25 results on '"Bagal SK"'
Search Results
2. Discovery of a Series of Orally Bioavailable Androgen Receptor Degraders for the Treatment of Prostate Cancer.
- Author
-
Bagal SK, Astles PC, Diène C, Argyrou A, Crafter C, Cassar DJ, Fallan C, Hock A, Jones T, Moreau K, Lamont GM, Lamont S, Michaloglou C, Packer MJ, Pike A, Ramos-Montoya A, Scott JS, Shaw J, and Shologu Z
- Subjects
- Male, Animals, Humans, Administration, Oral, Mice, Cell Line, Tumor, Structure-Activity Relationship, Antineoplastic Agents pharmacology, Antineoplastic Agents chemistry, Antineoplastic Agents therapeutic use, Antineoplastic Agents pharmacokinetics, Androgen Receptor Antagonists pharmacology, Androgen Receptor Antagonists chemistry, Androgen Receptor Antagonists therapeutic use, Androgen Receptor Antagonists pharmacokinetics, Drug Discovery, Xenograft Model Antitumor Assays, Rats, Receptors, Androgen metabolism, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology, Prostatic Neoplasms metabolism, Biological Availability
- Abstract
Androgen receptor (AR) signaling plays a key role in the progression of prostate cancer. This study describes the discovery and optimization of a novel series of AR PROTAC degraders that recruit the Cereblon (CRBN) E3 ligase. Having identified a series of AR ligands based on 4-(4-phenyl-1-piperidyl)-2-(trifluoromethyl)benzonitrile, our PROTAC optimization strategy focused on linker connectivity and CRBN ligand SAR to deliver potent degradation of AR in LNCaP cells. This work culminated in compounds 11 and 16 which demonstrated good rodent oral bioavailability. Subsequent SAR around the AR binding region brought in an additional desirable feature, degradation of the important treatment resistance mutation L702H. Compound 22 (AZ'3137) possessed an attractive profile showing degradation of AR and L702H mutant AR with good oral bioavailability across species. The compound also inhibited AR signaling in vitro and tumor growth in vivo in a mouse prostate cancer xenograft model.
- Published
- 2024
- Full Text
- View/download PDF
3. Development of a Series of Pyrrolopyridone MAT2A Inhibitors.
- Author
-
Atkinson SJ, Bagal SK, Argyrou A, Askin S, Cheung T, Chiarparin E, Coen M, Collie IT, Dale IL, De Fusco C, Dillman K, Evans L, Feron LJ, Foster AJ, Grondine M, Kantae V, Lamont GM, Lamont S, Lynch JT, Nilsson Lill S, Robb GR, Saeh J, Schimpl M, Scott JS, Smith J, Srinivasan B, Tentarelli S, Vazquez-Chantada M, Wagner D, Walsh JJ, Watson D, and Williamson B
- Subjects
- Humans, Entropy, Methionine Adenosyltransferase metabolism, Neoplasms
- Abstract
The optimization of an allosteric fragment, discovered by differential scanning fluorimetry, to an in vivo MAT2a tool inhibitor is discussed. The structure-based drug discovery approach, aided by relative binding free energy calculations, resulted in AZ'9567 ( 21 ), a potent inhibitor in vitro with excellent preclinical pharmacokinetic properties. This tool showed a selective antiproliferative effect on methylthioadenosine phosphorylase (MTAP) KO cells, both in vitro and in vivo, providing further evidence to support the utility of MAT2a inhibitors as potential anticancer therapies for MTAP-deficient tumors.
