1
|
Mardikasari SA, Sipos B, Csóka I, Katona G. Nasal route for antibiotics delivery: Advances, challenges and future opportunities applying the quality by design concepts. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
2
|
Bors LA, Bajza Á, Mándoki M, Tasi BJ, Cserey G, Imre T, Szabó P, Erdő F. Modulation of nose-to-brain delivery of a P-glycoprotein (MDR1) substrate model drug (quinidine) in rats. Brain Res Bull 2020; 160:65-73. [PMID: 32344126 DOI: 10.1016/j.brainresbull.2020.04.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/07/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023]
Abstract
During the last decades several new drug formulations were developed to target the central nervous system (CNS) from the nasal cavity. However, in these studies less attention was paid to the possible drug-drug interactions in case of multi-drug therapy. In our pilot study first we compared a nasal solution and a nasal gel to demonstrate their distribution in the nasal cavity (3D printed rat skull model and histology). Due to the aspiration induced high mortality at administration of nasal solution the study was continued only with the gel formulation of quinidine. The aim of our experiments was to identify the possible functional role of P-glycoprotein (P-gp) in the drug absorption in nasal cavity and to test drug-drug interactions at nose-to-brain delivery. Therefore, a P-gp substrate model drug, quinidine was tested by intranasal (IN) administration in presence of PSC-833 (specific P-gp inhibitor) given intravenously (IV) or IN and adrenaline (IN) at low (50 ng) or high (20 μg) dose. In control animals the brain penetration of quinidine was at the level of detection limit, but in combination therapy with IV PSC-833 the brain levels increased dramatically, similarly to high dose IN adrenalin, where due to vasoconstriction peripheral distribution was blocked. These results indicate that P-gp has an important role in drug absorption and efflux at nasal cavity, while adrenaline is also able to modify the penetration profile of the P-gp substrate model drug at nasal application as it decreases nose-to-blood absorption, letting more quinidine to reach the brain along with the nasal nerves.
Collapse
Affiliation(s)
- Luca Anna Bors
- Pázmány Péter Catholic University, Faculty of Information Technology and Bionics, Budapest, Hungary.
| | - Ágnes Bajza
- Pázmány Péter Catholic University, Faculty of Information Technology and Bionics, Budapest, Hungary.
| | - Míra Mándoki
- University of Veterinary Medicine, Department of Pathology, Budapest, Hungary.
| | - Benedek József Tasi
- Pázmány Péter Catholic University, Faculty of Information Technology and Bionics, Budapest, Hungary.
| | - György Cserey
- Pázmány Péter Catholic University, Faculty of Information Technology and Bionics, Budapest, Hungary.
| | - Tímea Imre
- Research Centre for Natural Sciences, Instrumentation Centre, Budapest, Hungary.
| | - Pál Szabó
- Research Centre for Natural Sciences, Instrumentation Centre, Budapest, Hungary.
| | - Franciska Erdő
- Pázmány Péter Catholic University, Faculty of Information Technology and Bionics, Budapest, Hungary.
| |
Collapse
|
3
|
Revisiting the blood-brain barrier: A hard nut to crack in the transportation of drug molecules. Brain Res Bull 2020; 160:121-140. [PMID: 32315731 DOI: 10.1016/j.brainresbull.2020.03.018] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/19/2020] [Accepted: 03/26/2020] [Indexed: 12/21/2022]
Abstract
Barriers are the hallmark of a healthy physiology, blood-brain barrier (BBB) being a tough nut to crack for most of the antigens and chemical substances. The presence of tight junctions plays a remarkable role in defending the brain from antigenic and pathogenic attacks. BBB constitutes a diverse assemblage of multiple physical and chemical barriers that judiciously restrict the flux of blood solutes into and out of the brain. Restrictions through the paracellular pathway and the tight junctions between intercellular clefts, together create well regulated metabolic and transport barricades, critical to brain pathophysiology. The brain being impermeable to many essential metabolites and nutrients regulates transportation via specialized transport systems across the endothelial abluminal and luminal membranes. The epithelial cells enveloping capillaries of the choroid plexus regulates the transport of complement, growth factors, hormones, microelements, peptides and trace elements into ventricles. Nerve terminals, microglia, and pericytes associated with the endothelium support barrier induction and function, ensuring an optimally stable ionic microenvironment that facilitates neurotransmission, orchestrated by multiple ion channels (Na+, K+ Mg2+, Ca2+) and transporters. Brain pathology which can develop due to genetic mutations or secondary to other cerebrovascular, neurodegenerative diseases can cause aberration in the microvasculature of CNS which is the uniqueness of BBB. This can also alter BBB permeation and result in BBB breakdown and other neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis. The concluding section outlines contemporary trends in drug discovery, focusing on molecular determinants of BBB permeation and novel drug-delivery systems, such as dendrimers, liposomes, nanoparticles, nanogels, etc.
