1
|
Jorgensen C, Linville RM, Galea I, Lambden E, Vögele M, Chen C, Troendle EP, Ruggiu F, Ulmschneider MB, Schiøtt B, Lorenz CD. Permeability Benchmarking: Guidelines for Comparing in Silico, in Vitro, and in Vivo Measurements. J Chem Inf Model 2025; 65:1067-1084. [PMID: 39823383 PMCID: PMC11815851 DOI: 10.1021/acs.jcim.4c01815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/04/2025] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
Permeability is a measure of the degree to which cells can transport molecules across biological barriers. Units of permeability are distance per unit time (typically cm/s), where accurate measurements are needed to define drug delivery in homeostasis and to model dysfunction occurring during disease. This perspective offers a set of community-led guidelines to benchmark permeability data across multidisciplinary approaches and different biological contexts. First, we lay out the analytical framework for three methodologies to calculate permeability: in silico assays using either transition-based counting or the inhomogeneous-solubility diffusion approaches, in vitro permeability assays using cells cultured in 2D or 3D geometries, and in vivo assays utilizing in situ brain perfusion or multiple time-point regression analysis. Then, we demonstrate a systematic benchmarking of in silico to both in vitro and in vivo, depicting the ways in which each benchmarking is sensitive to the choices of assay design. Finally, we outline seven recommendations for best practices in permeability benchmarking and underscore the significance of tailored permeability assays in driving advancements in drug delivery research and development. Our exploration encompasses a discussion of "generic" and tissue-specific biological barriers, including the blood-brain barrier (BBB), which is a major hurdle for the delivery of therapeutic agents into the brain. By addressing challenges in reconciling simulated data with experimental assays, we aim to provide insights essential for optimizing accuracy and reliability in permeability modeling.
Collapse
Affiliation(s)
- Christian Jorgensen
- School
of Medicine, Pharmacy and Biomedical Sciences, Faculty of Science
& Health, University of Portsmouth, Portsmouth PO1 2DT, Hampshire, U.K.
- Dept.
of Chemistry, Aarhus University, Langelandsgade, 140 8000 Aarhus C, Denmark
| | - Raleigh M. Linville
- The
Picower Institute for Learning and Memory, Massachusetts Institute of Technology, 43 Vassar Street, Cambridge, Massachusetts 02139, United States
| | - Ian Galea
- Clinical
Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, U.K.
| | - Edward Lambden
- Dept.
of Chemistry, King’s College London, London WC2R 2LS, U.K.
| | - Martin Vögele
- Department
of Computer Science, Stanford University, Stanford, California 94305, United States
- Department
of Molecular and Cellular Physiology, Stanford
University, Stanford, California 94305, United States
- Institute
for Computational and Mathematical Engineering, Stanford University, Stanford, California 94305, United States
| | - Charles Chen
- Synthetic
Biology Group, Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Evan P. Troendle
- Wellcome−Wolfson
Institute for Experimental Medicine, School of Medicine, Dentistry
and Biomedical Sciences, Queen’s
University Belfast, Belfast, County
Antrim, BT9 7BL, Northern Ireland, U.K.
| | - Fiorella Ruggiu
- Kimia
Therapeutics, 740 Heinz
Avenue, Berkeley, California 94710, United States
| | | | - Birgit Schiøtt
- Dept.
of Chemistry, Aarhus University, Langelandsgade, 140 8000 Aarhus C, Denmark
| | | |
Collapse
|
2
|
Smith QR, Mandula H, Parepally JMR, Oki J, Thomas F, Thorsheim HR, Al-Ahmad AJ, Abbruscato TJ, Ask P, Hage DS, Robinson PJ. Brain endothelial permeability, transport, and flow assessed over 10 orders of magnitude using the in situ brain perfusion technique. Fluids Barriers CNS 2024; 21:100. [PMID: 39690422 DOI: 10.1186/s12987-024-00584-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/06/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Cerebral blood flow normally places a limit on the magnitude of brain vascular permeability (P) that can be measured in vivo. At normal cerebral blood flow, this limit falls at the lower end of lipophilicity for most FDA-approved CNS drugs. In this study, we report on two methods that can be used to overcome this limitation and measure brain vascular permeability values that are up to ~1000 times higher using the in situ brain perfusion technique. METHODS Rat brain was perfused with physiological saline at increased flow rate and in the presence of various concentrations of plasma protein, serum albumin or alpha-acid glycoprotein. Plasma protein was added to the saline perfusion fluid to lower extraction into the measurable range using the Crone Renkin "diffusion-flow" equation to calculate brain PoS. RESULTS Cerebrovascular Po was determined for 125 solutes, of which 78 showed little or no evidence of active efflux transport. Fifty of the solutes were in the lipophilicity zone (Log Poct 1-5) of most FDA-approved CNS drugs. Care was taken to ensure the integrity of the brain vasculature during perfusion and to measure flow accurately using markers that had been verified for the flow rates. The results showed a linear relationship between Log Po and Log Poct over ~10 orders of magnitude with values for diazepam, estradiol, testosterone, and other agents that exceed prior published values by fivefold to 200-fold. CONCLUSIONS The results show that brain vascular permeability can be measured directly in vivo for highly lipophilic solutes and the PS values obtained match reasonably with that predicted by the Crone-Renkin flow diffusion equation with care taken to validate the accuracy for the component measurements and with no need to invoke "enhanced" or "induced" dissociation.
Collapse
Affiliation(s)
- Quentin R Smith
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
- National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
| | - Haritha Mandula
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Jagan Mohan R Parepally
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Jun Oki
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Fancy Thomas
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Helen R Thorsheim
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Abraham J Al-Ahmad
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Per Ask
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
| | - David S Hage
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Peter J Robinson
- National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
3
|
Moussaoui M, Baammi S, Soufi H, Baassi M, Salah M, Allali AEL, Mohammed BE, Daoud R, Belaaouad S. Design and Optimization of Quinazoline Derivatives as Potent EGFR Inhibitors for Lung Cancer Treatment: A Comprehensive QSAR, ADMET, and Molecular Modeling Investigation. ACS OMEGA 2024; 9:45842-45857. [PMID: 39583714 PMCID: PMC11579736 DOI: 10.1021/acsomega.4c04639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 11/26/2024]
Abstract
The epidermal growth factor receptor (EGFR) is part of a protein family that controls cell growth and development. Due to its importance, EGFR has been identified as a suitable target for creating novel drugs. For this research, we conducted a 2D-QSAR analysis on a set of 31 molecules derived from quinazoline, which exhibited inhibitory activity against human lung cancer. This investigation incorporated principal component analysis (PCA) and multiple linear regression (MLR), leading to the development of QSAR models with strong predictive capabilities (R 2 = 0.745, R 2_adj = 0.723, MSE = 0.061, R 2_test = 0.941, and Q 2_cv = 0.669). The reliability of these models was confirmed through internal, external, Y-randomization, and applicability domain validations. Leveraging the predictions from the QSAR model, we designed 18 new molecules based on the modifications at the N-3 and C-6 positions of the quinazoline ring, with electronegative substituents at these positions fostering optimal polar interactions and hydrophobic contacts within the ATP-binding site of EGFR, significantly enhancing the inhibitory activity against the lung cancer cell line. Subsequently, ADMET predictions were conducted for these 18 compounds, revealing outstanding ADMET profiles. Molecular docking analyses were performed to investigate the interactions between the newly designed molecules-Pred15, Pred17, Pred20, Pred21-and the EGFR protein, indicating high affinity of these proposed compounds to EGFR. Furthermore, molecular dynamics (MD) simulations were utilized to assess the stability and binding modes of compounds Pred17, Pred20, and Pred21, reinforcing their potential as novel inhibitors against human lung cancer. Overall, our findings suggest that these investigated compounds can serve as effective inhibitors, showcasing the utility of our analytical and design approach in the identification of promising therapeutic agents.
Collapse
Affiliation(s)
- Mohamed Moussaoui
- Laboratory
of Physical Chemistry of Materials, Faculty of Sciences Ben M’Sick, Hassan II University of Casablanca, Casablanca 20670, Morocco
| | - Soukayna Baammi
- Bioinformatics
Laboratory, College of Computing, Mohammed
VI Polytechnic University, Ben Guerir 20670, Morocco
| | - Hatim Soufi
- Laboratory
of Physical Chemistry of Materials, Faculty of Sciences Ben M’Sick, Hassan II University of Casablanca, Casablanca 20670, Morocco
| | - Mouna Baassi
- Laboratory
of Physical Chemistry of Materials, Faculty of Sciences Ben M’Sick, Hassan II University of Casablanca, Casablanca 20670, Morocco
| | - Mohammed Salah
- Team
of Chemoinformatics Research and Spectroscopy and Quantum Chemistry,
Department of Chemistry, Faculty of Science, University Chouaib Doukkali, B. P. 20, El Jadida 2300, Morocco
| | - Achraf EL Allali
- Bioinformatics
Laboratory, College of Computing, Mohammed
VI Polytechnic University, Ben Guerir 20670, Morocco
| | - Belghiti Elalaoui Mohammed
- Laboratory
of Physical Chemistry of Materials, Faculty of Sciences Ben M’Sick, Hassan II University of Casablanca, Casablanca 20670, Morocco
- Laboratory
of Nernest Technology, 163 Willington Street, Sherbrook, Quebec J1H5C7, Canada
| | - Rachid Daoud
- Chemical
and Biochemical Sciences-Green Processing Engineering, Mohammed VI Polytechnic University, Ben Guerir 43150, Morocco
| | - Said Belaaouad
- Laboratory
of Physical Chemistry of Materials, Faculty of Sciences Ben M’Sick, Hassan II University of Casablanca, Casablanca 20670, Morocco
| |
Collapse
|
4
|
Yang L, Lin Z, Mu R, Wu W, Zhi H, Liu X, Yang H, Liu L. Neurons enhance blood-brain barrier function via upregulating claudin-5 and VE-cadherin expression due to glial cell line-derived neurotrophic factor secretion. eLife 2024; 13:RP96161. [PMID: 39475379 PMCID: PMC11524583 DOI: 10.7554/elife.96161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Abstract
Blood-brain barrier (BBB) prevents neurotoxins from entering central nervous system. We aimed to establish and characterize an in vitro triple co-culture BBB model consisting of brain endothelial cells hCMEC/D3, astrocytoma U251 cells, and neuroblastoma SH-SY5Y cells. Co-culture of SH-SY5Y and U251 cells markedly enhanced claudin-5 and VE-cadherin expression in hCMEC/D3 cells, accompanied by increased transendothelial electrical resistance and decreased permeability. Conditioned medium (CM) from SH-SY5Y cells (S-CM), U251 cells (U-CM), and co-culture of SH-SY5Y and U251 cells (US-CM) also promoted claudin-5 and VE-cadherin expression. Glial cell line-derived neurotrophic factor (GDNF) levels in S-CM and US-CM were significantly higher than CMs from hCMEC/D3 and U-CM. Both GDNF and US-CM upregulated claudin-5 and VE-cadherin expression, which were attenuated by anti-GDNF antibody and GDNF signaling inhibitors. GDNF increased claudin-5 expression via the PI3K/AKT/FOXO1 and MAPK/ERK pathways. Meanwhile, GDNF promoted VE-cadherin expression by activating PI3K/AKT/ETS1 and MAPK/ERK/ETS1 signaling. The roles of GDNF in BBB integrity were validated using brain-specific Gdnf silencing mice. The developed triple co-culture BBB model was successfully applied to predict BBB permeability. In conclusion, neurons enhance BBB integrity by upregulating claudin-5 and VE-cadherin expression through GDNF secretion and established triple co-culture BBB model may be used to predict drugs' BBB permeability.
