1
|
Järvinen J, Montaser AB, Adla SK, Leppänen J, Lehtonen M, Vellonen KS, Laitinen T, Jalkanen A, Elmquist WF, Timonen J, Huttunen KM, Rautio J. Altering distribution profile of palbociclib by its prodrugs. Eur J Pharm Sci 2024; 192:106637. [PMID: 37967656 DOI: 10.1016/j.ejps.2023.106637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/17/2023]
Abstract
Palbociclib, a cyclin-dependent kinase (CDK) 4/6 inhibitor, is currently used clinically for treating hormone receptor-positive and human epidermal growth factor receptor 2 negative breast cancer. Additionally, it has the potential to be utilized in the treatment of various tumors, including malignant glioblastoma. Previous research has indicated that palbociclib is a substrate for two efflux transporters, P-glycoprotein (P-gp; MDR1) and breast cancer-resistant protein (BCRP), which restrict the brain exposure of palbociclib. In the present study, our objective was to alter the brain distribution pattern of palbociclib by creating and assessing two novel prodrugs through in vitro, in situ, and in vivo evaluations. To this end, we synthesized two prodrugs of palbociclib by attaching it to the tyrosine promoiety at the para- (PD1) and meta-(PD2) position via a carbamate bond. We hypothesized that the prodrugs could bypass efflux transporter-mediated drug resistance by leveraging the l-type amino acid transporter (LAT1) to facilitate their transport across the blood-brain barrier (BBB) and into cancer cells, such as glioma cells that express LAT1. The compounds PD1 and PD2 did not show selective binding and had limited inhibitory effects on LAT1 in three cell lines (MCF-7, U87-MG, HEK-hLAT1). However, PD1 and PD2 demonstrated the ability to evade efflux mechanisms, and their in vitro uptake profiles were comparable to that of palbociclib, indicating their potential for effective cellular transport. In in situ and in vivo studies, brain uptake was not significantly improved compared to palbociclib, but the pharmacokinetic profiles showed encouraging enhancements. PD1 exhibited a higher AUCbrain/plasma ratio, suggesting safer dosing, while PD2 showed favorable long-acting pharmacokinetics. Although our prodrug design did not significantly improve palbociclib brain delivery due to the potential size limitation of the prodrugs, the study provides valuable insights for future prodrug development and drug delivery strategies targeting specific transporters.
Collapse
Affiliation(s)
- Juulia Järvinen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| | - Ahmed B Montaser
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Santosh Kumar Adla
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Jukka Leppänen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Marko Lehtonen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Kati-Sisko Vellonen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Tuomo Laitinen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Aaro Jalkanen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - William F Elmquist
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Juri Timonen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, P.O. Box 56, 00014 Helsinki, Finland
| | - Kristiina M Huttunen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Jarkko Rautio
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| |
Collapse
|
2
|
Kim J, Shin SA, Lee CS, Chung HJ. An Improved In Vitro Blood-Brain Barrier Model for the Evaluation of Drug Permeability Using Transwell with Shear Stress. Pharmaceutics 2023; 16:48. [PMID: 38258059 PMCID: PMC10820479 DOI: 10.3390/pharmaceutics16010048] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
The development of drugs targeting the central nervous system (CNS) is challenging because of the presence of the Blood-Brain barrier (BBB). Developing physiologically relevant in vitro BBB models for evaluating drug permeability and predicting the activity of drug candidates is crucial. The transwell model is one of the most widely used in vitro BBB models. However, this model has limitations in mimicking in vivo conditions, particularly in the absence of shear stress. This study aimed to overcome the limitations of the transwell model using immortalized human endothelial cells (hCMEC/D3) by developing a novel dish design for an orbital shaker, providing shear stress. During optimization, we assessed cell layer integrity using trans-endothelial electrical resistance measurements and the % diffusion of lucifer yellow. The efflux transporter activity and mRNA expression of junctional proteins (claudin-5, occludin, and VE-cadherin) in the newly optimized model were verified. Additionally, the permeability of 14 compounds was evaluated and compared with published in vivo data. The cell-layer integrity was substantially increased using the newly designed annular shaking-dish model. The results demonstrate that our model provided robust conditions for evaluating the permeability of CNS drug candidates, potentially improving the reliability of in vitro BBB models in drug development.
Collapse
Affiliation(s)
- Junhyeong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.K.); (S.-A.S.); (C.S.L.)
- Anti-Aging Bio Cell factory Regional Leading Research Center (ABC-RLRC), Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Seong-Ah Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.K.); (S.-A.S.); (C.S.L.)
| | - Chang Sup Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.K.); (S.-A.S.); (C.S.L.)
| | - Hye Jin Chung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.K.); (S.-A.S.); (C.S.L.)
| |
Collapse
|
3
|
Parvez MM, Sadighi A, Ahn Y, Keller SF, Enoru JO. Uptake Transporters at the Blood-Brain Barrier and Their Role in Brain Drug Disposition. Pharmaceutics 2023; 15:2473. [PMID: 37896233 PMCID: PMC10610385 DOI: 10.3390/pharmaceutics15102473] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Uptake drug transporters play a significant role in the pharmacokinetic of drugs within the brain, facilitating their entry into the central nervous system (CNS). Understanding brain drug disposition is always challenging, especially with respect to preclinical to clinical translation. These transporters are members of the solute carrier (SLC) superfamily, which includes organic anion transporter polypeptides (OATPs), organic anion transporters (OATs), organic cation transporters (OCTs), and amino acid transporters. In this systematic review, we provide an overview of the current knowledge of uptake drug transporters in the brain and their contribution to drug disposition. Here, we also assemble currently available proteomics-based expression levels of uptake transporters in the human brain and their application in translational drug development. Proteomics data suggest that in association with efflux transporters, uptake drug transporters present at the BBB play a significant role in brain drug disposition. It is noteworthy that a significant level of species differences in uptake drug transporters activity exists, and this may contribute toward a disconnect in inter-species scaling. Taken together, uptake drug transporters at the BBB could play a significant role in pharmacokinetics (PK) and pharmacodynamics (PD). Continuous research is crucial for advancing our understanding of active uptake across the BBB.
