1
|
Matteucci F, Pavletić P, Bonifazi A, Del Bello F, Giorgioni G, Piergentili A, Amantini C, Zeppa L, Sabato E, Vistoli G, Garland R, Yano H, Castagna M, Mammoli V, Cappellacci L, Piergentili A, Quaglia W. New Arylpiperazines as Potent and Selective Dopamine D4 Receptor Ligands Potentially Useful to Treat Glioblastoma. J Med Chem 2025; 68:7441-7458. [PMID: 40156554 DOI: 10.1021/acs.jmedchem.4c03150] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
The dopamine D4 receptor (D4R) has recently been proposed as an emerging target for treating glioblastoma (GBM). In this article, new piperazine ligands, analogues of the potent and selective D4R lead compounds 9 and 10, were prepared and evaluated for their affinity at D2-like receptor subtypes. The most promising results were obtained by replacing the N4-phenyl terminal of 9 with a naphthyl group. Indeed, α-naphthyl derivative 15 proved to have four times higher affinity for D4R than lead 9, whereas β-naphthyl compound 16 was about tenfold more selective for D4R than 9. These compounds behaved as D4R antagonists in both Gi/Go activation and β-arrestin2 recruitment assays. Interestingly, both decreased cell viability dose-dependently and altered the cell cycle of U87 MG, T98G, and U251 MG human GBM cell lines after 48 h treatment, inducing an increase in ROS levels and time-dependent mitochondrial depolarization.
Collapse
Affiliation(s)
- Federica Matteucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri (ChIP), 62032 Camerino, Italy
| | - Pegi Pavletić
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri (ChIP), 62032 Camerino, Italy
- Faculty of Biotechnology and Drug Development, University of Rijeka, Radmile Matejcic 2, 51000 Rijeka, Croatia
| | - Alessandro Bonifazi
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri (ChIP), 62032 Camerino, Italy
| | - Gianfabio Giorgioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri (ChIP), 62032 Camerino, Italy
| | - Alessandro Piergentili
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri (ChIP), 62032 Camerino, Italy
| | - Consuelo Amantini
- School of Biosciences and Veterinary Medicine, Immunopathology and Molecular Medicine Unit, University of Camerino, via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - Laura Zeppa
- School of Biosciences and Veterinary Medicine, Immunopathology and Molecular Medicine Unit, University of Camerino, via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - Emanuela Sabato
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milano, Italy
| | - Giulio Vistoli
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milano, Italy
| | - Rian Garland
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Hideaki Yano
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Monica Castagna
- Center for Drug Discovery and Development-DMPK, Aptuit, an Evotec company, via A. Fleming, 4, 37135 Verona, Italy
| | - Valerio Mammoli
- Center for Drug Discovery and Development-DMPK, Aptuit, an Evotec company, via A. Fleming, 4, 37135 Verona, Italy
| | - Loredana Cappellacci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri (ChIP), 62032 Camerino, Italy
| | - Alessia Piergentili
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri (ChIP), 62032 Camerino, Italy
| | - Wilma Quaglia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri (ChIP), 62032 Camerino, Italy
| |
Collapse
|
2
|
Baldassarri C, Giorgioni G, Piergentili A, Quaglia W, Fontana S, Mammoli V, Minazzato G, Marangoni E, Gasparrini M, Sorci L, Raffaelli N, Cappellacci L, Petrelli R, Del Bello F. Properly Substituted Benzimidazoles as a New Promising Class of Nicotinate Phosphoribosyltransferase (NAPRT) Modulators. Pharmaceuticals (Basel) 2023; 16:189. [PMID: 37259338 PMCID: PMC9967085 DOI: 10.3390/ph16020189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 09/10/2024] Open
Abstract
The prevention of nicotinamide adenine dinucleotide (NAD) biosynthesis is considered an attractive therapeutic approach against cancer, considering that tumor cells are characterized by an increased need for NAD to fuel their reprogrammed metabolism. On the other hand, the decline of NAD is a hallmark of some pathological conditions, including neurodegeneration and metabolic diseases, and boosting NAD biosynthesis has proven to be of therapeutic relevance. Therefore, targeting the enzymes nicotinamide phosphoribosyltransferase (NAMPT) and nicotinate phosphoribosyltransferase (NAPRT), which regulate NAD biosynthesis from nicotinamide (NAM) and nicotinic acid (NA), respectively, is considered a promising strategy to modulate intracellular NAD pool. While potent NAMPT inhibitors and activators have been developed, the search for NAPRT modulators is still in its infancy. In this work, we report on the identification of a new class of NAPRT modulators bearing the 1,2-dimethylbenzimidazole scaffold properly substituted in position 5. In particular, compounds 24, 31, and 32 emerged as the first NAPRT activators reported so far, while 18 behaved as a noncompetitive inhibitor toward NA (Ki = 338 µM) and a mixed inhibitor toward phosphoribosyl pyrophosphate (PRPP) (Ki = 134 µM). From in vitro pharmacokinetic studies, compound 18 showed an overall good ADME profile. To rationalize the obtained results, docking studies were performed on the NAPRT structure. Moreover, a preliminary pharmacophore model was built to shed light on the shift from inhibitors to activators.
