1
|
Chowdhury EA, Noorani B, Alqahtani F, Bhalerao A, Raut S, Sivandzade F, Cucullo L. Understanding the brain uptake and permeability of small molecules through the BBB: A technical overview. J Cereb Blood Flow Metab 2021; 41:1797-1820. [PMID: 33444097 PMCID: PMC8327119 DOI: 10.1177/0271678x20985946] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The brain is the most important organ in our body requiring its unique microenvironment. By the virtue of its function, the blood-brain barrier poses a significant hurdle in drug delivery for the treatment of neurological diseases. There are also different theories regarding how molecules are typically effluxed from the brain. In this review, we comprehensively discuss how the different pharmacokinetic techniques used for measuring brain uptake/permeability of small molecules have evolved with time. We also discuss the advantages and disadvantages associated with these different techniques as well as the importance to utilize the right method to properly assess CNS exposure to drug molecules. Even though very strong advances have been made we still have a long way to go to ensure a reduction in failures in central nervous system drug development programs.
Collapse
Affiliation(s)
- Ekram Ahmed Chowdhury
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, USA
| | - Behnam Noorani
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, USA
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Aditya Bhalerao
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, USA
| | - Snehal Raut
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, USA
| | - Farzane Sivandzade
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, USA
| | - Luca Cucullo
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, USA
| |
Collapse
|
2
|
Campos-Arroyo D, Maldonado V, Bahena I, Quintanar V, Patiño N, Carlos Martinez-Lazcano J, Melendez-Zajgla J. Probenecid Sensitizes Neuroblastoma Cancer Stem Cells to Cisplatin. Cancer Invest 2016; 34:155-66. [PMID: 26963048 DOI: 10.3109/07357907.2016.1139717] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We used both in vitro cultures of neuroblastoma cell lines and nude-mice xenotransplants to explore the effects of co-administration of cisplatin and probenecid. Probenecid sensitized neuroblastoma cells, including tumor cells with stem features, to the effects of cisplatin, both in vitro and in vivo. This effect was mediated by an increase in the apoptotic cell death and a concomitant decrease in cell proliferation. This effect is accompanied by modulation of the mRNA and protein of the drug efflux transporters MDR1, MRP2, and BCRP. The co-administration of probenecid with cisplatin should be explored as a possible therapeutic strategy.
Collapse
Affiliation(s)
- Denise Campos-Arroyo
- a Functional Genomics Laboratory , Instituto Nacional de Medicina Genomica , Mexico City 14610 , Mexico
| | - Vilma Maldonado
- a Functional Genomics Laboratory , Instituto Nacional de Medicina Genomica , Mexico City 14610 , Mexico
| | - Ivan Bahena
- a Functional Genomics Laboratory , Instituto Nacional de Medicina Genomica , Mexico City 14610 , Mexico
| | - Valeria Quintanar
- a Functional Genomics Laboratory , Instituto Nacional de Medicina Genomica , Mexico City 14610 , Mexico
| | - Nelly Patiño
- a Functional Genomics Laboratory , Instituto Nacional de Medicina Genomica , Mexico City 14610 , Mexico
| | - Juan Carlos Martinez-Lazcano
- b Department of Neurophysiology , Instituto Nacional de Neurologia y Neurocirugia Manuel Velasco Suarez , Mexico City , Mexico
| | - Jorge Melendez-Zajgla
- a Functional Genomics Laboratory , Instituto Nacional de Medicina Genomica , Mexico City 14610 , Mexico
| |
Collapse
|
3
|
PKPD Aspects of Brain Drug Delivery in a Translational Perspective. DRUG DELIVERY TO THE BRAIN 2014. [DOI: 10.1007/978-1-4614-9105-7_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
4
|
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: 3.1] [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
|
5
|
Abstract
Severe HIV-associated neurocognitive disorders (HAND), such as HIV-associated dementia, and opportunistic CNS infections are now rare complications of HIV infection due to comprehensive highly active antiretroviral therapy (HAART). By contrast, mild to moderate neurocognitive disorders remain prevalent, despite good viral control in peripheral compartments. HIV infection seems to provoke chronic CNS injury that may evade systemic HAART. Penetration of antiretroviral drugs across the blood-brain barrier might be crucial for the treatment of HAND. This review identifies and evaluates the available clinical evidence on CSF penetration properties of antiretroviral drugs, addressing methodological issues and discussing the clinical relevance of drug concentration assessment. Although a substantial number of studies examined CSF concentrations of antiretroviral drugs, there is a need for adequate, well designed trials to provide more valid drug distribution profiles. Neuropsychological benefits and neurotoxicity of potentially CNS-active drugs require further investigation before penetration characteristics will regularly influence therapeutic strategies and outcome.