Collapse
Affiliation(s)
- Lu Yang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| | - Zijin Lin
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| | - Ruijing Mu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| | - Wenhan Wu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| | - Hao Zhi
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| | - Xiaodong Liu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| | - Hanyu Yang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| | - Li Liu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| |
Collapse
|
5
|
Ebert A, Dahley C. Can membrane permeability of zwitterionic compounds be predicted by the solubility-diffusion model? Eur J Pharm Sci 2024; 199:106819. [PMID: 38815700 DOI: 10.1016/j.ejps.2024.106819] [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: 02/22/2024] [Revised: 04/23/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Zwitterions contain both positively and negatively charged functional groups, resulting in an overall net neutral charge. Nevertheless, the membrane permeability of the zwitterionic form of a compound is assumed to be much lower than the permeability of the uncharged neutral form. Although a significant proportion of pharmaceuticals are zwitterionic, it has not been clear so far whether their permeability is dominated by the permeation of the zwitterionic or the neutral form, since neutral fractions are often quite low as compared to the zwitterionic fraction. This complicates the in silico prediction of the permeability of zwitterionic compounds. In this work, we re-evaluated existing in vitro permeability data from literature measured with Caco-2/MDCK cell assays, using more strict exclusion criteria for effects like diffusion limitation by the aqueous boundary layers, paracellular transport, active transport and retention. Using this re-evaluated data set, we show that extracted intrinsic permeabilities of the neutral fraction are well predicted by the solubility-diffusion model (RMSE = 1.21; n = 18) if the permeability of the zwitterionic species is assumed negligible. Our work thus suggests that only the neutral species is relevant for the membrane permeability of zwitterionic compounds, and that membrane permeability of zwitterionic compounds is indeed predictable by the solubility-diffusion model.
Collapse
Affiliation(s)
- Andrea Ebert
- Department of Computational Biology & Chemistry, Helmholtz Centre for Environmental Research (UFZ), Permoserstraße 15, 04318 Leipzig, Germany.
| | - Carolin Dahley
- Department of Computational Biology & Chemistry, Helmholtz Centre for Environmental Research (UFZ), Permoserstraße 15, 04318 Leipzig, Germany
| |
Collapse
|
6
|
Ebert A, Dahley C, Goss KU. Pitfalls in evaluating permeability experiments with Caco-2/MDCK cell monolayers. Eur J Pharm Sci 2024; 194:106699. [PMID: 38232636 DOI: 10.1016/j.ejps.2024.106699] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/19/2024]
Abstract
When studying the transport of molecules across biological membranes, intrinsic membrane permeability (P0) is more informative than apparent permeability (Papp), because it eliminates external (setup-specific) factors, provides consistency across experiments and mechanistic insight. It is thus an important building block for modeling the total permeability in any given scenario. However, extracting P0 is often difficult, if not impossible, when the membrane is not the dominant transport resistance. In this work, we set out to analyze Papp values measured with Caco-2/MDCK cell monolayers of 69 literature references. We checked the Papp values for a total of 318 different compounds for the extractability of P0, considering possible limitations by aqueous boundary layers, paracellular transport, recovery issues, active transport, a possible proton flux limitation, and sink conditions. Overall, we were able to extract 77 reliable P0 values, which corresponds to about one quarter of the total compounds analyzed, while about half were limited by the diffusion through the aqueous layers. Compared to an existing data set of P0 values published by Avdeef, our approach resulted in a much higher exclusion of compounds. This is a consequence of stricter compound- and reference-specific exclusion criteria, but also because we considered possible concentration-shift effects due to different pH values in the aqueous layers, an effect only recently described in literature. We thus provide a consistent and reliable set of P0, e.g. as a basis for future modeling.
Collapse
Affiliation(s)
- Andrea Ebert
- Department of Analytical Environmental Chemistry, Helmholtz Centre for Environmental Research (UFZ), Permoserstraße 15, Leipzig 04318, Federal Republic of Germany.
| | - Carolin Dahley
- Department of Analytical Environmental Chemistry, Helmholtz Centre for Environmental Research (UFZ), Permoserstraße 15, Leipzig 04318, Federal Republic of Germany
| | - Kai-Uwe Goss
- Department of Analytical Environmental Chemistry, Helmholtz Centre for Environmental Research (UFZ), Permoserstraße 15, Leipzig 04318, Federal Republic of Germany; Institute of Chemistry, University of Halle-Wittenberg, Kurt-Mothes-Straße 2, Halle 06120, Federal Republic of Germany
| |
Collapse
|
7
|
Dichiara M, Cosentino G, Giordano G, Pasquinucci L, Marrazzo A, Costanzo G, Amata E. Designing drugs optimized for both blood-brain barrier permeation and intra-cerebral partition. Expert Opin Drug Discov 2024; 19:317-329. [PMID: 38145409 DOI: 10.1080/17460441.2023.2294118] [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: 10/08/2023] [Accepted: 12/07/2023] [Indexed: 12/26/2023]
Abstract
INTRODUCTION With the increasing incidence and prevalence of neurological disorders globally, there is a paramount need for new pharmacotherapies. BBB effectively protects the brain but raises a profound challenge to drug permeation, with less than 2% of most drugs reaching the CNS. AREAS COVERED This article reviews aspects of the most recent design strategies, providing insights into ideas and concepts in CNS drug discovery. An overview of the products available on the market is given and why clinical trials are continuously failing is discussed. EXPERT OPINION Among the available CNS drugs, small molecules account for most successful CNS therapeutics due to their ability to penetrate the BBB through passive or carrier-mediated mechanisms. The development of new CNS drugs is very difficult. To date, there is a lack of effective drugs for alleviating or even reversing the progression of brain diseases. Particularly, the use of artificial intelligence strategies, together with more appropriate animal models, may enable the design of molecules with appropriate permeation, to elicit a biological response from the neurotherapeutic target.
Collapse
Affiliation(s)
- Maria Dichiara
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Catania, Italy
| | - Giuseppe Cosentino
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Catania, Italy
| | - Giorgia Giordano
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Catania, Italy
| | - Lorella Pasquinucci
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Catania, Italy
| | - Agostino Marrazzo
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Catania, Italy
| | - Giuliana Costanzo
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Catania, Italy
| | - Emanuele Amata
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Catania, Italy
| |
Collapse
|
8
|
Rahman MT, Guan D, Chaminda Lakmal HH, Decker AM, Imler GH, Kerr AT, Harris DL, Jin C. Design, Synthesis, and Structure-Activity Relationship Studies of Novel GPR88 Agonists (4-Substituted-phenyl)acetamides Based on the Reversed Amide Scaffold. ACS Chem Neurosci 2024; 15:169-192. [PMID: 38086012 PMCID: PMC10843732 DOI: 10.1021/acschemneuro.3c00684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024] Open
Abstract
The development of synthetic agonists for the orphan receptor GPR88 has recently attracted significant interest, given the promise of GPR88 as a novel drug target for psychiatric and neurodegenerative disorders. Examination of structure-activity relationships of two known agonist scaffolds 2-PCCA and 2-AMPP, as well as the recently resolved cryo-EM structure of 2-PCCA-bound GPR88, led to the design of a new scaffold based on the "reversed amide" strategy of 2-AMPP. A series of novel (4-substituted-phenyl)acetamides were synthesized and assessed in cAMP accumulation assays as GPR88 agonists, which led to the discovery of several compounds with better or comparable potencies to 2-AMPP. Computational docking studies suggest that these novel GPR88 agonists bind to the same allosteric site of GPR88 that 2-PCCA occupies. Collectively, our findings provide structural insight and SAR requirement at the allosteric site of GPR88 and a new scaffold for further development of GPR88 allosteric agonists.
Collapse
Affiliation(s)
- Md Toufiqur Rahman
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Dongliang Guan
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Hetti Handi Chaminda Lakmal
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Ann M Decker
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Gregory H Imler
- Center for Biomolecular Science and Engineering, Naval Research Laboratory, Code 6920, Washington, District of Columbia 20375, United States
| | - Andrew T Kerr
- Center for Biomolecular Science and Engineering, Naval Research Laboratory, Code 6920, Washington, District of Columbia 20375, United States
| | - Danni L Harris
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Chunyang Jin
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
9
|
Jorgensen C, Ulmschneider MB, Searson PC. Modeling Substrate Entry into the P-Glycoprotein Efflux Pump at the Blood-Brain Barrier. J Med Chem 2023; 66:16615-16627. [PMID: 38097510 PMCID: PMC12036829 DOI: 10.1021/acs.jmedchem.3c01069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
We report molecular dynamics simulations of rhodamine entry into the central binding cavity of P-gp in the inward open conformation. Rhodamine can enter the inner volume via passive transport across the luminal membrane or lateral diffusion in the lipid bilayer. Entry into the inner volume is determined by the aperture angle at the apex of the protein, with a critical angle of 27° for rhodamine. The central binding cavity has an aqueous phase with a few lipids, which significantly reduces substrate diffusion. Within the central binding cavity, we identified regions with relatively weak binding, suggesting that the combination of reduced mobility and weak substrate binding confines rhodamine to enable the completion of the efflux cycle. Tariquidar, a P-gp inhibitor, aggregates at the lower arms of the P-gp, suggesting that inhibition involves steric hindrance of entry into the inner volume and/or steric hindrance of access of ATP to the nucleotide-binding domains.
Collapse
Affiliation(s)
- Christian Jorgensen
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | - Peter C. Searson
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
10
|
Dahley C, Goss KU, Ebert A. Revisiting the pK a-Flux method for determining intrinsic membrane permeability. Eur J Pharm Sci 2023; 191:106592. [PMID: 37751809 DOI: 10.1016/j.ejps.2023.106592] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/11/2023] [Accepted: 09/23/2023] [Indexed: 09/28/2023]
Abstract
Intrinsic membrane permeability is one of several factors that critically determine the intestinal absorption of a chemical. The intrinsic membrane permeability of a chemical is usually extracted from transwell experiments with Caco-2 or MDCK cells, preferably by the pKa-Flux method, which is considered the method of choice when aqueous boundary layer effects need to be excluded. The pKa-Flux method has two variants, the iso-pH method, where apical and basolateral pH are equal, and the gradient-pH method, where apical and basolateral pH are different. The most commonly used method is the gradient-pH method, as it is intended to reflect the pH-conditions in the gastrointestinal tract. However, concentration-shift effects caused by the applied pH-difference between apical and basolateral compartment in the gradient-pH method have not been considered in the evaluation of the experimental data in the past. Consequently, incorrect intrinsic membrane permeabilities have been determined. In this work, we present a revised method for extracting the intrinsic membrane permeability from gradient-pH data that considers concentration-shift effects in the basolateral aqueous boundary layer and filter as well as in the cytosol. Furthermore, we propose the use of the iso-pH method, where only concentration-shift effects in the cytosol need to be considered, as an alternative to the gradient-pH method. We use the five lipophilic bases amantadine, chloroquine, propranolol, venlafaxine and verapamil as examples to compare gradient-pH method and iso-pH method with regard to the extractability of the intrinsic membrane permeability. For lipophilic bases, the iso-pH method proves to be advantageous. All intrinsic membrane permeabilities determined in this work were substantially higher than the intrinsic membrane permeabilities reported in literature.