Collapse
Affiliation(s)
- Md Masud Parvez
- Department of Quantitative, Translational & ADME Sciences (QTAS), AbbVie Biotherapeutics, San Francisco, CA 94080, USA; (M.M.P.)
| | - Armin Sadighi
- Department of Quantitative, Translational & ADME Sciences (QTAS), AbbVie Biotherapeutics, San Francisco, CA 94080, USA; (M.M.P.)
| | - Yeseul Ahn
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S Coulter St., Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Steve F. Keller
- Department of Quantitative, Translational & ADME Sciences (QTAS), AbbVie Biotherapeutics, San Francisco, CA 94080, USA; (M.M.P.)
| | - Julius O. Enoru
- Department of Quantitative, Translational & ADME Sciences (QTAS), AbbVie Biotherapeutics, San Francisco, CA 94080, USA; (M.M.P.)
| |
Collapse
|
4
|
Prantil-Baun R, Novak R, Das D, Somayaji MR, Przekwas A, Ingber DE. Physiologically Based Pharmacokinetic and Pharmacodynamic Analysis Enabled by Microfluidically Linked Organs-on-Chips. Annu Rev Pharmacol Toxicol 2019; 58:37-64. [PMID: 29309256 DOI: 10.1146/annurev-pharmtox-010716-104748] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Physiologically based pharmacokinetic (PBPK) modeling and simulation approaches are beginning to be integrated into drug development and approval processes because they enable key pharmacokinetic (PK) parameters to be predicted from in vitro data. However, these approaches are hampered by many limitations, including an inability to incorporate organ-specific differentials in drug clearance, distribution, and absorption that result from differences in cell uptake, transport, and metabolism. Moreover, such approaches are generally unable to provide insight into pharmacodynamic (PD) parameters. Recent development of microfluidic Organ-on-a-Chip (Organ Chip) cell culture devices that recapitulate tissue-tissue interfaces, vascular perfusion, and organ-level functionality offer the ability to overcome these limitations when multiple Organ Chips are linked via their endothelium-lined vascular channels. Here, we discuss successes and challenges in the use of existing culture models and vascularized Organ Chips for PBPK and PD modeling of human drug responses, as well as in vitro to in vivo extrapolation (IVIVE) of these results, and how these approaches might advance drug development and regulatory review processes in the future.
Collapse
Affiliation(s)
- Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, USA;
| | - Richard Novak
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, USA;
| | - Debarun Das
- CFD Research Corporation, Huntsville, Alabama 35806, USA
| | | | | | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, USA; .,Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.,Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
5
|
Saito K, Abe N, Toyama H, Ejima Y, Yamauchi M, Mushiake H, Kazama I. Second-Generation Histamine H1 Receptor Antagonists Suppress Delayed Rectifier K +-Channel Currents in Murine Thymocytes. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6261951. [PMID: 31183371 PMCID: PMC6515180 DOI: 10.1155/2019/6261951] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/31/2019] [Accepted: 04/18/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND/AIMS Voltage-dependent potassium channels (Kv1.3) are predominantly expressed in lymphocyte plasma membranes. These channels are critical for the activation and proliferation of lymphocytes. Since second-generation antihistamines are lipophilic and exert immunomodulatory effects, they are thought to affect the lymphocyte Kv1.3-channel currents. METHODS Using the patch-clamp whole-cell recording technique in murine thymocytes, we tested the effects of second-generation antihistamines, such as cetirizine, fexofenadine, azelastine, and terfenadine, on the channel currents and the membrane capacitance. RESULTS These drugs suppressed the peak and the pulse-end currents of the channels, although the effects of azelastine and terfenadine on the peak currents were more marked than those of cetirizine and fexofenadine. Both azelastine and terfenadine significantly lowered the membrane capacitance. Since these drugs did not affect the process of endocytosis in lymphocytes, they were thought to have interacted directly with the plasma membranes. CONCLUSIONS Our study revealed for the first time that second-generation antihistamines, including cetirizine, fexofenadine, azelastine, and terfenadine, exert suppressive effects on lymphocyte Kv1.3-channels. The efficacy of these drugs may be related to their immunomodulatory mechanisms that reduce the synthesis of inflammatory cytokine.
