51
|
Cressier D, Dhilly M, Cao Pham TT, Fillesoye F, Gourand F, Maïza A, Martins AF, Morfin JF, Geraldes CFGC, Tóth É, Barré L. Gallium-68 Complexes Conjugated to Pittsburgh Compound B: Radiolabeling and Biological Evaluation. Mol Imaging Biol 2017; 18:334-43. [PMID: 26543029 DOI: 10.1007/s11307-015-0906-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
PURPOSE The aim of this work is to develop an efficient and fully automated radiosynthesis of three derivatives of the Pittsburgh compound B labeled with gallium-68 for the detection of amyloid plaques. PROCEDURES The radiolabeling of the precursors and purification of the radiolabeled agents by high pressure liquid chromatography has been studied prior to their in vitro and in vivo evaluations. RESULTS The complete process led, in 50 min, to pure Ga-68 products in a 12-38 % yield and with appreciable specific radioactivity (SRA, 85-168 GBq/μmol) which enabled us to demonstrate a considerable in vivo stability of the products. Unfortunately, this result was associated with a poor blood-brain barrier (BBB) permeability and a limited uptake of our compounds by amyloid deposits was observed by in vitro autoradiography. CONCLUSION Although we have not yet identified a compound able to significantly mark cerebral amyloidosis, this present investigation will likely contribute to the development of more successful Ga-68 radiotracers.
Collapse
Affiliation(s)
- Damien Cressier
- CEA, I2BM, LDM-TEP, UMR 6301 ISTCT, GIP Cyceron, 14074, Caen, France. .,CNRS, UMR 6301 ISTCT, LDM-TEP, GIP Cyceron, 14074, Caen, France. .,Université de Caen Normandie, UMR 6301 ISTCT, LDM-TEP, GIP Cyceron, 14074, Caen, France.
| | - Martine Dhilly
- CEA, I2BM, LDM-TEP, UMR 6301 ISTCT, GIP Cyceron, 14074, Caen, France.,CNRS, UMR 6301 ISTCT, LDM-TEP, GIP Cyceron, 14074, Caen, France.,Université de Caen Normandie, UMR 6301 ISTCT, LDM-TEP, GIP Cyceron, 14074, Caen, France
| | - Thang T Cao Pham
- CEA, I2BM, LDM-TEP, UMR 6301 ISTCT, GIP Cyceron, 14074, Caen, France.,CNRS, UMR 6301 ISTCT, LDM-TEP, GIP Cyceron, 14074, Caen, France.,Université de Caen Normandie, UMR 6301 ISTCT, LDM-TEP, GIP Cyceron, 14074, Caen, France
| | - Fabien Fillesoye
- CEA, I2BM, LDM-TEP, UMR 6301 ISTCT, GIP Cyceron, 14074, Caen, France.,CNRS, UMR 6301 ISTCT, LDM-TEP, GIP Cyceron, 14074, Caen, France.,Université de Caen Normandie, UMR 6301 ISTCT, LDM-TEP, GIP Cyceron, 14074, Caen, France
| | - Fabienne Gourand
- CEA, I2BM, LDM-TEP, UMR 6301 ISTCT, GIP Cyceron, 14074, Caen, France.,CNRS, UMR 6301 ISTCT, LDM-TEP, GIP Cyceron, 14074, Caen, France.,Université de Caen Normandie, UMR 6301 ISTCT, LDM-TEP, GIP Cyceron, 14074, Caen, France
| | - Auriane Maïza
- CEA, I2BM, LDM-TEP, UMR 6301 ISTCT, GIP Cyceron, 14074, Caen, France.,CNRS, UMR 6301 ISTCT, LDM-TEP, GIP Cyceron, 14074, Caen, France.,Université de Caen Normandie, UMR 6301 ISTCT, LDM-TEP, GIP Cyceron, 14074, Caen, France
| | - André F Martins
- Centre de Biophysique Moléculaire UPR 4301, CNRS, Université d'Orléans, 45071, Orléans, France.,Department of Life Sciences and Coimbra Chemistry Center, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Jean-François Morfin
- Centre de Biophysique Moléculaire UPR 4301, CNRS, Université d'Orléans, 45071, Orléans, France
| | - Carlos F G C Geraldes
- Department of Life Sciences and Coimbra Chemistry Center, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Éva Tóth
- Centre de Biophysique Moléculaire UPR 4301, CNRS, Université d'Orléans, 45071, Orléans, France
| | - Louisa Barré
- CEA, I2BM, LDM-TEP, UMR 6301 ISTCT, GIP Cyceron, 14074, Caen, France.,CNRS, UMR 6301 ISTCT, LDM-TEP, GIP Cyceron, 14074, Caen, France.,Université de Caen Normandie, UMR 6301 ISTCT, LDM-TEP, GIP Cyceron, 14074, Caen, France
| |
Collapse
|
52
|
Ciura K, Belka M, Kawczak P, Bączek T, Markuszewski MJ, Nowakowska J. Combined computational-experimental approach to predict blood-brain barrier (BBB) permeation based on "green" salting-out thin layer chromatography supported by simple molecular descriptors. J Pharm Biomed Anal 2017. [PMID: 28641198 DOI: 10.1016/j.jpba.2017.05.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The objective of this paper is to build QSRR/QSAR model for predicting the blood-brain barrier (BBB) permeability. The obtained models are based on salting-out thin layer chromatography (SOTLC) constants and calculated molecular descriptors. Among chromatographic methods SOTLC was chosen, since the mobile phases are free of organic solvent. As consequences, there are less toxic, and have lower environmental impact compared to classical reserved phases liquid chromatography (RPLC). During the study three stationary phase silica gel, cellulose plates and neutral aluminum oxide were examined. The model set of solutes presents a wide range of log BB values, containing compounds which cross the BBB readily and molecules poorly distributed to the brain including drugs acting on the nervous system as well as peripheral acting drugs. Additionally, the comparison of three regression models: multiple linear regression (MLR), partial least-squares (PLS) and orthogonal partial least squares (OPLS) were performed. The designed QSRR/QSAR models could be useful to predict BBB of systematically synthesized newly compounds in the drug development pipeline and are attractive alternatives of time-consuming and demanding directed methods for log BB measurement. The study also shown that among several regression techniques, significant differences can be obtained in models performance, measured by R2 and Q2, hence it is strongly suggested to evaluate all available options as MLR, PLS and OPLS.
Collapse
Affiliation(s)
- Krzesimir Ciura
- Medical University of Gdańsk, Faculty of Pharmacy, Department of Physical Chemistry, Al. Gen. J. Hallera 107, 80-416, Gdańsk, Poland.
