrospinal fluid exposures. Drug Metab Dispos 2007; 35:1341-9. [PMID: 17470526 DOI: 10.1124/dmd.106.013953] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The central nervous system (CNS) distribution and transport mechanisms of the investigational drug candidate CP-615,003 (N-[3-fluoro-4-[2-(propylamino)ethoxy]phenyl]-4,5,6,7-tetrahydro-4-oxo-1H-indole-3-carboxamide) and its active metabolite CP-900,725 have been characterized. Brain distribution of CP-615,003 and CP-900,725 was low in rats and mice (brain-to-serum ratio < 0.2). Cerebrospinal fluid (CSF)-to-serum ratios of CP-615,003 were 6- to 8-fold lower than the plasma unbound fraction in rats and dogs. In vitro, CP-615,003 displayed quinidine-like efflux in MDR1-expressing Madin-Darby canine kidney II cells. The brain-to-serum ratio of CP-615,003 in mdr1a/1b (-/-) mice was approximately 7 times that in their wild-type counterparts, confirming that impaired CNS distribution was explained by P-gp efflux transport. In contrast, P-gp efflux did not explain the impaired CNS penetration of CP-900,725. Intracerebral microdialysis was used to characterize rat brain extracellular fluid (ECF) distribution. Interestingly, the ECF-to-serum ratio of the P-gp substrate CP-615,003 was 7-fold below the CSF-to-serum ratio, whereas this disequilibrium was not observed for CP-900,725. In a clinical study, steady-state CSF exposures were measured after administration of 100 mg of CP-615,003 b.i.d. The human CSF-to-plasma ratios of CP-615,003 and CP-900,725 were both approximately 10-fold below their ex vivo plasma unbound fractions, confirming impaired human CNS penetration. Preliminary estimates of CNS receptor occupancy from human CSF concentrations were sensitive to assumptions regarding the magnitude of the CSF-ECF gradient for CP-615,003 in humans. In summary, this case provides an example of intersite differences in CNS pharmacokinetics of a P-gp substrate and potential implications for projection of human CNS receptor occupancy of transporter substrates from CSF pharmacokinetic data when direct imaging-based approaches are not feasible.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP-Binding Cassette Transporters/genetics
- ATP-Binding Cassette Transporters/metabolism
- Animals
- Area Under Curve
- Biological Transport
- Brain/metabolism
- Brain Chemistry
- Cell Line
- Central Nervous System/metabolism
- Cerebrospinal Fluid/metabolism
- Dogs
- Extracellular Fluid/metabolism
- GABA-A Receptor Agonists
- Humans
- Indoles/blood
- Indoles/metabolism
- Indoles/pharmacokinetics
- Male
- Mice
- Mice, Inbred Strains
- Mice, Knockout
- Microdialysis
- Rats
- Rats, Sprague-Dawley
- Receptors, GABA-A/metabolism
- ATP-Binding Cassette Sub-Family B Member 4
Collapse
Affiliation(s)
- Karthik Venkatakrishnan
- Department of Clinical Pharmacology, Pfizer Global Research and Development, Groton, Connecticut 06340, USA.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Summerfield SG, Stevens AJ, Cutler L, del Carmen Osuna M, Hammond B, Tang SP, Hersey A, Spalding DJ, Jeffrey P. Improving the in vitro prediction of in vivo central nervous system penetration: integrating permeability, P-glycoprotein efflux, and free fractions in blood and brain. J Pharmacol Exp Ther 2005; 316:1282-90. [PMID: 16330496 DOI: 10.1124/jpet.105.092916] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
This work examines the inter-relationship between the unbound drug fractions in blood and brain homogenate, passive membrane permeability, P-glycoprotein (Pgp) efflux ratio, and log octanol/water partition coefficients (cLogP) in determining the extent of central nervous system (CNS) penetration observed in vivo. The present results demonstrate that compounds often considered to be Pgp substrates in rodents (efflux ratio greater than 5 in multidrug resistant Madin-Darby canine kidney cells) with poor passive permeability may still exhibit reasonable CNS penetration in vivo; i.e., where the unbound fractions and nonspecific tissue binding act as a compensating force. In these instances, the efflux ratio and in vitro blood-brain partition ratio may be used to predict the in vivo blood-brain ratio. This relationship may be extended to account for the differences in CNS penetration observed in vivo between mdr1a/b wild type and knockout mice. In some instances, cross-species differences that might initially seem to be related to differing transporter expression can be rationalized from knowledge of unbound fractions alone. The results presented in this article suggest that the information exists to provide a coherent picture of the nature of CNS penetration in the drug discovery setting, allowing the focus to be shifted away from understanding CNS penetration toward the more important aspect of understanding CNS efficacy.
Collapse
Affiliation(s)
- Scott G Summerfield
- Drug Metabolism and Pharmacokinetics Department, Neurology, and Gastrointestinal Centre of Excellence for Drug Discovery, GlaxoSmithKline R&D, New Frontiers Science Park, Third Avenue, Harlow, Essex CM19 5AW, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|