Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Drug Metab Pharmacokinet ; 42: 100430, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34896751

ABSTRACT

Breast cancer resistance protein (BCRP) expressed in the blood-brain barrier plays a major role in limiting drug distribution into the central nervous system (CNS). However, functional involvement of BCRP in drug distribution into the brain and cerebrospinal fluid (CSF) remains unclear. The aim of present study was to reveal the role and quantitative impact of BCRP on CNS distribution. The brain-to-plasma unbound concentration ratio (Kp,uu,brain) and CSF-to-plasma unbound concentration ratio (Kp,uu,CSF) values of BCRP-specific substrates were determined in rats. The Kp,uu,brain values decreased, as the in vitro BCRP corrected flux ratio (CFR) increased. The Kp,uu,CSF values of BCRP-specific substrates were greater than the Kp,uu,brain values. Increase in the Kp,uu,brain values induced by co-administration of BCRP inhibitor correlated with the in vitro BCRP CFR and were greater than the increase in Kp,uu,CSF values induced by BCRP inhibitor except nebicapone. The contribution of BCRP to the brain and CSF distribution of the dual P-glycoprotein/BCRP substrates, imatinib and prazosin, was similar to that of BCRP-specific substrates. Thus, we revealed that the impact of in vivo BCRP on CNS distribution is correlated with in vitro BCRP CFR, and that BCRP limits drug distribution into the brain more strongly than into the CSF.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2 , Blood-Brain Barrier , Pharmaceutical Preparations , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Pharmaceutical Preparations/cerebrospinal fluid , Pharmacokinetics , Rats
2.
Drug Metab Dispos ; 49(2): 142-151, 2021 02.
Article in English | MEDLINE | ID: mdl-33262223

ABSTRACT

In drug discovery, the extent of brain penetration as measured by free brain/plasma concentration ratio (Kp,uu) is normally determined from one experiment after constant intravenous infusion, and pharmacokinetics (PK) parameters, including clearance (CL), volume of distribution at steady state (Vss), and effective half-life (t 1/2 ,eff) are determined from another experiment after a single intravenous bolus injection. The objective of the present study was to develop and verify a method to simultaneously determine Kp,uu and PK parameters from a single intravenous infusion experiment. In this study, nine compounds (atenolol, loperamide, minoxidil, N-[3-(4'-fluorophenyl)-3-(4'-phenylphenoxy)propyl]sarcosine, sulpiride, and four proprietary compounds) were intravenously infused for 4 hours at 1 mg/kg or 24 hours at 1 or 6 mg/kg or bolus injected at 1 mg/kg. Plasma samples were serially collected, and brain and cerebrospinal fluid samples were collected at the end of infusion. The PK parameters were obtained using noncompartmental analysis (NCA) and compartmental analysis. The Kp,uu,brain values of those compounds increased up to 2.86-fold from 4 to 24 hours. The CL calculated from infusion rate over steady-state concentration from the 24-hour infusion studies was more consistent with the CL from the intravenous bolus studies than that from 4-hour infusion studies (CL avg. fold of difference 1.19-1.44 vs. 2.10). The compartmental analysis using one- and two-compartment models demonstrated better performance than NCA regardless of study design. In addition, volume of distribution at steady state and t 1/2,eff could be accurately obtained by one-compartment analysis within 2-fold difference. In conclusion, both unbound brain-to-plasma ratio and PK parameters can be successfully estimated from a 24-hour intravenous infusion study design. SIGNIFICANCE STATEMENT: We demonstrated that the extent of brain penetration and pharmacokinetic parameters (such as clearance, Vss, and effective t 1/2) can be determined from a single constant intravenous infusion study in rats.


Subject(s)
Brain/metabolism , Pharmaceutical Preparations/administration & dosage , Pharmacokinetics , Animals , Blood-Brain Barrier , Infusions, Intravenous , Male , Pharmaceutical Preparations/cerebrospinal fluid , Pharmaceutical Preparations/metabolism , Rats , Rats, Sprague-Dawley
3.
Pharmacol Res Perspect ; 8(2): e00575, 2020 04.
Article in English | MEDLINE | ID: mdl-32266794