- Published
- 2024
- Full Text
- View/download PDF
4. Discovery of AZD4747, a Potent and Selective Inhibitor of Mutant GTPase KRAS G12C with Demonstrable CNS Penetration.
- Author
-
Kettle JG, Bagal SK, Barratt D, Bodnarchuk MS, Boyd S, Braybrooke E, Breed J, Cassar DJ, Cosulich S, Davies M, Davies NL, Deng C, Eatherton A, Evans L, Feron LJ, Fillery S, Gleave ES, Goldberg FW, Cortés González MA, Guerot C, Haider A, Harlfinger S, Howells R, Jackson A, Johnström P, Kemmitt PD, Koers A, Kondrashov M, Lamont GM, Lamont S, Lewis HJ, Liu L, Mylrea M, Nash S, Niedbala MJ, Peter A, Phillips C, Pike K, Raubo P, Robb GR, Ross S, Sanders MG, Schou M, Simpson I, and Steward O
- Subjects
- Animals, Humans, Proto-Oncogene Proteins p21(ras) genetics, Drug Design, Glycine therapeutic use, Mutation, Antineoplastic Agents pharmacology, Neoplasms drug therapy, Lung Neoplasms drug therapy
- Abstract
The glycine to cysteine mutation at codon 12 of Kirsten rat sarcoma (KRAS) represents an Achilles heel that has now rendered this important GTPase druggable. Herein, we report our structure-based drug design approach that led to the identification of 14 , AZD4747, a clinical development candidate for the treatment of KRAS
G12C -positive tumors, including the treatment of central nervous system (CNS) metastases. Building on our earlier discovery of C5-tethered quinazoline AZD4625, excision of a usually critical pyrimidine ring yielded a weak but brain-penetrant start point which was optimized for potency and DMPK. Key design principles and measured parameters that give high confidence in CNS exposure are discussed. During optimization, divergence between rodent and non-rodent species was observed in CNS exposure, with primate PET studies ultimately giving high confidence in the expected translation to patients. AZD4747 is a highly potent and selective inhibitor of KRASG12C with an anticipated low clearance and high oral bioavailability profile in humans.- Published
- 2023
- Full Text
- View/download PDF
5. Discovery of AZD4625, a Covalent Allosteric Inhibitor of the Mutant GTPase KRAS G12C .
- Author
-
Kettle JG, Bagal SK, Bickerton S, Bodnarchuk MS, Boyd S, Breed J, Carbajo RJ, Cassar DJ, Chakraborty A, Cosulich S, Cumming I, Davies M, Davies NL, Eatherton A, Evans L, Feron L, Fillery S, Gleave ES, Goldberg FW, Hanson L, Harlfinger S, Howard M, Howells R, Jackson A, Kemmitt P, Lamont G, Lamont S, Lewis HJ, Liu L, Niedbala MJ, Phillips C, Polanski R, Raubo P, Robb G, Robinson DM, Ross S, Sanders MG, Tonge M, Whiteley R, Wilkinson S, Yang J, and Zhang W
- Subjects
- Drug Design, Humans, Mutation, Proto-Oncogene Proteins p21(ras) genetics, Quinazolines pharmacology, Structure-Activity Relationship, Antineoplastic Agents pharmacology, Lung Neoplasms drug therapy
- Abstract
KRAS is an archetypal high-value intractable oncology drug target. The glycine to cysteine mutation at codon 12 represents an Achilles heel that has now rendered this important GTPase druggable. Herein, we report our structure-based drug design approach that led to the identification of 21 , AZD4625, a clinical development candidate for the treatment of KRAS
G12C positive tumors. Highlights include a quinazoline tethering strategy to lock out a bio-relevant binding conformation and an optimization strategy focused on the reduction of extrahepatic clearance mechanisms seen in preclinical species. Crystallographic analysis was also key in helping to rationalize unusual structure-activity relationship in terms of ring size and enantio-preference. AZD4625 is a highly potent and selective inhibitor of KRASG12C with an anticipated low clearance and high oral bioavailability profile in humans.- Published
- 2022
- Full Text
- View/download PDF
6. Diverse, Potent, and Efficacious Inhibitors That Target the EED Subunit of the Polycomb Repressive Complex 2 Methyltransferase.