Collapse
Affiliation(s)
- Kazutomo Saito
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Nozomu Abe
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Hiroaki Toyama
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Yutaka Ejima
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Masanori Yamauchi
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Hajime Mushiake
- Department of Physiology, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Itsuro Kazama
- Department of Physiology, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
- Miyagi University, School of Nursing, Gakuen, Taiwa-cho, Kurokawa-gun, Miyagi, Japan
| |
Collapse
|
6
|
Chernykh IV, Shchul’kin AV, Myl’nikov PY, Gatsanoga MV, Popova NM, Yakusheva EN. A Method of Analysis of the Functional Activity of P-Glycoprotein in the Blood–Brain Barrier. NEUROCHEM J+ 2019. [DOI: 10.1134/s1819712419010069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
7
|
Mihajlica N, Betsholtz C, Hammarlund-Udenaes M. Rate of small-molecular drug transport across the blood-brain barrier in a pericyte-deficient state. Eur J Pharm Sci 2018; 124:182-187. [DOI: 10.1016/j.ejps.2018.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/08/2018] [Accepted: 08/08/2018] [Indexed: 01/23/2023]
|
8
|
Gealageas R, Devineau A, So PPL, Kim CMJ, Surendradoss J, Buchwalder C, Heller M, Goebeler V, Dullaghan EM, Grierson DS, Putnins EE. Development of Novel Monoamine Oxidase-B (MAO-B) Inhibitors with Reduced Blood-Brain Barrier Permeability for the Potential Management of Noncentral Nervous System (CNS) Diseases. J Med Chem 2018; 61:7043-7064. [PMID: 30016860 DOI: 10.1021/acs.jmedchem.7b01588] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Studies indicate that MAO-B is induced in peripheral inflammatory diseases. To target peripheral tissues using MAO-B inhibitors that do not permeate the blood-brain barrier (BBB) the MAO-B-selective inhibitor deprenyl was remodeled by replacing the terminal acetylene with a CO2H function, and incorporating a para-OCH2Ar motif (compounds 10a-s). Further, in compound 32 the C-2 side chain corresponded to CH2CN. In vitro, 10c, 10j, 10k, and 32 were identified as potent reversible MAO-B inhibitors, and all four compounds were more stable than deprenyl in plasma, liver microsomal, and hepatocyte stability assays. In vivo, they demonstrated greater plasma bioavailability. Assessment of in vitro BBB permeability showed that compound 10k is a P-glycoprotein (P-gp) substrate and 10j displayed mild interaction. Importantly, compounds 10c, 10j, 10k, and 32 displayed significantly reduced BBB permeability after intravenous, subcutaneous, and oral administration. These polar MAO-B inhibitors are pertinent leads for evaluation of efficacy in noncentral nervous system (CNS) inflammatory disease models.
Collapse
Affiliation(s)
- Ronan Gealageas
- Faculty of Pharmaceutical Sciences , The University of British Columbia , 2405 Wesbrook Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| | - Alice Devineau
- Faculty of Pharmaceutical Sciences , The University of British Columbia , 2405 Wesbrook Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| | - Pauline P L So
- Centre for Drug Research and Development , 2405 Wesbrook Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| | - Catrina M J Kim
- Centre for Drug Research and Development , 2405 Wesbrook Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| | - Jayakumar Surendradoss
- Centre for Drug Research and Development , 2405 Wesbrook Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| | - Christian Buchwalder
- Faculty of Pharmaceutical Sciences , The University of British Columbia , 2405 Wesbrook Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| | - Markus Heller
- Centre for Drug Research and Development , 2405 Wesbrook Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| | - Verena Goebeler
- Faculty of Dentistry , The University of British Columbia , 2199 Wesbrook Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| | - Edith M Dullaghan
- Centre for Drug Research and Development , 2405 Wesbrook Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| | - David S Grierson
- Faculty of Pharmaceutical Sciences , The University of British Columbia , 2405 Wesbrook Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| | - Edward E Putnins
- Faculty of Dentistry , The University of British Columbia , 2199 Wesbrook Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| |
Collapse
|
9
|
Ganguly S, Panetta JC, Roberts JK, Schuetz EG. Ketamine Pharmacokinetics and Pharmacodynamics Are Altered by P-Glycoprotein and Breast Cancer Resistance Protein Efflux Transporters in Mice. Drug Metab Dispos 2018; 46:1014-1022. [PMID: 29674491 PMCID: PMC5992966 DOI: 10.1124/dmd.117.078360] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 04/16/2018] [Indexed: 01/16/2023] Open
Abstract
To understand the systemic impact of breast cancer resistance protein (Bcrp) and P-glycoprotein (Pgp) deletion, untargeted metabolomics was performed on cerebral spinal fluid (CSF) and plasma of wild-type (WT) and Pgp and Bcrp double-knockout (dKO) rats anesthetized with ketamine-xylazine. We unexpectedly found elevated ketamine levels in both CSF and plasma of dKO versus WT rats. Therefore, the effect of these transporters was investigated on the 1) oral and intraperitoneal serum pharmacokinetics (PK) of ketamine, using a liquid chromatography method (high-performance liquid chromatography with ultraviolet detection), and 2) the anesthetic effect of ketamine using a duration of loss-of-righting reflex (dLORR) test in WT, Bcrp knockout (KO), Pgp KO, and Pgp/Bcrp dKO mice. The PK data demonstrated a significantly increased oral bioavailability and serum exposure of ketamine in dKO > Pgp KO > Bcrp KO mice compared with WT mice. Intraperitoneal ketamine-induced dLORR was significantly longer in dKO > Pgp KO > Bcrp KO > WT mice compared with WT mice. Inhibition of Bcrp and Pgp in WT mice using the dual Pgp/Bcrp inhibitor elacridar increased the ketamine-induced dLORR compared with vehicle-treated mice. The ketamine intracellular concentration was significantly decreased in Madin-Darby canine kidney II BCRP/PGP cells compared with the parental cells. In total, these results demonstrate that ketamine appears to be a dual Pgp/Bcrp substrate whose PK and pharmacodynamics are affected by Pgp and Bcrp-mediated efflux.