ABSTRACT

In clinical pharmacology, the free drug hypothesis has been widely applied in the interpretation of the relationship between pharmacokinetics and pharmacodynamics (PK/PD). The free drug hypothesis assumes that the unbound drug concentration in blood is the same as that in the site of action at steady state. The objective of this study is to demonstrate whether the free drug hypothesis is universally applicable for all drugs. The unbound concentrations of the 18 compounds in blood and in brain interstitial fluids (ISF) at steady state following constant intravenous infusion were simultaneously monitored up to 6 hours via in vivo microdialysis technique. Based on the permeability and efflux ratio (ER), the test compounds can be divided into two classes. Class I includes the compounds with good membrane permeability that are not substrates of efflux transporters (eg, P-gp, BCRP, and MRPs), whereas Class II includes the compounds that are substrates of efflux transporters. The steady-state unbound drug concentrations in blood, brain, and CSF are quantitatively very similar for Class I compounds, whereas the steady-state unbound concentrations in the brain and CSF are significantly lower than those in blood for Class II compounds. These results strongly suggest that the free drug hypothesis is not universal for all drugs but is only applicable for drugs with good permeability that are not substrates of efflux transporters.


Subject(s)
Brain/metabolism , Cell Membrane Permeability , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/cerebrospinal fluid , Animals , Caco-2 Cells , Dogs , Humans , Madin Darby Canine Kidney Cells , Male , Membrane Transport Proteins/metabolism , Microdialysis , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Rats, Sprague-Dawley
4.
Anal Chem ; 92(4): 3189-3197, 2020 02 18.
Article in English | MEDLINE | ID: mdl-31986881

ABSTRACT

Current methods for liquid-liquid extractions generally require microliter to milliliter volumes of solvents and sample, long equilibration times, and manual procedures. Extraction methods for samples in microfluidic systems have been limited as this tool is difficult to implement on the nanoliter or smaller scale. We have developed slug-flow nanoextraction (SFNE), a method based on droplet microfluidics that allows multiple liquid-liquid extractions to be performed simultaneously in a capillary tube, using only 5 nL of sample and extraction solvent per extraction. Each two-phase slug is segmented from the others by immiscible carrier fluid. We found rapid extractions (<5 s), partition coefficients matching those achieved at larger scale extractions, and potential to preconcentrate samples through volume manipulation. This method was used to accurately and rapidly determine octanol-water partition coefficients (Kow), determining identical Kow as the shake-flask method for acetaminophen, Kow = 2.48 ± 0.02. The measurement, along with calibration and 12 replicates, was complete within 5 min, consuming under 150 nL of solvent and sample. The method was also applied to extract analytes from complex biological samples prior to electrospray ionization-tandem mass spectrometry (ESI-MS/MS) at a rate of 6 s per sample, allowing for simultaneous determination of five different drugs spiked into human plasma, synthetic urine (SU), and artificial cerebral spinal fluid (aCSF) using ethyl acetate as the extraction phase. The signal-to-noise (S/N) for analytes improved up to 19-fold compared to direct ESI-MS of single-phase droplets (aqueous plugs segmented by carrier fluid), with limits of detection down to 7 nM (35 amol).


Subject(s)
Limit of Detection , Liquid-Liquid Extraction/methods , Calibration , Humans , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/cerebrospinal fluid , Pharmaceutical Preparations/isolation & purification , Pharmaceutical Preparations/urine
5.
AAPS J ; 21(4): 67, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31140038

ABSTRACT

Available data on human brain P-glycoprotein ontogeny during infancy and childhood are limited. This review discusses the current body of data relating to maturation of human brain P-glycoprotein including transporter expression levels in post-mortem human brain samples, in vivo transporter activity using probe substrates, surrogate marker endpoints, and extrapolations from animal models. Overall, the data tend to confirm that human brain P-glycoprotein activity keeps developing after birth, although with a developmental time frame that remains unclear. This knowledge gap is a concern given the critical role of brain P-glycoprotein in drug safety and efficacy, and the vulnerable nature of the pediatric population. Future research could include the measurement of brain P-glycoprotein activity across age groups using positron emission tomography or central pharmacodynamic responses. For now, caution is advised when extrapolating adult data to children aged younger than 2 years for drugs with P-glycoprotein-dependent central nervous system activity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Aging/metabolism , Brain/metabolism , Drug-Related Side Effects and Adverse Reactions/metabolism , Pharmaceutical Preparations/metabolism , Animals , Brain/growth & development , Child , Humans , Organ Specificity , Pharmaceutical Preparations/cerebrospinal fluid
6.
Nat Commun ; 8(1): 220, 2017 08 09.
Article in English | MEDLINE | ID: mdl-28790311