Collapse
|
4
|
Puscas I, Bernard-Patrzynski F, Jutras M, Lécuyer MA, Bourbonnière L, Prat A, Leclair G, Roullin VG. IVIVC Assessment of Two Mouse Brain Endothelial Cell Models for Drug Screening. Pharmaceutics 2019; 11:pharmaceutics11110587. [PMID: 31717321 PMCID: PMC6920823 DOI: 10.3390/pharmaceutics11110587] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 01/10/2023] Open
Abstract
Since most preclinical drug permeability assays across the blood-brain barrier (BBB) are still evaluated in rodents, we compared an in vitro mouse primary endothelial cell model to the mouse b.End3 and the acellular parallel artificial membrane permeability assay (PAMPA) models for drug screening purposes. The mRNA expression of key feature membrane proteins of primary and bEnd.3 mouse brain endothelial cells were compared. Transwell® monolayer models were further characterized in terms of tightness and integrity. The in vitro in vivo correlation (IVIVC) was obtained by the correlation of the in vitro permeability data with log BB values obtained in mice for seven drugs. The mouse primary model showed higher monolayer integrity and levels of mRNA expression of BBB tight junction (TJ) proteins and membrane transporters (MBRT), especially for the efflux transporter Pgp. The IVIVC and drug ranking underlined the superiority of the primary model (r2 = 0.765) when compared to the PAMPA-BBB (r2 = 0.391) and bEnd.3 cell line (r2 = 0.019) models. The primary monolayer mouse model came out as a simple and reliable candidate for the prediction of drug permeability across the BBB. This model encompasses a rapid set-up, a fair reproduction of BBB tissue characteristics, and an accurate drug screening.
Collapse
Affiliation(s)
- Ina Puscas
- Faculty of Pharmacy, Université de Montréal, CP6128 Succursale Centre-ville, Montreal, QC H3C 3J7, Canada; (I.P.); (F.B.-P.); (M.J.)
| | - Florian Bernard-Patrzynski
- Faculty of Pharmacy, Université de Montréal, CP6128 Succursale Centre-ville, Montreal, QC H3C 3J7, Canada; (I.P.); (F.B.-P.); (M.J.)
| | - Martin Jutras
- Faculty of Pharmacy, Université de Montréal, CP6128 Succursale Centre-ville, Montreal, QC H3C 3J7, Canada; (I.P.); (F.B.-P.); (M.J.)
| | - Marc-André Lécuyer
- Department of Neuroscience, Faculty of Medicine, Université de Montréal and Centre de Recherc and du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada; (M.-A.L.); (L.B.); (A.P.)
- Centre for Biostructural Imaging of Neurodegeneration, Institute for Multiple Sclerosis Research and Neuroimmunology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Lyne Bourbonnière
- Department of Neuroscience, Faculty of Medicine, Université de Montréal and Centre de Recherc and du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada; (M.-A.L.); (L.B.); (A.P.)
| | - Alexandre Prat
- Department of Neuroscience, Faculty of Medicine, Université de Montréal and Centre de Recherc and du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada; (M.-A.L.); (L.B.); (A.P.)
| | - Grégoire Leclair
- Faculty of Pharmacy, Université de Montréal, CP6128 Succursale Centre-ville, Montreal, QC H3C 3J7, Canada; (I.P.); (F.B.-P.); (M.J.)
- Correspondence: (G.L.); (V.G.R.)
| | - V. Gaëlle Roullin
- Faculty of Pharmacy, Université de Montréal, CP6128 Succursale Centre-ville, Montreal, QC H3C 3J7, Canada; (I.P.); (F.B.-P.); (M.J.)