Collapse
Affiliation(s)
- Carolin Dahley
- Department of Analytical Environmental Chemistry, Helmholtz Centre for Environmental Research (UFZ), Permoserstraße 15, Leipzig 04318, Germany
| | - Kai-Uwe Goss
- Department of Analytical Environmental Chemistry, Helmholtz Centre for Environmental Research (UFZ), Permoserstraße 15, Leipzig 04318, Germany; Institute of Chemistry, University of Halle-Wittenberg, Kurt-Mothes-Straße 2, Halle 06120, Germany
| | - Andrea Ebert
- Department of Analytical Environmental Chemistry, Helmholtz Centre for Environmental Research (UFZ), Permoserstraße 15, Leipzig 04318, Germany.
| |
Collapse
|
11
|
Jorgensen C, Troendle EP, Ulmschneider JP, Searson PC, Ulmschneider MB. A least-squares-fitting procedure for an efficient preclinical ranking of passive transport across the blood-brain barrier endothelium. J Comput Aided Mol Des 2023; 37:537-549. [PMID: 37573260 PMCID: PMC10505096 DOI: 10.1007/s10822-023-00525-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 07/24/2023] [Indexed: 08/14/2023]
Abstract
The treatment of various disorders of the central nervous system (CNS) is often impeded by the limited brain exposure of drugs, which is regulated by the human blood-brain barrier (BBB). The screening of lead compounds for CNS penetration is challenging due to the biochemical complexity of the BBB, while experimental determination of permeability is not feasible for all types of compounds. Here we present a novel method for rapid preclinical screening of libraries of compounds by utilizing advancements in computing hardware, with its foundation in transition-based counting of the flux. This method has been experimentally validated for in vitro permeabilities and provides atomic-level insights into transport mechanisms. Our approach only requires a single high-temperature simulation to rank a compound relative to a library, with a typical simulation time converging within 24 to 72 h. The method offers unbiased thermodynamic and kinetic information to interpret the passive transport of small-molecule drugs across the BBB.
Collapse
Affiliation(s)
- Christian Jorgensen
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Chemistry, Aarhus University, Langelandsgade 140, 8000, Aarhus C, Denmark.
| | | | | | - Peter C Searson
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
12
|
Langthaler K, Jones CR, Brodin B, Bundgaard C. Assessing extent of brain penetration in vivo (K p,uu,brain) in Göttingen minipig using a diverse set of reference drugs. Eur J Pharm Sci 2023; 190:106554. [PMID: 37543065 DOI: 10.1016/j.ejps.2023.106554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
The application of Göttingen minipigs for non-rodent pharmacokinetics (PK) and drug safety testing has seen a dramatic increase in recent years. The aim of this study was to determine the total and unbound brain-to-plasma ratios (Kp,brain and Kp,uu,brain) for a diverse set of reference compounds in female Göttingen minipigs and compare these with Kp,uu,brain values from other species to assess the suitability of Göttingen minipigs as a model for CNS drug safety testing and brain PK in clinical translation. The reference set consisted of 17 compounds with varying physico-chemical properties and included known human P-glycoprotein (P-gp) substrates. The results of the study showed, that minipig Kp,brain and Kp,uu,brain values for the tested compounds were in the range 0.03-86 and 0.02-2.4 (n = 3-4) respectively. The Kp,uu,brain values were comparable between minipig and rat for a large proportion of the compounds (71% within 2-fold, n = 17). Comparisons of brain penetration across several species for a subset of reference compounds revealed that minipig values were quite similar to those of rat, dog, monkey and human. The study highlighted that the largest Kp,uu,brain species differences were observed for compounds classified as transporter substrates (e.g. cimetidine, risperidone, Way-100635 and altanserin). In conclusion these brain penetration data add substantially to the available literature on PK and drug disposition for minipigs and support use of Göttingen minipig as a non-rodent drug safety model for CNS drug candidates and as a brain PK model for clinical translation.
Collapse
Affiliation(s)
- Kristine Langthaler
- Translational DMPK, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark; CNS Drug Delivery and Barrier Modelling, University of Copenhagen, Universitetsparken 2, 2100 København Ø, Denmark
| | - Christopher R Jones
- Translational DMPK, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark; PKPD Modelling & Simulation, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark.
| | - Birger Brodin
- CNS Drug Delivery and Barrier Modelling, University of Copenhagen, Universitetsparken 2, 2100 København Ø, Denmark
| | | |
Collapse
|
13
|
Zhao N, Chung TD, Guo Z, Jamieson JJ, Liang L, Linville RM, Pessell AF, Wang L, Searson PC. The influence of physiological and pathological perturbations on blood-brain barrier function. Front Neurosci 2023; 17:1289894. [PMID: 37937070 PMCID: PMC10626523 DOI: 10.3389/fnins.2023.1289894] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Abstract
The blood-brain barrier (BBB) is located at the interface between the vascular system and the brain parenchyma, and is responsible for communication with systemic circulation and peripheral tissues. During life, the BBB can be subjected to a wide range of perturbations or stresses that may be endogenous or exogenous, pathological or therapeutic, or intended or unintended. The risk factors for many diseases of the brain are multifactorial and involve perturbations that may occur simultaneously (e.g., two-hit model for Alzheimer's disease) and result in different outcomes. Therefore, it is important to understand the influence of individual perturbations on BBB function in isolation. Here we review the effects of eight perturbations: mechanical forces, temperature, electromagnetic radiation, hypoxia, endogenous factors, exogenous factors, chemical factors, and pathogens. While some perturbations may result in acute or chronic BBB disruption, many are also exploited for diagnostic or therapeutic purposes. The resultant outcome on BBB function depends on the dose (or magnitude) and duration of the perturbation. Homeostasis may be restored by self-repair, for example, via processes such as proliferation of affected cells or angiogenesis to create new vasculature. Transient or sustained BBB dysfunction may result in acute or pathological symptoms, for example, microhemorrhages or hypoperfusion. In more extreme cases, perturbations may lead to cytotoxicity and cell death, for example, through exposure to cytotoxic plaques.
Collapse
Affiliation(s)
- Nan Zhao
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
| | - Tracy D. Chung
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
| | - John J. Jamieson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Lily Liang
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Raleigh M. Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Alex F. Pessell
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Linus Wang
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Peter C. Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
14
|
Noh K, Liu X, Wei C. Optimizing transcardial perfusion of small molecules and biologics for brain penetration and biodistribution studies in rodents. Biopharm Drug Dispos 2023; 44:71-83. [PMID: 35508078 DOI: 10.1002/bdd.2317] [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: 03/18/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 11/06/2022]
Abstract
Efficiently removing blood from the brain vasculature is critical to evaluate accurately the brain penetration and biodistribution of drug candidates, especially for biologics as their blood concentrations are substantially higher than the brain concentrations. Transcardial perfusion has been used widely to remove residual blood in the brain; however, the perfusion conditions (such as the perfusion rate and time) reported in the literature are quite varied, and the performance of these methods on blood removal has not been investigated thoroughly. In this study, the effectiveness of the perfusion conditions was assessed by measuring brain hemoglobin levels. Sodium nitrite (NaNO2 ) as an additive in the perfusate was evaluated at different concentrations. Blood removal was significantly improved with 2% NaNO2 over a 20 min perfusion in mouse without disrupting the integrity of the blood-brain barrier (BBB). In mice, the optimized perfusion method significantly lowered the measured brain-to-plasma ratio (Kp,brain ) for monoclonal antibodies due to the removal of blood contamination and small molecules with a moderate-to-high BBB permeability and with a high brain-unbound-fraction (fu,brain ) presumably due to flux out of the brain during perfusion. Perfusion with or without NaNO2 clearly removed the residual blood in rat brain but with no difference observed in Kp,brain between the perfusion groups with or without 2% NaNO2 . In conclusion, a perfusion method was successfully developed to evaluate the brain penetration of small molecules and biologics in rodents for the first time. The transcardial perfusion with 2% NaNO2 effectively removed the residual blood in the brain and significantly improved the assessment of brain penetration of biologics. For small molecules, however, transcardial perfusion may not be performed, as small molecule compounds could be washed away from the brain by the perfusion procedure.
Collapse
Affiliation(s)
- Keumhan Noh
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, Massachusetts, USA
| | - Xingrong Liu
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, Massachusetts, USA
| | - Cong Wei
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, Massachusetts, USA
| |
Collapse
|
15
|
Ayoup MS, Mansour AF, Abdel-Hamid H, Abu-Serie MM, Mohyeldin SM, Teleb M. Nature-inspired new isoindole-based Passerini adducts as efficient tumor-selective apoptotic inducers via caspase-3/7 activation. Eur J Med Chem 2023; 245:114865. [DOI: 10.1016/j.ejmech.2022.114865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 11/29/2022]
|
16
|
ER-RAJY M, EL FADILI M, MRABTI NN, ZAROUGUI S, ELHALLAOUI M. QSAR, molecular docking, ADMET properties in silico studies for a series of 7-propanamide benzoxaboroles as potent anti-cancer agents. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2022. [DOI: 10.1016/j.cjac.2022.100163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
17
|
Linville RM, Nerenberg RF, Grifno G, Arevalo D, Guo Z, Searson PC. Brain microvascular endothelial cell dysfunction in an isogenic juvenile iPSC model of Huntington's disease. Fluids Barriers CNS 2022; 19:54. [PMID: 35773691 PMCID: PMC9245306 DOI: 10.1186/s12987-022-00347-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 05/13/2022] [Indexed: 11/10/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disease caused by expansion of cytosine-adenine-guanine (CAG) repeats in the huntingtin gene, which leads to neuronal loss and decline in cognitive and motor function. Increasing evidence suggests that blood-brain barrier (BBB) dysfunction may contribute to progression of the disease. Studies in animal models, in vitro models, and post-mortem tissue find that disease progression is associated with increased microvascular density, altered cerebral blood flow, and loss of paracellular and transcellular barrier function. Here, we report on changes in BBB phenotype due to expansion of CAG repeats using an isogenic pair of induced pluripotent stem cells (iPSCs) differentiated into brain microvascular endothelial-like cells (iBMECs). We show that CAG expansion associated with juvenile HD alters the trajectory of iBMEC differentiation, producing cells with ~ two-fold lower percentage of adherent endothelial cells. CAG expansion is associated with diminished transendothelial electrical resistance and reduced tight junction protein expression, but no significant changes in paracellular permeability. While mutant huntingtin protein (mHTT) aggregates were not observed in HD iBMECs, widespread transcriptional dysregulation was observed in iBMECs compared to iPSCs. In addition, CAG expansion in iBMECs results in distinct responses to pathological and therapeutic perturbations including angiogenic factors, oxidative stress, and osmotic stress. In a tissue-engineered BBB model, iBMECs show subtle changes in phenotype, including differences in cell turnover and immune cell adhesion. Our results further support that CAG expansion in BMECs contributes to BBB dysfunction during HD.
Collapse
Affiliation(s)
- Raleigh M Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Renée F Nerenberg
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Gabrielle Grifno
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Diego Arevalo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
18
|
Hu J, Cheng Y, Chen P, Huang Z, Yang L. Caffeine Citrate Protects Against Sepsis-Associated Encephalopathy and Inhibits the UCP2/NLRP3 Axis in Astrocytes. J Interferon Cytokine Res 2022; 42:267-278. [PMID: 35420462 DOI: 10.1089/jir.2021.0241] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a diffuse brain dysfunction without overt central nervous system infection. Caffeine citrate has therapeutic effect on different brain diseases, while its role in SAE remains unclear. The expression levels of interleukin (IL)-18 and IL-1β were upregulated in the cerebrospinal fluid of the subjects. In this study, a rat model of SAE was established by cecal ligation and puncture. Caffeine citrate inhibited SAE-induced neuronal apoptosis and astrocytic activation, decreased reactive oxygen species (ROS) generation, and elevated mitochondrial membrane potential (MMP) level in the cerebral cortex. In vitro, primary astrocytes were isolated from rat cerebral cortex and incubated with lipopolysaccharide (LPS) and interferon-γ (IFN-γ). Caffeine citrate reduced ROS and MMP levels and mitochondrial complex enzyme activities in LPS plus IFN-γ-induced astrocytes. Moreover, caffeine citrate inhibited the activation of nucleotide-binding and oligomerization domain (NOD)-like receptor (NLRP3) inflammasome and decreased the production of IL-1β and IL-18 in vivo and in vitro. Notably, caffeine citrate promoted UCP2 expression in astrocytes. The neuroprotective role of UCP2 has been reported in several experimental brain diseases. These results suggest that caffeine citrate inhibits neuronal apoptosis, astrocytic activation, mitochondrial dysfunction in rat cerebral cortex, thereby alleviating SAE. The protection of caffeine citrate against SAE may be achieved by the UCP2-mediated NLRP3 pathway inhibition in astrocytes.