ABSTRACT

In-vitro metabolite and drug detection rely on designed materials-based analytical platforms, which are universally used in biomedical research and clinical practice. However, metabolic analysis in bio-samples needs tedious sample preparation, due to the sample complexity and low molecular abundance. A further challenge is to construct diagnostic tools. Herein, we developed a platform using silver nanoshells. We synthesized SiO2@Ag with tunable shell structures by multi-cycled silver mirror reactions. Optimized nanoshells achieved direct laser desorption/ionization mass spectrometry in 0.5 µL of bio-fluids. We applied these nanoshells for disease diagnosis and therapeutic evaluation. We identified patients with postoperative brain infection through daily monitoring and glucose quantitation in cerebrospinal fluid. We measured drug distribution in blood and cerebrospinal fluid systems and validated the function of blood-brain/cerebrospinal fluid-barriers for pharmacokinetics. Our work sheds light on the design of materials for advanced metabolic analysis and precision diagnostics.Preparation of samples for diagnosis can affect the detection of biomarkers and metabolites. Here, the authors use a silver nanoparticle plasmonics approach for the detection of biomarkers in patients as well as investigate the distribution of drugs in serum and cerebral spinal fluid.


Subject(s)
Clinical Laboratory Techniques , Nanoshells/chemistry , Silver , Brain Diseases/cerebrospinal fluid , Brain Diseases/diagnosis , Glucose/cerebrospinal fluid , Humans , Infections/cerebrospinal fluid , Infections/diagnosis , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/cerebrospinal fluid , Pharmacokinetics , Postoperative Complications/cerebrospinal fluid , Postoperative Complications/diagnosis
7.
Rapid Commun Mass Spectrom ; 30(7): 854-8, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26969926

ABSTRACT

RATIONALE: In recent years, several liquid chromatography/tandem mass spectrometry (LC/MS/MS) methods have been reported for the quantitative determination of drugs and metabolites in cerebral spinal fluid (CSF). Artificial CSF (aCSF) is often used as a surrogate for preparing calibration curves and quality control samples in these methods. However, aCSF does not accurately represent the composition of real CSF because it is missing all of the proteins and lipids, which may alter the electrospray ionization (ESI) response when performing LC/MS/MS analyses. In the current study we compared the mass spectral response of several compounds with a range of physiochemical properties in aCSF (essentially a mixture of salts and buffers), diluted plasma (ranging from 1:5 to 1:200) and real CSF to find the best surrogate for CSF in LC/MS/MS methods. METHODS: A number of analytes from polar to non-polar, high protein binding to low protein binding, employing different sample preparation methods, were prepared in diluted plasma, actual CSF or aCSF and tested using LC/MS/MS. The analytes included cotinine and its metabolites, quetiapine, norquetiapine, chlorpromazine, efavirenz and lamivudine. The similarity of MS responses from these compounds in aCSF and diluted plasma to CSF was assessed by comparing the slopes of the calibration curves generated from using linear regression modeling. RESULTS: For all compounds, the lowest percent difference in response ratio (0 to 17%) was observed from 1:200 diluted plasma. Our results indicated that, irrespective of the inherent physiochemical properties of the analytes or the method of sample preparation, 1:200 diluted plasma performed as the best surrogate for CSF in LC/MS/MS methods. CONCLUSIONS: The percent difference in response ratio has been established to demonstrate how different compounds behave between CSF, aCSF and dilute plasma. Although among the compounds tested some of them showed a very similar MS response in actual and aCSF, there were analytes that demonstrated significant differences in ESI-MS signal when sprayed from these two matrices. However, even in such cases, 1:200 diluted plasma generated results with no significant difference from CSF. Therefore, we recommend that in order to develop robust and dependable bioanalytical LC/MS methods from CSF samples, it is more appropriate to prepare calibration curves and quality control samples in diluted plasma instead of aCSF. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Biomarkers/cerebrospinal fluid , Chromatography, Liquid/methods , Pharmaceutical Preparations/cerebrospinal fluid , Tandem Mass Spectrometry/methods , Calibration , Humans
8.
Drug Metab Dispos ; 43(7): 1123-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25943358

ABSTRACT

This study was designed to increase the throughput of rat brain microdialysis studies by administration of compounds as a cassette as opposed to discrete study. Eight compounds (carbamazepine, citalopram, desmethylclozapine, diphenhydramine, gabapentin, metoclopramide, naltrexone, and stavudine) were selected and administered as an intravenous bolus dose at 0.5-3.3 mg/kg each followed by an intravenous infusion at 1 mg/kg per hour for 6 hours in rats in a cassette or discrete dosing. The dialysate, plasma, brain, and cerebrospinal fluid were collected and analyzed using liquid chromatography-tandem mass spectrometry. The microdialysis probe recovery was determined by an in vitro gain method. The recovery between the cassette and discrete dosing was similar, with an average of 1.0 ± 0.10-fold difference. The stavudine interstitial fluid (ISF) concentration, as measured by brain microdialysis, was below the low limit of quantitation and was excluded from the analyses. The ratios of ISF concentration to unbound plasma concentration were within 2-fold for six of the remaining seven compounds, with an average of 0.92 ± 0.51-fold difference between the cassette and discrete methods. The ratios of ISF concentration to unbound brain concentration, as measured by the brain homogenate method, were also similar, with a 1.1 ± 0.7-fold difference. In addition, the ratios of ISF to cerebrospinal fluid concentrations were similar, with a 1.5 ± 0.6-fold difference. The results from this study support the use of a cassette dosing approach to enhance the throughput of rat brain microdialysis studies in drug discovery.