| | - Mariusz Belka
- Medical University of Gdansk, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| | - Piotr Kawczak
- Medical University of Gdansk, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| | - Tomasz Bączek
- Medical University of Gdansk, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| | - Michał J Markuszewski
- Medical University of Gdańsk, Faculty of Pharmacy, Department of Biopharmaceutics and Pharmacodynamics, Al. Gen. J. Hallera 107, PL 80-416, Gdańsk, Poland
| | - Joanna Nowakowska
- Medical University of Gdańsk, Faculty of Pharmacy, Department of Physical Chemistry, Al. Gen. J. Hallera 107, 80-416, Gdańsk, Poland
| |
Collapse
|
53
|
Xia H, Cheng Z, Cheng Y, Xu Y. Investigating the passage of tetramethylpyrazine-loaded liposomes across blood-brain barrier models in vitro and ex vivo. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 69:1010-7. [DOI: 10.1016/j.msec.2016.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 07/14/2016] [Accepted: 08/02/2016] [Indexed: 02/03/2023]
|
54
|
Multitarget-directed oxoisoaporphine derivatives: Anti-acetylcholinesterase, anti-β-amyloid aggregation and enhanced autophagy activity against Alzheimer’s disease. Bioorg Med Chem 2016; 24:6031-6039. [DOI: 10.1016/j.bmc.2016.09.061] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/23/2016] [Accepted: 09/24/2016] [Indexed: 02/01/2023]
|
55
|
Rabal O, Sánchez-Arias JA, Cuadrado-Tejedor M, de Miguel I, Pérez-González M, García-Barroso C, Ugarte A, Estella-Hermoso de Mendoza A, Sáez E, Espelosin M, Ursua S, Haizhong T, Wei W, Musheng X, Garcia-Osta A, Oyarzabal J. Design, Synthesis, and Biological Evaluation of First-in-Class Dual Acting Histone Deacetylases (HDACs) and Phosphodiesterase 5 (PDE5) Inhibitors for the Treatment of Alzheimer’s Disease. J Med Chem 2016; 59:8967-9004. [DOI: 10.1021/acs.jmedchem.6b00908] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
| | | | - Mar Cuadrado-Tejedor
- Anatomy Department, School of Medicine, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| | | | | | | | | | | | | | | | | | - Tan Haizhong
- WuXi Apptec (Tianjin) Co. Ltd.,
TEDA, No. 111 HuangHai Road, Fourth
Avenue, Tianjin 300456, PR China
| | - Wu Wei
- WuXi Apptec (Tianjin) Co. Ltd.,
TEDA, No. 111 HuangHai Road, Fourth
Avenue, Tianjin 300456, PR China
| | - Xu Musheng
- WuXi Apptec (Tianjin) Co. Ltd.,
TEDA, No. 111 HuangHai Road, Fourth
Avenue, Tianjin 300456, PR China
| | | | | |
Collapse
|
56
|
Li X, Wang H, Lu Z, Zheng X, Ni W, Zhu J, Fu Y, Lian F, Zhang N, Li J, Zhang H, Mao F. Development of Multifunctional Pyrimidinylthiourea Derivatives as Potential Anti-Alzheimer Agents. J Med Chem 2016; 59:8326-44. [DOI: 10.1021/acs.jmedchem.6b00636] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Xiaokang Li
- Shanghai
Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Huan Wang
- CAS
Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Zhengyu Lu
- Shanghai
Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Xinyu Zheng
- Shanghai
Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Wei Ni
- Shanghai
Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Jin Zhu
- Shanghai
Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Yan Fu
- CAS
Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Fulin Lian
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Naixia Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Jian Li
- Shanghai
Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| | - Haiyan Zhang
- CAS
Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Fei Mao
- Shanghai
Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| |
Collapse
|
57
|
Gómez-Vallejo V, Ugarte A, García-Barroso C, Cuadrado-Tejedor M, Szczupak B, Dopeso-Reyes IG, Lanciego JL, García-Osta A, Llop J, Oyarzabal J, Franco R. Pharmacokinetic investigation of sildenafil using positron emission tomography and determination of its effect on cerebrospinal fluid cGMP levels. J Neurochem 2016; 136:403-15. [PMID: 26641206 DOI: 10.1111/jnc.13454] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/17/2015] [Accepted: 11/20/2015] [Indexed: 11/29/2022]
Abstract
Sildenafil (Viagra) is a selective inhibitor of phosphodiesterase type 5 (PDE5), which degrades cyclic guanosine monophosphate to the linear nucleotide. Sildenafil is acutely used in erectile dysfunction and chronically in pulmonary hypertension. Evidence in the last decade shows that sildenafil may have potential as a therapeutic option for Alzheimer's disease or other neurodegenerative disorders. The purpose of this work was to explore whether sildenafil crosses the blood-brain barrier. Pharmacokinetic properties of sildenafil in rodents were investigated using (11) C-radiolabeling followed by in vivo positron emission tomography (PET) and ex vivo tissue dissection and gamma counting. PET results in rats suggest penetration into the central nervous system. Ex vivo data in perfused animals suggest that trapping of [(11) C]sildenafil within the cerebral vascular endothelium limits accumulation in the central nervous system parenchyma. Peroral sildenafil administration to Macaca fascicularis and subsequent chemical analysis of plasma and cerebrospinal fluid (CSF) using liquid chromatography coupled with tandem mass spectrometry showed that drug content in the CSF was high enough to achieve PDE5 inhibition, which was also demonstrated by the significant increases in CSF cyclic guanosine monophosphate levels. Central actions of sildenafil include both relaxation of the cerebral vasculature and inhibition of PDE5 in neurons and glia. This central action of sildenafil may underlie its efficacy in neuroprotection models, and may justify the continued search for a PDE5 ligand suitable for PET imaging. Sildenafil interacts with phosphodiesterase type 5 (PDE5) expressed in the endothelium and/or smooth muscle cells of brain vessels and also crosses the blood-brain barrier to interact with PDE5 expressed in brain cells. At therapeutic doses, the concentration of sildenafil in the cerebrospinal fluid (CSF) is high enough to inhibit PDE5 in the neural cells (neurons and glia). In turn, the concentration of cGMP likely increases in parenchymal cells and, as shown in this report, in the CSF. Read the Editorial Highlight for this article on page 220. Cover Image for this issue: doi: 10.1111/jnc.13302.
Collapse
Affiliation(s)
| | - Ana Ugarte
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Carolina García-Barroso
- Neurobiology of Alzheimer's disease, Neurosciences Division, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Mar Cuadrado-Tejedor
- Neurobiology of Alzheimer's disease, Neurosciences Division, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Anatomy Department, School of Medicine, University of Navarra, Pamplona, Spain
| | | | - Iria G Dopeso-Reyes
- Neurosciences Division, Centre for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Pamplona, Spain
| | - José L Lanciego
- Neurosciences Division, Centre for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Pamplona, Spain.,Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Pamplona, Spain
| | - Ana García-Osta
- Neurobiology of Alzheimer's disease, Neurosciences Division, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Jordi Llop
- Radiochemistry and Nuclear Imaging Group, CIC biomaGUNE, San Sebastian, Spain
| | - Julen Oyarzabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Rafael Franco
- Neurobiology of Alzheimer's disease, Neurosciences Division, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| |
Collapse
|
58
|
Patel YT, Jacus MO, Davis AD, Boulos N, Turner DC, Vuppala PK, Freeman BB, Gilbertson RJ, Stewart CF. Simvastatin Hydroxy Acid Fails to Attain Sufficient Central Nervous System Tumor Exposure to Achieve a Cytotoxic Effect: Results of a Preclinical Cerebral Microdialysis Study. Drug Metab Dispos 2016; 44:591-4. [PMID: 26802130 PMCID: PMC4810761 DOI: 10.1124/dmd.115.068445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/21/2016] [Indexed: 11/22/2022] Open
Abstract
3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors were potent hits against a mouse ependymoma cell line, but their effectiveness against central nervous system tumors will depend on their ability to cross the blood-brain barrier and attain a sufficient exposure at the tumor. Among 3-hydroxy-3-methylglutaryl coenzyme A inhibitors that had activity in vitro, we prioritized simvastatin (SV) as the lead compound for preclinical pharmacokinetic studies based on its potential for central nervous system penetration as determined from in silico models. Furthermore, we performed systemic plasma disposition and cerebral microdialysis studies of SV (100 mg/kg, p.o.) in a murine model of ependymoma to characterize plasma and tumor extracellular fluid (tECF) pharmacokinetic properties. The murine dosage of SV (100 mg/kg, p.o.) was equivalent to the maximum tolerated dose in patients (7.5 mg/kg, p.o.) based on equivalent plasma exposure of simvastatin acid (SVA) between the two species. SV is rapidly metabolized in murine plasma with 15 times lower exposure compared with human plasma. SVA exposure in tECF was <33.8 ± 11.9 µg/l per hour, whereas the tumor to plasma partition coefficient of SVA was <0.084 ± 0.008. Compared with in vitro washout IC50 values, we did not achieve sufficient exposure of SVA in tECF to suggest tumor growth inhibition; therefore, SV was not carried forward in subsequent preclinical efficacy studies.
Collapse
Affiliation(s)
- Yogesh T Patel
- Departments of Pharmaceutical Sciences (Y.T.P., M.O.J., A.D.D., D.C.T. C.F.S.), Hematology (N.B.), and Developmental Neurobiology (R.J.G.), Preclinical Pharmacokinetic Shared Resource (P.K.V., B.B.F.), St. Jude Children's Research Hospital, Memphis, Tennessee; Merck, Rahway, New Jersey (D.C.T.); and Cambridge Cancer Centre, Cambridge, United Kingdom (R.J.G.)
| | - Megan O Jacus
- Departments of Pharmaceutical Sciences (Y.T.P., M.O.J., A.D.D., D.C.T. C.F.S.), Hematology (N.B.), and Developmental Neurobiology (R.J.G.), Preclinical Pharmacokinetic Shared Resource (P.K.V., B.B.F.), St. Jude Children's Research Hospital, Memphis, Tennessee; Merck, Rahway, New Jersey (D.C.T.); and Cambridge Cancer Centre, Cambridge, United Kingdom (R.J.G.)
| | - Abigail D Davis
- Departments of Pharmaceutical Sciences (Y.T.P., M.O.J., A.D.D., D.C.T. C.F.S.), Hematology (N.B.), and Developmental Neurobiology (R.J.G.), Preclinical Pharmacokinetic Shared Resource (P.K.V., B.B.F.), St. Jude Children's Research Hospital, Memphis, Tennessee; Merck, Rahway, New Jersey (D.C.T.); and Cambridge Cancer Centre, Cambridge, United Kingdom (R.J.G.)
| | - Nidal Boulos
- Departments of Pharmaceutical Sciences (Y.T.P., M.O.J., A.D.D., D.C.T. C.F.S.), Hematology (N.B.), and Developmental Neurobiology (R.J.G.), Preclinical Pharmacokinetic Shared Resource (P.K.V., B.B.F.), St. Jude Children's Research Hospital, Memphis, Tennessee; Merck, Rahway, New Jersey (D.C.T.); and Cambridge Cancer Centre, Cambridge, United Kingdom (R.J.G.)