Collapse
Affiliation(s)
- Jing Hu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Yan Cheng
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Ping Chen
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Zhaoqi Huang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Liqi Yang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
19
|
Gay EA, Guan D, Van Voorhies K, Vasukuttan V, Mathews KM, Besheer J, Jin C. Discovery and Characterization of the First Nonpeptide Antagonists for the Relaxin-3/RXFP3 System. J Med Chem 2022; 65:7959-7974. [PMID: 35594150 DOI: 10.1021/acs.jmedchem.2c00508] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The neuropeptide relaxin-3/RXFP3 system is involved in many important physiological processes such as stress responses, appetite control, and motivation for reward. To date, pharmacological studies of RXFP3 have been limited to peptide ligands. In this study, we report the discovery of the first small-molecule antagonists of RXFP3 through a high-throughput screening campaign. Focused structure-activity relationship studies of the hit compound resulted in RLX-33 (33) that was able to inhibit relaxin-3 activity in a battery of functional assays. RLX-33 is selective for RXFP3 over RXFP1 and RXFP4, two related members in the relaxin/insulin superfamily, and has favorable pharmacokinetic properties for behavioral assessment. When administered to rats intraperitoneally, RLX-33 blocked food intake induced by the RXFP3-selective agonist R3/I5. Collectively, our findings demonstrated that RLX-33 represents a promising antagonist scaffold for the development of drugs targeting the relaxin-3/RXFP3 system.
Collapse
Affiliation(s)
- Elaine A Gay
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Dongliang Guan
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Kalynn Van Voorhies
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Vineetha Vasukuttan
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Kelly M Mathews
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Joyce Besheer
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States.,Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Chunyang Jin
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
20
|
Targeting Transporters for Drug Delivery to the Brain: Can We Do Better? Pharm Res 2022; 39:1415-1455. [PMID: 35359241 PMCID: PMC9246765 DOI: 10.1007/s11095-022-03241-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/21/2022] [Indexed: 12/11/2022]
Abstract
Limited drug delivery to the brain is one of the major reasons for high failure rates of central nervous system (CNS) drug candidates. The blood–brain barrier (BBB) with its tight junctions, membrane transporters, receptors and metabolizing enzymes is a main player in drug delivery to the brain, restricting the entrance of the drugs and other xenobiotics. Current knowledge about the uptake transporters expressed at the BBB and brain parenchymal cells has been used for delivery of CNS drugs to the brain via targeting transporters. Although many transporter-utilizing (pro)drugs and nanocarriers have been developed to improve the uptake of drugs to the brain, their success rate of translation from preclinical development to humans is negligible. In the present review, we provide a systematic summary of the current progress in development of transporter-utilizing (pro)drugs and nanocarriers for delivery of drugs to the brain. In addition, we applied CNS pharmacokinetic concepts for evaluation of the limitations and gaps in investigation of the developed transporter-utilizing (pro)drugs and nanocarriers. Finally, we give recommendations for a rational development of transporter-utilizing drug delivery systems targeting the brain based on CNS pharmacokinetic principles.
Collapse
|
21
|
Husain A, Makadia V, Valicherla GR, Riyazuddin M, Gayen JR. Approaches to minimize the effects of P-glycoprotein in drug transport: A review. Drug Dev Res 2022; 83:825-841. [PMID: 35103340 DOI: 10.1002/ddr.21918] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/21/2021] [Accepted: 01/13/2022] [Indexed: 12/20/2022]
Abstract
P-glycoprotein (P-gp) is a transporter protein that is come under the ATP binding cassette family of proteins. It is situated on the surface of the intestine epithelium, where P-gp substrate binds to the transporter and is pumped into the intestine lumen by the ATP-driven energy-dependent process. In this review, we summarize the role of the P-gp efflux transporter situated on the intestine, the clinical importance of P-gp related drug interactions, and approaches to minimize the effect of P-gp in drug transport. This review also focuses on the impact of P-gp on the bioavailability of the orally administered drug. Many drug's oral bioavailabilities can improve by concomitant use of P-gp inhibitors. Multidrug resistance are reduced by using some naturally occurring compounds obtained from plants and several synthetic P-gp inhibitors. Formulation strategies, one of the most important approaches to mimic the P-gp transporter's action, finally enhancing the oral bioavailability of the drug by inhibiting its P-gp efflux. Vitamin E TPGS, Gelucire 44/14 and other pharmaceutical/formulation excipients inhibit the P-gp efflux. A prodrug approach might be a useful strategy to overcome drug resistance. Prodrug helps to enhance the solubility or alter the pharmacokinetic properties but does not diminish the pharmacological action.
Collapse
Affiliation(s)
- Athar Husain
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vishal Makadia
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India.,Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Raibarelly, India
| | - Guru R Valicherla
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Mohammed Riyazuddin
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Jiaur R Gayen
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
22
|
Jorgensen C, Ulmschneider MB, Searson PC. Atomistic Model of Solute Transport across the Blood-Brain Barrier. ACS OMEGA 2022; 7:1100-1112. [PMID: 35036773 PMCID: PMC8757349 DOI: 10.1021/acsomega.1c05679] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/25/2021] [Indexed: 06/14/2023]
Abstract
The blood-brain barrier remains a major roadblock to the delivery of drugs to the brain. While in vitro and in vivo measurements of permeability are widely used to predict brain penetration, very little is known about the mechanisms of passive transport. Detailed insight into interactions between solutes and cell membranes could provide new insight into drug design and screening. Here, we perform unbiased atomistic MD simulations to visualize translocation of a library of 24 solutes across a lipid bilayer representative of brain microvascular endothelial cells. A temperature bias is used to achieve steady state of all solutes, including those with low permeability. Based on free-energy surface profiles, we show that the solutes can be classified into three groups that describe distinct mechanisms of transport across the bilayer. Simulations down to 310 K for solutes with fast permeability were used to justify the extrapolation of values at 310 K from higher temperatures. Comparison of permeabilities at 310 K to experimental values obtained from in vitro transwell measurements and in situ brain perfusion revealed that permeabilities obtained from simulations vary from close to the experimental values to more than 3 orders of magnitude faster. The magnitude of the difference was dependent on the group defined by free-energy surface profiles. Overall, these results show that MD simulations can provide new insight into the mechanistic details of brain penetration and provide a new approach for drug discovery.
Collapse
Affiliation(s)
- Christian Jorgensen
- Institute
for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | | | - Peter C. Searson
- Institute
for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department
of Materials Science and Engineering, Johns
Hopkins University, Baltimore, Maryland 21218, United States
| |
Collapse
|
23
|
Guan D, Rahman MT, Gay EA, Vasukuttan V, Mathews KM, Decker AM, Williams AH, Zhan CG, Jin C. Indole-Containing Amidinohydrazones as Nonpeptide, Dual RXFP3/4 Agonists: Synthesis, Structure-Activity Relationship, and Molecular Modeling Studies. J Med Chem 2021; 64:17866-17886. [PMID: 34855388 DOI: 10.1021/acs.jmedchem.1c01081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The central relaxin-3/RXFP3 system plays important roles in stress responses, feeding, and motivation for reward. However, exploration of its therapeutic applications has been hampered by the lack of small molecule ligands and the cross-activation of RXFP1 in the brain and RXFP4 in the periphery. Herein, we report the first structure-activity relationship studies of a series of novel nonpeptide amidinohydrazone-based agonists, which were characterized by RXFP3 functional and radioligand binding assays. Several potent and efficacious RXFP3 agonists (e.g., 10d) were identified with EC50 values <10 nM. These compounds also had high potency at RXFP4 but no agonist activity at RXFP1, demonstrating > 100-fold selectivity for RXFP3/4 over RXFP1. In vitro ADME and pharmacokinetic assessments revealed that the amidinohydrazone derivatives may have limited brain permeability. Collectively, our findings provide the basis for further optimization of lead compounds to develop a suitable agonist to probe RXFP3 functions in the brain.
Collapse
Affiliation(s)
- Dongliang Guan
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Md Toufiqur Rahman
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Elaine A Gay
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Vineetha Vasukuttan
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Kelly M Mathews
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Ann M Decker
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Alexander H Williams
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Chunyang Jin
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
24
|
Neumaier F, Zlatopolskiy BD, Neumaier B. Drug Penetration into the Central Nervous System: Pharmacokinetic Concepts and In Vitro Model Systems. Pharmaceutics 2021; 13:1542. [PMID: 34683835 PMCID: PMC8538549 DOI: 10.3390/pharmaceutics13101542] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/22/2022] Open
Abstract
Delivery of most drugs into the central nervous system (CNS) is restricted by the blood-brain barrier (BBB), which remains a significant bottleneck for development of novel CNS-targeted therapeutics or molecular tracers for neuroimaging. Consistent failure to reliably predict drug efficiency based on single measures for the rate or extent of brain penetration has led to the emergence of a more holistic framework that integrates data from various in vivo, in situ and in vitro assays to obtain a comprehensive description of drug delivery to and distribution within the brain. Coupled with ongoing development of suitable in vitro BBB models, this integrated approach promises to reduce the incidence of costly late-stage failures in CNS drug development, and could help to overcome some of the technical, economic and ethical issues associated with in vivo studies in animal models. Here, we provide an overview of BBB structure and function in vivo, and a summary of the pharmacokinetic parameters that can be used to determine and predict the rate and extent of drug penetration into the brain. We also review different in vitro models with regard to their inherent shortcomings and potential usefulness for development of fast-acting drugs or neurotracers labeled with short-lived radionuclides. In this regard, a special focus has been set on those systems that are sufficiently well established to be used in laboratories without significant bioengineering expertise.
Collapse
Affiliation(s)
- Felix Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (B.D.Z.); (B.N.)
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428 Jülich, Germany
| | - Boris D. Zlatopolskiy
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (B.D.Z.); (B.N.)
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428 Jülich, Germany
| | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (B.D.Z.); (B.N.)
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428 Jülich, Germany
| |
Collapse
|
25
|
Rahman MT, Decker AM, Laudermilk L, Maitra R, Ma W, Ben Hamida S, Darcq E, Kieffer BL, Jin C. Evaluation of Amide Bioisosteres Leading to 1,2,3-Triazole Containing Compounds as GPR88 Agonists: Design, Synthesis, and Structure-Activity Relationship Studies. J Med Chem 2021; 64:12397-12413. [PMID: 34387471 PMCID: PMC8395584 DOI: 10.1021/acs.jmedchem.1c01075] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The orphan receptor GPR88 has been implicated in a number of striatal-associated disorders, yet its endogenous ligand has not been discovered. We have previously reported that the amine functionality in the 2-AMPP-derived GPR88 agonists can be replaced with an amide (e.g., 4) without losing activity. Later, we have found that the amide can be replaced with a bioisosteric 1,3,4-oxadiazole with improved potency. Here, we report a further study of amide bioisosteric replacement with a variety of azoles containing three heteroatoms, followed by a focused structure-activity relationship study, leading to the discovery of a series of novel 1,4-disubstituted 1H-1,2,3-triazoles as GPR88 agonists. Collectively, our medicinal chemistry efforts have resulted in a potent, efficacious, and brain-penetrant GPR88 agonist 53 (cAMP EC50 = 14 nM), which is a suitable probe to study GPR88 functions in the brain.