Collapse
Affiliation(s)
- Christine Eisfeld
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | | | | | | |
Collapse
|
6
|
Campos-Arroyo D, Martínez-Lazcano JC, Melendez-Zajgla J. Probenecid is a chemosensitizer in cancer cell lines. Cancer Chemother Pharmacol 2011; 69:495-504. [DOI: 10.1007/s00280-011-1725-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 08/08/2011] [Indexed: 02/08/2023]
|
7
|
Eyal S, Hsiao P, Unadkat JD. Drug interactions at the blood-brain barrier: fact or fantasy? Pharmacol Ther 2009; 123:80-104. [PMID: 19393264 DOI: 10.1016/j.pharmthera.2009.03.017] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 03/20/2009] [Indexed: 12/24/2022]
Abstract
There is considerable interest in the therapeutic and adverse outcomes of drug interactions at the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB). These include altered efficacy of drugs used in the treatment of CNS disorders, such as AIDS dementia and malignant tumors, and enhanced neurotoxicity of drugs that normally penetrate poorly into the brain. BBB- and BCSFB-mediated interactions are possible because these interfaces are not only passive anatomical barriers, but are also dynamic in that they express a variety of influx and efflux transporters and drug metabolizing enzymes. Based on studies in rodents, it has been widely postulated that efflux transporters play an important role at the human BBB in terms of drug delivery. Furthermore, it is assumed that chemical inhibition of transporters or their genetic ablation in rodents is predictive of the magnitude of interaction to be expected at the human BBB. However, studies in humans challenge this well-established paradigm and claim that such drug interactions will be lesser in magnitude but yet may be clinically significant. This review focuses on current known mechanisms of drug interactions at the blood-brain and blood-CSF barriers and the potential impact of such interactions in humans. We also explore whether such drug interactions can be predicted from preclinical studies. Defining the mechanisms and the impact of drug-drug interactions at the BBB is important for improving efficacy of drugs used in the treatment of CNS disorders while minimizing their toxicity as well as minimizing neurotoxicity of non-CNS drugs.
Collapse
Affiliation(s)
- Sara Eyal
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195, USA
| | | | | |
Collapse
|
8
|
Rayner CR, Chanu P, Gieschke R, Boak LM, Jonsson EN. Population pharmacokinetics of oseltamivir when coadministered with probenecid. J Clin Pharmacol 2008; 48:935-47. [PMID: 18524996 DOI: 10.1177/0091270008320317] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Oseltamivir is a potent, selective, oral neuraminidase inhibitor for the treatment and prophylaxis of influenza. Plasma concentrations of the active metabolite, oseltamivir carboxylate, are increased in the presence of probenecid, suggesting that the combination could allow for the use of reduced doses of oseltamivir. To investigate this proposal, we developed a population pharmacokinetic model and simulated the pharmacokinetics of candidate combination regimens of oral oseltamivir (45 mg and 30 mg twice a day) plus oral probenecid (500 mg/6 hourly). Probenecid plus oseltamivir 45 mg achieved all the pharmacokinetic parameters expected of oseltamivir alone, but combination with oseltamivir 30 mg and dose interval extension approaches did not. An oseltamivir-probenecid combination may compromise tolerability and enhance the potential for drug interactions. In addition, increased dosing requirements may affect compliance and attainment of optimal oseltamivir exposure, potentially facilitating the emergence of viral strains with reduced susceptibility to oseltamivir. These factors, set alongside increased capacity for oseltamivir production, should be carefully considered before an oseltamivir-probenecid combination is used.
Collapse
Affiliation(s)
- Craig R Rayner
- F. Hoffmann-La Roche Ltd, Pharmaceuticals Division, Bldg. 015/1.006, CH-4070 Basel, Switzerland.
| | | | | | | | | |
Collapse
|
9
|
Giri N, Shaik N, Pan G, Terasaki T, Mukai C, Kitagaki S, Miyakoshi N, Elmquist WF. Investigation of the role of breast cancer resistance protein (Bcrp/Abcg2) on pharmacokinetics and central nervous system penetration of abacavir and zidovudine in the mouse. Drug Metab Dispos 2008; 36:1476-84. [PMID: 18443033 DOI: 10.1124/dmd.108.020974] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Many anti-human immunodeficiency virus 1 nucleoside reverse-transcriptase inhibitors have low central nervous system (CNS) distribution due in part to active efflux transport at the blood-brain barrier. We have previously shown that zidovudine (AZT) and abacavir (ABC) are in vitro substrates for the efflux transport protein breast cancer resistance protein (Bcrp) 1. We evaluated the influence of Bcrp1 on plasma pharmacokinetics and brain penetration of zidovudine and abacavir in wild-type and Bcrp1-deficient (Bcrp1-/-) FVB mice. There was no difference in either area under the concentration-time profiles for plasma (AUC(plasma)) or brain (AUC(brain)) for zidovudine between the wild-type and Bcrp1-/- mice. The AUC(plasma) of abacavir was 20% lower in the Bcrp1-/- mice, whereas the AUC(brain) was 20% greater. This difference resulted in a 1.5-fold increase in abacavir brain exposure in the Bcrp1-/- mice. The effect of selective and nonselective transport inhibitors on the ABC brain/plasma ratio at a single time point was evaluated. 3-(6-Isobutyl-9-methoxy-1,4-dioxo-1,2,3,4,6, 7,12,12a-octahydropyrazino[1',2':1,6]pyrido[3,4-b]indol-3-yl)-propionicacid tert-butyl ester (Ko143), N[4[2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)ethyl]phenyl]-5-methoxy-9-oxo-10H-acridine-4-carboxamide (GF120918), probenecid, and Pluronic P85 increased abacavir plasma concentrations in the wild-type mice. Abacavir plasma concentrations in Bcrp1-/- mice were increased by (R)-4-((1aR,6R,10bS)-1,2-difluoro-1,1a,6,10b-tetrahydrodibenzo (a,e)cyclopropa(c)cycloheptan-6-yl)-alpha-((5-quinoloyloxy)methyl)-1-piperazineethanol trihydrochloride (LY335979), GF120918, and probenecid, but not by Ko143. Brain/plasma concentration ratios in both the wild-type and Bcrp1-/- mice were increased by the P-glycoprotein inhibitors LY335979 and GF120918, but not by BCRP-selective inhibitors. These data indicate that deletion of Bcrp1 has little influence on the pharmacokinetics or brain penetration of AZT. However, for abacavir, deletion of Bcrp1 reduces plasma exposure and enhances brain penetration. These findings suggest that Bcrp1 does not play a significant role in limiting the CNS distribution of zidovudine and abacavir; however, brain penetration of abacavir is dependent on P-glycoprotein-mediated efflux.