- Author
-
Bagal SK, Gregson C, O' Donovan DH, Pike KG, Bloecher A, Barton P, Borodovsky A, Code E, Fillery SM, Hsu JH, Kawatkar SP, Li C, Longmire D, Nai Y, Nash SC, Pike A, Robinson J, Read JA, Rawlins PB, Shen M, Tang J, Wang P, Woods H, and Williamson B
- Subjects
- Allosteric Regulation, Animals, Catalytic Domain, Cell Line, Cell Proliferation drug effects, Enhancer of Zeste Homolog 2 Protein chemistry, Enhancer of Zeste Homolog 2 Protein drug effects, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacokinetics, Heterocyclic Compounds chemistry, Humans, Ligands, Polycomb Repressive Complex 2 chemistry, Rats, Structure-Activity Relationship, Enzyme Inhibitors pharmacology, Polycomb Repressive Complex 2 antagonists & inhibitors
- Abstract
Aberrant activity of the histone methyltransferase polycomb repressive complex 2 (PRC2) has been linked to several cancers, with small-molecule inhibitors of the catalytic subunit of the PRC2 enhancer of zeste homologue 2 (EZH2) being recently approved for the treatment of epithelioid sarcoma (ES) and follicular lymphoma (FL). Compounds binding to the EED subunit of PRC2 have recently emerged as allosteric inhibitors of PRC2 methyltransferase activity. In contrast to orthosteric inhibitors that target EZH2, small molecules that bind to EED retain their efficacy in EZH2 inhibitor-resistant cell lines. In this paper we disclose the discovery of potent and orally bioavailable EED ligands with good solubilities. The solubility of the EED ligands was optimized through a variety of design tactics, with the resulting compounds exhibiting in vivo efficacy in EZH2-driven tumors.
- Published
- 2021
- Full Text
- View/download PDF
7. Fragment-Based Design of a Potent MAT2a Inhibitor and in Vivo Evaluation in an MTAP Null Xenograft Model.
- Author
-
De Fusco C, Schimpl M, Börjesson U, Cheung T, Collie I, Evans L, Narasimhan P, Stubbs C, Vazquez-Chantada M, Wagner DJ, Grondine M, Sanders MG, Tentarelli S, Underwood E, Argyrou A, Smith JM, Lynch JT, Chiarparin E, Robb G, Bagal SK, and Scott JS
- Subjects
- Allosteric Site, Animals, Cell Proliferation, Enzyme Inhibitors metabolism, Enzyme Inhibitors pharmacology, Enzyme Inhibitors therapeutic use, Gene Knockout Techniques, HCT116 Cells, Half-Life, Humans, Methionine Adenosyltransferase genetics, Methionine Adenosyltransferase metabolism, Mice, Molecular Dynamics Simulation, Neoplasms drug therapy, Neoplasms pathology, Quinazolines chemistry, Quinazolines metabolism, Quinazolines pharmacology, Quinazolines therapeutic use, Rats, S-Adenosylmethionine metabolism, Structure-Activity Relationship, Transplantation, Heterologous, Drug Design, Enzyme Inhibitors chemistry, Methionine Adenosyltransferase antagonists & inhibitors
- Abstract
MAT2a is a methionine adenosyltransferase that synthesizes the essential metabolite S -adenosylmethionine (SAM) from methionine and ATP. Tumors bearing the co-deletion of p16 and MTAP genes have been shown to be sensitive to MAT2a inhibition, making it an attractive target for treatment of MTAP-deleted cancers. A fragment-based lead generation campaign identified weak but efficient hits binding in a known allosteric site. By use of structure-guided design and systematic SAR exploration, the hits were elaborated through a merging and growing strategy into an arylquinazolinone series of potent MAT2a inhibitors. The selected in vivo tool compound 28 reduced SAM-dependent methylation events in cells and inhibited proliferation of MTAP-null cells in vitro . In vivo studies showed that 28 was able to induce antitumor response in an MTAP knockout HCT116 xenograft model.
- Published
- 2021
- Full Text
- View/download PDF
8. Free energy perturbation in the design of EED ligands as inhibitors of polycomb repressive complex 2 (PRC2) methyltransferase.