- Correspondence: (G.L.); (V.G.R.)
| |
Collapse
|
5
|
Bors L, Tóth K, Tóth EZ, Bajza Á, Csorba A, Szigeti K, Máthé D, Perlaki G, Orsi G, Tóth GK, Erdő F. Age-dependent changes at the blood-brain barrier. A Comparative structural and functional study in young adult and middle aged rats. Brain Res Bull 2018. [PMID: 29522862 DOI: 10.1016/j.brainresbull.2018.03.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Decreased beta-amyloid clearance in Alzheimer's disease and increased blood-brain barrier permeability in aged subjects have been reported in several articles. However, morphological and functional characterization of blood-brain barrier and its membrane transporter activity have not been described in physiological aging yet. The aim of our study was to explore the structural changes in the brain microvessels and possible functional alterations of P-glycoprotein at the blood-brain barrier with aging. Our approach included MR imaging for anatomical orientation in middle aged rats, electronmicroscopy and immunohistochemistry to analyse the alterations at cellular level, dual or triple-probe microdialysis and SPECT to test P-glycoprotein functionality in young and middle aged rats. Our results indicate that the thickness of basal lamina increases, the number of tight junctions decreases and the size of astrocyte endfeet extends with advanced age. On the basis of microdialysis and SPECT results the P-gp function is reduced in old rats. With our multiparametric approach a complex regulation can be suggested which includes elements leading to increased permeability of blood-brain barrier by enhanced paracellular and transcellular transport, and factors working against it. To verify the role of P-gp pumps in brain aging further studies are warranted.
Collapse
Affiliation(s)
- Luca Bors
- Pázmány Péter Catholic University, Faculty of Information Technology and Bionics, Práter u. 50a, H-1083 Budapest
| | - Kinga Tóth
- Hungarian Academy of Sciences, Institute of Cognitive Neuroscience and Psychology, Magyar tudósok körútja 2. H-1117 Budapest
| | - Estilla Zsófia Tóth
- Hungarian Academy of Sciences, Institute of Cognitive Neuroscience and Psychology, Magyar tudósok körútja 2. H-1117 Budapest
| | - Ágnes Bajza
- Pázmány Péter Catholic University, Faculty of Information Technology and Bionics, Práter u. 50a, H-1083 Budapest
| | - Attila Csorba
- University of Szeged, Faculty of Pharmacy, Deparment of Pharmacognosy, Eötvös u. 6, H-6720 Szeged
| | - Krisztián Szigeti
- Semmelweis University, Faculty of Medicine, Department of Biophysics and Radiation Biology, Tűzoltó u. 37-47, H-1094 Budapest
| | - Domokos Máthé
- Semmelweis University, Faculty of Medicine, Department of Biophysics and Radiation Biology, Tűzoltó u. 37-47, H-1094 Budapest,; CROmed Translational Research Ltd. Budapest
| | - Gábor Perlaki
- MTA-PTE Clinical Neuroscience MR Research Group, Ret u. 2, H-7623 Pecs, Hungary; Department of Neurosurgery, University of Pecs, Medical School, Ret u. 2, H-7623 Pecs, Hungary
| | - Gergely Orsi
- MTA-PTE Clinical Neuroscience MR Research Group, Ret u. 2, H-7623 Pecs, Hungary; Department of Neurosurgery, University of Pecs, Medical School, Ret u. 2, H-7623 Pecs, Hungary
| | - Gábor K Tóth
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
| | - Franciska Erdő
- Pázmány Péter Catholic University, Faculty of Information Technology and Bionics, Práter u. 50a, H-1083 Budapest.
| |
Collapse
|
6
|
Hartz AMS, Pekcec A, Soldner ELB, Zhong Y, Schlichtiger J, Bauer B. P-gp Protein Expression and Transport Activity in Rodent Seizure Models and Human Epilepsy. Mol Pharm 2017; 14:999-1011. [PMID: 28195743 DOI: 10.1021/acs.molpharmaceut.6b00770] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A cure for epilepsy is currently not available, and seizure genesis, seizure recurrence, and resistance to antiseizure drugs remain serious clinical problems. Studies show that the blood-brain barrier is altered in animal models of epilepsy and in epileptic patients. In this regard, seizures increase expression of blood-brain barrier efflux transporters such as P-glycoprotein (P-gp), which is thought to reduce brain uptake of antiseizure drugs, and thus, contribute to antiseizure drug resistance. The goal of the current study was to assess the viability of combining in vivo and ex vivo preparations of isolated brain capillaries from animal models of seizures and epilepsy as well as from patients with epilepsy to study P-gp at the blood-brain barrier. Exposing isolated rat brain capillaries to glutamate ex vivo upregulated P-gp expression to levels that were similar to those in capillaries isolated from rats that had status epilepticus or chronic epilepsy. Moreover, the fold-increase in P-gp protein expression seen in animal models is consistent with the fold-increase in P-gp observed in human brain capillaries isolated from patients with epilepsy compared to age-matched control individuals. Overall, the in vivo/ex vivo approach presented here allows detailed analysis of the mechanisms underlying seizure-induced changes of P-gp expression and transport activity at the blood-brain barrier. This approach can be extended to other blood-brain barrier proteins that might contribute to drug-resistant epilepsy or other CNS disorders as well.