Collapse
Affiliation(s)
- Md Toufiqur Rahman
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Ann M Decker
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Lucas Laudermilk
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Rangan Maitra
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Weiya Ma
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec H4H 1R3, Canada
| | - Sami Ben Hamida
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec H4H 1R3, Canada
- INSERM U1114, University of Strasbourg, Strasbourg 67085, France
| | - Emmanuel Darcq
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec H4H 1R3, Canada
- INSERM U1114, University of Strasbourg, Strasbourg 67085, France
| | - Brigitte L Kieffer
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec H4H 1R3, Canada
- INSERM U1114, University of Strasbourg, Strasbourg 67085, France
| | - Chunyang Jin
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
26
|
Castellino S, Lareau NM, Groseclose MR. The emergence of imaging mass spectrometry in drug discovery and development: Making a difference by driving decision making. JOURNAL OF MASS SPECTROMETRY : JMS 2021; 56:e4717. [PMID: 33724654 PMCID: PMC8365693 DOI: 10.1002/jms.4717] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/25/2021] [Accepted: 03/02/2021] [Indexed: 05/10/2023]
Abstract
The pharmaceutical industry is a dynamic, science-driven business constantly under pressure to innovate and morph into a higher performing organization. Innovations can include the implementation of new technologies, adopting new scientific methods, changing the decision-making process, compressing timelines, or making changes to the organizational structure. The drivers for the constant focus on performance improvement are the high cost of R&D as well as the lengthy timelines required to deliver new medicines for unmet needs. Successful innovations are measured against both the quality and quantity of potential new medicines in the pipeline and the delivery to patients. In this special feature article, we share our collective experience implementing matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS) technology as an innovative approach to better understand the tissue biodistribution of drugs in the early phases of drug discovery to establish pharmacokinetic-pharmacodynamic (PK-PD) relationships, as well as in the development phase to understand pharmacology, toxicology, and disease pathogenesis. In our experience, successful implementation of MALDI IMS in support of therapeutic programs can be measured by the impact IMS studies have on driving decision making in pipeline progression. This provides a direct quantifiable measurement of the return to the organization for the investment in IMS. We have included discussion not only on the technical merits of IMS study conduct but also the key elements of setting study objectives, building collaborations, data integration into the medicine progression milestones, and potential pitfalls when trying to establish IMS in the pharmaceutical arena. We categorized IMS study types into five groups that parallel pipeline progression from the earliest phases of discovery to late stages of preclinical development. We conclude the article with some perspectives on how we see MALDI IMS maintaining relevance and becoming further embedded as an essential tool in the constantly changing environment of the pharmaceutical industry.
Collapse
Affiliation(s)
- Stephen Castellino
- GlaxoSmithKline BioimagingCollegevillePennsylvania19426USA
- Xenovista LLCChapel HillNorth Carolina27516USA
| | | | | |
Collapse
|
27
|
Lumbar cerebrospinal fluid-to-brain extracellular fluid surrogacy is context-specific: insights from LeiCNS-PK3.0 simulations. J Pharmacokinet Pharmacodyn 2021; 48:725-741. [PMID: 34142308 PMCID: PMC8405486 DOI: 10.1007/s10928-021-09768-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 06/01/2021] [Indexed: 11/01/2022]
Abstract
Predicting brain pharmacokinetics is critical for central nervous system (CNS) drug development yet difficult due to ethical restrictions of human brain sampling. CNS pharmacokinetic (PK) profiles are often altered in CNS diseases due to disease-specific pathophysiology. We previously published a comprehensive CNS physiologically-based PK (PBPK) model that predicted the PK profiles of small drugs at brain and cerebrospinal fluid compartments. Here, we improved this model with brain non-specific binding and pH effect on drug ionization and passive transport. We refer to this improved model as Leiden CNS PBPK predictor V3.0 (LeiCNS-PK3.0). LeiCNS-PK3.0 predicted the unbound drug concentrations of brain ECF and CSF compartments in rats and humans with less than two-fold error. We then applied LeiCNS-PK3.0 to study the effect of altered cerebrospinal fluid (CSF) dynamics, CSF volume and flow, on brain extracellular fluid (ECF) pharmacokinetics. The effect of altered CSF dynamics was simulated using LeiCNS-PK3.0 for six drugs and the resulting drug exposure at brain ECF and lumbar CSF were compared. Simulation results showed that altered CSF dynamics changed the CSF PK profiles, but not the brain ECF profiles, irrespective of the drug's physicochemical properties. Our analysis supports the notion that lumbar CSF drug concentration is not an accurate surrogate of brain ECF, particularly in CNS diseases. Systems approaches account for multiple levels of CNS complexity and are better suited to predict brain PK.
Collapse
|
28
|
Islam Y, Leach AG, Smith J, Pluchino S, Coxon CR, Sivakumaran M, Downing J, Fatokun AA, Teixidò M, Ehtezazi T. Physiological and Pathological Factors Affecting Drug Delivery to the Brain by Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2002085. [PMID: 34105297 PMCID: PMC8188209 DOI: 10.1002/advs.202002085] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/06/2021] [Indexed: 05/04/2023]
Abstract
The prevalence of neurological/neurodegenerative diseases, such as Alzheimer's disease is known to be increasing due to an aging population and is anticipated to further grow in the decades ahead. The treatment of brain diseases is challenging partly due to the inaccessibility of therapeutic agents to the brain. An increasingly important observation is that the physiology of the brain alters during many brain diseases, and aging adds even more to the complexity of the disease. There is a notion that the permeability of the blood-brain barrier (BBB) increases with aging or disease, however, the body has a defense mechanism that still retains the separation of the brain from harmful chemicals in the blood. This makes drug delivery to the diseased brain, even more challenging and complex task. Here, the physiological changes to the diseased brain and aged brain are covered in the context of drug delivery to the brain using nanoparticles. Also, recent and novel approaches are discussed for the delivery of therapeutic agents to the diseased brain using nanoparticle based or magnetic resonance imaging guided systems. Furthermore, the complement activation, toxicity, and immunogenicity of brain targeting nanoparticles as well as novel in vitro BBB models are discussed.
Collapse
Affiliation(s)
- Yamir Islam
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Andrew G. Leach
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- Division of Pharmacy and OptometryThe University of ManchesterStopford Building, Oxford RoadManchesterM13 9PTUK
| | - Jayden Smith
- Cambridge Innovation Technologies Consulting (CITC) LimitedSt. John's Innovation CentreCowley RoadCambridgeCB4 0WSUK
| | - Stefano Pluchino
- Department of Clinical NeurosciencesClifford Allbutt Building – Cambridge Biosciences Campus and NIHR Biomedical Research CentreUniversity of CambridgeHills RoadCambridgeCB2 0HAUK
| | - Christopher R. Coxon
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- School of Engineering and Physical SciencesHeriot‐Watt UniversityWilliam Perkin BuildingEdinburghEH14 4ASUK
| | - Muttuswamy Sivakumaran
- Department of HaematologyPeterborough City HospitalEdith Cavell CampusBretton Gate PeterboroughPeterboroughPE3 9GZUK
| | - James Downing
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Amos A. Fatokun
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Meritxell Teixidò
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)Baldiri Reixac 10Barcelona08028Spain
| | - Touraj Ehtezazi
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| |
Collapse
|
29
|
Abdel-Tawab M. Considerations to Be Taken When Carrying Out Medicinal Plant Research-What We Learn from an Insight into the IC 50 Values, Bioavailability and Clinical Efficacy of Exemplary Anti-Inflammatory Herbal Components. Pharmaceuticals (Basel) 2021; 14:437. [PMID: 34066427 PMCID: PMC8148151 DOI: 10.3390/ph14050437] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/23/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Medicinal plants represent a big reservoir for discovering new drugs against all kinds of diseases including inflammation. In spite the large number of promising anti-inflammatory plant extracts and isolated components, research on medicinal plants proves to be very difficult. Based on that background this review aims to provide a summarized insight into the hitherto known pharmacologically active concentrations, bioavailability, and clinical efficacy of boswellic acids, curcumin, quercetin and resveratrol. These examples have in common that the achieved plasma concentrations were found to be often far below the determined IC50 values in vitro. On the other hand demonstrated therapeutic effects suggest a necessity of rethinking our pharmacokinetic understanding. In this light this review discusses the value of plasma levels as pharmacokinetic surrogates in comparison to the more informative value of tissue concentrations. Furthermore the need for new methodological approaches is addressed like the application of combinatorial approaches for identifying and pharmacokinetic investigations of active multi-components. Also the physiological relevance of exemplary in vitro assays and absorption studies in cell-line based models is discussed. All these topics should be ideally considered to avoid inaccurate predictions for the efficacy of herbal components in vivo and to unlock the "black box" of herbal mixtures.
Collapse
Affiliation(s)
- Mona Abdel-Tawab
- Central Laboratory of German Pharmacists, Carl-Mannich-Str. 20, 65760 Eschborn, Germany; ; Tel.: +49-6196-937-955
- Institute of Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| |
Collapse
|
30
|
Najmi A, Wang S, Huang Y, Seefeldt T, Alqahtani Y, Guan X. 2-(2-Cholesteroxyethoxyl)ethyl 3'-S-glutathionylpropionate and its self-assembled micelles for brain delivery: Design, synthesis and evaluation. Int J Pharm 2021; 600:120520. [PMID: 33775725 DOI: 10.1016/j.ijpharm.2021.120520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/07/2021] [Accepted: 03/21/2021] [Indexed: 12/11/2022]
Abstract
The blood-brain barrier (BBB) is a barrier that prevents almost all large and most small exogenous molecules from reaching the brain. The barrier is the major cause of treatment failure for most brain diseases. Extensive efforts have been made to facilitate drug molecules to cross the BBB. One of the approaches is to employ an endogenous ligand or ligand analogue that can enter the brain through its transporter or receptor at the BBB as a brain-targeting agent. Glutathione (GSH) transporters are richly expressed at the BBB with limited presence in other tissues except kidneys. 2-(2-Cholesteroxyethoxyl)ethyl 3'-S-glutathionylpropionate (COXP), formed by connecting GSH with cholesterol through a linker, was designed as a GSH transporter-mediated brain targeting molecule. The amphiphilic nature of COXP enables the molecule to self-assemble to form micelles with a CMC value of 3.9 μM. By using DiR as a fluorescence tracking agent and the whole-body fluorescence imaging technique, the brain distribution of DiR delivered by COXP micelles in mice was 20 folds higher when compared with free DiR. Interestingly, the brain targeting effect was further enhanced by co-administration of GSH. The low CMC value and effective brain targeting make COXP micelles a promising drug delivery system to the brain.