Collapse
Affiliation(s)
- Samit Ganguly
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, (S.G., J.C.P., J.K.R., E.G.S.); and Cancer and Developmental Biology Track, University of Tennessee Health Science Center, Memphis, Tennessee (S.G.)
| | - John C Panetta
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, (S.G., J.C.P., J.K.R., E.G.S.); and Cancer and Developmental Biology Track, University of Tennessee Health Science Center, Memphis, Tennessee (S.G.)
| | - Jessica K Roberts
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, (S.G., J.C.P., J.K.R., E.G.S.); and Cancer and Developmental Biology Track, University of Tennessee Health Science Center, Memphis, Tennessee (S.G.)
| | - Erin G Schuetz
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, (S.G., J.C.P., J.K.R., E.G.S.); and Cancer and Developmental Biology Track, University of Tennessee Health Science Center, Memphis, Tennessee (S.G.)
| |
Collapse
|
10
|
Samra GK, Dang K, Ho H, Baranwal A, Mukherjee J. Dual targeting agents for Aβ plaque/P-glycoprotein and Aβ plaque/nicotinic acetylcholine α4β2* receptors-potential approaches to facilitate Aβ plaque removal in Alzheimer's disease brain. Med Chem Res 2018; 27:1634-1646. [PMID: 29937677 PMCID: PMC6013267 DOI: 10.1007/s00044-018-2178-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/31/2018] [Indexed: 12/28/2022]
Abstract
Alzheimer's disease (AD) affects 10% of people older than 65 and is characterized by a progressive loss of cognitive function with an abnormal accumulation of amyloid β (Aβ ) plaques and neurofibrillary tangles (NFT) in the brain. Efforts to reduce brain Aβ plaques continue to be investigated as a therapeutic approach for AD. We report here development of dual targeting agents with affinity for Aβ plaque/P-glycoprotein (Pgp) and Aβ plaque/α4β 2* nicotinic acetylcholine receptors (nAChR). These novel dual agents may be able to efflux Aβ plaques via the paravascular (glymphatic) pathways. Ferulic acid (FA), ferulic acid ethyl ester (FAEE), and curcumin (CUR) were used for Aβ plaques, fexofenadine (FEX) was used as substrate for Pgp and nifrolidine (NIF) was used for α4β 2* nAChRs. Aβ plaque/α4β 2* nAChR dual agent, FA-NIF (GKS-007) exhibited IC50 = 3-6 nM for α4β 2* nAChRs in [3H]cytisine-radiolabeled thalamus and frontal cortex in rat brain slices. In postmortem human AD frontal cortex, Aβ plaques labeled with [3H]PIB, FEX-CUR showed a 35% reduction in gray matter (GM)/white matter (WM) [3H]PIB binding, while CUR alone showed a 50% reduction. In vivo biodistribution studies are required of the Aβ-Pgp and Aβ-α4β 2* nAChRs dual targeting agents in order to evaluate their potential as therapeutic approaches for reducing brain Aβ plaques.
Collapse
Affiliation(s)
- Gurleen K Samra
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA
| | - Kenneth Dang
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA
| | - Heather Ho
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA
| | - Aparna Baranwal
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA
| | - Jogeshwar Mukherjee
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA
- Department of Biomedical Engineering, University of California-Irvine, Irvine, CA 92697, USA
| |
Collapse
|
11
|
Abstract
Antipsychotics, risperidone, and risperidone’s active metabolite, paliperidone (9-hydroxyrisperidone), are related molecules used for the treatment of schizophrenia and related disorders. Differences in receptor binding, 5-HT2A/D2 (serotonin/dopamine) binding ratios, and mitochondrial proteomics suggest that the effects of risperidone and paliperidone on neuronal firing, regulation of mitochondrial function, and movement are different. This review seeks to explore the most significant differences at the molecular level between risperidone and paliperidone, as reported in preclinical studies. Although risperidone shows higher affinity for 5-HT receptors, paliperidone does not fit this profile. Thus, the risperidone 5-HT2A/D2 binding ratio is significantly lower than the paliperidone 5-HT2A/D2 binding ratio. Paliperidone, similar to lithium and valproate, affects expression levels and phosphorylation of complex I and V proteins in synaptoneurosomal preparations of rat prefrontal cortex, suggesting that paliperidone behaves as a mood stabilizer. It is apparent that the presence of a hydroxyl group in the paliperidone molecule confers increased hydrophilicity to this drug compared with its parent, risperidone; thus, this contributes to differential effects on mitochondrial movement, protein expression, and phosphorylation. These differences are reflected in synaptic plasticity and neuronal firing and have only recently been implicated in the mechanisms of mitochondrial function and movement.
Collapse
|
12
|
Ohura K, Nakada Y, Kotani S, Imai T. Design of Fexofenadine Prodrugs Based on Tissue-Specific Esterase Activity and Their Dissimilar Recognition by P-Glycoprotein. J Pharm Sci 2015; 104:3076-83. [DOI: 10.1002/jps.24467] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 04/07/2015] [Accepted: 04/07/2015] [Indexed: 11/11/2022]
|
13
|
Sander K, Galante E, Gendron T, Yiannaki E, Patel N, Kalber TL, Badar A, Robson M, Johnson SP, Bauer F, Mairinger S, Stanek J, Wanek T, Kuntner C, Kottke T, Weizel L, Dickens D, Erlandsson K, Hutton BF, Lythgoe MF, Stark H, Langer O, Koepp M, Årstad E. Development of Fluorine-18 Labeled Metabolically Activated Tracers for Imaging of Drug Efflux Transporters with Positron Emission Tomography. J Med Chem 2015; 58:6058-80. [PMID: 26161456 DOI: 10.1021/acs.jmedchem.5b00652] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Increased activity of efflux transporters, e.g., P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP), at the blood-brain barrier is a pathological hallmark of many neurological diseases, and the resulting multiple drug resistance represents a major clinical challenge. Noninvasive imaging of transporter activity can help to clarify the underlying mechanisms of drug resistance and facilitate diagnosis, patient stratification, and treatment monitoring. We have developed a metabolically activated radiotracer for functional imaging of P-gp/BCRP activity with positron emission tomography (PET). In preclinical studies, the tracer showed excellent initial brain uptake and clean conversion to the desired metabolite, although at a sluggish rate. Blocking with P-gp/BCRP modulators led to increased levels of brain radioactivity; however, dynamic PET did not show differential clearance rates between treatment and control groups. Our results provide proof-of-concept for development of prodrug tracers for imaging of P-gp/BCRP function in vivo but also highlight some challenges associated with this strategy.