Collapse
Affiliation(s)
- Anika M S Hartz
- Sanders-Brown Center on Aging, University of Kentucky , Lexington, Kentucky 40536, United States.,Department of Pharmacology and Nutritional Sciences, University of Kentucky , Lexington, Kentucky 40536, United States
| | - Anton Pekcec
- Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota , Duluth, Minnesota 55812, United States
| | - Emma L B Soldner
- Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota , Duluth, Minnesota 55812, United States
| | - Yu Zhong
- Sanders-Brown Center on Aging, University of Kentucky , Lexington, Kentucky 40536, United States
| | - Juli Schlichtiger
- Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota , Duluth, Minnesota 55812, United States
| | - Bjoern Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky , Lexington, Kentucky 40536, United States.,Epilepsy Center, University of Kentucky , Lexington, Kentucky 40536, United States
| |
Collapse
|
7
|
Åslund AKO, Berg S, Hak S, Mørch Ý, Torp SH, Sandvig A, Widerøe M, Hansen R, de Lange Davies C. Nanoparticle delivery to the brain--By focused ultrasound and self-assembled nanoparticle-stabilized microbubbles. J Control Release 2015; 220:287-294. [PMID: 26518721 DOI: 10.1016/j.jconrel.2015.10.047] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 10/26/2015] [Indexed: 01/04/2023]
Abstract
The blood-brain barrier (BBB) constitutes a significant obstacle for the delivery of drugs into the central nervous system (CNS). Nanoparticles have been able to partly overcome this obstacle and can thus improve drug delivery across the BBB. Furthermore, focused ultrasound in combination with gas filled microbubbles has opened the BBB in a temporospatial manner in animal models, thus facilitating drug delivery across the BBB. In the current study we combine these two approaches in our quest to develop a novel, generic method for drug delivery across the BBB and into the CNS. Nanoparticles were synthesized using the polymer poly(butyl cyanoacrylate) (PBCA), and such nanoparticles have been reported to cross the BBB to some extent. Together with proteins, these nanoparticles self-assemble into microbubbles. Using these novel microbubbles in combination with focused ultrasound, we successfully and safely opened the BBB transiently in healthy rats. Furthermore, we also demonstrated that the nanoparticles could cross the BBB and deliver a model drug into the CNS.
Collapse
Affiliation(s)
- Andreas K O Åslund
- Department of Physics, Norwegian University of Science and Technology, 7491 Trondheim, Norway.
| | - Sigrid Berg
- SINTEF Technology and Society, P.O. box 4760 Sluppen, 7465 Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Sjoerd Hak
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Ýrr Mørch
- SINTEF Materials and Chemistry, P.O. box 4760 Sluppen, 7465 Trondheim, Norway
| | - Sverre H Torp
- Department of Pathology and Medical Genetics, St.Olavs University Hospital, Trondheim, Norway; Department of Neuroscience, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Axel Sandvig
- Department of Neuroscience, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Division of Pharmacology and Clinical Neuroscience, Department of Neurosurgery, Umeå University Hospital, Umeå, Sweden
| | - Marius Widerøe
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Rune Hansen
- SINTEF Technology and Society, P.O. box 4760 Sluppen, 7465 Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | | |
Collapse
|
8
|
Toledo-Sherman LM, Prime ME, Mrzljak L, Beconi MG, Beresford A, Brookfield FA, Brown CJ, Cardaun I, Courtney SM, Dijkman U, Hamelin-Flegg E, Johnson PD, Kempf V, Lyons K, Matthews K, Mitchell WL, O'Connell C, Pena P, Powell K, Rassoulpour A, Reed L, Reindl W, Selvaratnam S, Friley WW, Weddell DA, Went NE, Wheelan P, Winkler C, Winkler D, Wityak J, Yarnold CJ, Yates D, Munoz-Sanjuan I, Dominguez C. Development of a series of aryl pyrimidine kynurenine monooxygenase inhibitors as potential therapeutic agents for the treatment of Huntington's disease. J Med Chem 2015; 58:1159-83. [PMID: 25590515 DOI: 10.1021/jm501350y] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We report on the development of a series of pyrimidine carboxylic acids that are potent and selective inhibitors of kynurenine monooxygenase and competitive for kynurenine. We describe the SAR for this novel series and report on their inhibition of KMO activity in biochemical and cellular assays and their selectivity against other kynurenine pathway enzymes. We describe the optimization process that led to the identification of a program lead compound with a suitable ADME/PK profile for therapeutic development. We demonstrate that systemic inhibition of KMO in vivo with this lead compound provides pharmacodynamic evidence for modulation of kynurenine pathway metabolites both in the periphery and in the central nervous system.