Subject(s)
Brain Chemistry/physiology , High-Throughput Screening Assays/methods , Microdialysis/methods , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Animals , Blood Proteins/metabolism , Blood-Brain Barrier , Infusions, Intravenous , Male , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/cerebrospinal fluid , Protein Binding , Rats , Rats, Sprague-Dawley
9.
Drug Metab Pharmacokinet ; 29(5): 419-26, 2014.
Article in English | MEDLINE | ID: mdl-24806821

ABSTRACT

In central nervous system drug discovery, cerebrospinal fluid (CSF) drug concentration (C(CSF)) has been widely used as a surrogate for unbound brain concentrations (C(u,brain)). However, previous rodent studies demonstrated that when drugs undergo active efflux by transporters, such as P-glycoprotein (P-gp), at the blood-brain barrier, the C(CSF) overestimates the corresponding C(u,brain). To investigate the utility of C(CSF) as a surrogate for interstitial fluid (ISF) concentration (C(ISF)) in nonhuman primates, this study simultaneously determined the C(CSF) and C(ISF) of 12 compounds, including P-gp substrates, under steady-state conditions in cynomolgus monkeys using intracerebral microdialysis coupled with cisternal CSF sampling. Unbound plasma concentrations of non- or weak P-gp substrates were within 2.2-fold of the C(ISF) or C(CSF), whereas typical P-gp substrates (risperidone, verapamil, desloratadine, and quinidine) showed ISF-to-plasma unbound (K(p,uu,ISF)) and CSF-to-plasma unbound concentration ratios (K(p,uu,CSF)) that were appreciably lower than unity. Although the K(p,uu,CSF) of quinidine, verapamil, and desloratadine showed a trend of overestimating the K(p,uu,ISF), K(p,uu,CSF) showed a good agreement with K(p,uu,ISF) within 3-fold variations for all compounds examined. C(u,brain) of some basic compounds, as determined using brain homogenates, overestimated the C(ISF) and C(CSF). Therefore, C(CSF) could be used as a surrogate for C(ISF) in nonhuman primates.


Subject(s)
Brain/metabolism , Macaca fascicularis/cerebrospinal fluid , Macaca fascicularis/metabolism , Pharmaceutical Preparations/analysis , Pharmaceutical Preparations/cerebrospinal fluid , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Blood-Brain Barrier/metabolism , Male , Microdialysis
10.
Drug Metab Dispos ; 42(6): 983-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24644297

ABSTRACT

A pharmacokinetic model was constructed to explain the difference in brain- and cerebrospinal fluid (CSF)-to-plasma and brain-to-CSF unbound drug concentration ratios (Kp,uu,brain, Kp,uu,CSF, and Kp,uu,CSF/brain, respectively) of drugs under steady-state conditions in rats. The passive permeability across the blood-brain barrier (BBB), PS1, was predicted by two methods using log(D/molecular weight(0.5)) for PS1(1) or the partition coefficient in octanol/water at pH 7.4 (LogD), topologic van der Waals polar surface area, and van der Waals surface area of the basic atoms for PS1(2). The coefficients of each parameter were determined using previously reported in situ rat BBB permeability. Active transport of drugs by P-glycoprotein (P-gp) and breast cancer resistance protein (Bcrp) measured in P-gp- and Bcrp-overexpressing cells was extrapolated to in vivo by introducing scaling factors. Brain- and CSF-to-plasma unbound concentration ratios (Kp,uu,brain and Kp,uu,CSF, respectively) of 19 compounds, including P-gp and Bcrp substrates (daidzein, dantrolene, flavopiridol, genistein, loperamide, quinidine, and verapamil), were simultaneously fitted to the equations in a three-compartment model comprising blood, brain, and CSF compartments. The calculated Kp,uu,brain and Kp,uu,CSF of 17 compounds were within a factor of three of experimental values. Kp,uu,CSF values of genistein and loperamide were outliers of the prediction, and Kp,uu,brain of dantrolene also became an outlier when PS1(2) was used. Kp,uu,CSF/brain of the 19 compounds was within a factor of three of experimental values. In conclusion, the Kp,uu,CSF/brain of drugs, including P-gp and Bcrp substrates, could be successfully explained by a kinetic model using scaling factors combined with in vitro evaluation of P-gp and Bcrp activities.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Models, Biological , Pharmaceutical Preparations/cerebrospinal fluid , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Biological Transport/physiology , Pharmaceutical Preparations/metabolism , Protein Binding/physiology , Rats
11.
Drug Metab Dispos ; 42(4): 482-91, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24398459