Collapse
Affiliation(s)
- Cecilia Baldassarri
- Medicinal Chemistry Unit, School of Pharmacy, Chemistry Interdisciplinary Project (ChIP), University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Gianfabio Giorgioni
- Medicinal Chemistry Unit, School of Pharmacy, Chemistry Interdisciplinary Project (ChIP), University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Alessandro Piergentili
- Medicinal Chemistry Unit, School of Pharmacy, Chemistry Interdisciplinary Project (ChIP), University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Wilma Quaglia
- Medicinal Chemistry Unit, School of Pharmacy, Chemistry Interdisciplinary Project (ChIP), University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Stefano Fontana
- Center for Drug Discovery and Development-DMPK, Aptuit, an Evotec Company, Via A. Fleming 4, 37135 Verona, Italy
| | - Valerio Mammoli
- Center for Drug Discovery and Development-DMPK, Aptuit, an Evotec Company, Via A. Fleming 4, 37135 Verona, Italy
| | - Gabriele Minazzato
- Department of Agriculture, Food and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche 10, 60131 Ancona, Italy
| | - Elisa Marangoni
- Department of Agriculture, Food and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche 10, 60131 Ancona, Italy
| | - Massimiliano Gasparrini
- Department of Agriculture, Food and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche 10, 60131 Ancona, Italy
| | - Leonardo Sorci
- Division of Bioinformatics and Biochemistry, Department of Materials, Environmental Sciences and Urban Planning, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Nadia Raffaelli
- Department of Agriculture, Food and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche 10, 60131 Ancona, Italy
| | - Loredana Cappellacci
- Medicinal Chemistry Unit, School of Pharmacy, Chemistry Interdisciplinary Project (ChIP), University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Riccardo Petrelli
- Medicinal Chemistry Unit, School of Pharmacy, Chemistry Interdisciplinary Project (ChIP), University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Fabio Del Bello
- Medicinal Chemistry Unit, School of Pharmacy, Chemistry Interdisciplinary Project (ChIP), University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| |
Collapse
|
3
|
Del Bello F, Bonifazi A, Giorgioni G, Cifani C, Micioni Di Bonaventura MV, Petrelli R, Piergentili A, Fontana S, Mammoli V, Yano H, Matucci R, Vistoli G, Quaglia W. 1-[3-(4-Butylpiperidin-1-yl)propyl]-1,2,3,4-tetrahydroquinolin-2-one (77-LH-28-1) as a Model for the Rational Design of a Novel Class of Brain Penetrant Ligands with High Affinity and Selectivity for Dopamine D4 Receptor. J Med Chem 2018; 61:3712-3725. [DOI: 10.1021/acs.jmedchem.8b00265] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Fabio Del Bello
- Scuola di Scienze del Farmaco e dei Prodotti della Salute, Università di Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Alessandro Bonifazi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse−Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Gianfabio Giorgioni
- Scuola di Scienze del Farmaco e dei Prodotti della Salute, Università di Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Carlo Cifani
- Scuola di Scienze del Farmaco e dei Prodotti della Salute, Università di Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | | | - Riccardo Petrelli