Collapse
|
9
|
Cyclosporin A affects the bioavailability of ginkgolic acids via inhibition of P-gp and BCRP. Eur J Pharm Biopharm 2014; 88:759-67. [DOI: 10.1016/j.ejpb.2014.06.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 06/13/2014] [Accepted: 06/17/2014] [Indexed: 01/16/2023]
|
10
|
Chan GNY, Saldivia V, Yang Y, Pang H, de Lannoy I, Bendayan R. In vivo induction of P-glycoprotein expression at the mouse blood-brain barrier: an intracerebral microdialysis study. J Neurochem 2013; 127:342-52. [PMID: 23777437 DOI: 10.1111/jnc.12344] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 06/10/2013] [Accepted: 06/13/2013] [Indexed: 11/27/2022]
Abstract
Intracerebral microdialysis was utilized to investigate the effect of P-glycoprotein (a drug efflux transporter) induction at the mouse blood-brain barrier (BBB) on brain extracellular fluid concentrations of quinidine, an established substrate of P-glycoprotein. Induction was achieved by treating male CD-1 mice for 3 days with 5 mg/kg/day dexamethasone (DEX), a ligand of the nuclear receptor, pregnane X receptor, and a P-glycoprotein inducer. Tandem liquid chromatography mass spectrometric method was used to quantify analytes in dialysate, blood and plasma. P-glycoprotein, pregnane X receptor and Cyp3a11 (metabolizing enzyme for quinidine) protein expression in capillaries and brain homogenates was measured by immunoblot analysis. Following quinidine i.v. administration, the average ratio of unbound quinidine concentrations in brain extracellular fluid (determined from dialysate samples) to plasma at steady state (375-495 min) or Kp, uu, ECF /Plasma in the DEX-treated animals was 2.5-fold lower compared with vehicle-treated animals. In DEX-treated animals, P-glycoprotein expression in brain capillaries was 1.5-fold higher compared with vehicle-treated animals while Cyp3a11 expression in brain capillaries was not significantly different between the two groups. These data demonstrate that P-gp induction mediated by DEX at the BBB can significantly reduce quinidine brain extracellular fluid concentrations by decreasing its brain permeability and further suggest that drug-drug interactions as a result of P-gp induction at the BBB are possible. Applying microdialysis, distribution of quinidine, a P-gp substrate, in mouse brain extracellular fluid (ECF) was investigated following ligand-mediated P-glycoprotein (P-gp) induction at the blood-brain barrier (BBB). We demonstrated that a PXR agonist (dexamethasone) significantly up-regulated P-gp in brain capillaries and reduced quinidine brain ECF concentrations. Our data suggest that drug-drug interactions as a result of P-gp induction at the BBB are possible.
Collapse
Affiliation(s)
- Gary N Y Chan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
11
|
Ball K, Bouzom F, Scherrmann JM, Walther B, Declèves X. Physiologically based pharmacokinetic modelling of drug penetration across the blood-brain barrier--towards a mechanistic IVIVE-based approach. AAPS JOURNAL 2013; 15:913-32. [PMID: 23784110 DOI: 10.1208/s12248-013-9496-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 05/09/2013] [Indexed: 01/09/2023]
Abstract
Predicting the penetration of drugs across the human blood-brain barrier (BBB) is a significant challenge during their development. A variety of in vitro systems representing the BBB have been described, but the optimal use of these data in terms of extrapolation to human unbound brain concentration profiles remains to be fully exploited. Physiologically based pharmacokinetic (PBPK) modelling of drug disposition in the central nervous system (CNS) currently consists of fitting preclinical in vivo data to compartmental models in order to estimate the permeability and efflux of drugs across the BBB. The increasingly popular approach of using in vitro-in vivo extrapolation (IVIVE) to generate PBPK model input parameters could provide a more mechanistic basis for the interspecies translation of preclinical models of the CNS. However, a major hurdle exists in verifying these predictions with observed data, since human brain concentrations can't be directly measured. Therefore a combination of IVIVE-based and empirical modelling approaches based on preclinical data are currently required. In this review, we summarise the existing PBPK models of the CNS in the literature, and we evaluate the current opportunities and limitations of potential IVIVE strategies for PBPK modelling of BBB penetration.