Collapse
Affiliation(s)
- Asim Najmi
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, Box 2202C, South Dakota State University, Brookings, SD 57007, United States
| | - Shenggang Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, Box 2202C, South Dakota State University, Brookings, SD 57007, United States
| | - Yue Huang
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, Box 2202C, South Dakota State University, Brookings, SD 57007, United States
| | - Teresa Seefeldt
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, Box 2202C, South Dakota State University, Brookings, SD 57007, United States
| | - Yahya Alqahtani
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, Box 2202C, South Dakota State University, Brookings, SD 57007, United States
| | - Xiangming Guan
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, Box 2202C, South Dakota State University, Brookings, SD 57007, United States.
| |
Collapse
|
31
|
Saleh MAA, de Lange ECM. Impact of CNS Diseases on Drug Delivery to Brain Extracellular and Intracellular Target Sites in Human: A "WHAT-IF" Simulation Study. Pharmaceutics 2021; 13:95. [PMID: 33451111 PMCID: PMC7828633 DOI: 10.3390/pharmaceutics13010095] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 12/23/2022] Open
Abstract
The blood-brain barrier (BBB) is equipped with unique physical and functional processes that control central nervous system (CNS) drug transport and the resulting concentration-time profiles (PK). In CNS diseases, the altered BBB and CNS pathophysiology may affect the CNS PK at the drug target sites in the brain extracellular fluid (brainECF) and intracellular fluid (brainICF) that may result in changes in CNS drug effects. Here, we used our human CNS physiologically-based PK model (LeiCNS-PK3.0) to investigate the impact of altered cerebral blood flow (CBF), tight junction paracellular pore radius (pararadius), brainECF volume, and pH of brainECF (pHECF) and of brainICF (pHICF) on brainECF and brainICF PK for 46 small drugs with distinct physicochemical properties. LeiCNS-PK3.0 simulations showed a drug-dependent effect of the pathophysiological changes on the rate and extent of BBB transport and on brainECF and brainICF PK. Altered pararadius, pHECF, and pHICF affected both the rate and extent of BBB drug transport, whereas changes in CBF and brainECF volume modestly affected the rate of BBB drug transport. While the focus is often on BBB paracellular and active transport processes, this study indicates that also changes in pH should be considered for their important implications on brainECF and brainICF target site PK.
Collapse
Affiliation(s)
| | - Elizabeth C. M. de Lange
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands;
| |
Collapse
|
32
|
Rahman MT, Decker AM, Langston TL, Mathews KM, Laudermilk L, Maitra R, Ma W, Darcq E, Kieffer BL, Jin C. Design, Synthesis, and Structure-Activity Relationship Studies of (4-Alkoxyphenyl)glycinamides and Bioisosteric 1,3,4-Oxadiazoles as GPR88 Agonists. J Med Chem 2020; 63:14989-15012. [PMID: 33205975 PMCID: PMC7737621 DOI: 10.1021/acs.jmedchem.0c01581] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Increasing evidence implicates the orphan G protein-coupled receptor 88 (GPR88) in a number of striatal-associated disorders. In this study, we report the design and synthesis of a series of novel (4-alkoxyphenyl)glycinamides (e.g., 31) and the corresponding 1,3,4-oxadiazole bioisosteres derived from the 2-AMPP scaffold (1) as GPR88 agonists. The 5-amino-1,3,4-oxadiazole derivatives (84, 88-90) had significantly improved potency and lower lipophilicity compared to 2-AMPP. Compound 84 had an EC50 of 59 nM in the GPR88 overexpressing cell-based cAMP assay. In addition, 84 had an EC50 of 942 nM in the [35S]GTPγS binding assay using mouse striatal membranes but was inactive in membranes from GPR88 knockout mice, even at a concentration of 100 μM. In vivo pharmacokinetic testing of 90 in rats revealed that the 5-amino-1,3,4-oxadiazole analogues may have limited brain permeability. Taken together, these results provide the basis for further optimization to develop a suitable agonist to probe GPR88 functions in the brain.
Collapse
Affiliation(s)
- Md Toufiqur Rahman
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina, 27709, United States
| | - Ann M. Decker
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina, 27709, United States
| | - Tiffany L. Langston
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina, 27709, United States
| | - Kelly M. Mathews
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina, 27709, United States
| | - Lucas Laudermilk
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina, 27709, United States
| | - Rangan Maitra
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina, 27709, United States
| | - Weiya Ma
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec H4H 1R3, Canada
| | - Emmanuel Darcq
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec H4H 1R3, Canada,INSERM U1114, University of Strasbourg, Strasbourg 67085, France
| | - Brigitte L. Kieffer
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec H4H 1R3, Canada,INSERM U1114, University of Strasbourg, Strasbourg 67085, France
| | - Chunyang Jin
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina, 27709, United States,Corresponding author: Dr. Chunyang Jin, Research Triangle Institute, Post Office Box 12194, Research Triangle Park, NC 27709, Telephone: 919 541-6328, Fax: 919 541-8868,
| |
Collapse
|
33
|
Vendel E, Rottschäfer V, de Lange ECM. A 3D brain unit model to further improve prediction of local drug distribution within the brain. PLoS One 2020; 15:e0238397. [PMID: 32966285 PMCID: PMC7511021 DOI: 10.1371/journal.pone.0238397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 08/15/2020] [Indexed: 12/14/2022] Open
Abstract
The development of drugs targeting the brain still faces a high failure rate. One of the reasons is a lack of quantitative understanding of the complex processes that govern the pharmacokinetics (PK) of a drug within the brain. While a number of models on drug distribution into and within the brain is available, none of these addresses the combination of factors that affect local drug concentrations in brain extracellular fluid (brain ECF). Here, we develop a 3D brain unit model, which builds on our previous proof-of-concept 2D brain unit model, to understand the factors that govern local unbound and bound drug PK within the brain. The 3D brain unit is a cube, in which the brain capillaries surround the brain ECF. Drug concentration-time profiles are described in both a blood-plasma-domain and a brain-ECF-domain by a set of differential equations. The model includes descriptions of blood plasma PK, transport through the blood-brain barrier (BBB), by passive transport via paracellular and transcellular routes, and by active transport, and drug binding kinetics. The impact of all these factors on ultimate local brain ECF unbound and bound drug concentrations is assessed. In this article we show that all the above mentioned factors affect brain ECF PK in an interdependent manner. This indicates that for a quantitative understanding of local drug concentrations within the brain ECF, interdependencies of all transport and binding processes should be understood. To that end, the 3D brain unit model is an excellent tool, and can be used to build a larger network of 3D brain units, in which the properties for each unit can be defined independently to reflect local differences in characteristics of the brain.
Collapse
Affiliation(s)
- Esmée Vendel
- Mathematical Institute, Leiden University, Leiden, The Netherlands
| | - Vivi Rottschäfer
- Mathematical Institute, Leiden University, Leiden, The Netherlands
- * E-mail: (VR); (EL)
| | - Elizabeth C. M. de Lange
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- * E-mail: (VR); (EL)
| |
Collapse
|
34
|
Linville RM, DeStefano JG, Nerenberg RF, Grifno GN, Ye R, Gallagher E, Searson PC. Long-Term Cryopreservation Preserves Blood-Brain Barrier Phenotype of iPSC-Derived Brain Microvascular Endothelial Cells and Three-Dimensional Microvessels. Mol Pharm 2020; 17:3425-3434. [PMID: 32787285 PMCID: PMC9923881 DOI: 10.1021/acs.molpharmaceut.0c00484] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Brain microvascular endothelial cells derived from induced pluripotent stem cells (dhBMECs) are a scalable and reproducible resource for studies of the human blood-brain barrier, including mechanisms and strategies for drug delivery. Confluent monolayers of dhBMECs recapitulate key in vivo functions including tight junctions to limit paracellular permeability and efflux and nutrient transport to regulate transcellular permeability. Techniques for cryopreservation of dhBMECs have been reported; however, functional validation studies after long-term cryopreservation have not been extensively performed. Here, we characterize dhBMECs after 1 year of cryopreservation using selective purification on extracellular matrix-treated surfaces and ROCK inhibition. One-year cryopreserved dhBMECs maintain functionality of tight junctions, efflux pumps, and nutrient transporters with stable protein localization and gene expression. Cryopreservation is associated with a decrease in the yield of adherent cells and unique responses to cell stress, resulting in altered paracellular permeability of Lucifer yellow. Additionally, cryopreserved dhBMECs reliably form functional three-dimensional microvessels independent of cryopreservation length, with permeabilities lower than non-cryopreserved two-dimensional models. Long-term cryopreservation of dhBMECs offers key advantages including increased scalability, reduced batch-to-batch effects, the ability to conduct well-controlled follow up studies, and support of multisite collaboration from the same cell stock, all while maintaining phenotype for screening pharmaceutical agents.
Collapse
Affiliation(s)
- Raleigh M. Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
| | - Jackson G. DeStefano
- Institute for Nanobiotechnology and Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Reneé F. Nerenberg
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
| | - Gabrielle N. Grifno
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
| | - Robert Ye
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Erin Gallagher
- Institute for Nanobiotechnology and Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Peter C. Searson
- Institute for Nanobiotechnology and Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
| |
Collapse
|
35
|
Nicolaï J, Chapy H, Gillent E, Saunders K, Ungell AL, Nicolas JM, Chanteux H. Impact of In Vitro Passive Permeability in a P-gp-transfected LLC-PK1 Model on the Prediction of the Rat and Human Unbound Brain-to-Plasma Concentration Ratio. Pharm Res 2020; 37:175. [PMID: 32856111 DOI: 10.1007/s11095-020-02867-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/24/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE More accurate prediction of the extent of drug brain exposure in early drug discovery and understanding potential species differences could help to guide medicinal chemistry and avoid unnecessary animal studies. Hence, the aim of the current study was to validate the use of a P-gp transfected LLC-PK1 model to predict the unbound brain-to-plasma concentration ratio (Kpuu,brain) in rats and humans. METHODS MOCK-, Mdr1a- and MDR1-transfected LLC-PK1 monolayers were applied in a transwell setup to quantify the bidirectional transport for 12 specific P-gp substrates, 48 UCB drug discovery compounds, 11 compounds with reported rat in situ brain perfusion data and 6 compounds with reported human Kpuu,brain values. The in vitro transport data were introduced in a minimal PBPK model (SIVA®) to determine the transport parameters. These parameters were combined with the differences between in vitro and in vivo passive permeability as well as P-gp expression levels (as determined by LC-MS/MS), to predict the Kpuu,brain. RESULTS A 10-fold difference between in vitro and in vivo passive permeability was observed. Incorporation of the differences between in vitro and in vivo passive permeability and P-gp expression levels resulted in an improved prediction of rat (AAFE 2.17) and human Kpuu,brain (AAFE 2.10). CONCLUSIONS We have succesfully validated a methodology to use a P-gp overexpressing LLC-PK1 cell line to predict both rat and human Kpuu,brain by correcting for both passive permeability and P-gp expression levels.
Collapse
Affiliation(s)
- Johan Nicolaï
- Development Science, UCB Biopharma SRL, Chemin du Foriest, B1420, Braine-l'Alleud, Belgium.
| | - Hélène Chapy
- Development Science, UCB Biopharma SRL, Chemin du Foriest, B1420, Braine-l'Alleud, Belgium
| | - Eric Gillent
- Development Science, UCB Biopharma SRL, Chemin du Foriest, B1420, Braine-l'Alleud, Belgium
| | - Kenneth Saunders
- Development Science, UCB Biopharma SRL, Chemin du Foriest, B1420, Braine-l'Alleud, Belgium
| | - Anna-Lena Ungell
- Development Science, UCB Biopharma SRL, Chemin du Foriest, B1420, Braine-l'Alleud, Belgium
| | - Jean-Marie Nicolas
- Development Science, UCB Biopharma SRL, Chemin du Foriest, B1420, Braine-l'Alleud, Belgium
| | - Hugues Chanteux
- Development Science, UCB Biopharma SRL, Chemin du Foriest, B1420, Braine-l'Alleud, Belgium
| |
Collapse
|
36
|
Yamazaki M, Honda S, Tamaki K, Irie M, Mihara T. Effects of (+)-bicuculline, a GABAa receptor antagonist, on auditory steady state response in free-moving rats. PLoS One 2020; 15:e0236363. [PMID: 32706815 PMCID: PMC7380603 DOI: 10.1371/journal.pone.0236363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 07/03/2020] [Indexed: 12/11/2022] Open
Abstract
Auditory steady-state responses (ASSRs) are states in which the electrical activity of the brain reacts steadily to repeated auditory stimuli. They are known to be useful for testing the functional integrity of neural circuits in the cortex, as well as for their capacity to generate synchronous activity in both human and animal models. Furthermore, abnormal gamma oscillations on ASSR are typically observed in patients with schizophrenia (SZ). Changes in neural synchrony may reflect aberrations in cortical gamma-aminobutyric acid (GABA) neurotransmission. However, GABA’s impact and effects related to ASSR are still unclear. Here, we examined the effect of a GABAa receptor antagonist, (+)-bicuculline, on ASSR in free-moving rats. (+)-Bicuculline (1, 2 and 4 mg/kg, sc) markedly and dose-dependently reduced ASSR signals, consistent with current hypotheses. In particular, (+)-bicuculline significantly reduced event-related spectral perturbations (ERSPs) at 2 and 4 mg/kg between 10 and 30 minutes post-dose. Further, bicuculline (2 and 4 mg/kg) significantly and dose-dependently increased baseline gamma power. Furthermore, the occurrence of convulsions was consistent with the drug’s pharmacokinetics. For example, high doses of (+)-bicuculline such as those greater than 880 ng/g in the brain induced convulsion. Additionally, time-dependent changes in ERSP with (+)-bicuculline were observed in accordance with drug concentration. This study partially unraveled the contribution of GABAa receptor signals to the generation of ASSR.
Collapse
Affiliation(s)
- Mayako Yamazaki
- Department of Neuroscience, Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
- * E-mail:
| | - Sokichi Honda
- Department of Neuroscience, Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - Keisuke Tamaki
- Department of Neuroscience, Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - Megumi Irie
- Analysis & Pharmacokinetics Research Labs., Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - Takuma Mihara
- Department of Neuroscience, Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| |
Collapse
|
37
|
Nirogi R, Molgara P, Bhyrapuneni G, Manoharan A, Padala NP, Palacharla VRC. The use of inactivated brain homogenate to determine the in vitro fraction unbound in brain for unstable compounds. Xenobiotica 2020; 50:1228-1235. [DOI: 10.1080/00498254.2020.1771795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Ramakrishna Nirogi
- Drug Metabolism and Pharmacokinetics, Suven Life Sciences Limited, Hyderabad, India
| | | | - Gopinadh Bhyrapuneni
- Drug Metabolism and Pharmacokinetics, Suven Life Sciences Limited, Hyderabad, India
| | - Arunkumar Manoharan
- Drug Metabolism and Pharmacokinetics, Suven Life Sciences Limited, Hyderabad, India
| | | | | |
Collapse
|
38
|
Komin A, Bogorad MI, Lin R, Cui H, Searson PC, Hristova K. A peptide for transcellular cargo delivery: Structure-function relationship and mechanism of action. J Control Release 2020; 324:633-643. [PMID: 32474121 DOI: 10.1016/j.jconrel.2020.05.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/20/2020] [Indexed: 12/23/2022]
Abstract
The rate of transport of small molecule drugs across biological barriers, such as the blood-brain barrier, is often a limiting factor in achieving a therapeutic dose. One proposed strategy to enhance delivery across endothelial or epithelial monolayers is conjugation to cell-penetrating peptides (CPPs); however, very little is known about the design of CPPs for efficient transcellular transport. Here, we report on transcellular transport of a CPP, designated the CL peptide, that increases the delivery of small-molecule cargoes across model epithelium approximately 10-fold. The CL peptide contains a helix-like motif and a polyarginine tail. We investigated the effect of cargo, helix-like motif sequence, polyarginine tail length, and peptide stereochemistry on cargo delivery. We showed that there is an optimal helix-like motif sequence (RLLRLLR) and polyarginine tail length (R7) for cargo delivery. Furthermore, we demonstrated that the peptide-cargo conjugate is cleaved by cells in the epithelium at the site of a two-amino acid linker. The cleavage releases the cargo with the N-terminal linker amino acid from the peptide prior to transport out of the epithelium. These studies provide new insight into the sequence requirements for developing novel CPPs for transcellular delivery of cargo.
Collapse
Affiliation(s)
- Alexander Komin
- Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Maxim I Bogorad
- Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Ran Lin
- Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Honggang Cui
- Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA.
| | - Kalina Hristova
- Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA.
| |
Collapse
|
39
|
GM1 Oligosaccharide Crosses the Human Blood-Brain Barrier In Vitro by a Paracellular Route. Int J Mol Sci 2020; 21:ijms21082858. [PMID: 32325905 PMCID: PMC7215935 DOI: 10.3390/ijms21082858] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 01/08/2023] Open
Abstract
Ganglioside GM1 (GM1) has been reported to functionally recover degenerated nervous system in vitro and in vivo, but the possibility to translate GM1′s potential in clinical settings is counteracted by its low ability to overcome the blood–brain barrier (BBB) due to its amphiphilic nature. Interestingly, the soluble and hydrophilic GM1-oligosaccharide (OligoGM1) is able to punctually replace GM1 neurotrophic functions alone, both in vitro and in vivo. In order to take advantage of OligoGM1 properties, which overcome GM1′s pharmacological limitations, here we characterize the OligoGM1 brain transport by using a human in vitro BBB model. OligoGM1 showed a 20-fold higher crossing rate than GM1 and time–concentration-dependent transport. Additionally, OligoGM1 crossed the barrier at 4 °C and in inverse transport experiments, allowing consideration of the passive paracellular route. This was confirmed by the exclusion of a direct interaction with the active ATP-binding cassette (ABC) transporters using the “pump out” system. Finally, after barrier crossing, OligoGM1 remained intact and able to induce Neuro2a cell neuritogenesis by activating the TrkA pathway. Importantly, these in vitro data demonstrated that OligoGM1, lacking the hydrophobic ceramide, can advantageously cross the BBB in comparison with GM1, while maintaining its neuroproperties. This study has improved the knowledge about OligoGM1′s pharmacological potential, offering a tangible therapeutic strategy.
Collapse
|
40
|
Tsume Y, Igawa N, Drelich AJ, Ruan H, Amidon GE, Amidon GL. The in vivo predictive dissolution for immediate release dosage of donepezil and danazol, BCS class IIc drugs, with the GIS and the USP II with biphasic dissolution apparatus. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.01.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
41
|
Transport Studies Using Blood-Brain Barrier In Vitro Models: A Critical Review and Guidelines. Handb Exp Pharmacol 2020; 273:187-204. [PMID: 33037909 DOI: 10.1007/164_2020_394] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Permeation is one of the most evaluated parameters using preclinical in vitro blood-brain barrier models, as it has long been considered to be one of the major factors influencing central nervous system drug delivery. Blood-brain barrier permeability can be defined as the speed at which a compound crosses the brain endothelial cell barrier and is employed to assess barrier tightness, which is a crucial feature of brain capillaries in vivo. In addition, it is used to assess brain drug penetration. We review traditionally used methods to assess blood-brain barrier permeability in vitro and summarize often neglected in vivo (e.g., plasma protein and brain tissue binding) or in vitro (e.g., culture insert materials or methodology) factors that influence this property. These factors are crucial to consider when performing BBB permeability assessments, and especially when comparing permeability data obtained from different models, since model diversification significantly complicates inter-study comparisons. Finally, measuring transendothelial electrical resistance can be used to describe blood-brain barrier tightness; however, several parameters should be considered while comparing these measurements to the blood-brain barrier permeability to paracellular markers.
Collapse
|
42
|
Quantification and assessment of detection capability in imaging mass spectrometry using a revised mimetic tissue model. Bioanalysis 2019; 11:1099-1116. [PMID: 31251106 DOI: 10.4155/bio-2019-0035] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: A revised method of preparing the mimetic tissue model for quantitative imaging mass spectrometry (IMS) is evaluated. Concepts of assessing detection capability are adapted from other imaging or mass spectrometry (MS)-based technologies to improve upon the reliability of IMS quantification. Materials & methods: The mimetic tissue model is prepared by serially freezing spiked-tissue homogenates into a cylindrical mold to create a plug of tissue with a stepped concentration gradient of matrix-matched standards. Weighted least squares (WLS) linear regression is applied due to the heteroscedastisity (change in variance with intensity) of most MS data. Results & conclusions: Imaging poses several caveats for quantification which are unique compared with other MS-based methods. Aspects of the design, construction, application, and evaluation of the matrix-matched standard curve for the mimetic tissue model are discussed. In addition, the criticality of the ion distribution in the design of a purposeful liquid chromatography coupled to mass spectrometry (LC-MS) validation is reviewed.
Collapse
|
43
|
Kim M, Laramy JK, Gampa G, Parrish KE, Brundage R, Sarkaria JN, Elmquist WF. Brain Distributional Kinetics of a Novel MDM2 Inhibitor SAR405838: Implications for Use in Brain Tumor Therapy. Drug Metab Dispos 2019; 47:1403-1414. [PMID: 31619389 PMCID: PMC7042719 DOI: 10.1124/dmd.119.088716] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 10/01/2019] [Indexed: 01/10/2023] Open
Abstract
Achieving an effective drug concentration in the brain is as important as targeting the right pathway when developing targeted agents for brain tumors. SAR405838 is a novel molecularly targeted agent that is in clinical trials for various solid tumors. Its application for tumors in the brain has not yet been examined, even though the target, the MDM2-p53 interaction, is attractive for tumors that could occur in the brain, including glioblastoma and brain metastases. In vitro and in vivo studies indicate that SAR405838 is a substrate of P-glycoprotein (P-gp). P-gp mediated active efflux at the blood-brain barrier plays a dominant role in limiting SAR405838 brain distribution. Even though the absence of P-gp significantly increases the drug exposure in the brain, the systemic exposure, including absorption and clearance processes, were unaffected by P-gp deletion. Model-based parameters of SAR405838 distribution across the blood-brain barrier indicate the CLout of the brain was approximately 40-fold greater than the CLin The free fraction of SAR405838 in plasma and brain were found to be low, and subsequent Kpuu values were less than unity, even in P-gp/Bcrp knockout mice. These results indicate additional efflux transporters other than P-gp and Bcrp may be limiting distribution of SAR405838 to the brain. Concomitant administration of elacridar significantly increased brain exposure, also without affecting the systemic exposure. This study characterized the brain distributional kinetics of SAR405838, a novel MDM2 inhibitor, to evaluate its potential in the treatment of primary and metastatic brain tumors. SIGNIFICANCE STATEMENT: This paper examined the brain distributional kinetics of a novel MDM2-p53 targeted agent, SAR405838, to see its possible application for brain tumors by using in vitro, in vivo, and in silico approaches. SAR405838 is found to be a substrate of P-glycoprotein (P-gp), which limits its distribution to the brain. Based on the findings in the paper, manipulation of the function of P-gp can significantly increase the brain exposure of SAR405838, which may give an insight on its potential benefit as a treatment for primary and metastatic brain cancer.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- Animals
- Biological Transport
- Blood-Air Barrier/metabolism
- Brain/metabolism
- Brain Neoplasms/drug therapy
- Brain Neoplasms/metabolism
- Dogs
- Humans
- Indoles/blood
- Indoles/pharmacokinetics
- Indoles/pharmacology
- Indoles/therapeutic use
- Madin Darby Canine Kidney Cells
- Mice, Transgenic
- Models, Biological
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors
- Spiro Compounds/blood
- Spiro Compounds/pharmacokinetics
- Spiro Compounds/pharmacology
- Spiro Compounds/therapeutic use
- Tissue Distribution
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Minjee Kim
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (M.K., J.K.L., G.G., K.E.P., W.F.E.); Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota (R.B.); and Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Janice K Laramy
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (M.K., J.K.L., G.G., K.E.P., W.F.E.); Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota (R.B.); and Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Gautham Gampa
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (M.K., J.K.L., G.G., K.E.P., W.F.E.); Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota (R.B.); and Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Karen E Parrish
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (M.K., J.K.L., G.G., K.E.P., W.F.E.); Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota (R.B.); and Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Richard Brundage
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (M.K., J.K.L., G.G., K.E.P., W.F.E.); Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota (R.B.); and Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Jann N Sarkaria
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (M.K., J.K.L., G.G., K.E.P., W.F.E.); Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota (R.B.); and Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - William F Elmquist
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (M.K., J.K.L., G.G., K.E.P., W.F.E.); Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota (R.B.); and Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| |
Collapse
|
44
|
Structure-activity relationship studies of (E)-3,4-dihydroxystyryl alkyl sulfones as novel neuroprotective agents based on improved antioxidant, anti-inflammatory activities and BBB permeability. Eur J Med Chem 2019; 171:420-433. [DOI: 10.1016/j.ejmech.2019.03.044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 02/25/2019] [Accepted: 03/17/2019] [Indexed: 01/30/2023]
|
45
|
Development of Human in vitro Brain-blood Barrier Model from Induced Pluripotent Stem Cell-derived Endothelial Cells to Predict the in vivo Permeability of Drugs. Neurosci Bull 2019; 35:996-1010. [PMID: 31079318 DOI: 10.1007/s12264-019-00384-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/02/2019] [Indexed: 10/26/2022] Open
Abstract
An in vitro blood-brain barrier (BBB) model is critical for enabling rapid screening of the BBB permeability of the drugs targeting on the central nervous system. Though many models have been developed, their reproducibility and renewability remain a challenge. Furthermore, drug transport data from many of the models do not correlate well with the data for in vivo BBB drug transport. Induced-pluripotent stem cell (iPSC) technology provides reproducible cell resources for in vitro BBB modeling. Here, we generated a human in vitro BBB model by differentiating the human iPSC (hiPSC) line GM25256 into brain endothelial-type cells. The model displayed BBB characteristics including tight junction proteins (ZO-1, claudin-5, and occludin) and endothelial markers (von Willebrand factor and Ulex), as well as high trans-endothelial electrical resistance (TEER) (1560 Ω.cm2 ± 230 Ω.cm2) and γ-GTPase activity. Co-culture with primary rat astrocytes significantly increased the TEER of the model (2970 Ω.cm2 to 4185 Ω.cm2). RNAseq analysis confirmed the expression of key BBB-related genes in the hiPSC-derived endothelial cells in comparison with primary human brain microvascular endothelial cells, including P-glycoprotein (Pgp) and breast cancer resistant protein (BCRP). Drug transport assays for nine CNS compounds showed that the permeability of non-Pgp/BCRP and Pgp/BCRP substrates across the model was strongly correlated with rodent in situ brain perfusion data for these compounds (R2 = 0.982 and R2 = 0.9973, respectively), demonstrating the functionality of the drug transporters in the model. Thus, this model may be used to rapidly screen CNS compounds, to predict the in vivo BBB permeability of these compounds and to study the biology of the BBB.
Collapse
|
46
|
Mattei C, Taly A, Soualah Z, Saulais O, Henrion D, Guérineau NC, Verleye M, Legros C. Involvement of the GABA A receptor α subunit in the mode of action of etifoxine. Pharmacol Res 2019; 145:104250. [PMID: 31059790 DOI: 10.1016/j.phrs.2019.04.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/21/2019] [Accepted: 04/27/2019] [Indexed: 12/31/2022]
Abstract
Etifoxine (EFX) is a non-benzodiazepine psychoactive drug which exhibits anxiolytic effects through a dual mechanism, by directly binding to GABAA receptors (GABAARs) and to the mitochondrial 18-kDa translocator protein, resulting in the potentiation of the GABAergic function. The β subunit subtype plays a key role in the EFX-GABAAR interaction, however this does not explain the anxiolytic effects of this drug. Here, we combined behavioral and electrophysiological experiments to challenge the role of the GABAAR α subunit in the EFX mode of action. After single administrations of anxiolytic doses (25-50 mg/kg, intraperitoneal), EFX did not induce any neurological nor locomotor impairments, unlike the benzodiazepine bromazepam (0.5-1 mg/kg, intraperitoneal). We established the EFX pharmacological profile on heteropentameric GABAARs constructed with α1 to α6 subunit expressed in Xenopus oocyte. Unlike what is known for benzodiazepines, neither the γ nor δ subunits influenced EFX-mediated potentiation of GABA-evoked currents. EFX acted first as a partial agonist on α2β3γ2S, α3β3γ2S, α6β3γ2S and α6β3δ GABAARs, but not on α1β3γ2S, α4β3γ2S, α4β3δ nor α5β3γ2S GABAARs. Moreover, EFX exhibited much higher positive allosteric modulation towards α2β3γ2S, α3β3γ2S and α6β3γ2S than for α1β3γ2S, α4β3γ2S and α5β3γ2S GABAARs. At 20 μM, corresponding to brain concentration at anxiolytic doses, EFX increased GABA potency to the highest extent for α3β3γ2S GABAARs. We built a docking model of EFX on α3β3γ2S GABAARs, which is consistent with a binding site located between α and β subunits in the extracellular domain. In conclusion, EFX preferentially potentiates α2β3γ2S and α3β3γ2S GABAARs, which might support its advantageous anxiolytic/sedative balance.
Collapse
Affiliation(s)
- César Mattei
- Institut MITOVASC, UMR CNRS 6015 - UMR INSERM U1083, Université d'Angers, 3 Rue Roger Amsler 49100 ANGERS, France.
| | - Antoine Taly
- Theoretical Biochemistry Laboratory, Institute of Physico-Chemical Biology, CNRS UPR9080, University of Paris Diderot Sorbonne Paris Cité, 75005 Paris, France
| | - Zineb Soualah
- Institut MITOVASC, UMR CNRS 6015 - UMR INSERM U1083, Université d'Angers, 3 Rue Roger Amsler 49100 ANGERS, France
| | - Ophélie Saulais
- Institut MITOVASC, UMR CNRS 6015 - UMR INSERM U1083, Université d'Angers, 3 Rue Roger Amsler 49100 ANGERS, France
| | - Daniel Henrion
- Institut MITOVASC, UMR CNRS 6015 - UMR INSERM U1083, Université d'Angers, 3 Rue Roger Amsler 49100 ANGERS, France
| | - Nathalie C Guérineau
- Institut MITOVASC, UMR CNRS 6015 - UMR INSERM U1083, Université d'Angers, 3 Rue Roger Amsler 49100 ANGERS, France
| | - Marc Verleye
- Biocodex, Department of Pharmacology, Zac de Mercières, 60200 Compiègne, France
| | - Christian Legros
- Institut MITOVASC, UMR CNRS 6015 - UMR INSERM U1083, Université d'Angers, 3 Rue Roger Amsler 49100 ANGERS, France.
| |
Collapse
|
47
|
Feng S, Zheng L, Tang S, Gu J, Jiang X, Wang L. In-vitro and in situ assessment of the efflux of five antidepressants by breast cancer resistance protein. J Pharm Pharmacol 2019; 71:1133-1141. [PMID: 31037729 DOI: 10.1111/jphp.13100] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 04/07/2019] [Indexed: 01/08/2023]
Abstract
Abstract
Objectives
Antidepressants need to penetrate the blood–brain barrier (BBB) to exert their functions in the central nervous system. Breast cancer resistance protein (BCRP), an efflux transporter abundantly expressed in the BBB, prevents the accumulation of many drugs in the brain. This study aimed to identify whether five commonly used antidepressants (sertraline, duloxetine, fluoxetine, amitriptyline and mirtazapine) are BCRP substrates.
Methods
A combination of bidirectional transport and intracellular accumulation experiments was conducted on BCRP-overexpressing MDCKII and wild-type (WT) cells, and in situ brain perfusion was conducted in rats.
Key findings
The bidirectional transport study revealed that the net efflux ratio (NER) of sertraline reached 2.08 but decreased to 1.06 when co-incubated with Ko143, a selective BCRP inhibitor. Conversely, the other four antidepressants did not appear to be BCRP substrates, due to their low NER values (<1.5). The accumulation of sertraline in MDCKII-BCRP cells was significantly lower than that in MDCKII-WT cells. The presence of Ko143 significantly increased the sertraline accumulation in MDCKII-BCRP cells but not in MDCKII-WT cells. Brain perfusion showed that the permeability of 1 and 5 μm sertraline was significantly higher in the presence of Ko143.
Conclusions
Taken together, BCRP is involved in sertraline efflux.
Collapse
Affiliation(s)
- Suqin Feng
- Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Liang Zheng
- Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Shiwei Tang
- Department of Pharmacy, People's Hospital of Dujiangyan City, Dujiangyan, China
| | - Juan Gu
- Department of Pharmacy, Affiliated Hospital of Zunyi Medical College, Zunyi, China
| | - Xuehua Jiang
- Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Ling Wang
- Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
48
|
An experimentally validated approach to calculate the blood-brain barrier permeability of small molecules. Sci Rep 2019; 9:6117. [PMID: 30992465 PMCID: PMC6467875 DOI: 10.1038/s41598-019-42272-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/18/2019] [Indexed: 01/31/2023] Open
Abstract
Drug development for the treatment of central nervous system (CNS) diseases is extremely challenging, in large part due to the difficulty in crossing the blood-brain barrier (BBB). Here we develop and experimentally validate a new in silico method to predict quantitatively the BBB permeability for small-molecule drugs. We show accurate prediction of solute permeabilities at physiological temperature using high-temperature unbiased atomic detail molecular dynamics simulations of spontaneous drug diffusion across BBB bilayers. These simulations provide atomic detail insights into the transport mechanisms, as well as converged kinetics and thermodynamics. The method is validated computationally against physiological temperature simulations for fast-diffusing compounds, as well as experimentally by direct determination of the compound permeabilities using a transwell assay as an in vitro BBB model. The overall agreement of the predicted values with both direct simulations at physiological temperatures and experimental data is excellent. This new tool has the potential to replace current semi-empirical in silico screening and in vitro permeability measurements in CNS drug discovery.
Collapse
|
49
|
Di Marco A, Gonzalez Paz O, Fini I, Vignone D, Cellucci A, Battista MR, Auciello G, Orsatti L, Zini M, Monteagudo E, Khetarpal V, Rose M, Dominguez C, Herbst T, Toledo-Sherman L, Summa V, Muñoz-Sanjuán I. Application of an in Vitro Blood–Brain Barrier Model in the Selection of Experimental Drug Candidates for the Treatment of Huntington’s Disease. Mol Pharm 2019; 16:2069-2082. [DOI: 10.1021/acs.molpharmaceut.9b00042] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Vinod Khetarpal
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | - Mark Rose
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | - Celia Dominguez
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | - Todd Herbst
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | - Leticia Toledo-Sherman
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | | | - Ignacio Muñoz-Sanjuán
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| |
Collapse
|
50
|
L-type amino acid transporter 1 utilizing prodrugs of ferulic acid revealed structural features supporting the design of prodrugs for brain delivery. Eur J Pharm Sci 2019; 129:99-109. [DOI: 10.1016/j.ejps.2019.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/18/2018] [Accepted: 01/04/2019] [Indexed: 12/28/2022]
|