Collapse
Affiliation(s)
- Nagdeep Giri
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Shaik N, Giri N, Pan G, Elmquist WF. P-glycoprotein-mediated active efflux of the anti-HIV1 nucleoside abacavir limits cellular accumulation and brain distribution. Drug Metab Dispos 2007; 35:2076-85. [PMID: 17709369 DOI: 10.1124/dmd.107.017723] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
P-glycoprotein (P-gp)-mediated efflux at the blood-brain barrier has been implicated in limiting the brain distribution of many anti-HIV1 drugs, primarily protease inhibitors, resulting in suboptimal concentrations in this important sanctuary site. The objective of this study was to characterize the interaction of abacavir with P-gp and determine whether P-gp is an important mechanism in limiting abacavir delivery to the central nervous system (CNS). In vitro and in vivo techniques were employed to characterize this interaction. Abacavir stimulated P-gp ATPase activity at high concentrations. The cellular accumulation of abacavir was significantly decreased by approximately 70% in Madin-Darby canine kidney II (MDCKII)-MDR1 monolayers compared with wild-type cells and was completely restored by the P-gp inhibitors ((R)-4-((1aR,6R,10bS)-1,2-difluoro-1,1a,6,10b-tetrahydrodibenzo(a,e)cyclopropa(c)cycloheptan-6-yl)-alpha-((5-quinoloyloxy)methyl)-1-piperazineethanol, trihydrochloride) (LY335979) and N-[4-[2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)ethyl]phenyl]-5-methoxy-9-oxo-10H-acridine-4-carboxamide (GF120918). Directional flux experiments indicated that abacavir had greater permeability in the basolateral-to-apical direction (1.58E-05 cm/s) than in the apical-to-basolateral direction (3.44E-06 cm/s) in MDR1-transfected monolayers. The directionality in net flux was abolished by both LY335979 and GF120918. In vivo brain distribution studies showed that the AUC(plasma) in mdr1a(-/-) CF-1 mutant mice was approximately 2-fold greater than the AUC(plasma) in the wild type, whereas the AUC(brain) in the mutant was 20-fold higher than that in the wild type. Therefore, the CNS drug targeting index, defined as the ratio of AUC brain-to-plasma for mutant over wild type, was greater than 10. These data are the first in vitro and in vivo evidence that a nucleoside reverse transcriptase inhibitor is a P-gp substrate. The remarkable increase in abacavir brain distribution in P-gp-deficient mutant mice over wild-type mice suggests that P-gp may play a significant role in restricting the abacavir distribution to the CNS.
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/antagonists & inhibitors
- 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
- Acridines/chemistry
- Acridines/metabolism
- Acridines/pharmacokinetics
- Adenosine Triphosphatases/metabolism
- Animals
- Anti-HIV Agents/chemistry
- Anti-HIV Agents/metabolism
- Anti-HIV Agents/pharmacokinetics
- Area Under Curve
- Biological Transport, Active
- Brain/metabolism
- Cell Line
- Cell Membrane/metabolism
- Dibenzocycloheptenes/chemistry
- Dibenzocycloheptenes/metabolism
- Dibenzocycloheptenes/pharmacokinetics
- Dideoxynucleosides/chemistry
- Dideoxynucleosides/metabolism
- Dideoxynucleosides/pharmacokinetics
- Humans
- Mice
- Mice, Inbred Strains
- Mice, Knockout
- Molecular Structure
- Quinolines/chemistry
- Quinolines/metabolism
- Quinolines/pharmacokinetics
- Reverse Transcriptase Inhibitors/chemistry
- Reverse Transcriptase Inhibitors/metabolism
- Reverse Transcriptase Inhibitors/pharmacokinetics
- Tetrahydroisoquinolines/chemistry
- Tetrahydroisoquinolines/metabolism
- Tetrahydroisoquinolines/pharmacokinetics
- Vinblastine/chemistry
- Vinblastine/metabolism
- Vinblastine/pharmacokinetics
- Zidovudine/chemistry
- Zidovudine/metabolism
- Zidovudine/pharmacokinetics
Collapse
Affiliation(s)
- Naveed Shaik
- Department of Pharmaceutics, University of Minnesota, 308 Harvard St. SE, Room 9-125, Weaver-Densford Hall, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
11
|
Pan G, Giri N, Elmquist WF. Abcg2/Bcrp1 mediates the polarized transport of antiretroviral nucleosides abacavir and zidovudine. Drug Metab Dispos 2007; 35:1165-73. [PMID: 17437964 DOI: 10.1124/dmd.106.014274] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The bioavailability and targeted distribution of abacavir (ABC) and zidovudine (AZT) to viral reservoirs may be influenced by efflux transporters. The purpose of this study was to characterize the interaction of these nucleoside reverse transcriptase inhibitors with the Abcg2/Bcrp1 transporter, the murine homolog of human breast cancer resistance protein (BCRP), using a Bcrp1-transfected Madin-Darby canine kidney II cell model. Intracellular accumulation of ABC and AZT was significantly reduced by approximately 90% and approximately 70%, respectively, in Bcrp1-transfected cells compared with the wild-type cells. Both ABC and AZT showed significantly increased basolateral-to-apical (B-to-A) and decreased apical-to-basolateral (A-to-B) transport in Bcrp1 cells compared with wild-type directional flux. The efflux ratio (ratio of B-to-A to A-to-B) in Bcrp1-transfected cells was 22 for ABC and 11 for AZT. 