- Scuola di Scienze del Farmaco e dei Prodotti della Salute, Università di Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Alessandro Piergentili
- Scuola di Scienze del Farmaco e dei Prodotti della Salute, Università di Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Stefano Fontana
- Center for Drug Discovery and Development-DMPK, Aptuit, an Evotec Company, Via A. Fleming, 4, 37135 Verona, Italy
| | - Valerio Mammoli
- Center for Drug Discovery and Development-DMPK, Aptuit, an Evotec Company, Via A. Fleming, 4, 37135 Verona, Italy
| | - Hideaki Yano
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse−Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Rosanna Matucci
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), Sezione di Farmacologia e Tossicologia, Università degli Studi di Firenze, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Giulio Vistoli
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano, Italy
| | - Wilma Quaglia
- Scuola di Scienze del Farmaco e dei Prodotti della Salute, Università di Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| |
Collapse
|
4
|
Varnäs K, Juréus A, Johnström P, Ahlgren C, Schött P, Schou M, Gruber S, Jerning E, Malmborg J, Halldin C, Afzelius L, Farde L. Integrated Strategy for Use of Positron Emission Tomography in Nonhuman Primates to Confirm Multitarget Occupancy of Novel Psychotropic Drugs: An Example with AZD3676. J Pharmacol Exp Ther 2016; 358:464-71. [PMID: 27402278 DOI: 10.1124/jpet.116.234146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 07/06/2016] [Indexed: 03/08/2025] Open
Abstract
Positron emission tomography (PET) is widely applied in central nervous system (CNS) drug development for assessment of target engagement in vivo. As the majority of PET investigations have addressed drug interaction at a single binding site, findings of multitarget engagement have been less frequently reported and have often been inconsistent with results obtained in vitro. AZD3676 [N,N-dimethyl-7-(4-(2-(pyridin-2-yl)ethyl)piperazin-1-yl) benzofuran-2-carboxamide] is a novel combined serotonin (5-hydroxytryptamine) 5-HT1A and 5-HT1B receptor antagonist that was developed for the treatment of cognitive impairment in Alzheimer's disease. Here, we evaluated the properties of AZD3676 as a CNS drug by combining in vitro and ex vivo radioligand binding techniques, behavioral pharmacology in rodents, and PET imaging in nonhuman primates. Target engagement in the nonhuman primate brain was assessed in PET studies by determination of drug-induced occupancy using receptor-selective radioligands. AZD3676 showed preclinical properties consistent with CNS drug potential, including nanomolar receptor affinity and efficacy in rodent models of learning and memory. In PET studies of the monkey brain, AZD3676 inhibited radioligand binding in a dose-dependent manner with similar affinity at both receptors. The equally high affinity at 5-HT1A and 5-HT1B receptors as determined in vivo was not predicted from corresponding estimates obtained in vitro, suggesting more than 10-fold selectivity for 5-HT1A versus 5-HT1B receptors. These findings support the further integrated use of PET for confirmation of multitarget occupancy of CNS drugs. Importantly, earlier introduction of PET studies in nonhuman primates may reduce future development costs and the requirement for animal experiments in preclinical CNS drug development programs.
Collapse
Affiliation(s)
- Katarina Varnäs
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Psychiatry Research, Stockholm (K.V., P.J., M.S., C.H., L.F.); AstraZeneca Translational Science Centre at Karolinska Institutet, PET CoE, Stockholm (P.J., M.S., L.F.); AstraZeneca R&D, Södertälje (A.J., C.A., P.S., S.G., E.J., J.M., L.A.), Sweden
| | - Anders Juréus
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Psychiatry Research, Stockholm (K.V., P.J., M.S., C.H., L.F.); AstraZeneca Translational Science Centre at Karolinska Institutet, PET CoE, Stockholm (P.J., M.S., L.F.); AstraZeneca R&D, Södertälje (A.J., C.A., P.S., S.G., E.J., J.M., L.A.), Sweden
| | - Peter Johnström
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Psychiatry Research, Stockholm (K.V., P.J., M.S., C.H., L.F.); AstraZeneca Translational Science Centre at Karolinska Institutet, PET CoE, Stockholm (P.J., M.S., L.F.); AstraZeneca R&D, Södertälje (A.J., C.A., P.S., S.G., E.J., J.M., L.A.), Sweden
| | - Charlotte Ahlgren
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Psychiatry Research, Stockholm (K.V., P.J., M.S., C.H., L.F.); AstraZeneca Translational Science Centre at Karolinska Institutet, PET CoE, Stockholm (P.J., M.S., L.F.); AstraZeneca R&D, Södertälje (A.J., C.A., P.S., S.G., E.J., J.M., L.A.), Sweden
| | - Pär Schött
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Psychiatry Research, Stockholm (K.V., P.J., M.S., C.H., L.F.); AstraZeneca Translational Science Centre at Karolinska Institutet, PET CoE, Stockholm (P.J., M.S., L.F.); AstraZeneca R&D, Södertälje (A.J., C.A., P.S., S.G., E.J., J.M., L.A.), Sweden
| | - Magnus Schou
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Psychiatry Research, Stockholm (K.V., P.J., M.S., C.H., L.F.); AstraZeneca Translational Science Centre at Karolinska Institutet, PET CoE, Stockholm (P.J., M.S., L.F.); AstraZeneca R&D, Södertälje (A.J., C.A., P.S., S.G., E.J., J.M., L.A.), Sweden
| | - Susanne Gruber
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Psychiatry Research, Stockholm (K.V., P.J., M.S., C.H., L.F.); AstraZeneca Translational Science Centre at Karolinska Institutet, PET CoE, Stockholm (P.J., M.S., L.F.); AstraZeneca R&D, Södertälje (A.J., C.A., P.S., S.G., E.J., J.M., L.A.), Sweden
| | - Eva Jerning
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Psychiatry Research, Stockholm (K.V., P.J., M.S., C.H., L.F.); AstraZeneca Translational Science Centre at Karolinska Institutet, PET CoE, Stockholm (P.J., M.S., L.F.); AstraZeneca R&D, Södertälje (A.J., C.A., P.S., S.G., E.J., J.M., L.A.), Sweden
| | - Jonas Malmborg
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Psychiatry Research, Stockholm (K.V., P.J., M.S., C.H., L.F.); AstraZeneca Translational Science Centre at Karolinska Institutet, PET CoE, Stockholm (P.J., M.S., L.F.); AstraZeneca R&D, Södertälje (A.J., C.A., P.S., S.G., E.J., J.M., L.A.), Sweden
| | - Christer Halldin
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Psychiatry Research, Stockholm (K.V., P.J., M.S., C.H., L.F.); AstraZeneca Translational Science Centre at Karolinska Institutet, PET CoE, Stockholm (P.J., M.S., L.F.); AstraZeneca R&D, Södertälje (A.J., C.A., P.S., S.G., E.J., J.M., L.A.), Sweden
| | - Lovisa Afzelius
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Psychiatry Research, Stockholm (K.V., P.J., M.S., C.H., L.F.); AstraZeneca Translational Science Centre at Karolinska Institutet, PET CoE, Stockholm (P.J., M.S., L.F.); AstraZeneca R&D, Södertälje (A.J., C.A., P.S., S.G., E.J., J.M., L.A.), Sweden
| | - Lars Farde
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Psychiatry Research, Stockholm (K.V., P.J., M.S., C.H., L.F.); AstraZeneca Translational Science Centre at Karolinska Institutet, PET CoE, Stockholm (P.J., M.S., L.F.); AstraZeneca R&D, Södertälje (A.J., C.A., P.S., S.G., E.J., J.M., L.A.), Sweden
| |
Collapse
|
5
|
WRING STEPHEN, GAUKEL ERIC, NARE BAKELA, JACOBS ROBERT, BEAUDET BETH, BOWLING TANA, MERCER LUKE, BACCHI CYRUS, YARLETT NIGEL, RANDOLPH RYAN, PARHAM ROBIN, REWERTS CINDY, PLATNER JACOB, DON ROBERT. Pharmacokinetics and pharmacodynamics utilizing unbound target tissue exposure as part of a disposition-based rationale for lead optimization of benzoxaboroles in the treatment of Stage 2 Human African Trypanosomiasis. Parasitology 2014; 141:104-18. [PMID: 24007596 PMCID: PMC3884839 DOI: 10.1017/s003118201300098x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 05/21/2013] [Accepted: 05/24/2013] [Indexed: 11/06/2022]
Abstract
SUMMARY This review presents a progression strategy for the discovery of new anti-parasitic drugs that uses in vitro susceptibility, time-kill and reversibility measures to define the therapeutically relevant exposure required in target tissues of animal infection models. The strategy is exemplified by the discovery of SCYX-7158 as a potential oral treatment for stage 2 (CNS) Human African Trypanosomiasis (HAT). A critique of current treatments for stage 2 HAT is included to provide context for the challenges of achieving target tissue disposition and the need for establishing pharmacokinetic-pharmacodynamic (PK-PD) measures early in the discovery paradigm. The strategy comprises 3 stages. Initially, compounds demonstrating promising in vitro activity and selectivity for the target organism over mammalian cells are advanced to in vitro metabolic stability, barrier permeability and tissue binding assays to establish that they will likely achieve and maintain therapeutic concentrations during in-life efficacy studies. Secondly, in vitro time-kill and reversibility kinetics are employed to correlate exposure (based on unbound concentrations) with in vitro activity, and to identify pharmacodynamic measures that would best predict efficacy. Lastly, this information is used to design dosing regimens for pivotal pharmacokinetic-pharmacodyamic studies in animal infection models.
Collapse
Affiliation(s)
- STEPHEN WRING
- SCYNEXIS Inc., Research Triangle Park, North Carolina, USA
| | - ERIC GAUKEL
- SCYNEXIS Inc., Research Triangle Park, North Carolina, USA
| | - BAKELA NARE
- SCYNEXIS Inc., Research Triangle Park, North Carolina, USA
| | - ROBERT JACOBS
- SCYNEXIS Inc., Research Triangle Park, North Carolina, USA
| | - BETH BEAUDET
- SCYNEXIS Inc., Research Triangle Park, North Carolina, USA
| | - TANA BOWLING
- SCYNEXIS Inc., Research Triangle Park, North Carolina, USA
| | - LUKE MERCER
- SCYNEXIS Inc., Research Triangle Park, North Carolina, USA
| | - CYRUS BACCHI
- Haskins Laboratory, Pace University, New York, New York, USA
| | - NIGEL YARLETT
- Haskins Laboratory, Pace University, New York, New York, USA
| | - RYAN RANDOLPH
- SCYNEXIS Inc., Research Triangle Park, North Carolina, USA
| | - ROBIN PARHAM
- SCYNEXIS Inc., Research Triangle Park, North Carolina, USA
| | - CINDY REWERTS
- SCYNEXIS Inc., Research Triangle Park, North Carolina, USA
| | - JACOB PLATNER
- Anacor Pharmaceuticals, Inc., Palo Alto, California, USA
| | - ROBERT DON
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| |
Collapse
|
6
|
de Lange EC. The mastermind approach to CNS drug therapy: translational prediction of human brain distribution, target site kinetics, and therapeutic effects. Fluids Barriers CNS 2013; 10:12. [PMID: 23432852 PMCID: PMC3602026 DOI: 10.1186/2045-8118-10-12] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 02/01/2013] [Indexed: 01/11/2023] Open
Abstract
Despite enormous advances in CNS research, CNS disorders remain the world's leading cause of disability. This accounts for more hospitalizations and prolonged care than almost all other diseases combined, and indicates a high unmet need for good CNS drugs and drug therapies.Following dosing, not only the chemical properties of the drug and blood-brain barrier (BBB) transport, but also many other processes will ultimately determine brain target site kinetics and consequently the CNS effects. The rate and extent of all these processes are regulated dynamically, and thus condition dependent. Therefore, heterogenious conditions such as species, gender, genetic background, tissue, age, diet, disease, drug treatment etc., result in considerable inter-individual and intra-individual variation, often encountered in CNS drug therapy.For effective therapy, drugs should access the CNS "at the right place, at the right time, and at the right concentration". To improve CNS therapies and drug development, details of inter-species and inter-condition variations are needed to enable target site pharmacokinetics and associated CNS effects to be translated between species and between disease states. Specifically, such studies need to include information about unbound drug concentrations which drive the effects. To date the only technique that can obtain unbound drug concentrations in brain is microdialysis. This (minimally) invasive technique cannot be readily applied to humans, and we need to rely on translational approaches to predict human brain distribution, target site kinetics, and therapeutic effects of CNS drugs.In this review the term "Mastermind approach" is introduced, for strategic and systematic CNS drug research using advanced preclinical experimental designs and mathematical modeling. In this way, knowledge can be obtained about the contributions and variability of individual processes on the causal path between drug dosing and CNS effect in animals that can be translated to the human situation. On the basis of a few advanced preclinical microdialysis based investigations it will be shown that the "Mastermind approach" has a high potential for the prediction of human CNS drug effects.
Collapse
Affiliation(s)
- Elizabeth Cm de Lange
- Division of Pharmacology, Leiden-Academic Center for Drug Research, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
7
|
Westerhout J, Danhof M, De Lange ECM. Preclinical prediction of human brain target site concentrations: considerations in extrapolating to the clinical setting. J Pharm Sci 2011; 100:3577-93. [PMID: 21544824 DOI: 10.1002/jps.22604] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 04/13/2011] [Accepted: 04/18/2011] [Indexed: 01/11/2023]
Abstract
The development of drugs for central nervous system (CNS) disorders has encountered high failure rates. In part, this has been due to the sole focus on blood-brain barrier permeability of drugs, without taking into account all other processes that determine drug concentrations at the brain target site. This review deals with an overview of the processes that determine the drug distribution into and within the CNS, followed by a description of in vivo techniques that can be used to provide information on CNS drug distribution. A plea follows for the need for more mechanistic understanding of the mechanisms involved in brain target site distribution, and the condition-dependent contributions of these mechanisms to ultimate drug effect. As future direction, such can be achieved by performing integrative cross-compare designed studies, in which mechanisms are systematically influenced (e.g., inhibition of an efflux transporter or induction of pathological state). With the use of advanced mathematical modeling procedures, we may dissect contributions of individual mechanisms in animals as links to the human situation.
Collapse
Affiliation(s)
- Joost Westerhout
- Department of Pharmacology, Leiden/Amsterdam Center for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | | | | |
Collapse
|
8
|
Zonzini L, Bianchi F, Cesari N, Sartori M. In vivo rat PK profiling in drug discovery: new challenges. Expert Opin Drug Discov 2010; 5:1031-7. [DOI: 10.1517/17460441.2010.509396] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Laura Zonzini
- GlaxoSmithKline SpA, Neurosciences Centre of Excellence for Drug Discovery, Medicine Research Centre, Verona, Italy
- Aptuit Srl, Medicine Research Centre, Via A. Fleming 4, 37135 Verona, Italy
| | - Federica Bianchi
- Aptuit Srl, Medicine Research Centre, Via A. Fleming 4, 37135 Verona, Italy
| | - Nicola Cesari
- Chiesi Farmaceutici SpA, Via Palermo, 26/A, 43100 Parma, Italy
| | - Matteo Sartori
- Aptuit Srl, Medicine Research Centre, Via A. Fleming 4, 37135 Verona, Italy
| |
Collapse
|
9
|
Reichel A. Addressing Central Nervous System (CNS) Penetration in Drug Discovery: Basics and Implications of the Evolving New Concept. Chem Biodivers 2009; 6:2030-49. [DOI: 10.1002/cbdv.200900103] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
10
|
The role of the blood-CNS barrier in CNS disorders and their treatment. Neurobiol Dis 2009; 37:3-12. [PMID: 19664711 DOI: 10.1016/j.nbd.2009.07.029] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 07/30/2009] [Accepted: 07/30/2009] [Indexed: 01/16/2023] Open
Abstract
The physical barrier between blood and the CNS (the blood-brain barrier, the blood-spinal cord barrier and the blood-CSF barrier) protects the CNS from both toxic and pathogenic agents in the blood. It is now clear that disruption of the blood-CNS barrier plays a key role in a number of CNS disorders, particularly those associated with neurodegeneration. Such disruption is inevitably accompanied by inflammatory change, as immune cells and immune mediators gain access to the brain or spinal cord. The blood-CNS barrier also presents a major obstacle for potential CNS medicines. Robust methods to assess CNS permeation are therefore essential for CNS drug discovery, particularly when brain pharmacokinetics are taken into account and especially when such measures are linked to neurochemical, physiological, behavioural or neuroimaging readouts of drug action. Drug candidates can be successfully designed to cross the blood-CNS barrier, but for those that can't there is the possibility of entry with a delivery system that facilitates the movement of drug candidate across the blood-CNS barrier.
Collapse
|
11
|
Summerfield S, Jeffrey P. Discovery DMPK: changing paradigms in the eighties, nineties and noughties. Expert Opin Drug Discov 2009; 4:207-18. [DOI: 10.1517/17460440902729405] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
12
|
Summerfield SG, Lucas AJ, Porter RA, Jeffrey P, Gunn RN, Read KR, Stevens AJ, Metcalf AC, Osuna MC, Kilford PJ, Passchier J, Ruffo AD. Toward an improved prediction of humanin vivobrain penetration. Xenobiotica 2008; 38:1518-35. [DOI: 10.1080/00498250802499459] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
13
|
Dobson PD, Kell DB. Carrier-mediated cellular uptake of pharmaceutical drugs: an exception or the rule? Nat Rev Drug Discov 2008; 7:205-20. [PMID: 18309312 DOI: 10.1038/nrd2438] [Citation(s) in RCA: 340] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
It is generally thought that many drug molecules are transported across biological membranes via passive diffusion at a rate related to their lipophilicity. However, the types of biophysical forces involved in the interaction of drugs with lipid membranes are no different from those involved in their interaction with proteins, and so arguments based on lipophilicity could also be applied to drug uptake by membrane transporters or carriers. In this article, we discuss the evidence supporting the idea that rather than being an exception, carrier-mediated and active uptake of drugs may be more common than is usually assumed - including a summary of specific cases in which drugs are known to be taken up into cells via defined carriers - and consider the implications for drug discovery and development.
Collapse
Affiliation(s)
- Paul D Dobson
- School of Chemistry and Manchester Interdisciplinary Biocentre, University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| | | |
Collapse
|
14
|
Jeffrey P, Summerfield SG. Challenges for blood-brain barrier (BBB) screening. Xenobiotica 2008; 37:1135-51. [PMID: 17968740 DOI: 10.1080/00498250701570285] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Whilst blood-brain barrier permeability is an important determinant in achieving efficacious central nervous system drug concentrations, it should not be viewed or measured in isolation. Recent studies have highlighted the need for an integrated approach where optimal central nervous system penetration is achieved through the correct balance of permeability, a low potential for active efflux, and the appropriate physicochemical properties that allow for drug partitioning and distribution into brain tissue. Integrating data from permeability studies performed incorporating an assessment of active efflux by P-glycoprotein in combination with drug-free fraction measurements in blood and brain has furthered the understanding of the impact of the blood-brain barrier on central nervous system uptake and the underlying physicochemical properties that contribute to central nervous system drug disposition. This approach moves away from screening and ranking compounds in assays designed to measure or predict central nervous system penetration in the somewhat arbitrary units of brain-blood (or plasma) ratios.
Collapse
Affiliation(s)
- P Jeffrey
- Neurology & GI Centre of Excellence for Drug Discovery, GlaxoSmithKline, Harlow, UK
| | | |
Collapse
|