| | - David C Turner
- Departments of Pharmaceutical Sciences (Y.T.P., M.O.J., A.D.D., D.C.T. C.F.S.), Hematology (N.B.), and Developmental Neurobiology (R.J.G.), Preclinical Pharmacokinetic Shared Resource (P.K.V., B.B.F.), St. Jude Children's Research Hospital, Memphis, Tennessee; Merck, Rahway, New Jersey (D.C.T.); and Cambridge Cancer Centre, Cambridge, United Kingdom (R.J.G.)
| | - Pradeep K Vuppala
- Departments of Pharmaceutical Sciences (Y.T.P., M.O.J., A.D.D., D.C.T. C.F.S.), Hematology (N.B.), and Developmental Neurobiology (R.J.G.), Preclinical Pharmacokinetic Shared Resource (P.K.V., B.B.F.), St. Jude Children's Research Hospital, Memphis, Tennessee; Merck, Rahway, New Jersey (D.C.T.); and Cambridge Cancer Centre, Cambridge, United Kingdom (R.J.G.)
| | - Burgess B Freeman
- Departments of Pharmaceutical Sciences (Y.T.P., M.O.J., A.D.D., D.C.T. C.F.S.), Hematology (N.B.), and Developmental Neurobiology (R.J.G.), Preclinical Pharmacokinetic Shared Resource (P.K.V., B.B.F.), St. Jude Children's Research Hospital, Memphis, Tennessee; Merck, Rahway, New Jersey (D.C.T.); and Cambridge Cancer Centre, Cambridge, United Kingdom (R.J.G.)
| | - Richard J Gilbertson
- Departments of Pharmaceutical Sciences (Y.T.P., M.O.J., A.D.D., D.C.T. C.F.S.), Hematology (N.B.), and Developmental Neurobiology (R.J.G.), Preclinical Pharmacokinetic Shared Resource (P.K.V., B.B.F.), St. Jude Children's Research Hospital, Memphis, Tennessee; Merck, Rahway, New Jersey (D.C.T.); and Cambridge Cancer Centre, Cambridge, United Kingdom (R.J.G.)
| | - Clinton F Stewart
- Departments of Pharmaceutical Sciences (Y.T.P., M.O.J., A.D.D., D.C.T. C.F.S.), Hematology (N.B.), and Developmental Neurobiology (R.J.G.), Preclinical Pharmacokinetic Shared Resource (P.K.V., B.B.F.), St. Jude Children's Research Hospital, Memphis, Tennessee; Merck, Rahway, New Jersey (D.C.T.); and Cambridge Cancer Centre, Cambridge, United Kingdom (R.J.G.)
| |
Collapse
|
59
|
Bicker J, Alves G, Fortuna A, Soares-da-Silva P, Falcão A. A new PAMPA model using an in-house brain lipid extract for screening the blood-brain barrier permeability of drug candidates. Int J Pharm 2016; 501:102-11. [PMID: 26836708 DOI: 10.1016/j.ijpharm.2016.01.074] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 01/27/2016] [Accepted: 01/28/2016] [Indexed: 01/16/2023]
Abstract
The determination of the permeability of drug candidates across the blood-brain barrier (BBB) is a fundamental step during drug discovery programs. The parallel artificial membrane permeability assay (PAMPA) is a high throughput screening tool applied to evaluate the passive permeability and adapted to predict BBB penetration. Herein, a new PAMPA model was developed using an in-house brain lipid extract capable of discriminating BBB permeable from non-permeable compounds. The apparent permeability (Papp) of 18 reference molecules and 10 test compounds was assessed and compared with phosphatidylcholine and commercial porcine polar brain lipid (PBL). The physicochemical selectivity of the in-house brain lipid extract was demonstrated by correlating Papp values with physicochemical properties and its predictive capacity estimated by establishing in vitro-in vivo correlations. The strong correlations achieved between 2% (w/v) in-house lipid extract and PBL for reference (r(2)=0.77) and test compounds (r(2)=0.94) support an equivalent discriminatory capacity and validate the presented model. Moreover, PAMPA studies performed with PBL and in-house lipid extract exhibited a higher correlation with the in vivo parameter logBB (r(2)=0.76 and r(2)=0.72, respectively) than phosphatidylcholine (r(2)=0.51). Overall, the applied lipid extraction process was reproducible, economical and provided lipid extracts that can be used to reliably assess BBB permeation.
Collapse
Affiliation(s)
- Joana Bicker
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Gilberto Alves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Patrício Soares-da-Silva
- Department of Research and Development, BIAL, Av. da Siderurgia Nacional, 4745- 457, S. Mamede do Coronado, Portugal; Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Amílcar Falcão
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal.
| |
Collapse
|
60
|
Insights From an Integrated Physiologically Based Pharmacokinetic Model for Brain Penetration. J Pharm Sci 2016; 105:965-971. [DOI: 10.1016/j.xphs.2015.12.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/05/2015] [Accepted: 10/20/2015] [Indexed: 01/01/2023]
|
61
|
Associating a negatively charged GdDOTA-derivative to the Pittsburgh compound B for targeting Aβ amyloid aggregates. J Biol Inorg Chem 2015; 21:83-99. [DOI: 10.1007/s00775-015-1316-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/13/2015] [Indexed: 01/26/2023]
|
62
|
Isomeric methoxy analogs of nimesulide for development of brain cyclooxygense-2 (COX-2)-targeted imaging agents: Synthesis, in vitro COX-2-inhibitory potency, and cellular transport properties. Bioorg Med Chem 2015; 23:6807-14. [DOI: 10.1016/j.bmc.2015.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/03/2015] [Accepted: 10/05/2015] [Indexed: 12/23/2022]
|
63
|
Jiang L, Chen J, He Y, Zhang Y, Li G. A method to predict different mechanisms for blood-brain barrier permeability of CNS activity compounds in Chinese herbs using support vector machine. J Bioinform Comput Biol 2015; 14:1650005. [PMID: 26632324 DOI: 10.1142/s0219720016500050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The blood-brain barrier (BBB), a highly selective barrier between central nervous system (CNS) and the blood stream, restricts and regulates the penetration of compounds from the blood into the brain. Drugs that affect the CNS interact with the BBB prior to their target site, so the prediction research on BBB permeability is a fundamental and significant research direction in neuropharmacology. In this study, we combed through the available data and then with the help of support vector machine (SVM), we established an experiment process for discovering potential CNS compounds and investigating the mechanisms of BBB permeability of them to advance the research in this field four types of prediction models, referring to CNS activity, BBB permeability, passive diffusion and efflux transport, were obtained in the experiment process. The first two models were used to discover compounds which may have CNS activity and also cross the BBB at the same time; the latter two were used to elucidate the mechanism of BBB permeability of those compounds. Three optimization parameter methods, Grid Search, Genetic Algorithm (GA), and Particle Swarm Optimization (PSO), were used to optimize the SVM models. Then, four optimal models were selected with excellent evaluation indexes (the accuracy, sensitivity and specificity of each model were all above 85%). Furthermore, discrimination models were utilized to study the BBB properties of the known CNS activity compounds in Chinese herbs and this may guide the CNS drug development. With the relatively systematic and quick approach, the application rationality of traditional Chinese medicines for treating nervous system disease in the clinical practice will be improved.
Collapse
Affiliation(s)
- Ludi Jiang
- 1 School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, P. R. China
| | - Jiahua Chen
- 1 School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, P. R. China
| | - Yusu He
- 1 School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, P. R. China
| | - Yanling Zhang
- 1 School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, P. R. China
| | - Gongyu Li
- 1 School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, P. R. China
| |
Collapse
|
64
|
In vitro prediction of human intestinal absorption and blood–brain barrier partitioning: development of a lipid analog for micellar liquid chromatography. Anal Bioanal Chem 2015; 407:7453-66. [DOI: 10.1007/s00216-015-8911-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/07/2015] [Accepted: 07/08/2015] [Indexed: 10/23/2022]
|
65
|
Brito-Sánchez Y, Marrero-Ponce Y, Barigye SJ, Yaber-Goenaga I, Morell Pérez C, Le-Thi-Thu H, Cherkasov A. Towards Better BBB Passage Prediction Using an Extensive and Curated Data Set. Mol Inform 2015; 34:308-30. [PMID: 27490276 DOI: 10.1002/minf.201400118] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 01/20/2015] [Indexed: 12/25/2022]
Abstract
In the present report, the challenging task of drug delivery across the blood-brain barrier (BBB) is addressed via a computational approach. The BBB passage was modeled using classification and regression schemes on a novel extensive and curated data set (the largest to the best of our knowledge) in terms of log BB. Prior to the model development, steps of data analysis that comprise chemical data curation, structural, cutoff and cluster analysis (CA) were conducted. Linear Discriminant Analysis (LDA) and Multiple Linear Regression (MLR) were used to fit classification and correlation functions. The best LDA-based model showed overall accuracies over 85 % and 83 % for the training and test sets, respectively. Also a MLR-based model with acceptable explanation of more than 69 % of the variance in the experimental log BB was developed. A brief and general interpretation of proposed models allowed the estimation on how 'near' our computational approach is to the factors that determine the passage of molecules through the BBB. In a final effort some popular and powerful Machine Learning methods were considered. Comparable or similar performance was observed respect to the simpler linear techniques. Most of the compounds with anomalous behavior were put aside into a set denoted as controversial set and discussion regarding to these compounds is provided. Finally, our results were compared with methodologies previously reported in the literature showing comparable to better results. The results could represent useful tools available and reproducible by all scientific community in the early stages of neuropharmaceutical drug discovery/development projects.