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) inhibited this difference in accumulation between the two cell variants with an EC(50) of 1.32 +/- 0.3 microM for ABC and 0.31 +/- 0.1 microM for AZT. Potent and highly cooperative inhibition by Ko143 (3-(6-isobutyl-9-methoxy-1,4-dioxo-1,2,3,4,6,7,12,12a-octahydropyrazino[1',2':1,6]pyrido[3,4-b]indol-3-yl)-propionic acid tert-butyl ester) was observed with an EC(50) of 121 +/- 5 nM for ABC and 19.2 +/- 1.5 nM for AZT (Hill coefficient approximately 3-6). Probenecid, an organic anion inhibitor known to influence AZT biodistribution, had no effect on cellular accumulation in the Bcrp1 model. These studies characterize the Bcrp1-mediated transport of ABC and AZT and show that prototypical BCRP inhibitors GF120918 and Ko143 can inhibit the Bcrp1-mediated transport of these important antiretroviral compounds. The functional expression of BCRP at critical barriers, such as the intestinal enterocytes, brain capillary endothelium, and target lymphocytes, could influence the bioavailability and targeted delivery of these drugs to sanctuary sites.
Collapse
Affiliation(s)
- Guoyu Pan
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
12
|
Strazielle N, Ghersi-Egea JF. Factors affecting delivery of antiviral drugs to the brain. Rev Med Virol 2005; 15:105-33. [PMID: 15546130 DOI: 10.1002/rmv.454] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Although the CNS is in part protected from peripheral insults by the blood-brain barrier and the blood-cerebrospinal fluid barrier, a number of human viruses gain access to the brain, replicate within this organ, or sustain latent infection. The efficacy of antiviral drugs towards the cerebral viral load is often limited as both blood-brain interfaces impede their cerebral distribution. For polar compounds, the major factor restricting their entry lies in the tight junctions that occlude the paracellular pathway across these barriers. For compounds with more favourable lipid solubility properties, CNS penetration will be function of a number of physicochemical factors that include the degree of lipophilicity, size and ability to bind to protein or red blood cells, as well as other factors inherent to the vascular and choroidal systems, such as the local cerebral blood flow and the surface area available for exchange. In addition, influx and efflux transport systems, or metabolic processes active in both capillary endothelial cells and choroid plexus epithelial cells, can greatly change the bioavailability of a drug in one or several compartments of the CNS. The relative importance of these various factors with respect to the CNS delivery of the different classes of antiviral drugs is illustrated and discussed.
Collapse
|
13
|
Liu X, Smith BJ, Chen C, Callegari E, Becker SL, Chen X, Cianfrogna J, Doran AC, Doran SD, Gibbs JP, Hosea N, Liu J, Nelson FR, Szewc MA, Van Deusen J. Use of a physiologically based pharmacokinetic model to study the time to reach brain equilibrium: an experimental analysis of the role of blood-brain barrier permeability, plasma protein binding, and brain tissue binding. J Pharmacol Exp Ther 2005; 313:1254-62. [PMID: 15743928 DOI: 10.1124/jpet.104.079319] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study was designed 1) to examine the effects of blood-brain barrier (BBB) permeability [quantified as permeability-surface area product (PS)], unbound fraction in plasma (f(u,plasma)), and brain tissue (f(u,brain)) on the time to reach equilibrium between brain and plasma and 2) to investigate the drug discovery strategies to design and select compounds that can rapidly penetrate the BBB and distribute to the site of action. The pharmacokinetics of seven model compounds: caffeine, CP-141938 [methoxy-3-[(2-phenyl-piperadinyl-3-amino)-methyl]-phenyl-N-methyl-methane-sulfonamide], fluoxetine, NFPS [N[3-(4'-fluorophenyl)-3-(4'-phenylphenoxy)propyl]sarcosine], propranolol, theobromine, and theophylline in rat brain and plasma after subcutaneous administration were studied. The in vivo log PS and log f(u,brain) calculated using a physiologically based pharmacokinetic model correlates with in situ log PS (R(2) = 0.83) and in vitro log f(u,brain) (R(2) = 0.69), where the in situ PS and in vitro f(u,brain) was determined using in situ brain perfusion and equilibrium dialysis using brain homogenate, respectively. The time to achieve brain equilibrium can be quantitated with a proposed parameter, intrinsic brain equilibrium half-life [t(1/2eq,in) = V(b)ln2/(PS . f(u,brain))], where V(b) is the physiological volume of brain. The in vivo log t(1/2eq,in) does not correlate with in situ log PS (R(2) < 0.01) but correlates inversely with log(PS . f(u,brain)) (R(2) = 0.85). The present study demonstrates that rapid brain equilibration requires a combination of high BBB permeability and low brain tissue binding. A high BBB permeability alone cannot guarantee a rapid equilibration. The strategy to select compounds with rapid brain equilibration in drug discovery should identify compounds with high BBB permeability and low nonspecific binding in brain tissue.