Collapse
Affiliation(s)
- Kathryn Ball
- Centre de Pharmacocinétique et Métabolisme, Groupe de Recherche Servier, Orléans, France
| | | | | | | | | |
Collapse
|
12
|
Lanevskij K, Japertas P, Didziapetris R. Improving the prediction of drug disposition in the brain. Expert Opin Drug Metab Toxicol 2013; 9:473-86. [PMID: 23294027 DOI: 10.1517/17425255.2013.754423] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Ability to cross the blood-brain barrier is one of the key ADME characteristics of all drug candidates regardless of their target location in the body. While good brain penetration is essential for CNS drugs, it may lead to serious side effects in case of peripherally-targeted molecules. Despite a high demand of computational methods for estimating brain transport early in drug discovery, achieving good prediction accuracy still remains a challenging task. AREAS COVERED This article reviews various measures employed to quantify brain delivery and recent advances in QSAR approaches for predicting these properties from the compound's structure. Additionally, the authors discuss the classification models attempting to distinguish between permeable and impermeable chemicals. EXPERT OPINION Recent research in the field of brain penetration modeling showed an increasing understanding of the processes involved in drug disposition, although most models of brain/plasma partitioning still rely on purely statistical considerations. Preferably, new models should incorporate mechanistic knowledge since it is the prerequisite for guiding drug design efforts in the desired direction. To increase the efficiency of computational tools, a broader view is necessary, involving rate and extent of brain penetration, as well as plasma and brain tissue binding strength, instead of relying on any single property.
Collapse
Affiliation(s)
- Kiril Lanevskij
- VšĮ Aukštieji algoritmai, A. Mickeviciaus 29, LT-08117 Vilnius, Lithuania.
| | | | | |
Collapse
|
13
|
Schnepf R, Zolk O. Effect of the ATP-binding cassette transporter ABCG2 on pharmacokinetics: experimental findings and clinical implications. Expert Opin Drug Metab Toxicol 2013; 9:287-306. [PMID: 23289909 DOI: 10.1517/17425255.2013.742063] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION The ATP-binding cassette transporter ABCG2 can actively extrude a broad range of endogenous and exogenous substrates across biological membranes. Thereby, ABCG2 limits oral drug bioavailability, mediates hepatobiliary and renal excretion and participates functionally in the blood-brain barrier. AREAS COVERED The paper provides a review of the clinical evidence of the role of ABCG2 in the bioavailability and brain disposition of drugs. It also sheds light on the value of experimental/preclinical data in predicting the role of ABCG2 in pharmacokinetics in humans. EXPERT OPINION Experimental studies indicate that ABCG2 may limit the oral bioavailability and brain penetration of many drugs. ABCG2 has also been recognized as an important determinant of the disposition of some drugs in humans. For example, loss-of-function variants of ABCG2 affect the pharmacokinetics and pharmacodynamics of rosuvastatin in a clinically significant manner. Moreover, clinically relevant pharmacokinetic drug-drug interactions have been attributed to ABCG2 inhibition. However, examples from human studies are still rare compared with the overwhelming evidence from experimental studies. The large degree of functional redundancy of ABCG2 with other transporters such as P-glycoprotein may explain the rare occurrence of ABCG2-dependent drug-drug interactions in humans. Providing clinicians with consolidated information on the clinically relevant interactions of drugs with ABCG2 remains a matter of future exploration.
Collapse
Affiliation(s)
- Rebecca Schnepf
- Friedrich-Alexander Universität Erlangen-Nürnberg, Institute of Experimental and Clinical Pharmacology and Toxicology, Fahrstr. 17, 91054 Erlangen, Germany.
| | | |
Collapse
|