Collapse
Affiliation(s)
- Yoan Brito-Sánchez
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada.,Unit of Computer-Aided Molecular "Biosilico" Discovery and Bioinformatic Research, International Network (CAMD-BIR International Network), Los Laureles L76MD, Nuevo Bosque, 130015, Cartagena de Indias, Bolivar, Colombia. Homepage: http://www.uv.es/yoma/ Homepage: http://sites.google.com/site/ymponce/home
| | - Yovani Marrero-Ponce
- Unit of Computer-Aided Molecular "Biosilico" Discovery and Bioinformatic Research, International Network (CAMD-BIR International Network), Los Laureles L76MD, Nuevo Bosque, 130015, Cartagena de Indias, Bolivar, Colombia. Homepage: http://www.uv.es/yoma/ Homepage: http://sites.google.com/site/ymponce/home. .,Grupo de Investigación en Estudios Químicos y Biológicos, Facultad de Ciencias Básicas, Universidad Tecnológica de Bolívar, Parque Industrial y Tecnológico Carlos Vélez Pombo Km 1 vía Turbaco, 130010, Cartagena de Indias, Bolívar, Colombia. .,Facultad de Química Farmacéutica, Universidad de Cartagena, Cartagena de Indias, Bolívar, Colombia.
| | - Stephen J Barigye
- Unit of Computer-Aided Molecular "Biosilico" Discovery and Bioinformatic Research, International Network (CAMD-BIR International Network), Los Laureles L76MD, Nuevo Bosque, 130015, Cartagena de Indias, Bolivar, Colombia. Homepage: http://www.uv.es/yoma/ Homepage: http://sites.google.com/site/ymponce/home.,Department of Chemistry, Federal University of Lavras, P.O. Box 3037, 37200-000, Lavras, MG, Brazil
| | - Iván Yaber-Goenaga
- Grupo de Investigación en Estudios Químicos y Biológicos, Facultad de Ciencias Básicas, Universidad Tecnológica de Bolívar, Parque Industrial y Tecnológico Carlos Vélez Pombo Km 1 vía Turbaco, 130010, Cartagena de Indias, Bolívar, Colombia
| | - Carlos Morell Pérez
- Center of Studies on Informatics, Universidad "Marta Abreu" de Las Villas, Santa Clara, 54830, Villa Clara, Cuba
| | - Huong Le-Thi-Thu
- School of Medicine and Pharmacy, Vietnam National University, Hanoi (VNU) 144 Xuan Thuy, CauGiay, Hanoi, Vietnam
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, V6H 3Z6, Canada
| |
Collapse
|
66
|
Hexum JK, Becker CM, Kempema AM, Ohlfest JR, Largaespada DA, Harki DA. Parthenolide prodrug LC-1 slows growth of intracranial glioma. Bioorg Med Chem Lett 2015; 25:2493-5. [PMID: 25978958 DOI: 10.1016/j.bmcl.2015.04.058] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 04/16/2015] [Accepted: 04/20/2015] [Indexed: 11/25/2022]
Abstract
LC-1 (also known as DMAPT or dimethylamino-parthenolide), a prodrug of parthenolide, was tested for anti-proliferative activity against glioma. LC-1 was found to have low micromolar cytotoxic activity against three glioma cell lines and was also found to be brain penetrant in healthy mice (2.1-3.0 brain-to-plasma ratio). In a syngeneic GL261 murine glioma model, LC-1 slowed tumor growth kinetics and extended the survival time of tumor-bearing mice in comparison to the vehicle control. Consequently, LC-1 represents a promising lead compound for further development as a glioma therapy.
Collapse
Affiliation(s)
- Joseph K Hexum
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Chani M Becker
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Aaron M Kempema
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - John R Ohlfest
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - David A Largaespada
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel A Harki
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
67
|
Bujak R, Struck-Lewicka W, Kaliszan M, Kaliszan R, Markuszewski MJ. Blood–brain barrier permeability mechanisms in view of quantitative structure–activity relationships (QSAR). J Pharm Biomed Anal 2015; 108:29-37. [DOI: 10.1016/j.jpba.2015.01.046] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/22/2015] [Accepted: 01/23/2015] [Indexed: 01/16/2023]
|
68
|
Le Douaron G, Schmidt F, Amar M, Kadar H, Debortoli L, Latini A, Séon-Méniel B, Ferrié L, Michel PP, Touboul D, Brunelle A, Raisman-Vozari R, Figadère B. Neuroprotective effects of a brain permeant 6-aminoquinoxaline derivative in cell culture conditions that model the loss of dopaminergic neurons in Parkinson disease. Eur J Med Chem 2015; 89:467-79. [DOI: 10.1016/j.ejmech.2014.10.067] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 10/22/2014] [Accepted: 10/23/2014] [Indexed: 12/26/2022]
|
69
|
Filipe HAL, Salvador A, Silvestre JM, Vaz WLC, Moreno MJ. Beyond Overton’s Rule: Quantitative Modeling of Passive Permeation through Tight Cell Monolayers. Mol Pharm 2014; 11:3696-706. [DOI: 10.1021/mp500437e] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- H. A. L. Filipe
- Centro de Química de Coimbra, Largo D. Dinis, Rua Larga, 3004-535 Coimbra, Portugal
- Centro
de Neurociências e Biologia Celular, Universidade de Coimbra, 3000-214 Coimbra, Portugal
| | - A. Salvador
- Centro
de Neurociências e Biologia Celular, Universidade de Coimbra, 3000-214 Coimbra, Portugal
- Chemistry
Department, FCTUC, Largo D. Dinis, Rua Larga, 3004-535 Coimbra, Portugal
| | - J. M. Silvestre
- Centro
de Neurociências e Biologia Celular, Universidade de Coimbra, 3000-214 Coimbra, Portugal
| | - W. L. C. Vaz
- Centro de Química de Coimbra, Largo D. Dinis, Rua Larga, 3004-535 Coimbra, Portugal
- Chemistry
Department, FCTUC, Largo D. Dinis, Rua Larga, 3004-535 Coimbra, Portugal
| | - M. J. Moreno
- Centro de Química de Coimbra, Largo D. Dinis, Rua Larga, 3004-535 Coimbra, Portugal
- Centro
de Neurociências e Biologia Celular, Universidade de Coimbra, 3000-214 Coimbra, Portugal
- Chemistry
Department, FCTUC, Largo D. Dinis, Rua Larga, 3004-535 Coimbra, Portugal
| |
Collapse
|
70
|
De Vrieze M, Verzele D, Szucs R, Sandra P, Lynen F. Evaluation of sphingomyelin, cholester, and phosphatidylcholine-based immobilized artificial membrane liquid chromatography to predict drug penetration across the blood-brain barrier. Anal Bioanal Chem 2014; 406:6179-88. [PMID: 25124450 DOI: 10.1007/s00216-014-8054-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/17/2014] [Accepted: 07/18/2014] [Indexed: 11/28/2022]
Abstract
Over the past decades, several in vitro methods have been tested for their ability to predict drug penetration across the blood-brain barrier. So far, in high-performance liquid chromatography, most attention has been paid to micellar liquid chromatography and immobilized artificial membrane (IAM) LC. IAMLC has been described as a viable approach, since the stationary phase emulates the lipid environment of a cell membrane. However, research in IAMLC has almost exclusively been limited to phosphatidylcholine (PC)-based stationary phases, even though PC is only one of the lipids present in cell membranes. In this article, sphingomyelin and cholester stationary phases have been tested for the first time towards their ability to predict drug penetration across the blood-brain barrier. Upon comparison with the PC stationary phase, the sphingomyelin- and cholester-based columns depict similar predictive performance. Combining data from the different stationary phases did not lead to improvements of the models.