Collapse
Affiliation(s)
- Xingrong Liu
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Global Research and Development, Groton, CT 06340, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Müller M, dela Peña A, Derendorf H. Issues in pharmacokinetics and pharmacodynamics of anti-infective agents: distribution in tissue. Antimicrob Agents Chemother 2004; 48:1441-53. [PMID: 15105091 PMCID: PMC400530 DOI: 10.1128/aac.48.5.1441-1453.2004] [Citation(s) in RCA: 190] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Markus Müller
- Health Science Center, Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida 32610-0494, USA
| | | | | |
Collapse
|
15
|
Lindén K, Ståhle L, Ljungdahl-Ståhle E, Borg N. Effect of Probenecid and Quinidine on the Transport of Alovudine (3'-Fluorothymidine) to the Rat Brain Studied by Microdialysis. ACTA ACUST UNITED AC 2003; 93:226-32. [PMID: 14629734 DOI: 10.1046/j.1600-0773.2003.pto930505.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Microdialysis was used to sample extracellular unbound concentrations of alovudine in order to study the influence of well-known transport inhibitors (probenecid and quinidine) on the transport of alovudine between the blood and the brain extracellular fluid or whole brain tissue. The AUC (area under the time versus concentration curve) ratio brain extracellular fluid/serum was 0.17+/-0.036 after a subcutaneous injection of alovudine 25 mg/kg in rats treated with probenecid 25 mg/kg subcutaneous (n=5), which was not significantly different from the control group (AUC ratio 0.24+/-0.039). Perfusion through the microdialysis probe with probenecid 100 microM (n=4) also had no effect on the brain extracellular fluid/serum AUC ratio after alovudine 25 mg/kg subcutaneous. The AUC ratio brain extracellular fluid/serum was 0.085+/-0.009 after subcutaneous injection of alovudine 25 mg/kg in rats treated with quinidine 25 mg/kg intraperitoneally (n=8), which was significantly lower than the control group. However, the whole brain tissue concentration was not significantly different between control rats (n=5) and rats treated with quinidine (n=4) 1 hr after subcutaneous injection of alovudine 25 mg/kg (brain to serum ratios being 0.11+/-0.006 and 0.10+/-0.005 respectively). Finally, the microdialysis recovery of alovudine increased with increasing concentrations (10, 50, 250, 1250 microM) of alovudine in the perfusion fluid. The recovery of alovudine was increased in quinidine-treated rats but not in those given probenecid. Thus, probenecid does not significantly influence the concentration gradient of alovudine over the blood-brain barrier in the rat after systemic or after local administration, while quinidine lowered brain extracellular fluid concentration of alovudine, but not total brain tissue concentration. The mechanism behind this phenomenon is not yet known.
Collapse
Affiliation(s)
- Karin Lindén
- Department of Clinical Pharmacology, Huddinge University Hospital, SE-14186 Huddinge, Sweden.
| | | | | | | |
Collapse
|
16
|
Abstract
Delivery of drugs across the blood-brain barrier has been shown to be altered during pathological states involving pain. Pain is a complex phenomenon involving immune and centrally mediated responses, as well as activation of the hypothalamic-pituitary-adrenal axis. Mediators released in response to pain have been shown to affect the structure and function of the blood-brain barrier in vitro and in vivo. These alterations in blood-brain barrier permeability and cytoarchitecture have implications in terms of drug delivery to the central nervous system, since pain and inflammation have the capacity to alter drug uptake and efflux across the blood-brain barrier. An understanding of how blood-brain barrier and central nervous system drug delivery mechanisms are altered during pathological conditions involving pain and/or inflammation is important in designing effective therapeutic regimens to treat disease.