Collapse
Affiliation(s)
- Kerstin Sander
- †Institute of Nuclear Medicine, University College London, 235 Euston Road, T5, London NW1 2BU, U.K
| | - Eva Galante
- †Institute of Nuclear Medicine, University College London, 235 Euston Road, T5, London NW1 2BU, U.K
| | - Thibault Gendron
- †Institute of Nuclear Medicine, University College London, 235 Euston Road, T5, London NW1 2BU, U.K
| | - Elena Yiannaki
- ‡Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K
| | - Niral Patel
- §Centre for Advanced Biomedical Imaging, University College London, 72 Huntley Street, London WC1E 6DD, U.K
| | - Tammy L Kalber
- §Centre for Advanced Biomedical Imaging, University College London, 72 Huntley Street, London WC1E 6DD, U.K
| | - Adam Badar
- §Centre for Advanced Biomedical Imaging, University College London, 72 Huntley Street, London WC1E 6DD, U.K
| | - Mathew Robson
- ∥Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, U.K
| | - Sean P Johnson
- ∥Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, U.K
| | - Florian Bauer
- ⊥Department of Medicinal Chemistry, Faculty of Life Sciences, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Severin Mairinger
- #Health and Environment Department, AIT Austrian Institute of Technology GmbH, A-2444 Seibersdorf, Austria
| | - Johann Stanek
- #Health and Environment Department, AIT Austrian Institute of Technology GmbH, A-2444 Seibersdorf, Austria
| | - Thomas Wanek
- #Health and Environment Department, AIT Austrian Institute of Technology GmbH, A-2444 Seibersdorf, Austria
| | - Claudia Kuntner
- #Health and Environment Department, AIT Austrian Institute of Technology GmbH, A-2444 Seibersdorf, Austria
| | - Tim Kottke
- ∇Institute of Pharmaceutical Chemistry, Biocenter, Johann Wolfgang Goethe University, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany
| | - Lilia Weizel
- ∇Institute of Pharmaceutical Chemistry, Biocenter, Johann Wolfgang Goethe University, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany
| | - David Dickens
- ○The Wolfson Centre for Personalised Medicine, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Block A Waterhouse Buildings, 1-5 Brownlow Street, Liverpool L69 3GL, U.K
| | - Kjell Erlandsson
- †Institute of Nuclear Medicine, University College London, 235 Euston Road, T5, London NW1 2BU, U.K
| | - Brian F Hutton
- †Institute of Nuclear Medicine, University College London, 235 Euston Road, T5, London NW1 2BU, U.K
| | - Mark F Lythgoe
- §Centre for Advanced Biomedical Imaging, University College London, 72 Huntley Street, London WC1E 6DD, U.K
| | - Holger Stark
- ∇Institute of Pharmaceutical Chemistry, Biocenter, Johann Wolfgang Goethe University, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany
| | - Oliver Langer
- ●Department of Clinical Pharmacology, Medical University of Vienna, Waehringer-Guertel 18-20, A-1090 Vienna, Austria
| | - Matthias Koepp
- ◆Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, U.K
| | - Erik Årstad
- †Institute of Nuclear Medicine, University College London, 235 Euston Road, T5, London NW1 2BU, U.K
- ‡Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K
| |
Collapse
|
14
|
Farré M, Pérez-Mañá C, Papaseit E, Menoyo E, Pérez M, Martin S, Bullich S, Rojas S, Herance JR, Trampal C, Labeaga L, Valiente R. Bilastine vs. hydroxyzine: occupation of brain histamine H1 -receptors evaluated by positron emission tomography in healthy volunteers. Br J Clin Pharmacol 2015; 78:970-80. [PMID: 24833043 PMCID: PMC4243871 DOI: 10.1111/bcp.12421] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 05/08/2014] [Indexed: 12/16/2022] Open
Abstract
AIM A close correlation exists between positron emission tomography (PET)-determined histamine H1 -receptor occupancy (H1 RO) and the incidence of sedation. Antihistamines with H1 RO <20% are classified as non-sedating. The objective was to compare the H1 RO of bilastine, a second generation antihistamine, with that of hydroxyzine. METHODS This randomized, double-blind, crossover study used PET imaging with [(11) C]-doxepin to evaluate H1 RO in 12 healthy males (mean age 26.2 years), after single oral administration of bilastine (20 mg), hydroxyzine (25 mg) or placebo. Binding potentials and H1 ROs were calculated in five cerebral cortex regions of interest: frontal, occipital, parietal, temporal, insula. Plasma bilastine concentrations, subjective sedation (visual analogue scale), objective psychomotor performance (digital symbol substitution test), physiological variables and safety (adverse events, AEs), were also evaluated. RESULTS The mean binding potential of all five regions of interest (total binding potential) was significantly greater with bilastine than hydroxyzine (mean value 0.26 vs. 0.13, P < 0.01; mean difference and 95% CI -0.130 [-0.155, 0.105]). There was no significant difference between bilastine and placebo. Overall H1 RO by bilastine was significantly lower than that by hydroxyzine (mean value -3.92% vs. 53.95%, P < 0.01; mean difference and 95% CI 57.870% [42.664%, 73.075%]). There was no significant linear relationship between individual bilastine plasma concentrations and total binding potential values. No significant between-treatment differences were observed for sedation and psychomotor performance. Twenty-six non-serious AEs were reported. Sleepiness or sedation was not reported with bilastine but appeared in some subjects with hydroxyzine. CONCLUSIONS A single oral dose of bilastine 20 mg had minimal H1 RO, was not associated with subjective sedation or objective impairment of psychomotor performance and was devoid of treatment-related sedative AEs, thus satisfying relevant subjective, objective and PET criteria as a non-sedating antihistamine.