Collapse
Affiliation(s)
- Mike De Vrieze
- Separation Science Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4bis, 9000, Ghent, Belgium
| | | | | | | | | |
Collapse
|
71
|
Andersen CA, Perfetti P, Nibbio M, Bellini M, Angelini R, Fornasier M. Brain penetration assessment in vivo: A reliable and simple method in anesthetized rats at steady state. J Neurosci Methods 2014; 232:199-206. [DOI: 10.1016/j.jneumeth.2014.04.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 03/25/2014] [Accepted: 04/10/2014] [Indexed: 10/25/2022]
|
72
|
Andersson O, Badisco L, Hansen AH, Hansen SH, Hellman K, Nielsen PA, Olsen LR, Verdonck R, Abbott NJ, Vanden Broeck J, Andersson G. Characterization of a novel brain barrier ex vivo insect-based P-glycoprotein screening model. Pharmacol Res Perspect 2014; 2:e00050. [PMID: 25505597 PMCID: PMC4186439 DOI: 10.1002/prp2.50] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 01/16/2023] Open
Abstract
In earlier studies insects were proposed as suitable models for vertebrate blood–brain barrier (BBB) permeability prediction and useful in early drug discovery. Here we provide transcriptome and functional data demonstrating the presence of a P-glycoprotein (Pgp) efflux transporter in the brain barrier of the desert locust (Schistocerca gregaria). In an in vivo study on the locust, we found an increased uptake of the two well-known Pgp substrates, rhodamine 123 and loperamide after co-administration with the Pgp inhibitors cyclosporine A or verapamil. Furthermore, ex vivo studies on isolated locust brains demonstrated differences in permeation of high and low permeability compounds. The vertebrate Pgp inhibitor verapamil did not affect the uptake of passively diffusing compounds but significantly increased the brain uptake of Pgp substrates in the ex vivo model. In addition, studies at 2°C and 30°C showed differences in brain uptake between Pgp-effluxed and passively diffusing compounds. The transcriptome data show a high degree of sequence identity of the locust Pgp transporter protein sequences to the human Pgp sequence (37%), as well as the presence of conserved domains. As in vertebrates, the locust brain–barrier function is morphologically confined to one specific cell layer and by using a whole-brain ex vivo drug exposure technique our locust model may retain the major cues that maintain and modulate the physiological function of the brain barrier. We show that the locust model has the potential to act as a robust and convenient model for assessing BBB permeability in early drug discovery.
Collapse
Affiliation(s)
- Olga Andersson
- EntomoPharm, R&D Medicon Village, S-223 81, Lund, Sweden
| | - Liesbeth Badisco
- Department of Animal Physiology and Neurobiology, Katholieke Universiteit Leuven Leuven, Belgium
| | | | - Steen Honoré Hansen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen DK-2100, Copenhagen, Denmark
| | - Karin Hellman
- EntomoPharm, R&D Medicon Village, S-223 81, Lund, Sweden
| | | | - Line Rørbæk Olsen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen DK-2100, Copenhagen, Denmark
| | - Rik Verdonck
- Department of Animal Physiology and Neurobiology, Katholieke Universiteit Leuven Leuven, Belgium
| | - N Joan Abbott
- BBB Group, Institute of Pharmaceutical Science, King's College London Franklin Wilkins Building, London, SE1 9NH, United Kingdom
| | - Jozef Vanden Broeck
- Department of Animal Physiology and Neurobiology, Katholieke Universiteit Leuven Leuven, Belgium
| | | |
Collapse
|
73
|
Spreafico M, Jacobson MP. In silico prediction of brain exposure: drug free fraction, unbound brain to plasma concentration ratio and equilibrium half-life. Curr Top Med Chem 2014; 13:813-20. [PMID: 23578025 DOI: 10.2174/1568026611313070004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 01/22/2013] [Accepted: 01/01/2013] [Indexed: 01/14/2023]
Abstract
The focus of CNS drug pharmacokinetics programs has recently shifted from determining the total concentrations in brain and blood to considering also unbound fractions and concentrations. Unfortunately, assessing unbound brain exposure experimentally requires demanding in vivo and in vitro studies. We propose a physical model, based on lipid binding and pH partitioning, to predict in silico the unbound volume of distribution in the brain. The model takes into account the partition of a drug into lipids, interstitial fluid and intracellular compartments of the brain. The results are in good agreement with the experimental data, suggesting that the contributions of lipid binding and pH partitioning are important in determining drug exposure in brain. The predicted values are used, together with predictions for plasma protein binding, as corrective terms in a second model to derive the unbound brain to plasma concentration ratio starting from experimental values of total concentration ratio. The calculated values of brain free fraction and passive permeability are also used to qualitatively determine the brain to plasma equilibration time in a model that shows promising results but is limited to a very small set of compounds. The models we propose are a step forward in understanding and predicting pharmacologically relevant exposure in brain starting from compounds' chemical structure and neuropharmacokinetics, by using experimental total brain to plasma ratios, in silico calculated properties and simple physics-based approaches. The models can be used in central nervous system drug discovery programs for a fast and cheap assessment of unbound brain exposure. For existing compounds, the unbound ratios can be derived from experimental values of total brain to plasma ratios. For both existing and hypothetical compounds, the unbound volume of distribution due to lipid binding and pH partitioning can be calculated starting only from the chemical structure.
Collapse
Affiliation(s)
- Morena Spreafico
- Department of Pharmaceutical Chemistry, University of California, San Francisco, 1700 4th Street, San Francisco, CA 94158, USA
| | | |
Collapse
|
74
|
Bicker J, Alves G, Fortuna A, Falcão A. Blood-brain barrier models and their relevance for a successful development of CNS drug delivery systems: a review. Eur J Pharm Biopharm 2014; 87:409-32. [PMID: 24686194 DOI: 10.1016/j.ejpb.2014.03.012] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2013] [Revised: 03/13/2014] [Accepted: 03/20/2014] [Indexed: 02/05/2023]
Abstract
During the research and development of new drugs directed at the central nervous system, there is a considerable attrition rate caused by their hampered access to the brain by the blood-brain barrier. Throughout the years, several in vitro models have been developed in an attempt to mimic critical functionalities of the blood-brain barrier and reliably predict the permeability of drug candidates. However, the current challenge lies in developing a model that retains fundamental blood-brain barrier characteristics and simultaneously remains compatible with the high throughput demands of pharmaceutical industries. This review firstly describes the roles of all elements of the neurovascular unit and their influence on drug brain penetration. In vitro models, including non-cell based and cell-based models, and in vivo models are herein presented, with a particular emphasis on their methodological aspects. Lastly, their contribution to the improvement of brain drug delivery strategies and drug transport across the blood-brain barrier is also discussed.
Collapse
Affiliation(s)
- Joana Bicker
- Laboratory of Pharmacology, University of Coimbra, Coimbra, Portugal; CNC - Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Gilberto Alves
- CNC - Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| | - Ana Fortuna
- Laboratory of Pharmacology, University of Coimbra, Coimbra, Portugal; CNC - Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Amílcar Falcão
- Laboratory of Pharmacology, University of Coimbra, Coimbra, Portugal; CNC - Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
75
|
Raevsky O, Solodova S, Lagunin A, Poroikov V. Computer modeling of blood brain barrier permeability of physiologically active compounds. ACTA ACUST UNITED AC 2014; 60:161-81. [DOI: 10.18097/pbmc20146002161] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
At present work discusses the current level of computer modeling the relationship structure of organic compounds and drugs and their ability to penetrate the BBB. All descriptors that influence to this permeability within classification and regression QSAR models are generalized and analyzed. The crucial role of H-bond in processes both passive, and active transport across BBB is observed. It is concluded that further research should be focused on interpretation the spatial structure of a full-size P-glycoprotein molecule with high resolution and the creation of QSAR models describing the quantitative relationship between structure and active transport of substances across BBB.
Collapse
Affiliation(s)
- O.A. Raevsky
- Institute of Physiologically Active Compounds, Russian Academy of Science
| | - S.L. Solodova
- Institute of Physiologically Active Compounds, Russian Academy of Science
| | - A.A. Lagunin
- Orekhovich Institute of Biomedical Chemistry of Russian Academy of Medical Sciences
| | - V.V. Poroikov
- Orekhovich Institute of Biomedical Chemistry of Russian Academy of Medical Sciences
| |
Collapse
|
76
|
Martins AF, Morfin JF, Geraldes CFGC, Tóth É. Gd3+ complexes conjugated to Pittsburgh compound B: potential MRI markers of β-amyloid plaques. J Biol Inorg Chem 2013; 19:281-95. [DOI: 10.1007/s00775-013-1055-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 10/09/2013] [Indexed: 11/29/2022]
|
77
|
Alata W, Paris-Robidas S, Emond V, Bourasset F, Calon F. Brain uptake of a fluorescent vector targeting the transferrin receptor: a novel application of in situ brain perfusion. Mol Pharm 2013; 11:243-53. [PMID: 24215184 DOI: 10.1021/mp400421a] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Monoclonal antibodies (mAbs) targeting blood-brain barrier (BBB) transporters are being developed for brain drug targeting. However, brain uptake quantification remains a challenge, particularly for large compounds, and often requires the use of radioactivity. In this work, we adapted an in situ brain perfusion technique for a fluorescent mAb raised against the mouse transferrin receptor (TfR) (clone Ri7). We first confirmed in vitro that the internalization of fluorolabeled Ri7 mAbs is saturable and dependent on the TfR in N2A and bEnd5 cells. We next showed that the brain uptake coefficient (Clup) of 100 μg (∼220 nM) of Ri7 mAbs fluorolabeled with Alexa Fluor 750 (AF750) was 0.27 ± 0.05 μL g(-1) s(-1) after subtraction of values obtained with a control IgG. A linear relationship was observed between the distribution volume VD (μL g(-1)) and the perfusion time (s) over 30-120 s (r(2) = 0.997), confirming the metabolic stability of the AF750-Ri7 mAbs during perfusion. Co-perfusion of increasing quantities of unlabeled Ri7 decreased the AF750-Ri7 Clup down to control IgG levels over 500 nM, consistent with a saturable mechanism. Fluorescence microscopy analysis showed a vascular distribution of perfused AF750-Ri7 in the brain and colocalization with a marker of basal lamina. To our knowledge, this is the first reported use of the in situ brain perfusion technique combined with quantification of compounds labeled with near-infrared fluorophores. Furthermore, this study confirms the accumulation of the antitransferrin receptor Ri7 mAb in the brain of mice through a saturable uptake mechanism.