Collapse
Affiliation(s)
- Anne M Wolka
- Department of Pharmacology, University of Arizona College of Medicine, 1501 N. Campbell Avenue, Tucson, AZ 85724, USA
| | | | | |
Collapse
|
17
|
Abstract
The central nervous system (CNS) contains important cellular barriers that maintain homeostasis by protecting the brain from circulating toxins and through the elimination of toxic metabolites generated in the brain. The barriers that limit the concentration of toxins and xenobiotics in the interstitial fluids of the CNS are the capillary endothelial cells of the blood-brain barrier (BBB) and the epithelial cells of the blood-cerebrospinal fluid barrier (BCSFB). Both of these barriers have cellular tight junctions and express transport systems which serve to actively transport nutrients into the brain, and actively efflux toxic metabolites and xenobiotics out of the brain. This review will focus on the expression and function of selected drug efflux transporters in these two barriers, specifically the multidrug resistance transporter, p-glycoprotein, and various organic anion transporters, such as multidrug resistance-associated proteins, organic anion transporter polypeptides, and organic anion transporters. These transport systems are increasingly recognized as important determinants of drug distribution to, and elimination from, different compartments of the CNS. Consequences of drug efflux transporters in barriers of the CNS include limiting the distribution of substrates that are beneficial to treat CNS diseases, and increasing the possibility of drug-drug interactions that may lead to untoward toxicities. Therefore, the study of these transporters is important in examining the various determinants of drug delivery to the CNS.
Collapse
Affiliation(s)
- Haiying Sun
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 986025 Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | |
Collapse
|
18
|
Adachi M, Reid G, Schuetz JD. Therapeutic and biological importance of getting nucleotides out of cells: a case for the ABC transporters, MRP4 and 5. Adv Drug Deliv Rev 2002; 54:1333-42. [PMID: 12406648 DOI: 10.1016/s0169-409x(02)00166-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The energy dependent transport of drugs contributes to cellular resistance and is undoubtedly a prime suspect in chemotherapeutic failure of a variety of disease processes. Early studies focused on a single gene, the multidrug resistance gene, MDR1, as a main contributor to chemotherapeutic failure. However, the multifaceted nature of cellular resistance lead to the discovery of the MRP gene. This pivotal finding and the concurrent rapid development of gene databases lead to the expansion of the MRP gene family. The purpose of this review is to discuss two of the recently described MRP family members that were orphans until their role in drug resistance was discovered. This review will provide an overview of the current state of our understanding of MRP4 and 5.
Collapse
Affiliation(s)
- Masashi Adachi
- Department of Pharmaceutical Sciences, St Jude Children's Research Hospital, 332 N Lauderdale Avenue, Memphis, TN 38105, USA
| | | | | |
Collapse
|
19
|
Taylor EM. The impact of efflux transporters in the brain on the development of drugs for CNS disorders. Clin Pharmacokinet 2002; 41:81-92. [PMID: 11888329 DOI: 10.2165/00003088-200241020-00001] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The development of drugs to treat disorders of the CNS requires consideration of achievable brain concentrations. Factors that influence the brain concentrations of drugs include the rate of transport into the brain across the blood-brain barrier (BBB), metabolic stability of the drug, and active transport out of the brain by efflux mechanisms. To date, three classes of transporter have been implicated in the efflux of drugs from the brain: multidrug resistance transporters, monocarboxylic acid transporters, and organic ion transporters. Each of the three classes comprises multiple transporters, each of which has multiple substrates, and the combined substrate profile of these transporters includes a large number of commonly used drugs. This system of transporters may therefore provide a mechanism through which the penetration of CNS-targeted drugs into the brain is effectively minimised. The action of these efflux transporters at the BBB may be reflected in the clinic as the minimal effectiveness of drugs targeted at CNS disorders, including HIV dementia, epilepsy, CNS-based pain, meningitis and brain cancers. Therefore, modulation of these efflux transporters by design of inhibitors and/or design of compounds that have minimal affinity for these transporters may well enhance the treatment of intractable CNS disorders.
Collapse
Affiliation(s)
- Eve M Taylor
- NeoTherapeutics Inc., Irvine, California 92618, USA.
| |
Collapse
|
20
|
Fox E, Bungay PM, Bacher J, McCully CL, Dedrick RL, Balis FM. Zidovudine concentration in brain extracellular fluid measured by microdialysis: steady-state and transient results in rhesus monkey. J Pharmacol Exp Ther 2002; 301:1003-11. [PMID: 12023531 DOI: 10.1124/jpet.301.3.1003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We measured zidovudine concentrations in blood, muscle, and brain extracellular fluid (ECF) by microdialysis and in serum ultrafiltrate and cerebrospinal fluid (CSF) samples during a continuous intravenous infusion (15 mg/kg/h) and after bolus dosing (50-80 mg/kg over 15 min) in nonhuman primates to determine whether CSF drug penetration is a valid surrogate for blood-brain barrier penetration. Recovery was estimated in vivo by zero net flux for the continuous infusion and retrodialysis for the bolus dosing. In vivo recovery was tissue-dependent and was lower in brain than in blood or muscle. Mean (+/-S.D.) steady-state blood, muscle, and brain zidovudine concentrations by microdialysis were 112 +/- 63.8, 105 +/- 51.1, and 13.8 +/- 10.4 microM, respectively; and steady-state serum ultrafiltrate and CSF concentrations were 81.2 +/- 40.2 and 14.1 +/- 8.0 microM, respectively. Brain ECF penetration (microdialysis brain/blood ratio) and CSF penetration (standard sampling CSF/serum ratio) at steady state were 0.13 +/- 0.06 and 0.17 +/- 0.02, respectively. With bolus dosing the mean (+/-S.D.) zidovudine area under concentration-time curve (AUC) normalized to a dose of 80 mg/kg was 577 +/- 103 microM. h in blood, 528 +/- 202 microM. h in muscle, and 108 +/- 74 microM. h in brain (brain/blood ratio of 0.18 +/- 0.10) by microdialysis. Serum ultrafiltrate AUC was 446 +/- 72 microM. h and the CSF AUC was 123 +/- 4.7 microM. h (CSF/serum ratio of 0.28 +/- 0.06). In conclusion, recovery was tissue-dependent. CSF and brain ECF zidovudine concentrations were comparable at steady state, and the corresponding AUCs were comparable after bolus injection. Thus, zidovudine penetration in brain ECF and CSF in nonhuman primates is limited to a similar extent, presumably by active transport, as in other species.