- Author
-
O' Donovan DH, Gregson C, Packer MJ, Greenwood R, Pike KG, Kawatkar S, Bloecher A, Robinson J, Read J, Code E, Hsu JH, Shen M, Woods H, Barton P, Fillery S, Williamson B, Rawlins PB, and Bagal SK
- Subjects
- Dose-Response Relationship, Drug, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors chemistry, Humans, Ligands, Microsomes, Liver chemistry, Microsomes, Liver metabolism, Molecular Structure, Polycomb Repressive Complex 2 metabolism, Purines chemical synthesis, Purines chemistry, Quantum Theory, Structure-Activity Relationship, Drug Design, Enzyme Inhibitors pharmacology, Polycomb Repressive Complex 2 antagonists & inhibitors, Purines pharmacology, Thermodynamics
- Abstract
Free Energy Perturbation (FEP) calculations can provide high-confidence predictions of the interaction strength between a ligand and its protein target. We sought to explore a series of triazolopyrimidines which bind to the EED subunit of the PRC2 complex as potential anticancer therapeutics, using FEP calculations to inform compound design. Combining FEP predictions with a late-stage functionalisation (LSF) inspired synthetic approach allowed us to rapidly evaluate structural modifications in a previously unexplored region of the EED binding site. This approach generated a series of novel triazolopyrimidine EED ligands with improved physicochemical properties and which inhibit PRC2 methyltransferase activity in a cancer-relevant G401 cell line., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
9. Structure-Based Design and Pharmacokinetic Optimization of Covalent Allosteric Inhibitors of the Mutant GTPase KRAS G12C .
- Author
-
Kettle JG, Bagal SK, Bickerton S, Bodnarchuk MS, Breed J, Carbajo RJ, Cassar DJ, Chakraborty A, Cosulich S, Cumming I, Davies M, Eatherton A, Evans L, Feron L, Fillery S, Gleave ES, Goldberg FW, Harlfinger S, Hanson L, Howard M, Howells R, Jackson A, Kemmitt P, Kingston JK, Lamont S, Lewis HJ, Li S, Liu L, Ogg D, Phillips C, Polanski R, Robb G, Robinson D, Ross S, Smith JM, Tonge M, Whiteley R, Yang J, Zhang L, and Zhao X
- Subjects
- Allosteric Regulation, Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents pharmacokinetics, Caco-2 Cells, Cell Line, Tumor, Drug Design, Humans, Male, Mice, Nude, Molecular Conformation, Mutation, Piperazines chemical synthesis, Piperazines pharmacokinetics, Proto-Oncogene Proteins p21(ras) genetics, Quinazolines chemical synthesis, Quinazolines pharmacokinetics, Quinolones chemical synthesis, Quinolones pharmacokinetics, Rats, Wistar, Structure-Activity Relationship, Xenograft Model Antitumor Assays, Antineoplastic Agents therapeutic use, Neoplasms drug therapy, Piperazines therapeutic use, Proto-Oncogene Proteins p21(ras) antagonists & inhibitors, Quinazolines therapeutic use, Quinolones therapeutic use
- Abstract
Attempts to directly drug the important oncogene KRAS have met with limited success despite numerous efforts across industry and academia. The KRAS
G12C mutant represents an "Achilles heel" and has recently yielded to covalent targeting with small molecules that bind the mutant cysteine and create an allosteric pocket on GDP-bound RAS, locking it in an inactive state. A weak inhibitor at this site was optimized through conformational locking of a piperazine-quinazoline motif and linker modification. Subsequent introduction of a key methyl group to the piperazine resulted in enhancements in potency, permeability, clearance, and reactivity, leading to identification of a potent KRASG12C inhibitor with high selectivity and excellent cross-species pharmacokinetic parameters and in vivo efficacy.- Published