ABSTRACT

The study objectives were 1) to test the hypothesis that the lack of P-glycoprotein (P-gp) and the inhibition of breast cancer resistance protein (Bcrp) at the blood-brain barrier after cassette dosing of potent P-gp and Bcrp inhibitors were due to low plasma concentrations of those inhibitors and 2) to examine the effects of P-gp on the unbound brain (C(u,brain)) and cerebrospinal fluid (CSF) concentrations (C(u,CSF)) of P-gp substrates in rats. In vitro inhibition of 11 compounds (amprenavir, citalopram, digoxin, elacridar, imatinib, Ko143 [(3S,6S,12aS)-1,2,3,4,6,7,12,12a-octahydro-9-methoxy-6-(2-methylpropyl)-1,4-dioxopyrazino[1',2':1,6]pyrido[3,4-b]indole-3-propanoic acid 1,1-dimethylethyl ester], loperamide, prazosin, quinidine, sulfasalazine, and verapamil) on P-gp and Bcrp was examined in P-gp- and Bcrp-expressing Madin-Darby canine kidney cells, respectively. An in vivo study was conducted in wild-type and Mdr1a(-/-) rats after subcutaneous cassette dosing of the 11 compounds at 1-3 mg/kg, and the brain, CSF, and plasma concentrations of these compounds were determined. At the maximal unbound concentrations observed in rats at 1-3 mg/kg, P-gp and Bcrp were not inhibited by a cassette of the 11 compounds. For non-P-gp/Bcrp substrates, similar C(u,brain), C(u,CSF), and unbound plasma concentrations (C(u,plasma)) were observed in wild-type and P-gp knockout rats. For P-gp/Bcrp substrates, C(u,brain) ≤ C(u,CSF) ≤ C(u,plasma) in wild-type rats, but C(u,brain) and C(u,CSF) increased in the P-gp knockout rats and were within 3-fold of C(u,plasma) for six of the seven P-gp substrates. These results indicate that P-gp and Bcrp inhibition at the blood-brain barrier is unlikely in cassette dosing and also suggest that P-gp and Bcrp activity at the blood-CSF barrier is functionally not important in determination of the CSF concentration for their substrates.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Brain/metabolism , Pharmaceutical Preparations , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Animals, Genetically Modified , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/drug effects , Cell Culture Techniques , Dogs , Gene Knockout Techniques , Madin Darby Canine Kidney Cells , Male , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/cerebrospinal fluid , Rats , Rats, Sprague-Dawley , Substrate Specificity
12.
Bioanalysis ; 5(15): 1831-41, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23905857

ABSTRACT

This White Paper is focused on the technical aspects regarding quantifying pharmaceutically derived inorganic elements in biomatrices in support of GLP nonclinical and clinical studies using inductively coupled plasma (ICP) techniques. For decades ICP has been used in support of Environmental Protection Agency analyses and has more recently been applied for use in the pharmaceutical industry. Current bioanalytical method validation and sample analysis regulatory guidance applies to chromatographic platforms used for analysis of large- and small-molecule PK and TK assessments; however, it is not directly applicable to all aspects of various ICP techniques. Increasingly, quadrupole and high-resolution ICP-MS methods of analysis are being used to quantify inorganic elements contained in pharmaceutical compounds and biomatrices. Many elements occur endogenously in biomatrices, affecting quantification of blanks, standard curve samples, QC samples, and the selection of appropriate levels for the LLOQ.


Subject(s)
Drug Discovery/instrumentation , Drug Discovery/methods , Pharmaceutical Preparations/analysis , Spectrophotometry, Atomic/instrumentation , Spectrophotometry, Atomic/methods , Biomarkers/analysis , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Biomarkers/urine , Calibration , Drug Discovery/standards , Elements , Guidelines as Topic , Limit of Detection , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/cerebrospinal fluid , Pharmaceutical Preparations/urine , Quality Control , Reference Standards , Specimen Handling , Spectrophotometry, Atomic/standards , Validation Studies as Topic
13.
J Cereb Blood Flow Metab ; 32(11): 1959-72, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22929442

ABSTRACT

The blood-brain barrier (BBB) prevents the brain uptake of most pharmaceuticals. This property arises from the epithelial-like tight junctions within the brain capillary endothelium. The BBB is anatomically and functionally distinct from the blood-cerebrospinal fluid barrier at the choroid plexus. Certain small molecule drugs may cross the BBB via lipid-mediated free diffusion, providing the drug has a molecular weight <400 Da and forms <8 hydrogen bonds. These chemical properties are lacking in the majority of small molecule drugs, and all large molecule drugs. Nevertheless, drugs can be reengineered for BBB transport, based on the knowledge of the endogenous transport systems within the BBB. Small molecule drugs can be synthesized that access carrier-mediated transport (CMT) systems within the BBB. Large molecule drugs can be reengineered with molecular Trojan horse delivery systems to access receptor-mediated transport (RMT) systems within the BBB. Peptide and antisense radiopharmaceuticals are made brain-penetrating with the combined use of RMT-based delivery systems and avidin-biotin technology. Knowledge on the endogenous CMT and RMT systems expressed at the BBB enable new solutions to the problem of BBB drug transport.


Subject(s)
Biological Transport, Active/physiology , Blood-Brain Barrier/metabolism , Pharmaceutical Preparations/metabolism , Animals , Carrier Proteins/metabolism , Humans , Lipid Metabolism/physiology , Neuroimaging , Pharmaceutical Preparations/cerebrospinal fluid
14.
Drug Metab Dispos ; 40(4): 779-87, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22266779

ABSTRACT

The objectives of the study were to characterize the selectivity of dantrolene to breast cancer resistance protein (Bcrp) and to evaluate whether cerebrospinal fluid (CSF) can be used as a surrogate to assess brain exposures of BCRP and P-glycoprotein (Pgp) substrates. The impact of Bcrp and Pgp on dantrolene exposures in brain and CSF was examined in Bcrp and Mdr1a/1b knockout mice and was further investigated in wild-type mice in the presence of the Bcrp inhibitor (3S,6S,12aS)-1,2,3,4,6,7,12,12a-octahydro-9-methoxy-6-(2-methylpropyl)-1,4-dioxopyrazino[1',2':1,6]pyrido[3,4-b]indole-3-propanoic acid 1,1-dimethylethyl ester (Ko143), the Pgp inhibitor 6-[(2S,4R,6E)-4-methyl-2-(methylamino)-3-oxo-6-octenoic acid]-7-l-valine-cyclosporine A (PSC833), and the dual inhibitor N-(4-[2-(1,2,3,4-tetrahydro-6,7-dimethoxy-2-isoquinolinyl)ethyl]-phenyl)-9,10-dihydro-5-methoxy-9-oxo-4-acridine carboxamide (GF120918). The effect of Bcrp and Pgp on digoxin exposures in brain and CSF was investigated in wild-type mice in the presence of the inhibitors. In vivo studies showed dantrolene exposures in brain and CSF, but not the blood, increased in Bcrp(-/-) and Mdr1a/1b(-/-)/Bcrp(-/-) mice, or in the presence of the Bcrp inhibitors Ko143 or GF120918. Inhibition of Pgp by GF120918 and PSC833 significantly increased digoxin exposures in brain, CSF, and blood to a lesser extent. Results from the present study demonstrated that inhibition of Bcrp and Pgp increased not only the exposures of dantrolene and digoxin in brain, but also the exposures in CSF. In addition, the change of exposures in CSF reflected the changes in brain. The present study strongly suggests that the dantrolene and digoxin exposures in CSF are primarily determined by the rapid transport from brain to CSF, and inhibition of Bcrp and Pgp exhibits little impact on using CSF as surrogates to assess brain exposures of Bcrp and Pgp substrates.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Blood-Brain Barrier/metabolism , Pharmaceutical Preparations , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Animals , Biological Transport , Blood-Brain Barrier/drug effects , Caco-2 Cells , Dantrolene/administration & dosage , Dantrolene/blood , Dantrolene/cerebrospinal fluid , Digoxin/administration & dosage , Digoxin/blood , Digoxin/cerebrospinal fluid , Dose-Response Relationship, Drug , Humans , Mice , Mice, Knockout , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/cerebrospinal fluid , Time Factors , Tissue Distribution
15.
Bioanalysis ; 3(2): 167-79, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21250846

ABSTRACT

BACKGROUND: plasma/cerebrospinal fluid (CSF) ratio of compound K was determined to be lower than predicted during the conduct of clinical study. This triggered the evaluation of nonspecific binding of drug K and an additional ten compounds in CSF to collection tubing. Physiochemical properties such as pKa and log D values were correlated to the quantified results. RESULTS: mean recoveries of compound K quality controls sampled within 24 h, as compared with spiked quality control, were 12.9, 44.4 and 77.7% from silicone, Tygon and Pharmed collection tubing, respectively. Eight out of ten compounds showed significant loss into silicone and seven out of ten compounds showed no significant loss into Pharmed. CONCLUSION: silicone tubing is not recommended and Pharmed appears to be most appropriate. CSF sample collection tubing must be evaluated prior to starting clinical studies.