Collapse
Affiliation(s)
- Elizabeth Fox
- Pharmacology and Experimental Therapeutics Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, 10/13C193, 10 Center Drive, MSC 1920, Bethesda, MD 20892, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Busidan Y, Shi X, Dow-Edwards DL. AZT distribution in the fetal and postnatal rat central nervous system. J Pharm Sci 2001; 90:1964-71. [PMID: 11745755 DOI: 10.1002/jps.1147] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The distribution of 3'-azido-3'-deoxythymidine (AZT, zidovudine), an antiviral drug used in the treatment of human immunodeficiency virus, was investigated in gestation day-20 (G-20) fetuses and in postnatal day-20 (PND-20) rats. At both ages, a single dose of 150 mg/kg (1.78 mmol/kg) AZT was administered orally along with tracer amounts of 14C-AZT, and rats were randomly killed at 15, 30, 60, 120, or 240 min after dosing. The fetuses, brains, and spinal cords were processed for autoradiography. The peak concentrations of AZT in plasma of G-20 and PND-20 rats were 92.2 microg/mL (0.345 micromol/mL) and 56.6 microg/mL (0.21 micromol/mL) at 15 and 30 min after intubation, respectively. The peak concentration of fetal tissue occurred in the colon at 60 min and was 205.8 microg/g tissue. In the G-20 rats, the brain showed higher levels of AZT than spinal cord only at the 30-min sample time, whereas in the PND-20 rats, greater radioactivity was found in the spinal cord up to the 240-min sample time. This pattern of AZT distribution in the central nervous system may hypothetically be attributed to the postnatal development of an organic anion carrier system believed to be responsible for transporting AZT from the brain to the blood, resulting in relatively greater overall exposure of the spinal cord to AZT than observed in the brain.
Collapse
Affiliation(s)
- Y Busidan
- Laboratory of Cerebral Metabolism, Department of Physiology & Pharmacology, State University of New York, Health Science Center, Box 29, 450 Clarkson Avenue, Brooklyn, New York 11203, USA
| | | | | |
Collapse
|
22
|
Abstract
Quantitative microdialysis in the central nervous system (CNS) has recently provided evidence for the existence of transporters as they relate to the brain distribution of a variety of drugs. Support for the existence of drug transporters in the blood-brain barrier (or in the blood-CSF barrier) comes from investigations that have found: unbound drug concentrations in brain fluids that are lower than corresponding levels in plasma; saturability of transport clearances across the blood-brain barrier and; the regulation of transport by putative inhibitors. Additional confirmatory evidence for the existence of active transport or carrier-mediated processes has also been derived from models that relate observed drug levels in the CNS with those in plasma or blood. The conclusion that reduced drug levels in brain fluids generally indicate the existence of active efflux transport is questioned. In the case of relatively polar compounds with modest blood-brain barrier permeability, lower unbound concentrations in brain may be a consequence of dilution by turnover of brain fluids. This review summarizes recent reports (grouped by class of compounds) where investigators have used microdialysis to examine the distribution of therapeutic agents to the CNS, and have reached conclusions regarding the functional presence of drug transporters in the brain.
Collapse
Affiliation(s)
- R J Sawchuk
- Department of Pharmaceutics, College of Pharmacy, Weaver-Densford Hall, Room 9-143B, 308 Harvard Street S.E., University of Minnesota, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
23
|
Stahle L, Borg N. Transport of alovudine (3'-fluorothymidine) into the brain and the cerebrospinal fluid of the rat, studied by microdialysis. Life Sci 2000; 66:1805-16. [PMID: 10809178 DOI: 10.1016/s0024-3205(00)00504-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Extracellular unbound concentrations of alovudine were sampled by microdialysis in order to study the transport of alovudine between the blood and the brain and the cerebrospinal fluid (CSF) in the rat. The AUC (area under the curve) ratio CSF/blood was higher than the brain/blood ratio after i.v. infusion of alovudine 25mg/kg/hr after a loading dose of 25 mg/kg in 5 minutes (n=4). Neither i.v. infusion of thymidine (25 mg/kg/hr, n=5; 100 mg/kg/hr, n=2) nor acetazolamide (50 mg/kg i.p. bolus followed by 25 mg/kg i.p. every second hour, n=3) influenced the brain/blood AUC ratio after alovudine 25 mg/kg s.c. injection compared to controls (n=5). Finally, perfusion through the microdialysis probe with thymidine (1000 microM, n=3) had also no effect on the brain/blood AUC ratio after alovudine 25 mg/kg s.c. Because neither thymidine nor acetazolamide has significant influence on the ability of alovudine to penetrate the blood-brain barrier in the rat, neither thymidine transport nor carboanhydrase dependent CSF production appear to be major determinants of the blood-brain concentration gradient. Thus, it is concluded that alovudine reaches the extracellular fluid of the brain not by cerebrospinal fluid, but via the cerebral capillaries and that the existence of a concentration gradient over both blood-brain and CSF-brain barrier can probably be explained by the presence of an active process pumping alovudine out from the brain.