Collapse
Affiliation(s)
- Magí Farré
- Human Pharmacology and Neuroscience Research Unit, Hospital del Mar Medical Research Institute-IMIM, and Universidad Autónoma de Barcelona-UAB, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Anoush M, Mohammad Khani MR. Evaluating the Anti-nociceptive and Anti-inflammatory Effects of Ketotifen and Fexofenadine in Rats. Adv Pharm Bull 2015; 5:217-22. [PMID: 26236660 DOI: 10.15171/apb.2015.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 07/20/2014] [Accepted: 07/21/2014] [Indexed: 11/09/2022] Open
Abstract
PURPOSE As H1 and H3 receptors' roles has been defined in peripheral pains in some papers and because histamine is known for its role in inflammatory responses; this study investigated the possible analgesic and anti-inflammatory effects of ketotifen and fexofenadine as relatively safe long acting anti histamines in both chronic chemical pain and acute inflammation in rats. METHODS In this study, male Sprague-Dawley rats weighing 225-250 grams were used. In order to evaluate the chemical chronic pain, sub-plantar injection of formalin applied and the pain scores were recorded every 15 seconds during 60 minutes. Carrageenan injection to the right hind paw was used for induction of acute inflammation and the paw edema was measured every 60 minutes for 4 hours. RESULTS Based on the results, both ketotifen and fexofenadine were able to significantly diminish chemical acute and chronic pain as well as inflammation in comparison with the control group and the effects were acceptable according to the standard treatment. Both effects for fexofenadine started later than those of ketotifen. CONCLUSION According to the outcomes of the study, ketotifen and fexofenadine demonstrated significant analgesic and anti-inflammatory characteristics in both models of chemical pain and acute inflammation in laboratory animals.
Collapse
Affiliation(s)
- Mahdieh Anoush
- Department of Pharmacology& Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Znjan, Iran. ; Applied Drug Research Center, Zanjan University of Medical Sciences, Znjan, Iran
| | | |
Collapse
|
16
|
Rankovic Z. CNS Drug Design: Balancing Physicochemical Properties for Optimal Brain Exposure. J Med Chem 2015; 58:2584-608. [DOI: 10.1021/jm501535r] [Citation(s) in RCA: 342] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Zoran Rankovic
- Eli Lilly and Company, 893 South
Delaware Street, Indianapolis, Indiana 46285, United States
| |
Collapse
|
17
|
Sjöstedt N, Kortejärvi H, Kidron H, Vellonen KS, Urtti A, Yliperttula M. Challenges of using in vitro data for modeling P-glycoprotein efflux in the blood-brain barrier. Pharm Res 2014; 31:1-19. [PMID: 23797466 DOI: 10.1007/s11095-013-1124-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 06/11/2013] [Indexed: 02/06/2023]
Abstract
The efficacy of central nervous system (CNS) drugs may be limited by their poor ability to cross the bloodbrain barrier (BBB). Transporters, such as p-glycoprotein, may affect the distribution of many drugs into the CNS in conjunction with the restricted paracellular pathway of the BBB. It is therefore important to gain information on unbound drug concentrations in the brain in drug development to ensure sufficient drug exposure from plasma at the target site in the CNS. In vitro methods are routinely used in drug development to study passive permeability and p-glycoprotein efflux of new drugs. This review discusses the challenges in the use of in vitro data as input parameters in physiologically based pharmacokinetic (PBPK) models of CNS drug disposition of p-glycoprotein substrates. Experience with quinidine demonstrates the variability in in vitro parameters of passive permeability and active pglycoprotein efflux. Further work is needed to generate parameter values that are independent of the model and assay. This is a prerequisite for reliable predictions of drug concentrations in the brain in vivo.
Collapse
|
18
|
Won CS, Lan T, VanderMolen KM, Dawson PA, Oberlies NH, Widmer WW, Scarlett YV, Paine MF. A modified grapefruit juice eliminates two compound classes as major mediators of the grapefruit juice-fexofenadine interaction: an in vitro-in vivo "connect". J Clin Pharmacol 2013; 53:982-90. [PMID: 23878024 PMCID: PMC4029847 DOI: 10.1002/jcph.136] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 06/18/2013] [Indexed: 02/02/2023]
Abstract
The grapefruit juice (GFJ)-fexofenadine interaction involves inhibition of intestinal organic anion transporting polypeptide (OATP)-mediated uptake. Only naringin has been shown clinically to inhibit intestinal OATP; other constituents have not been evaluated. The effects of a modified GFJ devoid of furanocoumarins (~99%) and polymethoxyflavones (~90%) on fexofenadine disposition were compared to effects of the original juice. Extracts of both juices inhibited estrone 3-sulfate and fexofenadine uptake by similar extents in OATP-transfected cells (~50% and ~25%, respectively). Healthy volunteers (n = 18) were administered fexofenadine (120 mg) with water, GFJ, or modified GFJ (240 mL) by randomized, three-way crossover design. Compared to water, both juices decreased fexofenadine geometric mean AUC and C(max) by ~25% (P ≤ .008 and P ≤ .011, respectively), with no effect on terminal half-life (P = .11). Similar effects by both juices on fexofenadine pharmacokinetics indicate furanocoumarins and polymethoxyflavones are not major mediators of the GFJ-fexofenadine interaction.
Collapse
Affiliation(s)
- Christina S. Won
- Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Tian Lan
- Department of Internal Medicine, Section on Gastroenterology, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Karen M. VanderMolen
- Department of Chemistry and Biochemistry, The University of North Carolina at Greensboro, Greensboro, NC
| | - Paul A. Dawson
- Department of Internal Medicine, Section on Gastroenterology, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Nicholas H. Oberlies
- Department of Chemistry and Biochemistry, The University of North Carolina at Greensboro, Greensboro, NC
| | - Wilbur W. Widmer
- Citrus and Subtropical Products Laboratory, United States Department of Agriculture, Winter Haven, FL
| | - Yolanda V. Scarlett
- School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Mary F. Paine
- Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
19
|
Hitchcock SA. Structural Modifications that Alter the P-Glycoprotein Efflux Properties of Compounds. J Med Chem 2012; 55:4877-95. [DOI: 10.1021/jm201136z] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
20
|
Yanai K, Rogala B, Chugh K, Paraskakis E, Pampura AN, Boev R. Safety considerations in the management of allergic diseases: focus on antihistamines. Curr Med Res Opin 2012; 28:623-42. [PMID: 22455874 DOI: 10.1185/03007995.2012.672405] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To conduct a systematic review of evidence supporting the safety profiles of frequently used oral H(1)-antihistamines (AHs) for the treatment of patients with histamine-release related allergic diseases, e.g. allergic rhinitis and urticaria, and to compare them to the safety profiles of other medications, mostly topical corticosteroids and leukotriene antagonists (LTRA). RESEARCH DESIGN AND METHODS Systematic search of the published literature (PubMed) and of the regulatory authorities databases (EMA and FDA) for oral AHs. RESULTS Similarly to histamine, antihistamines (AHs) have organ-specific efficacy and adverse effects. The peripheral H(1)-receptor (PrH1R) stimulation leads to allergic symptoms while the brain H(1)-receptor (BrH1R) blockade leads to somnolence, fatigue, increased appetite, decreased cognitive functions (impaired memory and learning), seizures, aggressive behaviour, etc. First-generation oral AHs (FGAHs) inhibit the effects of histamine not only peripherally but also in the brain, and additionally have potent antimuscarinic, anti-α-adrenergic and antiserotonin effects leading to symptoms such as visual disturbances (mydriasis, photophobia, and diplopia), dry mouth, tachycardia, constipation, urinary retention, agitation, and confusion. The somnolence caused by FGAHs interferes with the natural circadian sleep-wake cycle and therefore FGAHs are not suitable to be used as sleeping pills. Second-generation oral AHs (SGAHs) have proven better safety and tolerability profiles, much lower proportional impairment ratios, with at least similar if not better efficacy, than their predecessors. Only SGAHs, and especially those with a proven long-term (e.g., ≥12 months) clinical safety, should be prescribed for young children. Evidence exist that intranasally applied medications, like intranasal antihistamines, have the potential to reach the brain and cause somnolence. CONCLUSIONS Second-generation oral antihistamines are the preferred first-line treatment option for allergic rhinitis and urticaria. Patients taking SGAHs report relatively little and mild adverse events even after long-term continuous treatments. An antihistamine should ideally possess high selectivity for the H(1)-receptor, high PrH1R occupancy and low to no BrH1R occupancy.
Collapse
Affiliation(s)
- K Yanai
- Department of Pharmacology, Tohoku University School of Medicine, Sendai, Japan.
| | | | | | | | | | | |
Collapse
|
21
|
Wager TT, Liras JL, Mente S, Trapa P. Strategies to minimize CNS toxicity:in vitrohigh-throughput assays and computational modeling. Expert Opin Drug Metab Toxicol 2012; 8:531-42. [DOI: 10.1517/17425255.2012.677028] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
22
|
Improving the prediction of the brain disposition for orally administered drugs using BDDCS. Adv Drug Deliv Rev 2012; 64:95-109. [PMID: 22261306 DOI: 10.1016/j.addr.2011.12.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 12/08/2011] [Accepted: 12/12/2011] [Indexed: 01/16/2023]
Abstract
In modeling blood-brain barrier (BBB) passage, in silico models have yielded ~80% prediction accuracy, and are currently used in early drug discovery. Being derived from molecular structural information only, these models do not take into account the biological factors responsible for the in vivo outcome. Passive permeability and P-glycoprotein (Pgp, ABCB1) efflux have been successfully recognized to impact xenobiotic extrusion from the brain, as Pgp is known to play a role in limiting the BBB penetration of oral drugs in humans. However, these two properties alone fail to explain the BBB penetration for a significant number of marketed central nervous system (CNS) agents. The Biopharmaceutics Drug Disposition Classification System (BDDCS) has proved useful in predicting drug disposition in the human body, particularly in the liver and intestine. Here we discuss the value of using BDDCS to improve BBB predictions of oral drugs. BDDCS class membership was integrated with in vitro Pgp efflux and in silico permeability data to create a simple 3-step classification tree that accurately predicted CNS disposition for more than 90% of 153 drugs in our data set. About 98% of BDDCS class 1 drugs were found to markedly distribute throughout the brain; this includes a number of BDDCS class 1 drugs shown to be Pgp substrates. This new perspective provides a further interpretation of how Pgp influences the sedative effects of H1-histamine receptor antagonists.
Collapse
|
23
|
Eikel D, Vavrek M, Smith S, Bason C, Yeh S, Korfmacher WA, Henion JD. Liquid extraction surface analysis mass spectrometry (LESA-MS) as a novel profiling tool for drug distribution and metabolism analysis: the terfenadine example. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2011; 25:3587-96. [PMID: 22095508 DOI: 10.1002/rcm.5274] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Liquid extraction surface analysis mass spectrometry (LESA-MS) is a novel surface profiling technique that combines micro-liquid extraction from a solid surface with nano-electrospray mass spectrometry. One potential application is the examination of the distribution of drugs and their metabolites by analyzing ex vivo tissue sections, an area where quantitative whole body autoradiography (QWBA) is traditionally employed. However, QWBA relies on the use of radiolabeled drugs and is limited to total radioactivity measured whereas LESA-MS can provide drug- and metabolite-specific distribution information. Here, we evaluate LESA-MS, examining the distribution and biotransformation of unlabeled terfenadine in mice and compare our findings to QWBA, whole tissue LC/MS/MS and MALDI-MSI. The spatial resolution of LESA-MS can be optimized to ca. 1 mm on tissues such as brain, liver and kidney, also enabling drug profiling within a single organ. LESA-MS can readily identify the biotransformation of terfenadine to its major, active metabolite fexofenadine. Relative quantification can confirm the rapid absorption of terfendine after oral dosage, its extensive first pass metabolism and the distribution of both compounds into systemic tissues such as muscle, spleen and kidney. The elimination appears to be consistent with biliary excretion and only trace levels of fexofenadine could be confirmed in brain. We found LESA-MS to be more informative in terms of drug distribution than a comparable MALDI-MS imaging study, likely due to its favorable overall sensitivity due to the larger surface area sampled. LESA-MS appears to be a useful new profiling tool for examining the distribution of drugs and their metabolites in tissue sections.
Collapse
|
24
|
Padowski JM, Pollack GM. Influence of time to achieve substrate distribution equilibrium between brain tissue and blood on quantitation of the blood–brain barrier P-glycoprotein effect. Brain Res 2011; 1426:1-17. [DOI: 10.1016/j.brainres.2011.10.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 09/07/2011] [Accepted: 10/04/2011] [Indexed: 01/11/2023]
|
25
|
Sugiyama Y, Yamashita S. Impact of microdosing clinical study -- why necessary and how useful? Adv Drug Deliv Rev 2011; 63:494-502. [PMID: 20950660 DOI: 10.1016/j.addr.2010.09.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2010] [Revised: 09/17/2010] [Accepted: 09/21/2010] [Indexed: 01/07/2023]
Abstract
The microdose (MD) clinical study enables to select a "better" compound for new drug candidate that shows desirable PK profiles in human. This new methodology is highly expected to streamline the drug development and to increase the success probability in the clinical trial. Since only a small amount of the test compound (less than 100 μg) is administered, the risk of harmful events to a human subject is regarded as minimal in the MD clinical study. However, the low dose also incurs the arguments about the usefulness of this method, since it may result in different PK profiles of drugs from that at the therapeutic dose. In addition, information on the efficacy/safety of the test compound cannot be obtained from the MD clinical study. On the other hand, PBPK model analysis based on the data of both the MD clinical study and in vitro study on metabolism, transport and binding enables the accurate prediction of PK profiles in humans at the therapeutic dose. PET molecular imaging technology further enhances the usability and applicability of the MD clinical study by offering the information on efficacy/safety. These methodologies, if coordinated effectively, are expected to innovate the new drug discovery and development.
Collapse
Affiliation(s)
- Yuichi Sugiyama
- Laboratoryof Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyoku, Tokyo, 113-0033, Japan
| | | |
Collapse
|
26
|
Tsinman O, Tsinman K, Sun N, Avdeef A. Physicochemical selectivity of the BBB microenvironment governing passive diffusion--matching with a porcine brain lipid extract artificial membrane permeability model. Pharm Res 2011; 28:337-63. [PMID: 20945153 PMCID: PMC3034772 DOI: 10.1007/s11095-010-0280-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Accepted: 09/13/2010] [Indexed: 12/22/2022]
Abstract
PURPOSE To mimic the physicochemical selectivity of the blood-brain barrier (BBB) and to predict its passive permeability using a PAMPA model based on porcine brain lipid extract (PBLE 10%w/v in alkane). METHODS Three PAMPA (BD pre-coated and PBLE with 2 different lipid volumes) models were tested with 108 drugs. Abraham solvation descriptors were used to interpret the in vitro-in vivo correlation with 282 in situ brain perfusion measurements, spanning over 5 orders of magnitude. An in combo PAMPA model was developed from combining measured PAMPA permeability with one H-bond descriptor. RESULTS The in combo PAMPA predicted 93% of the variance of 197 largely efflux-inhibited in situ permeability training set. The model was cross-validated by the "leave-many-out" procedure, with q(2) = 0.92 ± 0.03. The PAMPA models indicated the presence of paramembrane water channels. Only the PBLE-based PAMPA-BBB model with sufficient lipid to fill all the internal pore space of the filter showed a wide dynamic range window, selectivity coefficient near 1, and was suitable for predicting BBB permeability. CONCLUSION BBB permeability can be predicted by in combo PAMPA. Its speed and substantially lower cost, compared to in vivo measurements, make it an attractive first-pass screening method for BBB passive permeability.
Collapse
Affiliation(s)
- Oksana Tsinman
- pION INC, 5 Constitution Way, Woburn, Massachusetts 01801, USA
| | | | | | | |
Collapse
|
27
|
Lappin G, Shishikura Y, Jochemsen R, Weaver RJ, Gesson C, Houston B, Oosterhuis B, Bjerrum OJ, Rowland M, Garner C. Pharmacokinetics of fexofenadine: Evaluation of a microdose and assessment of absolute oral bioavailability. Eur J Pharm Sci 2010; 40:125-31. [DOI: 10.1016/j.ejps.2010.03.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Revised: 02/27/2010] [Accepted: 03/12/2010] [Indexed: 12/01/2022]
|
28
|
Assessment of Blood–Brain Barrier Permeability Using the In Situ Mouse Brain Perfusion Technique. Pharm Res 2009; 26:1657-64. [DOI: 10.1007/s11095-009-9876-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Accepted: 03/16/2009] [Indexed: 01/16/2023]
|