Subject(s)
Cerebrospinal Fluid/chemistry , Chromatography, Liquid/methods , Pharmaceutical Preparations/cerebrospinal fluid , Tandem Mass Spectrometry/methods , Technology, Pharmaceutical/methods , Biomedical Research , Humans , Pharmaceutical Preparations/analysis , Quality Control , Silicones/chemistry
16.
J Pharmacokinet Pharmacodyn ; 37(6): 629-44, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21132572

ABSTRACT

We introduce how biophysical modeling in pharmaceutical research and development, combining physiological observations at the tissue, organ and system level with selected drug physiochemical properties, may contribute to a greater and non-intuitive understanding of drug pharmacokinetics and therapeutic design. Based on rich first-principle knowledge combined with experimental data at both conception and calibration stages, and leveraging our insights on disease processes and drug pharmacology, biophysical modeling may provide a novel and unique opportunity to interactively characterize detailed drug transport, distribution, and subsequent therapeutic effects. This innovative approach is exemplified through a three-dimensional (3D) computational fluid dynamics model of the spinal canal motivated by questions arising during pharmaceutical development of one molecular therapy for spinal cord injury. The model was based on actual geometry reconstructed from magnetic resonance imaging data subsequently transformed in a parametric 3D geometry and a corresponding finite-volume representation. With dynamics controlled by transient Navier-Stokes equations, the model was implemented in a commercial multi-physics software environment established in the automotive and aerospace industries. While predictions were performed in silico, the underlying biophysical models relied on multiple sources of experimental data and knowledge from scientific literature. The results have provided insights into the primary factors that can influence the intrathecal distribution of drug after lumbar administration. This example illustrates how the approach connects the causal chain underlying drug distribution, starting with the technical aspect of drug delivery systems, through physiology-driven drug transport, then eventually linking to tissue penetration, binding, residence, and ultimately clearance. Currently supporting our drug development projects with an improved understanding of systems physiology, biophysical models are being increasingly used to characterize drug transport and distribution in human tissues where pharmacokinetic measurements are difficult or impossible to perform. Importantly, biophysical models can describe emergent properties of a system, i.e. properties not identifiable through the study of the system's components taken in isolation.


Subject(s)
Models, Anatomic , Models, Biological , Pharmaceutical Preparations/cerebrospinal fluid , Pharmacokinetics , Spinal Canal/anatomy & histology , Spinal Canal/physiology , Animals , Computational Biology/methods , Computer Simulation , Humans , Hydrodynamics , Injections, Spinal , Pharmaceutical Preparations/administration & dosage , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Tissue Distribution
17.
Bioanalysis ; 2(4): 755-67, 2010 Apr.
Article in English | MEDLINE | ID: mdl-21083273

ABSTRACT

BACKGROUND: The distribution coefficient, D, is a physicochemical property used to determine the partitioning of compounds between aqueous and hydrophobic media at a given pH. RESULTS: A clear relationship was observed between the calculated pH-dependent distribution coefficient of six representative pharmaceutical probe compounds and their propensity to partition between a relatively hydrophobic polypropylene surface and the aqueous matrices, human urine or human cerebrospinal fluid (CSF). Compound log D cut-off values of 1.5 and 3.8 for urine and CSF, respectively, were determined using a threshold of less than 20% adsorption to the polypropylene surface. CONCLUSION: The ability to forecast the adsorption of a given compound to a polypropylene container with urine and CSF offers an effective means for screening potential issues and identifying when additional testing and corrective measures may need to be applied.