- 2020
- Full Text
- View/download PDF
10. Discovery of Allosteric, Potent, Subtype Selective, and Peripherally Restricted TrkA Kinase Inhibitors.
- Author
-
Bagal SK, Omoto K, Blakemore DC, Bungay PJ, Bilsland JG, Clarke PJ, Corbett MS, Cronin CN, Cui JJ, Dias R, Flanagan NJ, Greasley SE, Grimley R, Johnson E, Fengas D, Kitching L, Kraus ML, McAlpine I, Nagata A, Waldron GJ, and Warmus JS
- Subjects
- Allosteric Regulation, Amino Acid Sequence, Animals, Binding Sites, Crystallography, X-Ray, Drug Evaluation, Preclinical, Half-Life, High-Throughput Screening Assays, Humans, Ligands, Microsomes, Liver metabolism, Molecular Dynamics Simulation, Protein Binding, Protein Isoforms antagonists & inhibitors, Protein Isoforms metabolism, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors pharmacokinetics, Protein Structure, Tertiary, Pyrazoles chemical synthesis, Pyrazoles chemistry, Pyrazoles pharmacokinetics, Rats, Receptor, trkA metabolism, Sequence Alignment, Structure-Activity Relationship, Protein Kinase Inhibitors chemistry, Receptor, trkA antagonists & inhibitors
- Abstract
Tropomyosin receptor kinases (TrkA, TrkB, TrkC) are activated by hormones of the neurotrophin family: nerve growth factor (NGF), brain derived neurotrophic factor (BDNF), neurotrophin 3 (NT3), and neurotrophin 4 (NT4). Moreover, the NGF antibody tanezumab has provided clinical proof of concept for inhibition of the TrkA kinase pathway in pain leading to significant interest in the development of small molecule inhibitors of TrkA. However, achieving TrkA subtype selectivity over TrkB and TrkC via a Type I and Type II inhibitor binding mode has proven challenging and Type III or Type IV allosteric inhibitors may present a more promising selectivity design approach. Furthermore, TrkA inhibitors with minimal brain availability are required to deliver an appropriate safety profile. Herein, we describe the discovery of a highly potent, subtype selective, peripherally restricted, efficacious, and well-tolerated series of allosteric TrkA inhibitors that culminated in the delivery of candidate quality compound 23.
- Published
- 2019
- Full Text
- View/download PDF
11. The discovery and optimization of benzimidazoles as selective Na V 1.8 blockers for the treatment of pain.
- Author
-
Brown AD, Bagal SK, Blackwell P, Blakemore DC, Brown B, Bungay PJ, Corless M, Crawforth J, Fengas D, Fenwick DR, Gray V, Kemp M, Klute W, Malet Sanz L, Miller D, Murata Y, Payne CE, Skerratt S, Stevens EB, and Warmus JS
- Subjects
- Benzimidazoles chemical synthesis, Benzimidazoles chemistry, Benzimidazoles pharmacokinetics, Drug Design, HEK293 Cells, Humans, Molecular Structure, Solubility, Structure-Activity Relationship, Voltage-Gated Sodium Channel Blockers chemical synthesis, Voltage-Gated Sodium Channel Blockers chemistry, Voltage-Gated Sodium Channel Blockers pharmacokinetics, Benzimidazoles pharmacology, NAV1.8 Voltage-Gated Sodium Channel metabolism, Voltage-Gated Sodium Channel Blockers pharmacology
- Abstract
The voltage gated sodium channel Na
V 1.8 has been postulated to play a key role in the transmission of pain signals. Core hopping from our previously reported phenylimidazole leads has allowed the identification of a novel series of benzimidazole NaV 1.8 blockers. Subsequent optimization allowed the identification of compound 9, PF-06305591, as a potent, highly selective blocker with an excellent preclinical in vitro ADME and safety profile., (Copyright © 2018. Published by Elsevier Ltd.)- Published
- 2019
- Full Text
- View/download PDF
12. Discovery of Potent, Selective, and Peripherally Restricted Pan-Trk Kinase Inhibitors for the Treatment of Pain.
- Author
-
Bagal SK, Andrews M, Bechle BM, Bian J, Bilsland J, Blakemore DC, Braganza JF, Bungay PJ, Corbett MS, Cronin CN, Cui JJ, Dias R, Flanagan NJ, Greasley SE, Grimley R, James K, Johnson E, Kitching L, Kraus ML, McAlpine I, Nagata A, Ninkovic S, Omoto K, Scales S, Skerratt SE, Sun J, Tran-Dubé M, Waldron GJ, Wang F, and Warmus JS
- Subjects
- Animals, Humans, Ligands, Molecular Docking Simulation, Protein Conformation, Protein Kinase Inhibitors chemistry, Protein Kinase Inhibitors pharmacokinetics, Protein Kinase Inhibitors therapeutic use, Pyridines chemistry, Pyridines pharmacokinetics, Pyridines pharmacology, Pyridines therapeutic use, Rats, Receptor Protein-Tyrosine Kinases chemistry, Receptor Protein-Tyrosine Kinases metabolism, Solubility, Structure-Activity Relationship, Tissue Distribution, