101
|
Kawamura S, Ito Y, Hirokawa T, Hikiyama E, Yamada S, Shuto S. Ligand-Phospholipid Conjugation: A Versatile Strategy for Developing Long-Acting Ligands That Bind to Membrane Proteins by Restricting the Subcellular Localization of the Ligand. J Med Chem 2018; 61:4020-4029. [PMID: 29652494 DOI: 10.1021/acs.jmedchem.8b00041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We hypothesized that if drug localization can be restricted to a particular subcellular domain where their target proteins reside, the drugs could bind to their target proteins without being metabolized and/or excreted, which would significantly extend the half-life of the corresponding drug-target complex. Thus, we designed ligand-phospholipid conjugates in which the ligand is conjugated with a phospholipid through a polyethylene glycol linker to restrict the subcellular localization of the ligand in the vicinity of the lipid bilayer. Here, we present the design, synthesis, pharmacological activity, and binding mode analysis of ligand-phospholipid conjugates with muscarinic acetylcholine receptors as the target proteins. These results demonstrate that ligand-phospholipid conjugation can be a versatile strategy for developing long-acting ligands that bind to membrane proteins in drug discovery.
Collapse
Affiliation(s)
| | - Yoshihiko Ito
- Center for Pharma-Food Research, Department of Pharmacokinetics and Pharmacodynamics, Graduate School of Pharmaceutical Sciences , University of Shizuoka , 52-1 Yada , Suruga-ku, Shizuoka 422-8526 , Japan
| | - Takatsugu Hirokawa
- Molecular Profiling Research Center for Drug Discovery (MOLPROF) , National Institute of Advanced Industrial Science and Technology (AIST) , 2-4-7 Aomi , Koutou-ku, Tokyo 135-0064 , Japan
| | - Eriko Hikiyama
- Center for Pharma-Food Research, Department of Pharmacokinetics and Pharmacodynamics, Graduate School of Pharmaceutical Sciences , University of Shizuoka , 52-1 Yada , Suruga-ku, Shizuoka 422-8526 , Japan
| | - Shizuo Yamada
- Center for Pharma-Food Research, Department of Pharmacokinetics and Pharmacodynamics, Graduate School of Pharmaceutical Sciences , University of Shizuoka , 52-1 Yada , Suruga-ku, Shizuoka 422-8526 , Japan
| | | |
Collapse
|
102
|
C-547, a 6-methyluracil derivative with long-lasting binding and rebinding on acetylcholinesterase: Pharmacokinetic and pharmacodynamic studies. Neuropharmacology 2018; 131:304-315. [DOI: 10.1016/j.neuropharm.2017.12.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 12/07/2017] [Accepted: 12/19/2017] [Indexed: 01/09/2023]
|
103
|
Gherbi K, Briddon SJ, Charlton SJ. Micro-pharmacokinetics: Quantifying local drug concentration at live cell membranes. Sci Rep 2018; 8:3479. [PMID: 29472588 PMCID: PMC5823863 DOI: 10.1038/s41598-018-21100-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/24/2018] [Indexed: 12/13/2022] Open
Abstract
Fundamental equations for determining pharmacological parameters, such as the binding affinity of a ligand for its target receptor, assume a homogeneous distribution of ligand, with concentrations in the immediate vicinity of the receptor being the same as those in the bulk aqueous phase. It is, however, known that drugs are able to interact directly with the plasma membrane, potentially increasing local ligand concentrations around the receptor. We have previously reported an influence of ligand-phospholipid interactions on ligand binding kinetics at the β2-adrenoceptor, which resulted in distinct "micro-pharmacokinetic" ligand profiles. Here, we directly quantified the local concentration of BODIPY630/650-PEG8-S-propranolol (BY-propranolol), a fluorescent derivative of the classical β-blocker propranolol, at various distances above membranes of single living cells using fluorescence correlation spectroscopy. We show for the first time a significantly increased ligand concentration immediately adjacent to the cell membrane compared to the bulk aqueous phase. We further show a clear role of both the cell membrane and the β2-adrenoceptor in determining high local BY-propranolol concentrations at the cell surface. These data suggest that the true binding affinity of BY-propranolol for the β2-adrenoceptor is likely far lower than previously reported and highlights the critical importance of understanding the "micro-pharmacokinetic" profiles of ligands for membrane-associated proteins.
Collapse
Affiliation(s)
- Karolina Gherbi
- Division of Pharmacology, Physiology and Neuroscience, School of Life Sciences, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK.,Excellerate Bioscience Ltd, MediCity, Nottingham, NG90 6BH, UK
| | - Stephen J Briddon
- Division of Pharmacology, Physiology and Neuroscience, School of Life Sciences, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK.,Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, The Midlands, UK
| | - Steven J Charlton
- Division of Pharmacology, Physiology and Neuroscience, School of Life Sciences, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK. .,Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, The Midlands, UK. .,Excellerate Bioscience Ltd, MediCity, Nottingham, NG90 6BH, UK.
| |
Collapse
|
104
|
Stoddart LA, Kilpatrick LE, Hill SJ. NanoBRET Approaches to Study Ligand Binding to GPCRs and RTKs. Trends Pharmacol Sci 2018; 39:136-147. [PMID: 29132917 DOI: 10.1016/j.tips.2017.10.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/12/2017] [Accepted: 10/17/2017] [Indexed: 12/30/2022]
Abstract
Recent advances in the development of fluorescent ligands for G-protein-coupled receptors (GPCRs) and receptor tyrosine kinase receptors (RTKs) have facilitated the study of these receptors in living cells. A limitation of these ligands is potential uptake into cells and increased nonspecific binding. However, this can largely be overcome by using proximity approaches, such as bioluminescence resonance energy transfer (BRET), which localise the signal (within 10nm) to the specific receptor target. The recent engineering of NanoLuc has resulted in a luciferase variant that is smaller and significantly brighter (up to tenfold) than existing variants. Here, we review the use of BRET from N-terminal NanoLuc-tagged GPCRs or a RTK to a receptor-bound fluorescent ligand to provide quantitative pharmacology of ligand-receptor interactions in living cells in real time.
Collapse
Affiliation(s)
- Leigh A Stoddart
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK; These authors contributed equally to this work
| | - Laura E Kilpatrick
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK; These authors contributed equally to this work
| | - Stephen J Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK.
| |
Collapse
|
105
|
Stoddart LA, Vernall AJ, Bouzo-Lorenzo M, Bosma R, Kooistra AJ, de Graaf C, Vischer HF, Leurs R, Briddon SJ, Kellam B, Hill SJ. Development of novel fluorescent histamine H 1-receptor antagonists to study ligand-binding kinetics in living cells. Sci Rep 2018; 8:1572. [PMID: 29371669 PMCID: PMC5785503 DOI: 10.1038/s41598-018-19714-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/03/2018] [Indexed: 01/01/2023] Open
Abstract
The histamine H1-receptor (H1R) is an important mediator of allergy and inflammation. H1R antagonists have particular clinical utility in allergic rhinitis and urticaria. Here we have developed six novel fluorescent probes for this receptor that are very effective for high resolution confocal imaging, alongside bioluminescence resonance energy transfer approaches to monitor H1R ligand binding kinetics in living cells. The latter technology exploits the opportunities provided by the recently described bright bioluminescent protein NanoLuc when it is fused to the N-terminus of a receptor. Two different pharmacophores (mepyramine or the fragment VUF13816) were used to generate fluorescent H1R antagonists conjugated via peptide linkers to the fluorophore BODIPY630/650. Kinetic properties of the probes showed wide variation, with the VUF13816 analogues having much longer H1R residence times relative to their mepyramine-based counterparts. The kinetics of these fluorescent ligands could also be monitored in membrane preparations providing new opportunities for future drug discovery applications.
Collapse
Affiliation(s)
- Leigh A Stoddart
- Division of Pharmacology Physiology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Andrea J Vernall
- School of Pharmacy, Division of Biomolecular Science and Medicinal Chemistry, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Monica Bouzo-Lorenzo
- Division of Pharmacology Physiology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Reggie Bosma
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Albert J Kooistra
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Chris de Graaf
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Henry F Vischer
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Rob Leurs
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, PO Box 7161, Amsterdam, The Netherlands
| | - Stephen J Briddon
- Division of Pharmacology Physiology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Barrie Kellam
- School of Pharmacy, Division of Biomolecular Science and Medicinal Chemistry, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK.
| | - Stephen J Hill
- Division of Pharmacology Physiology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK.
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK.
| |
Collapse
|
106
|
A distributed delay approach for modeling delayed outcomes in pharmacokinetics and pharmacodynamics studies. J Pharmacokinet Pharmacodyn 2018; 45:285-308. [DOI: 10.1007/s10928-018-9570-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/10/2018] [Indexed: 12/13/2022]
|
107
|
Abstract
The development of therapies for the treatment of neurological cancer faces a number of major challenges including the synthesis of small molecule agents that can penetrate the blood-brain barrier (BBB). Given the likelihood that in many cases drug exposure will be lower in the CNS than in systemic circulation, it follows that strategies should be employed that can sustain target engagement at low drug concentration. Time dependent target occupancy is a function of both the drug and target concentration as well as the thermodynamic and kinetic parameters that describe the binding reaction coordinate, and sustained target occupancy can be achieved through structural modifications that increase target (re)binding and/or that decrease the rate of drug dissociation. The discovery and deployment of compounds with optimized kinetic effects requires information on the structure-kinetic relationships that modulate the kinetics of binding, and the molecular factors that control the translation of drug-target kinetics to time-dependent drug activity in the disease state. This Review first introduces the potential benefits of drug-target kinetics, such as the ability to delineate both thermodynamic and kinetic selectivity, and then describes factors, such as target vulnerability, that impact the utility of kinetic selectivity. The Review concludes with a description of a mechanistic PK/PD model that integrates drug-target kinetics into predictions of drug activity.
Collapse
Affiliation(s)
- Peter J. Tonge
- Institute for Chemical Biology & Drug Discovery, Departments of Chemistry and Radiology, Stony Brook University, Stony Brook, New York 11794-3400, United States
| |
Collapse
|
108
|
Żurawska-Płaksej E, Rorbach-Dolata A, Wiglusz K, Piwowar A. The effect of glycation on bovine serum albumin conformation and ligand binding properties with regard to gliclazide. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2018; 189:625-633. [PMID: 28888191 DOI: 10.1016/j.saa.2017.08.071] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 08/11/2017] [Accepted: 08/31/2017] [Indexed: 06/07/2023]
Abstract
Albumin, the major serum protein, plays a variety of functions, including binding and transporting endogenous and exogenous ligands. Its molecular structure is sensitive to different environmental modifiers, among which glucose is one of the most significant. In vivo albumin glycation occurs under physiological conditions, but it is increased in diabetes. Since bovine serum albumin (BSA) may serve as a model protein in in vitro experiments, we aimed to investigate the impact of glucose-mediated BSA glycation on the binding capacity towards gliclazide, as well as the ability of this drug to prevent glycation of the BSA molecule. To reflect normo- and hyperglycemia, the conditions of the glycation process were established. Structural changes of albumin after interaction with gliclazide (0-14μM) were determined using fluorescence quenching and circular dichroism spectroscopy. Moreover, thermodynamic parameters as well as energy transfer parameters were determined. Calculated Stern-Volmer quenching constants, as well as binding constants for the BSA-gliclazide complex, were lower for the glycated form of albumin than for the unmodified protein. The largest, over 2-fold, decrease in values of binding parameters was observed for the sample with 30mM of glucose, reflecting the poorly controlled diabetic state, which indicates that the degree of glycation had a critical influence on binding with gliclazide. In contrast to significant changes in the tertiary structure of BSA upon binding with gliclazide, only slight changes in the secondary structure were observed, which was reflected by about a 3% decrease of the α-helix content of glycated BSA (regardless of glucose concentration) in comparison to unmodified BSA. The presence of gliclazide during glycation did not affect its progress. The results of this study indicate that glycation significantly changed the binding ability of BSA towards gliclazide and the scale of these changes depended on glucose concentration. It may have a direct impact on the free drug fraction and its pharmacokinetic behavior, including the risk of hypoglycemic episodes or unexpected interactions with other ligands. The use of BSA in examining binding effects upon glycation seems to be good model for preliminary research and may be used to identify a potential drug response in a diabetic state.
Collapse
Affiliation(s)
- Ewa Żurawska-Płaksej
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland.
| | - Anna Rorbach-Dolata
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland
| | - Katarzyna Wiglusz
- Department of Analytical Chemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211 A, 50-566 Wrocław, Poland.
| | - Agnieszka Piwowar
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland.
| |
Collapse
|
109
|
Drug target residence time: a misleading concept. Drug Discov Today 2018; 23:12-16. [DOI: 10.1016/j.drudis.2017.07.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/13/2017] [Accepted: 07/27/2017] [Indexed: 01/03/2023]
|
110
|
Khalili H, Brocchini S, Khaw PT, Filippov SK. Comparative thermodynamic analysis in solution of a next generation antibody mimetic to VEGF. RSC Adv 2018; 8:35787-35793. [PMID: 35547916 PMCID: PMC9088213 DOI: 10.1039/c8ra07059h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/12/2018] [Indexed: 12/28/2022] Open
Abstract
An antibody mimetic known as Fab–PEG–Fab (FpF) is a stable bivalent molecule that may have some potential therapeutic advantages over IgG antibodies due to differences in their binding kinetics as determined by surface plasmon resonance. Here we describe the thermodynamic binding properties to vascular endothelial growth factor (VEGF) of the FpF antibody mimetics derived from bevacizumab and ranibizumab. Bevacizumab is an IgG antibody and ranibizumab is an antibody fragment (Fab). Both are used clinically to target VEGF to inhibit angiogenesis. FpFbeva displayed comparable binding affinity (KD) and binding thermodynamics (ΔH = −25.7 kcal mole−1 and ΔS = 14 kcal mole−1) to bevacizumab (ΔH = −25 kcal mole−1, ΔS = 13.3 kcal mole−1). FpFrani interactions with VEGF were characterised by large favourable enthalpy (ΔH = −42 kcal mole−1) and unfavourable entropy (ΔS = 31 kcal mole−1) changes compared to ranibizumab (ΔH = −18.5 kcal mole−1 and ΔS = 6.7 kcal mole−1), which being a Fab, is mono-valent. A large negative entropy change resulting in binding of bivalent FpF to homodimer VEGF might be due to the conformational change of the flexible regions of the FpF upon ligand binding. Mono-valent Fab (i.e. ranibizumab or the Fab derived from bevacizumab) displayed a larger degree of freedom (smaller unfavourable entropy) upon binding to homodimer VEGF. Our report describes the first comprehensive enthalpy and entropy compensation analysis for FpF antibody mimetics. While the FpFs displayed similar thermodynamics and binding affinity to the full IgG (i.e. bevacizumab), their enhanced protein stability, slower dissociation rate and lack of Fc effector functions could make FpF a potential next-generation therapy for local tissue-targeted indications. ITC illustrated similar binding thermodynamics for anti-VEGF IgG and FpFs. Bivalent FpFrani displayed larger enthalpy and entropy than monovalent ranibizumab.![]()
Collapse
Affiliation(s)
- Hanieh Khalili
- UEL School of Health
- Sport and Bioscience
- London
- UK
- UCL School of Pharmacy
| | - Steve Brocchini
- UCL School of Pharmacy
- London
- UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust
- UCL Institute of Ophthalmology
| | - Peng Tee Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust
- UCL Institute of Ophthalmology
- London
- UK
| | | |
Collapse
|
111
|
Calo' G, Rizzi A, Ruzza C, Ferrari F, Pacifico S, Gavioli EC, Salvadori S, Guerrini R. Peptide welding technology - A simple strategy for generating innovative ligands for G protein coupled receptors. Peptides 2018; 99:195-204. [PMID: 29031796 DOI: 10.1016/j.peptides.2017.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 09/20/2017] [Accepted: 10/11/2017] [Indexed: 12/19/2022]
Abstract
Based on their high selectivity of action and low toxicity, naturally occurring peptides have great potential in terms of drug development. However, the pharmacokinetic properties of peptides, in particular their half life, are poor. Among different strategies developed for reducing susceptibility to peptidases, and thus increasing the duration of action of peptides, the generation of branched peptides has been described. However, the synthesis and purification of branched peptides are extremely complicated thus limiting their druggability. Here we present a novel and facile synthesis of tetrabranched peptides acting as GPCR ligands and their in vitro and vivo pharmacological characterization. Tetrabranched derivatives of nociceptin/orphanin FQ (N/OFQ), N/OFQ related peptides, opioid peptides, tachykinins, and neuropeptide S were generated with the strategy named peptide welding technology (PWT) and characterized by high yield and purity of the desired final product. In general, PWT derivatives displayed a pharmacological profile similar to that of the natural sequence in terms of affinity, pharmacological activity, potency, and selectivity of action in vitro. More importantly, in vivo studies demonstrated that PWT peptides are characterized by increased potency associated with long lasting duration of action. In conclusion, PWT derivatives of biologically active peptides can be viewed as innovative pharmacological tools for investigating those conditions and states in which selective and prolonged receptor stimulation promotes beneficial effects.
Collapse
Affiliation(s)
- Girolamo Calo'
- Section of Pharmacology, Department of Medical Sciences, and National Institute of Neurosciences, University of Ferrara, Italy.
| | - Anna Rizzi
- Section of Pharmacology, Department of Medical Sciences, and National Institute of Neurosciences, University of Ferrara, Italy
| | - Chiara Ruzza
- Section of Pharmacology, Department of Medical Sciences, and National Institute of Neurosciences, University of Ferrara, Italy
| | - Federica Ferrari
- Section of Pharmacology, Department of Medical Sciences, and National Institute of Neurosciences, University of Ferrara, Italy
| | - Salvatore Pacifico
- Department of Chemical and Pharmaceutical Sciences and LTTA, University of Ferrara, Italy
| | - Elaine C Gavioli
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Severo Salvadori
- Department of Chemical and Pharmaceutical Sciences and LTTA, University of Ferrara, Italy
| | - Remo Guerrini
- Department of Chemical and Pharmaceutical Sciences and LTTA, University of Ferrara, Italy
| |
Collapse
|
112
|
Newman MR, Russell SG, Schmitt CS, Marozas IA, Sheu TJ, Puzas JE, Benoit DSW. Multivalent Presentation of Peptide Targeting Groups Alters Polymer Biodistribution to Target Tissues. Biomacromolecules 2017; 19:71-84. [PMID: 29227674 DOI: 10.1021/acs.biomac.7b01193] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Drug delivery to bone is challenging, whereby drug distribution is commonly <1% of injected dose, despite development of several bone-targeted drug delivery systems specific to hydroxyapatite. These bone-targeted drug delivery systems still suffer from poor target cell localization within bone, as at any given time overall bone volume is far greater than acutely remodeling bone volume, which harbors relevant cell targets (osteoclasts or osteoblasts). Thus, there exists a need to target bone-acting drugs specifically to sites of bone remodeling. To address this need, this study synthesized oligo(ethylene glycol) copolymers based on a peptide with high affinity to tartrate-resistant acid phosphatase (TRAP), an enzyme deposited by osteoclasts during the bone resorption phase of bone remodeling, which provides greater specificity relevant for bone cell drugging. Gradient and random peptide orientations, as well as polymer molecular weights, were investigated. TRAP-targeted, high molecular weight (Mn) random copolymers exhibited superior accumulation in remodeling bone, where fracture accumulation was observed for at least 1 week and accounted for 14% of tissue distribution. Intermediate and low Mn random copolymer accumulation was lower, indicating residence time depends on Mn. High Mn gradient polymers were cleared, with only 2% persisting at fractures after 1 week, suggesting TRAP binding depends on peptide density. Peptide density and Mn are easily modified in this versatile targeting platform, which can be applied to a range of bone drug delivery applications.
Collapse
Affiliation(s)
- Maureen R Newman
- Biomedical Engineering and ‡Chemical Engineering, University of Rochester , Rochester, New York 14627, United States.,Center for Musculoskeletal Research, ∥Department of Orthopaedics, ¶Center for Oral Biology, and ⊥Department of Biomedical Genetics, University of Rochester Medical Center , Rochester, New York 14642, United States
| | - Steven G Russell
- Biomedical Engineering and ‡Chemical Engineering, University of Rochester , Rochester, New York 14627, United States.,Center for Musculoskeletal Research, ∥Department of Orthopaedics, ¶Center for Oral Biology, and ⊥Department of Biomedical Genetics, University of Rochester Medical Center , Rochester, New York 14642, United States
| | - Christopher S Schmitt
- Biomedical Engineering and ‡Chemical Engineering, University of Rochester , Rochester, New York 14627, United States.,Center for Musculoskeletal Research, ∥Department of Orthopaedics, ¶Center for Oral Biology, and ⊥Department of Biomedical Genetics, University of Rochester Medical Center , Rochester, New York 14642, United States
| | - Ian A Marozas
- Biomedical Engineering and ‡Chemical Engineering, University of Rochester , Rochester, New York 14627, United States.,Center for Musculoskeletal Research, ∥Department of Orthopaedics, ¶Center for Oral Biology, and ⊥Department of Biomedical Genetics, University of Rochester Medical Center , Rochester, New York 14642, United States
| | - Tzong-Jen Sheu
- Biomedical Engineering and ‡Chemical Engineering, University of Rochester , Rochester, New York 14627, United States.,Center for Musculoskeletal Research, ∥Department of Orthopaedics, ¶Center for Oral Biology, and ⊥Department of Biomedical Genetics, University of Rochester Medical Center , Rochester, New York 14642, United States
| | - J Edward Puzas
- Biomedical Engineering and ‡Chemical Engineering, University of Rochester , Rochester, New York 14627, United States.,Center for Musculoskeletal Research, ∥Department of Orthopaedics, ¶Center for Oral Biology, and ⊥Department of Biomedical Genetics, University of Rochester Medical Center , Rochester, New York 14642, United States
| | - Danielle S W Benoit
- Biomedical Engineering and ‡Chemical Engineering, University of Rochester , Rochester, New York 14627, United States.,Center for Musculoskeletal Research, ∥Department of Orthopaedics, ¶Center for Oral Biology, and ⊥Department of Biomedical Genetics, University of Rochester Medical Center , Rochester, New York 14642, United States
| |
Collapse
|
113
|
Hendrickx R, Lamm Bergström E, Janzén DLI, Fridén M, Eriksson U, Grime K, Ferguson D. Translational model to predict pulmonary pharmacokinetics and efficacy in man for inhaled bronchodilators. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 7:147-157. [PMID: 29280349 PMCID: PMC5869554 DOI: 10.1002/psp4.12270] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/14/2017] [Accepted: 11/16/2017] [Indexed: 01/11/2023]
Abstract
Translational pharmacokinetic (PK) models are needed to describe and predict drug concentration‐time profiles in lung tissue at the site of action to enable animal‐to‐man translation and prediction of efficacy in humans for inhaled medicines. Current pulmonary PK models are generally descriptive rather than predictive, drug/compound specific, and fail to show successful cross‐species translation. The objective of this work was to develop a robust compartmental modeling approach that captures key features of lung and systemic PK after pulmonary administration of a set of 12 soluble drugs containing single basic, dibasic, or cationic functional groups. The model is shown to allow translation between animal species and predicts drug concentrations in human lungs that correlate with the forced expiratory volume for different classes of bronchodilators. Thus, the pulmonary modeling approach has potential to be a key component in the prediction of human PK, efficacy, and safety for future inhaled medicines.
Collapse
Affiliation(s)
- Ramon Hendrickx
- DMPK, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Eva Lamm Bergström
- DMPK, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - David L I Janzén
- DMPK, Cardiovascular and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Markus Fridén
- DMPK, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Ulf Eriksson
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Ken Grime
- DMPK, Respiratory, Inflammation, and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Douglas Ferguson
- DMPK, Oncology, IMED Biotech Unit, AstraZeneca, Boston, Massachusetts, USA
| |
Collapse
|
114
|
Vauquelin G. Distinct in vivo target occupancy by bivalent- and induced-fit-like binding drugs. Br J Pharmacol 2017; 174:4233-4246. [PMID: 28838028 PMCID: PMC5715606 DOI: 10.1111/bph.13989] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/20/2017] [Accepted: 07/27/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Optimal drug therapy often requires long-lasting target occupancy While this attribute was usually linked to the drug's pharmacokinetic properties, the dissociation rate is now increasingly recognized to contribute as well. Nearly all the earlier pharmacokinetic-pharmacodynamic (PK-PD) simulations encompassed single-step binding drugs and focused on koff . However, 'micro'-PK mechanisms and more complex binding mechanisms like bivalent- and induced-fit binding may contribute as well. Corresponding binding models are presently explored. EXPERIMENTAL APPROACH We compared the 24 h in vivo occupancy over time profiles of prototype bivalent- and induced-fit-like binding drugs (A and B) after one or repeated daily dosings, both without and with rebinding. Special attention was focused on the effect of each of the microscopic rate constants on the occupancy profiles and on the metrics to represent those profiles. KEY RESULTS Although both models can be represented by the same mathematical formulation, drugs A and B display quite different occupancy profiles, even though they have the same potency. These differences can be attributed to the different effects of their microscopic rate constants on their composite koff and also on their susceptibility to experience rebinding. This also affects how the occupancy profiles of bivalent- and induced-fit-like binders progress when repeating the dosings and by changing the dosage. CONCLUSIONS AND IMPLICATIONS Closer attention should be paid to more complex binding models in PK-PD simulations. This may help pharmacologists and medicinal chemists to improve the translation of in vitro kinetic measurements from preclinical screening programmes into clinical efficiency.
Collapse
Affiliation(s)
- Georges Vauquelin
- Department of Molecular and Biochemical PharmacologyVrije Universiteit BrusselBrusselsBelgium
| |
Collapse
|
115
|
Regional Differences in Serotonin Transporter Occupancy by Escitalopram: An [ 11C]DASB PK-PD Study. Clin Pharmacokinet 2017; 56:371-381. [PMID: 27557550 DOI: 10.1007/s40262-016-0444-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND OBJECTIVE Escitalopram is one of the most commonly prescribed selective serotonin reuptake inhibitors (SSRIs). It is thought to act by blocking the serotonin transporter (SERT). However, its dose-SERT occupancy relationship is not well known, so it is not clear what level of SERT blockade is achieved by currently approved doses. METHODS To determine the dose-occupancy relationship, we measured serial SERT occupancy using [11C]DASB [3-amino-4-(2-dimethylaminomethylphenylsulfanyl)-benzonitrile] positron emission tomography (PET) and plasma drug concentrations after the administration of escitalopram in 12 healthy volunteers. We then built a pharmacokinetic-pharmacodynamic model to characterize the dose-occupancy relationship in the putamen and the dorsal raphe nucleus. RESULTS Escitalopram at approved doses occupied less SERT than expected and the SERT occupancy showed regional effects [occupancy was higher in the dorsal raphe nucleus than in the putamen (p < 0.001)]. The drug concentration when 50 % of receptors are occupied (EC50) value and Hill coefficient were significantly different between the putamen (EC50 4.30, Hill coefficient 0.459) and the dorsal raphe nucleus (EC50 2.89, Hill coefficient 0.817). CONCLUSIONS Higher doses of escitalopram than 20 mg are needed to achieve 80 % or greater SERT occupancy. Higher occupancy by escitalopram in the dorsal raphe nucleus relative to the striatum may explain the delayed onset of action of SSRIs by modulating autoreceptor function. The prevention of the 5-HT1A autoreceptor-mediated negative feedback could be a strategy for accelerating the clinical antidepressant effects.
Collapse
|
116
|
de Witte W, Vauquelin G, van der Graaf P, de Lange E. The influence of drug distribution and drug-target binding on target occupancy: The rate-limiting step approximation. Eur J Pharm Sci 2017; 109S:S83-S89. [DOI: 10.1016/j.ejps.2017.05.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 05/11/2017] [Indexed: 12/21/2022]
|
117
|
Extrapyramidal side effects of antipsychotics are linked to their association kinetics at dopamine D 2 receptors. Nat Commun 2017; 8:763. [PMID: 28970469 PMCID: PMC5624946 DOI: 10.1038/s41467-017-00716-z] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 07/20/2017] [Indexed: 12/20/2022] Open
Abstract
Atypical antipsychotic drugs (APDs) have been hypothesized to show reduced extrapyramidal side effects (EPS) due to their rapid dissociation from the dopamine D2 receptor. However, support for this hypothesis is limited to a relatively small number of observations made across several decades and under different experimental conditions. Here we show that association rates, but not dissociation rates, correlate with EPS. We measured the kinetic binding properties of a series of typical and atypical APDs in a novel time-resolved fluorescence resonance energy transfer assay, and correlated these properties with their EPS and prolactin-elevating liabilities at therapeutic doses. EPS are robustly predicted by a rebinding model that considers the microenvironment of postsynaptic D2 receptors and integrates association and dissociation rates to calculate the net rate of reversal of receptor blockade. Thus, optimizing binding kinetics at the D2 receptor may result in APDs with improved therapeutic profile. Atypical antipsychotics show reduced extrapyramidal side effects compared to first generation drugs. Here the authors use time-resolved FRET to measure binding kinetics, and show that side effects correlate with drug association rates to the D2 receptor, while dissociation rates correlate with prolactin elevation.
Collapse
|
118
|
Abstract
Parasympathetic activity is increased in patients with chronic obstructive pulmonary disease (COPD) and asthma and appears to be the major reversible component of airway obstruction. Therefore, treatment with muscarinic receptor antagonists is an effective bronchodilator therapy in COPD and also in asthmatic patients. In recent years, the accumulating evidence that the cholinergic system controls not only contraction by airway smooth muscle but also the functions of inflammatory cells and airway epithelial cells has suggested that muscarinic receptor antagonists could exert other effects that may be of clinical relevance when we must treat a patient suffering from COPD or asthma. There are currently six muscarinic receptor antagonists licenced for use in the treatment of COPD, the short-acting muscarinic receptor antagonists (SAMAs) ipratropium bromide and oxitropium bromide and the long-acting muscarinic receptor antagonists (LAMAs) aclidinium bromide, tiotropium bromide, glycopyrronium bromide and umeclidinium bromide. Concerns have been raised about possible associations of muscarinic receptor antagonists with cardiovascular safety, but the most advanced compounds seem to have an improved safety profile. Further beneficial effects of SAMAs and LAMAs are seen when added to existing treatments, including LABAs, inhaled corticosteroids and phosphodiesterase 4 inhibitors. The importance of tiotropium bromide in the maintenance treatment of COPD, and likely in asthma, has spurred further research to identify new LAMAs. There are a number of molecules that are being identified, but only few have reached the clinical development.
Collapse
|
119
|
Wagner JR, Sørensen J, Hensley N, Wong C, Zhu C, Perison T, Amaro RE. POVME 3.0: Software for Mapping Binding Pocket Flexibility. J Chem Theory Comput 2017; 13:4584-4592. [PMID: 28800393 DOI: 10.1021/acs.jctc.7b00500] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We present a substantial update to the open-source POVME binding pocket analysis software. New capabilities of POVME 3.0 include a flexible chemical coloring scheme for feature identification, postanalysis tools for comparing large ensembles of pockets (e.g., from molecular dynamics simulations), and the introduction of scripts and methods that facilitate binding pocket comparison and analysis. We envision the use of this software for visualization of binding pocket dynamics, selection of representative structures for ensemble docking, and incorporation of molecular dynamics results into ligand design efforts.
Collapse
Affiliation(s)
- Jeffrey R Wagner
- Department of Chemistry and Biochemistry, University of California, San Diego , La Jolla, California 92093, United States
| | - Jesper Sørensen
- Department of Chemistry and Biochemistry, University of California, San Diego , La Jolla, California 92093, United States
| | - Nathan Hensley
- Department of Chemistry and Biochemistry, University of California, San Diego , La Jolla, California 92093, United States
| | - Celia Wong
- Department of Chemistry and Biochemistry, University of California, San Diego , La Jolla, California 92093, United States
| | - Clare Zhu
- Department of Chemistry and Biochemistry, University of California, San Diego , La Jolla, California 92093, United States
| | - Taylor Perison
- Department of Chemistry and Biochemistry, University of California, San Diego , La Jolla, California 92093, United States
| | - Rommie E Amaro
- Department of Chemistry and Biochemistry, University of California, San Diego , La Jolla, California 92093, United States.,National Biomedical Computation Resource, University of California, San Diego , La Jolla, California 92093, United States
| |
Collapse
|
120
|
Yan S, Shaw DE, Yang L, Sandham DA, Healy MP, Reilly J, Wang B. Interactions between β2-Adrenoceptor Ligands and Membrane: Atomic-Level Insights from Magic-Angle Spinning NMR. J Med Chem 2017; 60:6867-6879. [PMID: 28703592 DOI: 10.1021/acs.jmedchem.7b00205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
To understand the relationship between structural properties of the β2-adrenoceptor ligands and their interactions with membranes, we have investigated the location and distribution of five β2 agonists with distinct clinical durations and onsets of action (indacaterol, two indacaterol analogues, salmeterol and formoterol) in monounsaturated model membranes using magic angle spinning NMR to measure these interactions through both 1H nuclear Overhauser enhancement (NOE) and paramagnetic relaxation enhancement (PRE) techniques. The hydrophilic aromatic groups of all five β2 agonists show maximum distribution in the lipid/water interface, but distinct location and dynamic behavior were observed for the lipophilic aromatic rings. Our study elucidates at atomic level that the hydrophobicity and substitution geometry of lipophilic groups play important roles in compound-lipid interactions.
Collapse
Affiliation(s)
- Si Yan
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research , Cambridge, Massachusetts 02139, United States
| | - Duncan E Shaw
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research , Cambridge, Massachusetts 02139, United States
| | - Linhong Yang
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research , Cambridge, Massachusetts 02139, United States
| | - David A Sandham
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research , Cambridge, Massachusetts 02139, United States
| | - Mark P Healy
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research , Cambridge, Massachusetts 02139, United States
| | - John Reilly
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research , Cambridge, Massachusetts 02139, United States
| | - Bing Wang
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research , Cambridge, Massachusetts 02139, United States
| |
Collapse
|
121
|
A kinetic view of GPCR allostery and biased agonism. Nat Chem Biol 2017; 13:929-937. [DOI: 10.1038/nchembio.2431] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/31/2017] [Indexed: 12/21/2022]
|
122
|
Herrington-Symes A, Choi JW, Brocchini S. Interferon dimers: IFN-PEG-IFN. J Drug Target 2017; 25:881-890. [PMID: 28817988 DOI: 10.1080/1061186x.2017.1363214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Increasingly complex proteins can be made by a recombinant chemical approach where proteins that can be made easily can be combined by site-specific chemical conjugation to form multifunctional or more active protein therapeutics. Protein dimers may display increased avidity for cell surface receptors. The increased size of protein dimers may also increase circulation times. Cytokines bind to cell surface receptors that dimerise, so much of the solvent accessible surface of a cytokine is involved in binding to its target. Interferon (IFN) homo-dimers (IFN-PEG-IFN) were prepared by two methods: site-specific bis-alkylation conjugation of PEG to the two thiols of a native disulphide or to two imidazoles on a histidine tag of two His8-tagged IFN (His8IFN). Several control conjugates were also prepared to assess the relative activity of these IFN homo-dimers. The His8IFN-PEG20-His8IFN obtained by histidine-specific conjugation displayed marginally greater in vitro antiviral activity compared to the IFN-PEG20-IFN homo-dimer obtained by disulphide re-bridging conjugation. This result is consistent with previous observations in which enhanced retention of activity was made possible by conjugation to an N-terminal His-tag on the IFN. Comparison of the antiviral and antiproliferative activities of the two IFN homo-dimers prepared by disulphide re-bridging conjugation indicated that IFN-PEG10-IFN was more biologically active than IFN-PEG20-IFN. This result suggests that the size of PEG may influence the antiviral activity of IFN-PEG-IFN homo-dimers.
Collapse
Affiliation(s)
| | - Ji-Won Choi
- a Abzena , Babraham Research Campus , Babraham, Cambridge , UK
| | | |
Collapse
|
123
|
Doornbos MLJ, Cid JM, Haubrich J, Nunes A, van de Sande JW, Vermond SC, Mulder-Krieger T, Trabanco AA, Ahnaou A, Drinkenburg WH, Lavreysen H, Heitman LH, IJzerman AP, Tresadern G. Discovery and Kinetic Profiling of 7-Aryl-1,2,4-triazolo[4,3-a]pyridines: Positive Allosteric Modulators of the Metabotropic Glutamate Receptor 2. J Med Chem 2017; 60:6704-6720. [DOI: 10.1021/acs.jmedchem.7b00669] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Maarten L. J. Doornbos
- Division
of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O.
Box 9502, 2300RA Leiden, The Netherlands
| | - José María Cid
- Janssen Research and Development, Calle Jarama 75A, 45007, Toledo, Spain
| | - Jordi Haubrich
- Division
of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O.
Box 9502, 2300RA Leiden, The Netherlands
| | - Alexandro Nunes
- Division
of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O.
Box 9502, 2300RA Leiden, The Netherlands
| | - Jasper W. van de Sande
- Division
of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O.
Box 9502, 2300RA Leiden, The Netherlands
| | - Sophie C. Vermond
- Division
of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O.
Box 9502, 2300RA Leiden, The Netherlands
| | - Thea Mulder-Krieger
- Division
of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O.
Box 9502, 2300RA Leiden, The Netherlands
| | - Andrés A. Trabanco
- Janssen Research and Development, Calle Jarama 75A, 45007, Toledo, Spain
| | - Abdellah Ahnaou
- Janssen Research and Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | - Hilde Lavreysen
- Janssen Research and Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Laura H. Heitman
- Division
of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O.
Box 9502, 2300RA Leiden, The Netherlands
| | - Adriaan P. IJzerman
- Division
of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O.
Box 9502, 2300RA Leiden, The Netherlands
| | - Gary Tresadern
- Janssen Research and Development, Calle Jarama 75A, 45007, Toledo, Spain
| |
Collapse
|
124
|
Xiang Z, Liu J, Sun H, Wen X. Discovery of Novel Potent Muscarinic M3Receptor Antagonists with Proper Plasma Stability by Structural Recombination of Marketed M3Antagonists. ChemMedChem 2017; 12:1173-1182. [DOI: 10.1002/cmdc.201700189] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/03/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Zuojuan Xiang
- State Key Laboratory of Natural Medicine and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases; China Pharmaceutical University; 24 Tongjia Xiang Nanjing 210009 P.R. China
| | - Jun Liu
- State Key Laboratory of Natural Medicine and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases; China Pharmaceutical University; 24 Tongjia Xiang Nanjing 210009 P.R. China
| | - Hongbin Sun
- State Key Laboratory of Natural Medicine and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases; China Pharmaceutical University; 24 Tongjia Xiang Nanjing 210009 P.R. China
| | - Xiaoan Wen
- State Key Laboratory of Natural Medicine and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases; China Pharmaceutical University; 24 Tongjia Xiang Nanjing 210009 P.R. China
| |
Collapse
|
125
|
Johnstone S, Albert JS. Pharmacological property optimization for allosteric ligands: A medicinal chemistry perspective. Bioorg Med Chem Lett 2017; 27:2239-2258. [PMID: 28408223 DOI: 10.1016/j.bmcl.2017.03.084] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 03/26/2017] [Accepted: 03/27/2017] [Indexed: 12/11/2022]
Abstract
New strategies to potentially improve drug safety and efficacy emerge with allosteric programs. Biased allosteric modulators can be designed with high subtype selectivity and defined receptor signaling endpoints, however, selecting the most meaningful parameters for optimization can be perplexing. Historically, "potency hunting" at the expense of physicochemical and pharmacokinetic optimization has led to numerous tool compounds with excellent pharmacological properties but no path to drug development. Conversely, extensive physicochemical and pharmacokinetic screening with only post hoc bias and allosteric characterization has led to inefficacious compounds or compounds with on-target toxicities. This field is rapidly evolving with new mechanistic understanding, changes in terminology, and novel opportunities. The intent of this digest is to summarize current understanding and debates within the field. We aim to discuss, from a medicinal chemistry perspective, the parameter choices available to drive SAR.
Collapse
Affiliation(s)
- Shawn Johnstone
- Department of Chemistry, IntelliSyn Pharma, 7171 Frederick-Banting, Montreal, Quebec H4S 1Z9, Canada.
| | - Jeffrey S Albert
- Department of Chemistry, IntelliSyn Pharma, 7171 Frederick-Banting, Montreal, Quebec H4S 1Z9, Canada; Department of Chemistry, AviSyn Pharma, 4275 Executive Square, Suite 200, La Jolla, CA 92037, United States.
| |
Collapse
|
126
|
Ellisdon AM, Nold-Petry CA, D’Andrea L, Cho SX, Lao JC, Rudloff I, Ngo D, Lo CY, Soares da Costa TP, Perugini MA, Conroy PJ, Whisstock JC, Nold MF. Homodimerization attenuates the anti-inflammatory activity of interleukin-37. Sci Immunol 2017; 2:2/8/eaaj1548. [DOI: 10.1126/sciimmunol.aaj1548] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 11/29/2016] [Accepted: 01/19/2017] [Indexed: 12/14/2022]
|
127
|
Malerba M, Radaeli A, Montuschi P, Babu KS, Morjaria JB. Investigational beta-2 adrenergic agonists for the treatment of chronic obstructive pulmonary disease. Expert Opin Investig Drugs 2017; 26:319-329. [PMID: 28117615 DOI: 10.1080/13543784.2017.1287172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Long-acting bronchodilators are pivotal in the therapeutic management of COPD patients with moderate-to-severe airflow obstruction. New ultra-long-acting β2-agnoists (ultra-LABAs) have been developed, some of which have been licensed for use as monotherapy and/or in combination with other bronchodilators or inhaled corticosteroids, for use in COPD patients with persistent symptoms and worsening airflow limitation. These new agents are faster in onset and have a prolonged duration of action, with a similar safety profile to the traditional twice-daily bronchodilators which may have an impact on patient concordance. Areas covered: A number of these ultra-LABAs are still under development and bi-functional hybrid molecules containing regions functioning as β2-agonists, and as muscarinic agonists (MABAs) has been developed. This review summarizes these (excluding the licensed ultra-LABAs) with attention on phase II studies data available to-date on their pharmacological profiles, clinical efficacy and safety, and future perspectives. Expert opinion: Despite all the new agents' available, the challenges that persist include any differences in efficacy and safety between the various possible LAMA/LABA combinations, relative advantages of MABAs over fixed-dose LAMA/LABAs, and the impact of these new molecules in terms of long term safety, especially in certain populations in co-morbidities frequently associated with COPD.
Collapse
Affiliation(s)
- Mario Malerba
- a Department of Internal Medicine , University of Brescia and ASST Spedali Civili , Brescia , Italy
| | | | - Paolo Montuschi
- c Department of Pharmacology, Faculty of Medicine , University Hospital Agostino Gemelli Foundation Catholic University of the Sacred Heart, Pharmacology , Rome , Italy
| | - Kesavan S Babu
- d Department of Respiratory Medicine , Queen Alexandra Hospital, Cosham , Portsmouth , UK
| | - Jaymin B Morjaria
- e Department of Respiratory Medicine , Royal Brompton & Harefield NHS Trust, Harefield Hospital , Harefield , UK
| |
Collapse
|
128
|
Hothersall JD, Guo D, Sarda S, Sheppard RJ, Chen H, Keur W, Waring MJ, IJzerman AP, Hill SJ, Dale IL, Rawlins PB. Structure-Activity Relationships of the Sustained Effects of Adenosine A2A Receptor Agonists Driven by Slow Dissociation Kinetics. Mol Pharmacol 2017; 91:25-38. [PMID: 27803241 PMCID: PMC5198511 DOI: 10.1124/mol.116.105551] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 10/28/2016] [Indexed: 12/02/2022] Open
Abstract
The duration of action of adenosine A2A receptor (A2A) agonists is critical for their clinical efficacy, and we sought to better understand how this can be optimized. The in vitro temporal response profiles of a panel of A2A agonists were studied using cAMP assays in recombinantly (CHO) and endogenously (SH-SY5Y) expressing cells. Some agonists (e.g., 3cd; UK-432,097) but not others (e.g., 3ac; CGS-21680) demonstrated sustained wash-resistant agonism, where residual receptor activation continued after washout. The ability of an antagonist to reverse pre-established agonist responses was used as a surrogate read-out for agonist dissociation kinetics, and together with radioligand binding studies suggested a role for slow off-rate in driving sustained effects. One compound, 3ch, showed particularly marked sustained effects, with a reversal t1/2 > 6 hours and close to maximal effects that remained for at least 5 hours after washing. Based on the structure-activity relationship of these compounds, we suggest that lipophilic N6 and bulky C2 substituents can promote stable and long-lived binding events leading to sustained agonist responses, although a high compound logD is not necessary. This provides new insight into the binding interactions of these ligands and we anticipate that this information could facilitate the rational design of novel long-acting A2A agonists with improved clinical efficacy.
Collapse
Affiliation(s)
- J Daniel Hothersall
- AstraZeneca, Discovery Sciences, Alderley Park, United Kingdom (J.D.H., S.S.); AstraZeneca, Oncology, Cambridge, United Kingdom (R.J.S.); AstraZeneca, Discovery Sciences, Mölndal, Sweden (H.C.); AstraZeneca, Discovery Sciences, Cambridge Science Park, United Kingdom (I.L.D., P.B.R.); AstraZeneca, Oncology, Alderley Park, United Kingdom (M.J.W.); Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, The Netherlands (D.G., W.K., A.P.I.J.); and University of Nottingham, School of Life Sciences, United Kingdom (S.J.H.)
| | - Dong Guo
- AstraZeneca, Discovery Sciences, Alderley Park, United Kingdom (J.D.H., S.S.); AstraZeneca, Oncology, Cambridge, United Kingdom (R.J.S.); AstraZeneca, Discovery Sciences, Mölndal, Sweden (H.C.); AstraZeneca, Discovery Sciences, Cambridge Science Park, United Kingdom (I.L.D., P.B.R.); AstraZeneca, Oncology, Alderley Park, United Kingdom (M.J.W.); Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, The Netherlands (D.G., W.K., A.P.I.J.); and University of Nottingham, School of Life Sciences, United Kingdom (S.J.H.)
| | - Sunil Sarda
- AstraZeneca, Discovery Sciences, Alderley Park, United Kingdom (J.D.H., S.S.); AstraZeneca, Oncology, Cambridge, United Kingdom (R.J.S.); AstraZeneca, Discovery Sciences, Mölndal, Sweden (H.C.); AstraZeneca, Discovery Sciences, Cambridge Science Park, United Kingdom (I.L.D., P.B.R.); AstraZeneca, Oncology, Alderley Park, United Kingdom (M.J.W.); Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, The Netherlands (D.G., W.K., A.P.I.J.); and University of Nottingham, School of Life Sciences, United Kingdom (S.J.H.)
| | - Robert J Sheppard
- AstraZeneca, Discovery Sciences, Alderley Park, United Kingdom (J.D.H., S.S.); AstraZeneca, Oncology, Cambridge, United Kingdom (R.J.S.); AstraZeneca, Discovery Sciences, Mölndal, Sweden (H.C.); AstraZeneca, Discovery Sciences, Cambridge Science Park, United Kingdom (I.L.D., P.B.R.); AstraZeneca, Oncology, Alderley Park, United Kingdom (M.J.W.); Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, The Netherlands (D.G., W.K., A.P.I.J.); and University of Nottingham, School of Life Sciences, United Kingdom (S.J.H.)
| | - Hongming Chen
- AstraZeneca, Discovery Sciences, Alderley Park, United Kingdom (J.D.H., S.S.); AstraZeneca, Oncology, Cambridge, United Kingdom (R.J.S.); AstraZeneca, Discovery Sciences, Mölndal, Sweden (H.C.); AstraZeneca, Discovery Sciences, Cambridge Science Park, United Kingdom (I.L.D., P.B.R.); AstraZeneca, Oncology, Alderley Park, United Kingdom (M.J.W.); Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, The Netherlands (D.G., W.K., A.P.I.J.); and University of Nottingham, School of Life Sciences, United Kingdom (S.J.H.)
| | - Wesley Keur
- AstraZeneca, Discovery Sciences, Alderley Park, United Kingdom (J.D.H., S.S.); AstraZeneca, Oncology, Cambridge, United Kingdom (R.J.S.); AstraZeneca, Discovery Sciences, Mölndal, Sweden (H.C.); AstraZeneca, Discovery Sciences, Cambridge Science Park, United Kingdom (I.L.D., P.B.R.); AstraZeneca, Oncology, Alderley Park, United Kingdom (M.J.W.); Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, The Netherlands (D.G., W.K., A.P.I.J.); and University of Nottingham, School of Life Sciences, United Kingdom (S.J.H.)
| | - Michael J Waring
- AstraZeneca, Discovery Sciences, Alderley Park, United Kingdom (J.D.H., S.S.); AstraZeneca, Oncology, Cambridge, United Kingdom (R.J.S.); AstraZeneca, Discovery Sciences, Mölndal, Sweden (H.C.); AstraZeneca, Discovery Sciences, Cambridge Science Park, United Kingdom (I.L.D., P.B.R.); AstraZeneca, Oncology, Alderley Park, United Kingdom (M.J.W.); Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, The Netherlands (D.G., W.K., A.P.I.J.); and University of Nottingham, School of Life Sciences, United Kingdom (S.J.H.)
| | - Adriaan P IJzerman
- AstraZeneca, Discovery Sciences, Alderley Park, United Kingdom (J.D.H., S.S.); AstraZeneca, Oncology, Cambridge, United Kingdom (R.J.S.); AstraZeneca, Discovery Sciences, Mölndal, Sweden (H.C.); AstraZeneca, Discovery Sciences, Cambridge Science Park, United Kingdom (I.L.D., P.B.R.); AstraZeneca, Oncology, Alderley Park, United Kingdom (M.J.W.); Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, The Netherlands (D.G., W.K., A.P.I.J.); and University of Nottingham, School of Life Sciences, United Kingdom (S.J.H.)
| | - Stephen J Hill
- AstraZeneca, Discovery Sciences, Alderley Park, United Kingdom (J.D.H., S.S.); AstraZeneca, Oncology, Cambridge, United Kingdom (R.J.S.); AstraZeneca, Discovery Sciences, Mölndal, Sweden (H.C.); AstraZeneca, Discovery Sciences, Cambridge Science Park, United Kingdom (I.L.D., P.B.R.); AstraZeneca, Oncology, Alderley Park, United Kingdom (M.J.W.); Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, The Netherlands (D.G., W.K., A.P.I.J.); and University of Nottingham, School of Life Sciences, United Kingdom (S.J.H.)
| | - Ian L Dale
- AstraZeneca, Discovery Sciences, Alderley Park, United Kingdom (J.D.H., S.S.); AstraZeneca, Oncology, Cambridge, United Kingdom (R.J.S.); AstraZeneca, Discovery Sciences, Mölndal, Sweden (H.C.); AstraZeneca, Discovery Sciences, Cambridge Science Park, United Kingdom (I.L.D., P.B.R.); AstraZeneca, Oncology, Alderley Park, United Kingdom (M.J.W.); Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, The Netherlands (D.G., W.K., A.P.I.J.); and University of Nottingham, School of Life Sciences, United Kingdom (S.J.H.)
| | - Philip B Rawlins
- AstraZeneca, Discovery Sciences, Alderley Park, United Kingdom (J.D.H., S.S.); AstraZeneca, Oncology, Cambridge, United Kingdom (R.J.S.); AstraZeneca, Discovery Sciences, Mölndal, Sweden (H.C.); AstraZeneca, Discovery Sciences, Cambridge Science Park, United Kingdom (I.L.D., P.B.R.); AstraZeneca, Oncology, Alderley Park, United Kingdom (M.J.W.); Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, The Netherlands (D.G., W.K., A.P.I.J.); and University of Nottingham, School of Life Sciences, United Kingdom (S.J.H.)
| |
Collapse
|
129
|
Khalili H, Lee RW, Khaw PT, Brocchini S, Dick AD, Copland DA. An anti-TNF-α antibody mimetic to treat ocular inflammation. Sci Rep 2016; 6:36905. [PMID: 27874029 PMCID: PMC5118814 DOI: 10.1038/srep36905] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/20/2016] [Indexed: 12/14/2022] Open
Abstract
Infliximab is an antibody that neutralizes TNF-α and is used principally by systemic administration to treat many inflammatory disorders. We prepared the antibody mimetic Fab-PEG-Fab (FpFinfliximab) for direct intravitreal injection to assess whether such formulations have biological activity and potential utility for ocular use. FpFinfliximab was designed to address side effects caused by antibody degradation and the presence of the Fc region. Surface plasmon resonance analysis indicated that infliximab and FpFinfliximab maintained binding affinity for both human and murine recombinant TNF-α. No Fc mediated RPE cellular uptake was observed for FpFinfliximab. Both Infliximab and FpFinfliximab suppressed ocular inflammation by reducing the number of CD45+ infiltrate cells in the EAU mice after a single intravitreal injection at the onset of peak disease. These results offer an opportunity to develop and formulate for ocular use, FpF molecules designed for single and potentially multiple targets using bi-specific FpFs.
Collapse
Affiliation(s)
- Hanieh Khalili
- UCL School of Pharmacy, London, UK.,National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK.,University of East London, School of Health, Sport and Bioscience, Water lane, Stratford campus, London, E15 4LZ, UK
| | - Richard W Lee
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK.,School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Peng T Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Steve Brocchini
- UCL School of Pharmacy, London, UK.,National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Andrew D Dick
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK.,School of Clinical Sciences, University of Bristol, Bristol, UK
| | - David A Copland
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK.,School of Clinical Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
130
|
Zhang R, Wong K. High performance enzyme kinetics of turnover, activation and inhibition for translational drug discovery. Expert Opin Drug Discov 2016; 12:17-37. [PMID: 27784173 DOI: 10.1080/17460441.2017.1245721] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Enzymes are the macromolecular catalysts of many living processes and represent a sizable proportion of all druggable biological targets. Enzymology has been practiced just over a century during which much progress has been made in both the identification of new enzymes and the development of novel methodologies for enzyme kinetics. Areas covered: This review aims to address several key practical aspects in enzyme kinetics in reference to translational drug discovery research. The authors first define what constitutes a high performance enzyme kinetic assay. The authors then review the best practices for turnover, activation and inhibition kinetics to derive critical parameters guiding drug discovery. Notably, the authors recommend global progress curve analysis of dose/time dependence employing an integrated Michaelis-Menten equation and global curve fitting of dose/dose dependence. Expert opinion: The authors believe that in vivo enzyme and substrate abundance and their dynamics, binding modality, drug binding kinetics and enzyme's position in metabolic networks should be assessed to gauge the translational impact on drug efficacy and safety. Integrating these factors in a systems biology and systems pharmacology model should facilitate translational drug discovery.
Collapse
Affiliation(s)
- Rumin Zhang
- a Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc. , Kenilworth , NJ , USA
| | - Kenny Wong
- a Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc. , Kenilworth , NJ , USA
| |
Collapse
|
131
|
Guo D, Heitman LH, IJzerman AP. The Added Value of Assessing Ligand-Receptor Binding Kinetics in Drug Discovery. ACS Med Chem Lett 2016; 7:819-21. [PMID: 27660682 DOI: 10.1021/acsmedchemlett.6b00273] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In the past decade drug research community has started to appreciate the indispensable role of ligand-receptor binding kinetics (BK) in drug discovery. Next to the classical equilibrium-based drug evaluation process with affinity and potency values as outcomes, kinetic investigation of the ligand-receptor interaction can aid compound triage in the hit-to-lead campaign and provide additional information to understand the molecular mechanism of drug action. Translational models incorporating BK are emerging as well, which represent powerful tools for the prediction of in vivo effects. In this viewpoint we will summarize some recent findings and discuss and emphasize the added value of ligand-receptor binding kinetics in drug research.
Collapse
Affiliation(s)
- Dong Guo
- Jiangsu
Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, China
| | - Laura H. Heitman
- Division
of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O.
Box 9502, 2300 RA Leiden, The Netherlands
| | - Adriaan P. IJzerman
- Division
of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O.
Box 9502, 2300 RA Leiden, The Netherlands
| |
Collapse
|
132
|
Vauquelin G. Cell membranes… and how long drugs may exert beneficial pharmacological activity in vivo. Br J Clin Pharmacol 2016; 82:673-82. [PMID: 27135195 PMCID: PMC5338106 DOI: 10.1111/bcp.12996] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/28/2016] [Accepted: 04/28/2016] [Indexed: 12/14/2022] Open
Abstract
The time course of the beneficial pharmacological effect of a drug has long been considered to depend merely on the temporal fluctuation of its free concentration. Only in the last decade has it become widely accepted that target-binding kinetics can also affect in vivo pharmacological activity. Although current reviews still essentially focus on genuine dissociation rates, evidence is accumulating that additional micro-pharmacokinetic (PK) and -pharmacodynamic (PD) mechanisms, in which the cell membrane plays a central role, may also increase the residence time of a drug on its target. The present review provides a compilation of otherwise widely dispersed information on this topic. The cell membrane can intervene in drug binding via the following three major mechanisms: (i) by acting as a sink/repository for the drug; (ii) by modulating the conformation of the drug and even by participating in the binding process; and (iii) by facilitating the approach (and rebinding) of the drug to the target. To highlight these mechanisms, we focus on drugs that are currently used in clinical therapy, such as the antihypertensive angiotensin II type 1 receptor antagonist candesartan, the atypical antipsychotic agent clozapine and the bronchodilator salmeterol. Although the role of cell membranes in PK-PD modelling is gaining increasing interest, many issues remain unresolved. It is likely that novel biophysical and computational approaches will provide improved insights in the near future.
Collapse
Affiliation(s)
- Georges Vauquelin
- Department Molecular and Biochemical PharmacologyVrije Universiteit BrusselBrusselsBelgium
| |
Collapse
|
133
|
Soave M, Stoddart LA, Brown A, Woolard J, Hill SJ. Use of a new proximity assay (NanoBRET) to investigate the ligand-binding characteristics of three fluorescent ligands to the human β1-adrenoceptor expressed in HEK-293 cells. Pharmacol Res Perspect 2016; 4:e00250. [PMID: 27588207 PMCID: PMC4988514 DOI: 10.1002/prp2.250] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/27/2016] [Accepted: 07/01/2016] [Indexed: 12/12/2022] Open
Abstract
Previous research has indicated that allosteric interactions across the dimer interface of β1‐adrenoceptors may be responsible for a secondary low affinity binding conformation. Here we have investigated the potential for probe dependence, in the determination of antagonist pKi values at the human β1‐adenoceptor, which may result from such allosterism interactions. Three fluorescent β1‐adrenoceptor ligands were used to investigate this using bioluminescence energy transfer (BRET) between the receptor‐bound fluorescent ligand and the N‐terminal NanoLuc tag of a human β1‐adrenoceptor expressed in HEK 293 cells (NanoBRET). This proximity assay showed high‐affinity‐specific binding to the NanoLuc‐ β1‐adrenoceptor with each of the three fluorescent ligands yielding KD values of 87.1 ± 10 nmol/L (n = 8), 38.1 ± 12 nmol/L (n = 7), 13.4 ± 2 nmol/L (n = 14) for propranolol‐Peg8‐BY630, propranolol‐ β(Ala‐Ala)‐BY630 and CGP‐12177‐TMR, respectively. Parallel radioligand‐binding studies with 3H‐CGP12177 and TIRF microscopy, to monitor NanoLuc bioluminescence, confirmed a high cell surface expression of the NanoLuc‐ β1‐adrenoceptor in HEK 293 cells (circa 1500 fmol.mg protein−1). Following a 1 h incubation with fluorescent ligands and β1‐adrenoceptor competing antagonists, there were significant differences (P < 0.001) in the pKi values obtained for CGP20712a and CGP 12177 with the different fluorescent ligands and 3H‐CGP 12177. However, increasing the incubation time to 2 h removed these significant differences. The data obtained show that the NanoBRET assay can be applied successfully to study ligand‐receptor interactions at the human β1‐adrenoceptor. However, the study also emphasizes the importance of ensuring that both the fluorescent and competing ligands are in true equilibrium before interpretations regarding probe dependence can be made.
Collapse
Affiliation(s)
- Mark Soave
- Cell Signalling and Pharmacology Research Group School of Life Sciences University of Nottingham Nottingham NG7 2UH United Kingdom
| | - Leigh A Stoddart
- Cell Signalling and Pharmacology Research Group School of Life Sciences University of Nottingham Nottingham NG7 2UH United Kingdom
| | - Alastair Brown
- Heptares Therapeutics Ltd. Bio Park Welwyn Garden City AL7 3AX United Kingdom
| | - Jeanette Woolard
- Cell Signalling and Pharmacology Research Group School of Life Sciences University of Nottingham Nottingham NG7 2UH United Kingdom
| | - Stephen J Hill
- Cell Signalling and Pharmacology Research Group School of Life Sciences University of Nottingham Nottingham NG7 2UH United Kingdom
| |
Collapse
|
134
|
Hall ER, Bibby LI, Slack RJ. Characterisation of a novel, high affinity and selective αvβ6 integrin RGD-mimetic radioligand. Biochem Pharmacol 2016; 117:88-96. [PMID: 27501918 DOI: 10.1016/j.bcp.2016.08.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 08/02/2016] [Indexed: 11/25/2022]
Abstract
The alpha-v beta-6 (αvβ6) integrin has been identified as playing a key role in the activation of transforming growth factor-β (TGFβ) that is hypothesised to be pivotal in the development of cancer and fibrotic diseases. Therefore, the αvβ6 integrin is an attractive therapeutic target for these debilitating diseases and a drug discovery programme to identify small molecule αvβ6 selective arginyl-glycinyl-aspartic acid (RGD)-mimetics was initiated within GlaxoSmithKline. The primary aim of this study was to pharmacologically characterise the binding to αvβ6 of a novel clinical candidate, compound 1, using a radiolabelled form. Radioligand binding studies were completed with [(3)H]compound 1 against the human and mouse soluble protein forms of αvβ6 to determine accurate affinity estimates and binding kinetics. The selectivity of compound 1 for the RGD integrin family was also determined using saturation binding studies (αvβ1, αvβ3, αvβ5, αvβ8, α5β1 and α8β1 integrins) and fibrinogen-induced platelet aggregation (αIIbβ3 integrin). In addition, the relationship between divalent metal cation type and concentration and αvβ6 RGD site binding was also investigated. Compound 1 has been demonstrated to bind with extremely high affinity and selectivity for the αvβ6 integrin and has the potential as a clinical tool and therapeutic for investigating the role of αvβ6 in a range of disease states both pre-clinically and clinically. In addition, this is the first study that has successfully applied radioligand binding to the RGD integrin field to accurately determine the affinity and selectivity profile of a small molecule RGD-mimetic.
Collapse
Affiliation(s)
- Eleanor R Hall
- Fibrosis and Lung Injury Discovery Performance Unit, Respiratory TAU, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire, UK
| | - Lloyd I Bibby
- Fibrosis and Lung Injury Discovery Performance Unit, Respiratory TAU, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire, UK
| | - Robert J Slack
- Fibrosis and Lung Injury Discovery Performance Unit, Respiratory TAU, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire, UK.
| |
Collapse
|
135
|
Nederpelt I, Bleeker D, Tuijt B, IJzerman AP, Heitman LH. Kinetic binding and activation profiles of endogenous tachykinins targeting the NK1 receptor. Biochem Pharmacol 2016; 118:88-95. [PMID: 27501920 DOI: 10.1016/j.bcp.2016.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/02/2016] [Indexed: 02/01/2023]
Abstract
Ligand-receptor binding kinetics (i.e. association and dissociation rates) are emerging as important parameters for drug efficacy in vivo. Awareness of the kinetic behavior of endogenous ligands is pivotal, as drugs often have to compete with those. The binding kinetics of neurokinin 1 (NK1) receptor antagonists have been widely investigated while binding kinetics of endogenous tachykinins have hardly been reported, if at all. Therefore, the aim of this research was to investigate the binding kinetics of endogenous tachykinins and derivatives thereof and their role in the activation of the NK1 receptor. We determined the binding kinetics of seven tachykinins targeting the NK1 receptor. Dissociation rate constants (koff) ranged from 0.026±0.0029min-1 (Sar9,Met(O2)11-SP) to 0.21±0.015min-1 (septide). Association rate constants (kon) were more diverse: substance P (SP) associated the fastest with a kon value of 0.24±0.046nM-1min-1 while neurokinin A (NKA) had the slowest association rate constant of 0.001±0.0002nM-1min-1. Kinetic binding parameters were highly correlated with potency and maximal response values determined in label-free impedance-based experiments on U-251 MG cells. Our research demonstrates large variations in binding kinetics of tachykinins which correlate to receptor activation. These findings provide new insights into the ligand-receptor interactions of tachykinins and underline the importance of measuring binding kinetics of both drug candidates and competing endogenous ligands.
Collapse
Affiliation(s)
- I Nederpelt
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - D Bleeker
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - B Tuijt
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - A P IJzerman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - L H Heitman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| |
Collapse
|
136
|
Vauquelin G. Effects of target binding kinetics on in vivo drug efficacy: koff , kon and rebinding. Br J Pharmacol 2016; 173:2319-34. [PMID: 27129075 PMCID: PMC4945762 DOI: 10.1111/bph.13504] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/07/2016] [Accepted: 04/19/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Optimal drug therapy often requires continuing high levels of target occupancy. Besides the traditional pharmacokinetic contribution, target binding kinetics is increasingly considered to play an important role as well. While most attention has been focused on the dissociation rate of the complex, recent reports expressed doubt about the unreserved translatability of this pharmacodynamic property into clinical efficacy. 'Micro'-pharmacokinetic mechanisms like drug rebinding and partitioning into the cell membrane may constitute a potential fix. EXPERIMENTAL APPROACH Simulations were based on solving differential equations. KEY RESULTS Based on a selected range of association and dissociation rate constants, kon and koff , and rebinding potencies of the drugs as variables, their effects on the temporal in vivo occupancy profile of their targets, after one or multiple repetitive dosings, have here been simulated. CONCLUSIONS AND IMPLICATIONS Most strikingly, the simulations show that, when rebinding is also taken into account, increasing kon may produce closely the same outcome as decreasing koff when dosing is performed in accordance with the therapeutically most relevant constant [Lmax ]/KD ratio paradigm. Also, under certain conditions, rebinding may produce closely the same outcome as invoking slow diffusion of the drug between the plasma compartment and a target-containing 'effect' compartment. Although the present simulations should only be regarded as a 'proof of principle', these findings may help pharmacologists and medicinal chemists to devise ex vivo and in vitro binding kinetic assays that are more relevant and translatable to in vivo settings.
Collapse
Affiliation(s)
- Georges Vauquelin
- Department of Molecular and Biochemical PharmacologyVrije Universiteit BrusselBrusselsBelgium
| |
Collapse
|
137
|
BRET-based β-arrestin2 recruitment to the histamine H1 receptor for investigating antihistamine binding kinetics. Pharmacol Res 2016; 111:679-687. [PMID: 27468652 DOI: 10.1016/j.phrs.2016.07.034] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/14/2016] [Accepted: 07/24/2016] [Indexed: 01/23/2023]
Abstract
Ligand residence time is thought to be a critical parameter for optimizing the in vivo efficacy of drug candidates. For the histamine H1 receptor (H1R) and other G protein-coupled receptors, the kinetics of ligand binding are typically measured by low throughput radioligand binding experiments using homogenized cell membranes expressing the target receptor. In this study, a real-time proximity assay between H1R and β-arrestin2 in living cells was established to investigate the dynamics of antihistamine binding to the H1R. No receptor reserve was found for the histamine-induced recruitment of β-arrestin2 to the H1R and the transiently recruited β-arrestin2 therefore reflected occupancy of the receptor by histamine. Antihistamines displayed similar kinetic signatures on antagonizing histamine-induced β-arrestin2 recruitment as compared to displacing radioligand binding from the H1R. This homogeneous functional method unambiguously determined the fifty-fold difference in the dissociation rate constant between mepyramine and the long residence time antihistamines levocetirizine and desloratadine.
Collapse
|
138
|
Dickson CJ, Hornak V, Velez-Vega C, McKay DJJ, Reilly J, Sandham DA, Shaw D, Fairhurst RA, Charlton SJ, Sykes DA, Pearlstein RA, Duca JS. Uncoupling the Structure-Activity Relationships of β2 Adrenergic Receptor Ligands from Membrane Binding. J Med Chem 2016; 59:5780-9. [PMID: 27239696 DOI: 10.1021/acs.jmedchem.6b00358] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Ligand binding to membrane proteins may be significantly influenced by the interaction of ligands with the membrane. In particular, the microscopic ligand concentration within the membrane surface solvation layer may exceed that in bulk solvent, resulting in overestimation of the intrinsic protein-ligand binding contribution to the apparent/measured affinity. Using published binding data for a set of small molecules with the β2 adrenergic receptor, we demonstrate that deconvolution of membrane and protein binding contributions allows for improved structure-activity relationship analysis and structure-based drug design. Molecular dynamics simulations of ligand bound membrane protein complexes were used to validate binding poses, allowing analysis of key interactions and binding site solvation to develop structure-activity relationships of β2 ligand binding. The resulting relationships are consistent with intrinsic binding affinity (corrected for membrane interaction). The successful structure-based design of ligands targeting membrane proteins may require an assessment of membrane affinity to uncouple protein binding from membrane interactions.
Collapse
Affiliation(s)
- Callum J Dickson
- Computer-Aided Drug Discovery, Global Discovery Chemistry, Novartis Institutes for BioMedical Research , 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Viktor Hornak
- Computer-Aided Drug Discovery, Global Discovery Chemistry, Novartis Institutes for BioMedical Research , 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Camilo Velez-Vega
- Computer-Aided Drug Discovery, Global Discovery Chemistry, Novartis Institutes for BioMedical Research , 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Daniel J J McKay
- Computer-Aided Drug Discovery, Global Discovery Chemistry, Novartis Institutes for BioMedical Research , 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - John Reilly
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research , 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - David A Sandham
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research , 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Duncan Shaw
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research , 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Robin A Fairhurst
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG , Werk Klybeck, Postfach, CH-4002 Basel, Switzerland
| | - Steven J Charlton
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Queen's Medical Centre , Nottingham NG7 2UH, U.K
| | - David A Sykes
- Cell Signalling Research Group, School of Life Sciences, University of Nottingham, Queen's Medical Centre , Nottingham NG7 2UH, U.K
| | - Robert A Pearlstein
- Computer-Aided Drug Discovery, Global Discovery Chemistry, Novartis Institutes for BioMedical Research , 100 Technology Square, Cambridge, Massachusetts 02139, United States
| | - Jose S Duca
- Computer-Aided Drug Discovery, Global Discovery Chemistry, Novartis Institutes for BioMedical Research , 100 Technology Square, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
139
|
Shimizu Y, Ogawa K, Nakayama M. Characterization of Kinetic Binding Properties of Unlabeled Ligands via a Preincubation Endpoint Binding Approach. ACTA ACUST UNITED AC 2016; 21:729-37. [PMID: 27270099 DOI: 10.1177/1087057116652065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/06/2016] [Indexed: 11/16/2022]
Abstract
The dissociation rates of unlabeled drugs have been well studied by kinetic binding analyses. Since kinetic assays are laborious, we developed a simple method to determine the kinetic binding parameters of unlabeled competitors by a preincubation endpoint assay. The probe binding after preincubation of a competitor can be described by a single equation as a function of time. Simulations using the equation revealed the degree of IC50 change induced by preincubation of a competitor depended on the dissociation rate koff of the competitor but not on the association rate kon To validate the model, an in vitro binding assay was performed using a smoothened receptor (SMO) and [(3)H]TAK-441, a SMO antagonist. The equilibrium dissociation constants (KI) and koff of SMO antagonists determined by globally fitting the model to the concentration-response curves obtained with and without 24 h preincubation correlated well with those determined by other methods. This approach could be useful for early-stage optimization of drug candidates by enabling determination of binding kinetics in a high-throughput manner because it does not require kinetic measurements, an intermediate washout step during the reaction, or prior determination of competitors' KI values.
Collapse
Affiliation(s)
- Yuji Shimizu
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Kazumasa Ogawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Masaharu Nakayama
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| |
Collapse
|
140
|
Nederpelt I, Vergroesen R, IJzerman A, Heitman L. Persistent GnRH receptor activation in pituitary αT3-1 cells analyzed with a label-free technology. Biosens Bioelectron 2016; 79:721-7. [DOI: 10.1016/j.bios.2015.12.066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/10/2015] [Accepted: 12/20/2015] [Indexed: 12/21/2022]
|
141
|
Receptor residence time trumps drug-likeness and oral bioavailability in determining efficacy of complement C5a antagonists. Sci Rep 2016; 6:24575. [PMID: 27094554 PMCID: PMC4837355 DOI: 10.1038/srep24575] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 03/31/2016] [Indexed: 12/13/2022] Open
Abstract
Drug discovery and translation are normally based on optimizing efficacy by increasing receptor affinity, functional potency, drug-likeness (rule-of-five compliance) and oral bioavailability. Here we demonstrate that residence time of a compound on its receptor has an overriding influence on efficacy, exemplified for antagonists of inflammatory protein complement C5a that activates immune cells and promotes disease. Three equipotent antagonists (3D53, W54011, JJ47) of inflammatory responses to C5a (3 nM) were compared for drug-likeness, receptor affinity and antagonist potency in human macrophages, and anti-inflammatory efficacy in rats. Only the least drug-like antagonist (3D53) maintained potency in cells against higher C5a concentrations and had a much longer duration of action (t1/2 ~ 20 h) than W54011 or JJ47 (t1/2 ~ 1 -3 h) in inhibiting macrophage responses. The unusually long residence time of 3D53 on its receptor was mechanistically probed by molecular dynamics simulations, which revealed long-lasting interactions that trap the antagonist within the receptor. Despite negligible oral bioavailability, 3D53 was much more orally efficacious than W54011 or JJ47 in preventing repeated agonist insults to induce rat paw oedema over 24 h. Thus, residence time on a receptor can trump drug-likeness in determining efficacy, even oral efficacy, of pharmacological agents.
Collapse
|
142
|
Guo D, Heitman LH, IJzerman AP. Kinetic Aspects of the Interaction between Ligand and G Protein-Coupled Receptor: The Case of the Adenosine Receptors. Chem Rev 2016; 117:38-66. [DOI: 10.1021/acs.chemrev.6b00025] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Dong Guo
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Laura H. Heitman
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Adriaan P. IJzerman
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| |
Collapse
|
143
|
Abstract
Interest in the application of molecular dynamics (MD) simulations has increased in the field of protein kinase (PK) drug discovery. PKs belong to an important drug target class because they are directly involved in a number of diseases, including cancer. MD methods simulate dynamic biological and chemical events at an atomic level. This information can be combined with other in silico and experimental methods to efficiently target selected receptors. In this review, we present common and advanced methods of MD simulations and we focus on the recent applications of MD-based methodologies that provided significant insights into the elucidation of biological mechanisms involving PKs and into the discovery of novel kinase inhibitors.
Collapse
|
144
|
Vauquelin G, Van Liefde I, Swinney DC. On the different experimental manifestations of two-state 'induced-fit' binding of drugs to their cellular targets. Br J Pharmacol 2016; 173:1268-85. [PMID: 26808227 DOI: 10.1111/bph.13445] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 09/03/2015] [Accepted: 01/12/2016] [Indexed: 01/17/2023] Open
Abstract
'Induced-fit' binding of drugs to a target may lead to high affinity, selectivity and a long residence time, and this mechanism has been proposed to apply to many drugs with high clinical efficacy. It is a multistep process that initially involves the binding of a drug to its target to form a loose RL complex and a subsequent isomerization/conformational change to yield a tighter binding R'L state. Equations with the same mathematical form may also describe the binding of bivalent antibodies and related synthetic drugs. Based on a selected range of 'microscopic' rate constants and variables such as the ligand concentration and incubation time, we have simulated the experimental manifestations that may go along with induced-fit binding. Overall, they validate different experimental procedures that have been used over the years to identify such binding mechanisms. However, they also reveal that each of these manifestations only becomes perceptible at particular combinations of rate constants. The simulations also show that the durable nature of R'L and the propensity of R'L to be formed repeatedly before the ligand dissociates will increase the residence time. This review may help pharmacologists and medicinal chemists obtain preliminary indications for identifying an induced-fit mechanism.
Collapse
Affiliation(s)
- Georges Vauquelin
- Department of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Isabelle Van Liefde
- Department of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Brussels, Belgium
| | - David C Swinney
- Institute for Rare and Neglected Diseases Drug Discovery, Mountain View, CA, USA
| |
Collapse
|
145
|
Affiliation(s)
- Gwen Hughes
- Assistant professor in Physiology, University of Nottingham
| |
Collapse
|
146
|
Abstract
The Fc-fusion mimetic RpR 2̲ was prepared by disulfide bridging conjugation using PEG in the place of the Fc.
Collapse
Affiliation(s)
- H. Khalili
- UCL School of Pharmacy
- University College London
- London WC1N 1AX
- UK
- NIHR Biomedical Research Centre
| | - P. T. Khaw
- NIHR Biomedical Research Centre
- Moorfields Eye Hospital and UCL Institute of Ophthalmology
- London
- UK
| | - S. Brocchini
- UCL School of Pharmacy
- University College London
- London WC1N 1AX
- UK
- NIHR Biomedical Research Centre
| |
Collapse
|
147
|
Abstract
The drug-target residence time model was first introduced in 2006 and has been broadly adopted across the chemical biology, biotechnology and pharmaceutical communities. While traditional in vitro methods view drug-target interactions exclusively in terms of equilibrium affinity, the residence time model takes into account the conformational dynamics of target macromolecules that affect drug binding and dissociation. The key tenet of this model is that the lifetime (or residence time) of the binary drug-target complex, and not the binding affinity per se, dictates much of the in vivo pharmacological activity. Here, this model is revisited and key applications of it over the past 10 years are highlighted.
Collapse
|
148
|
The experimental power of FR900359 to study Gq-regulated biological processes. Nat Commun 2015; 6:10156. [PMID: 26658454 PMCID: PMC4682109 DOI: 10.1038/ncomms10156] [Citation(s) in RCA: 278] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 11/06/2015] [Indexed: 12/13/2022] Open
Abstract
Despite the discovery of heterotrimeric αβγ G proteins ∼25 years ago, their selective perturbation by cell-permeable inhibitors remains a fundamental challenge. Here we report that the plant-derived depsipeptide FR900359 (FR) is ideally suited to this task. Using a multifaceted approach we systematically characterize FR as a selective inhibitor of Gq/11/14 over all other mammalian Gα isoforms and elaborate its molecular mechanism of action. We also use FR to investigate whether inhibition of Gq proteins is an effective post-receptor strategy to target oncogenic signalling, using melanoma as a model system. FR suppresses many of the hallmark features that are central to the malignancy of melanoma cells, thereby providing new opportunities for therapeutic intervention. Just as pertussis toxin is used extensively to probe and inhibit the signalling of Gi/o proteins, we anticipate that FR will at least be its equivalent for investigating the biological relevance of Gq.
Collapse
|
149
|
de Witte WEA, Wong YC, Nederpelt I, Heitman LH, Danhof M, van der Graaf PH, Gilissen RAHJ, de Lange ECM. Mechanistic models enable the rational use of in vitro drug-target binding kinetics for better drug effects in patients. Expert Opin Drug Discov 2015; 11:45-63. [PMID: 26484747 DOI: 10.1517/17460441.2016.1100163] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Drug-target binding kinetics are major determinants of the time course of drug action for several drugs, as clearly described for the irreversible binders omeprazole and aspirin. This supports the increasing interest to incorporate newly developed high-throughput assays for drug-target binding kinetics in drug discovery. A meaningful application of in vitro drug-target binding kinetics in drug discovery requires insight into the relation between in vivo drug effect and in vitro measured drug-target binding kinetics. AREAS COVERED In this review, the authors discuss both the relation between in vitro and in vivo measured binding kinetics and the relation between in vivo binding kinetics, target occupancy and effect profiles. EXPERT OPINION More scientific evidence is required for the rational selection and development of drug-candidates on the basis of in vitro estimates of drug-target binding kinetics. To elucidate the value of in vitro binding kinetics measurements, it is necessary to obtain information on system-specific properties which influence the kinetics of target occupancy and drug effect. Mathematical integration of this information enables the identification of drug-specific properties which lead to optimal target occupancy and drug effect in patients.
Collapse
Affiliation(s)
- Wilhelmus E A de Witte
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Yin Cheong Wong
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Indira Nederpelt
- b Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Laura H Heitman
- b Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Meindert Danhof
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Piet H van der Graaf
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| | - Ron A H J Gilissen
- c A Division of Janssen Pharmaceutica N.V., Janssen Research and Development , Turnhoutseweg 30, Beerse 2340 , Belgium
| | - Elizabeth C M de Lange
- a Division of Pharmacology, Leiden Academic Centre for Drug Research , Leiden University , Einsteinweg 55, 2333 CC Leiden , The Netherlands
| |
Collapse
|
150
|
Vauquelin G, Van Liefde I, Swinney DC. Radioligand binding to intact cells as a tool for extended drug screening in a representative physiological context. DRUG DISCOVERY TODAY. TECHNOLOGIES 2015; 17:28-34. [PMID: 26724334 DOI: 10.1016/j.ddtec.2015.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 09/04/2015] [Accepted: 09/09/2015] [Indexed: 06/05/2023]
Abstract
Radioligand binding assays on intact cells offer distinct advantages to those on membrane suspensions. Major pharmacological properties like drug affinity and binding kinetics are more physiologically relevant. Complex mechanisms can be studied with a wider choice of experimental approaches and so provide insights into induced-fit type binding, receptor internalisation and even into pharmacomicrokinetic phenomena like drug rebinding and partitioning into the membrane. Hence, intact cell binding constitutes a valuable addition to the pharmacologist's toolbox.
Collapse
Affiliation(s)
- Georges Vauquelin
- Dept. of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| | - Isabelle Van Liefde
- Dept. of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - David C Swinney
- Institute for Rare and Neglected Diseases Drug Discovery, 897 Independence Ave, Suite 2C, Mountain View, CA 94043, United States
| |
Collapse
|