Subject(s)
Pharmaceutical Preparations/chemistry , Polypropylenes/chemistry , Adsorption , Cholic Acids/chemistry , Humans , Hydrogen-Ion Concentration , Pharmaceutical Preparations/cerebrospinal fluid , Pharmaceutical Preparations/urine , Solvents/chemistry
18.
Pharm Res ; 27(10): 2054-62, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20473558

ABSTRACT

In the last decade, there has been substantial progress in understanding vectorial ligand transport through rodent and human choroid plexus (CP), the locus of the blood-CSF interface. In this Review, we enumerate the experimental data required to establish vectorial transport through CP and describe transporters involved in vectorial transport across CP. We also note how these transporters differ from those at the blood-brain barrier. The ligand (substrate) examples presented are methyltetrahydrofolate, methotrexate, leukotriene C(4), nucleosides, thiamine monophosphate, prostaglandins, and digoxin. Our focus is on more definitive experiments, including animal and human transporter "knock-outs." Finally, we discuss the neurochemical implications of vectorial transport through CP and the clinical implications of transporter polymorphisms and knockouts. Examples include descriptions of how vectorial transport through the CP for several micronutrients (e.g., methyltetrahydrofolate) nourishes the brain and how knowledge of CP vectorial transport can lead to important treatments.


Subject(s)
Blood-Brain Barrier/metabolism , Choroid Plexus/metabolism , Membrane Transport Proteins/metabolism , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/cerebrospinal fluid , Animals , Biological Transport , Humans , Ligands , Substrate Specificity
19.
J Cereb Blood Flow Metab ; 30(1): 150-61, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19756019

ABSTRACT

A major challenge associated with the determination of the unbound brain-to-plasma concentration ratio of a drug (K(p,uu,brain)), is the error associated with correction for the drug in various vascular spaces of the brain, i.e., in residual blood. The apparent brain vascular spaces of plasma water (V(water), 10.3 microL/g brain), plasma proteins (V(protein), 7.99 microL/g brain), and the volume of erythrocytes (V(er), 2.13 microL/g brain) were determined and incorporated into a novel, drug-specific correction model that took the drug-unbound fraction in the plasma (f(u,p)) into account. The correction model was successfully applied for the determination of K(p,uu,brain) for indomethacin, loperamide, and moxalactam, which had potential problems associated with correction. The influence on correction of the drug associated with erythrocytes was shown to be minimal. Therefore, it is proposed that correction for residual blood can be performed using an effective plasma space in the brain (V(eff)), which is calculated from the measured f(u,p) of the particular drug as well as from the estimates of V(water) and V(protein), which are provided in this study. Furthermore, the results highlight the value of determining K(p,uu,brain) with statistical precision to enable appropriate interpretation of brain exposure for drugs that appear to be restricted to the brain vascular spaces.


Subject(s)
Blood-Brain Barrier/physiology , Brain/metabolism , Pharmaceutical Preparations/metabolism , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antidiarrheals/administration & dosage , Antidiarrheals/pharmacokinetics , Central Nervous System/metabolism , Cerebrovascular Circulation/physiology , Chromatography, Liquid , Erythrocyte Volume , Hematocrit , Indomethacin/administration & dosage , Indomethacin/pharmacokinetics , Loperamide/administration & dosage , Loperamide/pharmacokinetics , Male , Mass Spectrometry , Moxalactam/administration & dosage , Moxalactam/pharmacokinetics , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/cerebrospinal fluid , Pharmacokinetics , Plasma/physiology , Rats , Rats, Sprague-Dawley , Reproducibility of Results
20.
J Med Chem ; 52(20): 6233-43, 2009 Oct 22.
Article in English | MEDLINE | ID: mdl-19764786

ABSTRACT

New experimental methodologies were applied to measure the unbound brain-to-plasma concentration ratio (K(p,uu,brain)) and the unbound CSF-to-plasma concentration ratio (K(p,uu,CSF)) in rats for 43 structurally diverse drugs. The relationship between chemical structure and K(p,uu,brain) was dominated by hydrogen bonding. Contrary to popular understanding based on the total brain-to-plasma concentration ratio (logBB), lipophilicity was not a determinant of unbound brain exposure. Although changing the number of hydrogen bond acceptors is a useful design strategy for optimizing K(p,uu,brain), future improvement of in silico prediction models is dependent on the accommodation of active drug transport. The structure-brain exposure relationships found in the rat also hold for humans, since the rank order of the drugs was similar for human and rat K(p,uu,CSF). This cross-species comparison was supported by K(p,uu,CSF) being within 3-fold of K(p,uu,brain) in the rat for 33 of 39 drugs. It was, however, also observed that K(p,uu,CSF) overpredicts K(p,uu,brain) for highly effluxed drugs, indicating lower efflux capacity of the blood-cerebrospinal fluid barrier compared to the blood-brain barrier.


Subject(s)
Brain/metabolism , Extracellular Fluid/metabolism , Pharmaceutical Preparations/cerebrospinal fluid , Pharmaceutical Preparations/chemistry , Pharmacokinetics , Animals , Blood-Brain Barrier/metabolism , Humans , Linear Models , Models, Biological , Pharmaceutical Preparations/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...