Collapse
Affiliation(s)
- Wael Alata
- Faculty of Pharmacy, Université Laval , Québec, QC G1V 0A6, Canada
| | | | | | | | | |
Collapse
|
78
|
Hwang SR, Kim K. Nano-enabled delivery systems across the blood–brain barrier. Arch Pharm Res 2013; 37:24-30. [DOI: 10.1007/s12272-013-0272-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 10/21/2013] [Indexed: 10/26/2022]
|
79
|
Design of brain imaging agents for positron emission tomography: do large bioconjugates provide an opportunity for in vivo brain imaging? Future Med Chem 2013; 5:1621-34. [PMID: 24047268 DOI: 10.4155/fmc.13.128] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The development of brain imaging agents for positron emission tomography and other in vivo imaging modalities mostly relies on small compounds of low MW as a result of the restricted transport of larger molecules, such as peptides and proteins, across the blood–brain barrier. Besides passive transport, only a few active carrier mechanisms, such as glucose transporters and amino acid transporters, have so far been exploited to mediate the accumulation of imaging probes in the brain. An important question for the future is whether some of the abundant active carrier systems located at the blood–brain barrier can be used to shuttle potential, but non-crossing, imaging agents into the brain. What are the biological and chemical constrictions toward such bioconjugates and is it worthwhile to persue such a delivery strategy?
Collapse
|
80
|
Trapani A, Palazzo C, de Candia M, Lasorsa FM, Trapani G. Targeting of the Translocator Protein 18 kDa (TSPO): A Valuable Approach for Nuclear and Optical Imaging of Activated Microglia. Bioconjug Chem 2013; 24:1415-28. [DOI: 10.1021/bc300666f] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Adriana Trapani
- Department of Pharmacy and Drug
Sciences, University of Bari, Bari, 70125,
Italy
| | - Claudio Palazzo
- Department of Pharmacy and Drug
Sciences, University of Bari, Bari, 70125,
Italy
| | - Modesto de Candia
- Department of Pharmacy and Drug
Sciences, University of Bari, Bari, 70125,
Italy
| | | | - Giuseppe Trapani
- Department of Pharmacy and Drug
Sciences, University of Bari, Bari, 70125,
Italy
| |
Collapse
|
81
|
Raevsky OA, Solodova SL, Lagunin AA, Poroikov VV. Computer modeling of blood brain barrier permeability for physiologically active compounds. BIOCHEMISTRY MOSCOW-SUPPLEMENT SERIES B-BIOMEDICAL CHEMISTRY 2013. [DOI: 10.1134/s199075081302008x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
82
|
De Vrieze M, Lynen F, Chen K, Szucs R, Sandra P. Predicting drug penetration across the blood–brain barrier: comparison of micellar liquid chromatography and immobilized artificial membrane liquid chromatography. Anal Bioanal Chem 2013; 405:6029-41. [DOI: 10.1007/s00216-013-7015-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 04/20/2013] [Accepted: 04/23/2013] [Indexed: 12/01/2022]
|
83
|
Andersson O, Hansen SH, Hellman K, Olsen LR, Andersson G, Badolo L, Svenstrup N, Nielsen PA. The grasshopper: a novel model for assessing vertebrate brain uptake. J Pharmacol Exp Ther 2013; 346:211-8. [PMID: 23671124 DOI: 10.1124/jpet.113.205476] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The aim of the present study was to develop a blood-brain barrier (BBB) permeability model that is applicable in the drug discovery phase. The BBB ensures proper neural function, but it restricts many drugs from entering the brain, and this complicates the development of new drugs against central nervous system diseases. Many in vitro models have been developed to predict BBB permeability, but the permeability characteristics of the human BBB are notoriously complex and hard to predict. Consequently, one single suitable BBB permeability screening model, which is generally applicable in the early drug discovery phase, does not yet exist. A new refined ex vivo insect-based BBB screening model that uses an intact, viable whole brain under controlled in vitro-like exposure conditions is presented. This model uses intact brains from desert locusts, which are placed in a well containing the compound solubilized in an insect buffer. After a limited time, the brain is removed and the compound concentration in the brain is measured by conventional liquid chromatography-mass spectrometry. The data presented here include 25 known drugs, and the data show that the ex vivo insect model can be used to measure the brain uptake over the hemolymph-brain barrier of drugs and that the brain uptake shows linear correlation with in situ perfusion data obtained in vertebrates. Moreover, this study shows that the insect ex vivo model is able to identify P-glycoprotein (Pgp) substrates, and the model allows differentiation between low-permeability compounds and compounds that are Pgp substrates.
Collapse
|
84
|
Martins AF, Morfin JF, Kubíčková A, Kubíček V, Buron F, Suzenet F, Salerno M, Lazar AN, Duyckaerts C, Arlicot N, Guilloteau D, Geraldes CFGC, Tóth É. PiB-Conjugated, Metal-Based Imaging Probes: Multimodal Approaches for the Visualization of β-Amyloid Plaques. ACS Med Chem Lett 2013; 4:436-40. [PMID: 24900692 DOI: 10.1021/ml400042w] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 04/14/2013] [Indexed: 01/19/2023] Open
Abstract
In an effort toward the visualization of β-amyloid plaques by in vivo imaging techniques, we have conjugated an optimized derivative of the Pittsburgh compound B (PiB), a well-established marker of Aβ plaques, to DO3A-monoamide that is capable of forming stable, noncharged complexes with different trivalent metal ions including Gd(3+) for MRI and (111)In(3+) for SPECT applications. Proton relaxivity measurements evidenced binding of Gd(DO3A-PiB) to the amyloid peptide Aβ1-40 and to human serum albumin, resulting in a two- and four-fold relaxivity increase, respectively. Ex vivo immunohistochemical studies showed that the DO3A-PiB complexes selectively target Aβ plaques on Alzheimer's disease human brain tissue. Ex vivo biodistribution data obtained for the (111)In-analogue pointed to a moderate blood-brain barrier (BBB) penetration in adult male Swiss mice (without amyloid deposits) with 0.36% ID/g in the cortex at 2 min postinjection.
Collapse
Affiliation(s)
- André F. Martins
- Centre de Biophysique Moléculaire, CNRS, Rue Charles Sadron, 45071 Orléans Cedex
2, France
- Department of
Life Sciences,
Center of Neurosciences and Cell Biology (CNC), and Coimbra Chemistry
Center, University of Coimbra, Portugal
| | - Jean-François Morfin
- Centre de Biophysique Moléculaire, CNRS, Rue Charles Sadron, 45071 Orléans Cedex
2, France
| | - Anna Kubíčková
- Centre de Biophysique Moléculaire, CNRS, Rue Charles Sadron, 45071 Orléans Cedex
2, France
- Department of Analytical Chemistry, Charles University in Prague, Albertov 2030, 12840
Prague, Czech Republic
| | - Vojtěch Kubíček
- Centre de Biophysique Moléculaire, CNRS, Rue Charles Sadron, 45071 Orléans Cedex
2, France
| | - Frédéric Buron
- Institut de Chimie Organique
et Analytique, UMR 7311 CNRS/Université d’Orléans, rue de Chartres, 45067 Orléans, France
| | - Franck Suzenet
- Institut de Chimie Organique
et Analytique, UMR 7311 CNRS/Université d’Orléans, rue de Chartres, 45067 Orléans, France
| | - Milena Salerno
- Laboratoire CSPBAT, CNRS UMR
7244, UFR-SMBH, Université Paris 13, 74 rue Marcel Cachin, 93017 Bobigny, France
| | - Adina N. Lazar
- Centre de Recherche de l’Institut
du Cerveau et de la Moelle, CNRS UMR7225, INSERM, UMR975 and UPMC, Hôpital de la Pitié-Salpêtrière 47, Bd de l’Hôpital 75013 Paris, France
| | - Charles Duyckaerts
- Centre de Recherche de l’Institut
du Cerveau et de la Moelle, CNRS UMR7225, INSERM, UMR975 and UPMC, Hôpital de la Pitié-Salpêtrière 47, Bd de l’Hôpital 75013 Paris, France
| | - Nicolas Arlicot
- Inserm, U930, Université François Rabelais de Tours, CHRU de Tours, 37044
Tours Cedex 9, France
| | - Denis Guilloteau
- Inserm, U930, Université François Rabelais de Tours, CHRU de Tours, 37044
Tours Cedex 9, France
| | - Carlos F. G. C. Geraldes
- Department of
Life Sciences,
Center of Neurosciences and Cell Biology (CNC), and Coimbra Chemistry
Center, University of Coimbra, Portugal
| | - Éva Tóth
- Centre de Biophysique Moléculaire, CNRS, Rue Charles Sadron, 45071 Orléans Cedex
2, France
| |
Collapse
|
85
|
Structure-based ligand discovery for the Large-neutral Amino Acid Transporter 1, LAT-1. Proc Natl Acad Sci U S A 2013; 110:5480-5. [PMID: 23509259 DOI: 10.1073/pnas.1218165110] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The Large-neutral Amino Acid Transporter 1 (LAT-1)--a sodium-independent exchanger of amino acids, thyroid hormones, and prescription drugs--is highly expressed in the blood-brain barrier and various types of cancer. LAT-1 plays an important role in cancer development as well as in mediating drug and nutrient delivery across the blood-brain barrier, making it a key drug target. Here, we identify four LAT-1 ligands, including one chemically novel substrate, by comparative modeling, virtual screening, and experimental validation. These results may rationalize the enhanced brain permeability of two drugs, including the anticancer agent acivicin. Finally, two of our hits inhibited proliferation of a cancer cell line by distinct mechanisms, providing useful chemical tools to characterize the role of LAT-1 in cancer metabolism.
Collapse
|
86
|
Lanevskij K, Japertas P, Didziapetris R. Improving the prediction of drug disposition in the brain. Expert Opin Drug Metab Toxicol 2013; 9:473-86. [PMID: 23294027 DOI: 10.1517/17425255.2013.754423] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Ability to cross the blood-brain barrier is one of the key ADME characteristics of all drug candidates regardless of their target location in the body. While good brain penetration is essential for CNS drugs, it may lead to serious side effects in case of peripherally-targeted molecules. Despite a high demand of computational methods for estimating brain transport early in drug discovery, achieving good prediction accuracy still remains a challenging task. AREAS COVERED This article reviews various measures employed to quantify brain delivery and recent advances in QSAR approaches for predicting these properties from the compound's structure. Additionally, the authors discuss the classification models attempting to distinguish between permeable and impermeable chemicals. EXPERT OPINION Recent research in the field of brain penetration modeling showed an increasing understanding of the processes involved in drug disposition, although most models of brain/plasma partitioning still rely on purely statistical considerations. Preferably, new models should incorporate mechanistic knowledge since it is the prerequisite for guiding drug design efforts in the desired direction. To increase the efficiency of computational tools, a broader view is necessary, involving rate and extent of brain penetration, as well as plasma and brain tissue binding strength, instead of relying on any single property.
Collapse
Affiliation(s)
- Kiril Lanevskij
- VšĮ Aukštieji algoritmai, A. Mickeviciaus 29, LT-08117 Vilnius, Lithuania.
| | | | | |
Collapse
|
87
|
Hammarlund-Udenaes M. In Vivo Approaches to Assessing the Blood–Brain Barrier. TOPICS IN MEDICINAL CHEMISTRY 2013. [DOI: 10.1007/7355_2013_27] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
88
|
Ashraf T, Kis O, Banerjee N, Bendayan R. Drug Transporters At Brain Barriers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013. [DOI: 10.1007/978-1-4614-4711-5_2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
89
|
García-Osta A, Cuadrado-Tejedor M, García-Barroso C, Oyarzábal J, Franco R. Phosphodiesterases as therapeutic targets for Alzheimer's disease. ACS Chem Neurosci 2012; 3:832-44. [PMID: 23173065 DOI: 10.1021/cn3000907] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 10/01/2012] [Indexed: 01/10/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia among the elderly. In AD patients, memory loss is accompanied by the formation of beta-amyloid plaques and the appearance of tau in a pathological form. Given the lack of effective treatments for AD, the development of new management strategies for these patients is critical. The continued failure to find effective therapies using molecules aimed at addressing the anti-beta amyloid pathology has led researchers to focus on other non-amyloid-based approaches to restore memory function. Promising non-amyloid related candidate targets include phosphosdiesterases (PDEs), and indeed, Rolipram, a specific PDE4 inhibitor, was the first compound found to effectively restore cognitive deficits in animal models of AD. More recently, PDE5 inhibitors have also been shown to effectively restore memory function. Accordingly, inhibitors of other members of the PDE family may also improve memory performance in AD and non-AD animal models. Hence, in this review, we will summarize the data supporting the use of PDE inhibitors as cognitive enhancers and we will discuss the possible mechanisms of action underlying these effects. We shall also adopt a medicinal chemistry perspective that leads us to propose the most promising PDE candidates on the basis of inhibitor selectivity, brain distribution, and mechanism of action.
Collapse
Affiliation(s)
- Ana García-Osta
- Molecular and Cell Neuropharmacology
Laboratory, Neurosciences Division, CIMA (Centro de investigación Médica Aplicada), Avda Pio
XII, Pamplona, Spain
| | - Mar Cuadrado-Tejedor
- Molecular and Cell Neuropharmacology
Laboratory, Neurosciences Division, CIMA (Centro de investigación Médica Aplicada), Avda Pio
XII, Pamplona, Spain
| | - Carolina García-Barroso
- Molecular and Cell Neuropharmacology
Laboratory, Neurosciences Division, CIMA (Centro de investigación Médica Aplicada), Avda Pio
XII, Pamplona, Spain
| | - Julen Oyarzábal
- Molecular and Cell Neuropharmacology
Laboratory, Neurosciences Division, CIMA (Centro de investigación Médica Aplicada), Avda Pio
XII, Pamplona, Spain
- Small
Molecule Discovery Platform, CIMA (Centro
de investigación Médica
Aplicada), Avda Pio XII, Pamplona, Spain
| | - Rafael Franco
- Molecular and Cell Neuropharmacology
Laboratory, Neurosciences Division, CIMA (Centro de investigación Médica Aplicada), Avda Pio
XII, Pamplona, Spain
| |
Collapse
|
90
|
Di L, Rong H, Feng B. Demystifying Brain Penetration in Central Nervous System Drug Discovery. J Med Chem 2012; 56:2-12. [PMID: 23075026 DOI: 10.1021/jm301297f] [Citation(s) in RCA: 204] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Li Di
- Pfizer Inc., Groton, Connecticut
06340, United States
| | - Haojing Rong
- Pfizer Inc., Groton, Connecticut
06340, United States
| | - Bo Feng
- Pfizer Inc., Groton, Connecticut
06340, United States
| |
Collapse
|
91
|
Abstract
The brain is one of the most protected organs in the body. There are two key barriers that control the access of endogenous substances and xenobiotics (drugs or toxins) to the CNS. These physiological structures are the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier. The BBB represents the main determinant of the effective delivery of drugs to the CNS. Good access through the BBB is essential if the target site is located within the CNS or, in contrast, can be a disadvantage if adverse reactions occur at central level. The development of new drugs targeted to the CNS requires a better knowledge of the factors affecting BBB permeation as well as in vitro and in silico predictive tools to optimize screening, and to reduce the attrition rate at later stages of drug development. This review discusses the particular characteristics of the biology and physiology of the BBB with respect to the permeation and distribution of drugs into the brain. The factors affecting rate, extent and distribution into the brain are discussed and a brief description of the in silico, in vitro, in situ and in vivo methods used to measure BBB transport are presented. Finally, the lastest proposals and strategies to enhance transport across the BBB of new CNS drugs are summarized.
Collapse
|
92
|
Watanabe K, Nishimura Y, Nomoto T, Umemoto N, Zhang Z, Zhang B, Kuroyanagi J, Shimada Y, Shintou T, Okano M, Miyazaki T, Imamura T, Tanaka T. In vivo assessment of the permeability of the blood-brain barrier and blood-retinal barrier to fluorescent indoline derivatives in zebrafish. BMC Neurosci 2012; 13:101. [PMID: 22894547 PMCID: PMC3807752 DOI: 10.1186/1471-2202-13-101] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 08/10/2012] [Indexed: 01/05/2023] Open
Abstract
Background Successful delivery of compounds to the brain and retina is a challenge in the development of therapeutic drugs and imaging agents. This challenge arises because internalization of compounds into the brain and retina is restricted by the blood–brain barrier (BBB) and blood-retinal barrier (BRB), respectively. Simple and reliable in vivo assays are necessary to identify compounds that can easily cross the BBB and BRB. Methods We developed six fluorescent indoline derivatives (IDs) and examined their ability to cross the BBB and BRB in zebrafish by in vivo fluorescence imaging. These fluorescent IDs were administered to live zebrafish by immersing the zebrafish larvae at 7-8 days post fertilization in medium containing the ID, or by intracardiac injection. We also examined the effect of multidrug resistance proteins (MRPs) on the permeability of the BBB and BRB to the ID using MK571, a selective inhibitor of MRPs. Results The permeability of these barriers to fluorescent IDs administered by simple immersion was comparable to when administered by intracardiac injection. Thus, this finding supports the validity of drug administration by simple immersion for the assessment of BBB and BRB permeability to fluorescent IDs. Using this zebrafish model, we demonstrated that the length of the methylene chain in these fluorescent IDs significantly affected their ability to cross the BBB and BRB via MRPs. Conclusions We demonstrated that in vivo assessment of the permeability of the BBB and BRB to fluorescent IDs could be simply and reliably performed using zebrafish. The structure of fluorescent IDs can be flexibly modified and, thus, the permeability of the BBB and BRB to a large number of IDs can be assessed using this zebrafish-based assay. The large amount of data acquired might be useful for in silico analysis to elucidate the precise mechanisms underlying the interactions between chemical structure and the efflux transporters at the BBB and BRB. In turn, understanding these mechanisms may lead to the efficient design of compounds targeting the brain and retina.
Collapse
Affiliation(s)
- Kohei Watanabe
- Corporate R&D Headquarters, Canon Inc Tokyo, Ohta-ku, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Riebeling C, Hayess K, Peters AK, Steemans M, Spielmann H, Luch A, Seiler AEM. Assaying embryotoxicity in the test tube: current limitations of the embryonic stem cell test (EST) challenging its applicability domain. Crit Rev Toxicol 2012; 42:443-64. [PMID: 22512667 DOI: 10.3109/10408444.2012.674483] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Testing for embryotoxicity in vitro is an attractive alternative to animal experimentation. The embryonic stem cell test (EST) is such a method, and it has been formally validated by the European Centre for the Validation of Alternative Methods. A number of recent studies have underscored the potential of this method. However, the EST performed well below the 78% accuracy expected from the validation study using a new set of chemicals and pharmaceutical compounds, and also of toxicity criteria, tested to enlarge the database of the validated EST as part of the Work Package III of the ReProTect Project funded within the 6th Framework Programme of the European Union. To assess the performance and applicability domain of the EST we present a detailed review of the substances and their effects in the EST being nitrofen, ochratoxin A, D-penicillamine, methylazoxymethanol, lovastatin, papaverine, warfarin, β-aminopropionitrile, dinoseb, furosemide, doxylamine, pravastatin, and metoclopramide. By delineation of the molecular mechanisms of the substances we identify six categories of reasons for misclassifications. Some of these limitations might also affect other in vitro methods assessing embryotoxicity. Substances that fall into these categories need to be included in future validation sets and in validation guidelines for embryotoxicity testing. Most importantly, we suggest conceivable improvements and additions to the EST which will resolve most of the limitations.
Collapse
Affiliation(s)
- Christian Riebeling
- German Federal Institute for Risk Assessment (BfR), ZEBET - Alternative Methods to Animal Experiments, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
94
|
García-Barroso C, Ricobaraza A, Pascual-Lucas M, Unceta N, Rico AJ, Goicolea MA, Sallés J, Lanciego JL, Oyarzabal J, Franco R, Cuadrado-Tejedor M, García-Osta A. Tadalafil crosses the blood-brain barrier and reverses cognitive dysfunction in a mouse model of AD. Neuropharmacology 2012; 64:114-23. [PMID: 22776546 DOI: 10.1016/j.neuropharm.2012.06.052] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 06/13/2012] [Accepted: 06/27/2012] [Indexed: 11/27/2022]
Abstract
Previous studies have demonstrated that cognitive function can be restored in mouse models of Alzheimer's disease (AD) following administration of sildenafil, a specific PDE5 inhibitor (Puzzo et al., 2009; Cuadrado-Tejedor et al.). Another very potent PDE5 inhibitor with a longer half-life and safe in chronic treatments, tadalafil, may represent a better alternative candidate for AD therapy. However, tadalafil was proven unable to achieve similar benefits than those of sildenafil in AD animal models (Puzzo et al., 2009). The lack of efficacy was attributed to inability to cross the blood-brain barrier (BBB). In this paper we first measured the blood and brain levels of tadalafil to prove that the compound crosses BBB and that chronic treatment leads to accumulation in the brain of the J20 transgenic mouse model of AD. We demonstrated the presence of PDE5 mRNA in the brain of the mice and also in the human brain. After a 10 week treatment with either of these PDE5 inhibitors, the performance of the J20 mice in the Morris water maze test improved when compared with the transgenic mice that received vehicle. Biochemical analysis revealed that neither sildenafil nor tadalafil altered the amyloid burden, although both compounds reduced Tau phosphorylation in the mouse hippocampus. This study provides evidence of the potential benefits of a chronic tadalafil treatment in AD therapy. This article is part of a Special Issue entitled 'Cognitive Enhancers'.
Collapse
Affiliation(s)
- Carolina García-Barroso
- Cell and Molecular Neuropharmacology, Neurosciences Division, Center for Applied Medical Research, CIMA, University of Navarra, Av. Pio XII 55, 31008 Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Pabel J, Faust M, Prehn C, Wörlein B, Allmendinger L, Höfner G, Wanner KT. Development of an (S)-1-{2-[Tris(4-methoxyphenyl)methoxy]ethyl}piperidine-3-carboxylic acid [(S)-SNAP-5114] Carba Analogue Inhibitor for Murine γ-Aminobutyric Acid Transporter Type 4. ChemMedChem 2012; 7:1245-55. [DOI: 10.1002/cmdc.201200126] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 04/05/2012] [Indexed: 11/06/2022]
|
96
|
Low brain penetrant CB1 receptor agonists for the treatment of neuropathic pain. Bioorg Med Chem Lett 2012; 22:2932-7. [DOI: 10.1016/j.bmcl.2012.02.048] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 02/15/2012] [Accepted: 02/16/2012] [Indexed: 12/18/2022]
|
97
|
Krajcsi P, Jani M, Tóth B, Erdő F, Kis E, Beéry E, Sziráki I. Efflux transporters in the blood–brain interfaces –in vitroandin vivomethods and correlations. Expert Opin Drug Metab Toxicol 2012; 8:419-31. [DOI: 10.1517/17425255.2012.668184] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
98
|
Brainpeps: the blood-brain barrier peptide database. Brain Struct Funct 2011; 217:687-718. [PMID: 22205159 DOI: 10.1007/s00429-011-0375-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 12/12/2011] [Indexed: 01/13/2023]
Abstract
Peptides are able to cross the blood-brain barrier (BBB) through various mechanisms, opening new diagnostic and therapeutic avenues. However, their BBB transport data are scattered in the literature over different disciplines, using different methodologies reporting different influx or efflux aspects. Therefore, a comprehensive BBB peptide database (Brainpeps) was constructed to collect the BBB data available in the literature. Brainpeps currently contains BBB transport information with positive as well as negative results. The database is a useful tool to prioritize peptide choices for evaluating different BBB responses or studying quantitative structure-property (BBB behaviour) relationships of peptides. Because a multitude of methods have been used to assess the BBB behaviour of compounds, we classified these methods and their responses. Moreover, the relationships between the different BBB transport methods have been clarified and visualized.
Collapse
|
99
|
Trapani A, Denora N, Iacobellis G, Sitterberg J, Bakowsky U, Kissel T. Methotrexate-loaded chitosan- and glycol chitosan-based nanoparticles: a promising strategy for the administration of the anticancer drug to brain tumors. AAPS PharmSciTech 2011; 12:1302-11. [PMID: 21948322 DOI: 10.1208/s12249-011-9695-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 09/12/2011] [Indexed: 11/30/2022] Open
Abstract
Brain tumor treatment employing methotrexate (MTX) is limited by the efflux mechanism of Pg-p on the blood-brain barrier. We aimed to investigate MTX-loaded chitosan or glycol chitosan (GCS) nanoparticles (NPs) in the presence and in the absence of a coating layer of Tween 80 for brain delivery of MTX. The effect of a low Tween 80 concentration was evaluated. MTX NPs were formulated following the ionic gelation technique and size and zeta potential measurements were acquired. Transport across MDCKII-MDR1 monolayer and cytotoxicity studies against C6 glioma cell line were also performed. Cell/particles interaction was visualized by confocal microscopy. The particles were shown to be cytotoxic against C6 cells line and able to overcome MDCKII-MDR1 cell barrier. GCS-based NPs were the most cytotoxic NPs. Confocal observations highlighted the internalization of Tween 80-coated fluorescent NPs more than Tween 80-uncoated NPs. The results suggest that even a low concentration of Tween 80 is sufficient for enhancing the transport of MTX from the NPs across MDCKII-MDR1 cells. The nanocarriers represent a promising strategy for the administration of MTX to brain tumors which merits further investigations under in vivo conditions.
Collapse
|
100
|
Fujimoto T, Tomata Y, Kunitomo J, Hirozane M, Marui S. Discovery of spiropiperidine-based potent and selective Orexin-2 receptor antagonists. Bioorg Med Chem Lett 2011; 21:6409-13. [DOI: 10.1016/j.bmcl.2011.08.094] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 08/22/2011] [Accepted: 08/23/2011] [Indexed: 11/29/2022]
|