Collapse
Affiliation(s)
- L Stahle
- Department of Clinical Pharmacology, Huddinge University Hospital, Sweden
| | | |
Collapse
|
24
|
Johnson MD, Anderson BD. Use of cultured cerebral capillary endothelial cells in modeling the central nervous system availability of 2',3'-dideoxyinosine. J Pharm Sci 2000; 89:322-35. [PMID: 10707013 DOI: 10.1002/(sici)1520-6017(200003)89:3<322::aid-jps4>3.0.co;2-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The biochemical and physiological mechanisms responsible for the limited central nervous system (CNS) uptake of dideoxynucleoside reverse transcriptase inhibitors currently used to treat HIV-1 infection in humans are poorly understood. In vitro models of the blood-brain barrier (BBB) offer an attractive alternative to in vivo or in situ animal studies for understanding the role of the blood-brain barrier in regulating brain tissue concentrations of these agents. In the present study, the kinetics of 2', 3'-dideoxyinosine (ddI) uptake and purine nucleoside phosphorylase (PNP) mediated catabolism in primary cultures of bovine brain microvessel endothelial cells (BBMECs) were determined in order to ascertain the importance of both transport and metabolism governing the CNS availability of this purine dideoxynucleoside. Initial rates of ddI uptake as a function of ddI donor concentration suggest the involvement of both passive diffusion and carrier-mediated processes. These studies confirm earlier in vivo findings that transporters may play a role in regulating the CNS concentration of ddI. Analysis of ddI uptake and metabolite accumulation in BBMECs over longer time intervals (beyond the intial rate region) provide substantial in vitro evidence for an enzymatic BBB for ddI. Simulations of the CNS availability of ddI derived from in vitro estimates of parameters for passive diffusion, carrier-mediation, and metabolism indicate that the fraction of ddI entering the BBB cells which actually reaches the brain parenchyma may be quite low (< 2%) due to metabolism by PNP localized within the BBB, consistent with the low CNS delivery of ddI observed in vivo. Transporters and metabolic enzymes within the BBB may function in coordinated fashion to reduce the CNS concentrations of both rapidly metabolized and poorly metabolized dideoxynucleosides.
Collapse
Affiliation(s)
- M D Johnson
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, USA
| | | |
Collapse
|
25
|
Investigation of distribution, transport and uptake of anti-HIV drugs to the central nervous system. Adv Drug Deliv Rev 1999; 39:5-31. [PMID: 10837765 DOI: 10.1016/s0169-409x(99)00017-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The distribution of currently available anti-HIV drugs into the CNS is reviewed with a focus on transport mechanisms. Among these drugs, nucleoside analogs are most well studied for their CNS distribution. The average reported values of the CSF/plasma steady-state concentration or corresponding AUC ratios are 0.23 (AZT), 0.06 (ddI), 0.04 (ddC), 0.49 (d4T), and 0.08 (3TC). Active efflux transport out of the CNS appears to be a predominant mechanism limiting nucleoside access to the CNS, although poor penetration may contribute to some extent for some polar nucleosides. The nature of the efflux pump for these drugs is speculated to be MRP-like transporter(s) in blood-brain and blood-CSF barriers. For non-nucleoside and protease inhibitors, much research remains to be done on the extent, time course, and mechanisms of their CNS distribution. The CNS penetration of some protease inhibitors is restricted by P-glycoprotein. A better understanding of transport mechanisms of anti-HIV drugs in the CNS is essential to develop approaches to enhance CNS delivery of available drugs and to identify new drugs less subject to active efflux transporter(s) in the CNS.
Collapse
|
26
|
Abstract
During the last two decades, a number of methods have been developed for in vivo collection, separation and characterization of biological samples and analytes. The capability and reliability of the microdialysis technique for measuring endogenous substances (such as neurotransmitters and their metabolites) as well as exogenous therapeutic agents in various tissue systems have brought it to the forefront of the in vivo tissue sampling methods. The usability of this technique is demonstrated by its application as reported in almost 3600 scientific papers (as of January 1998). This paper describes the general aspects and various applications of this fast growing technique. Emphasis has been given to analytical considerations with regards to microdialysis probe recovery and newer HPLC techniques.
Collapse
Affiliation(s)
- C S Chaurasia
- Division of Bioequivalence, Food and Drug Administration, MPN II, Rm 123E, 7500 Standish Place, Rockville, MD 20855, USA
| |
Collapse
|
27
|
|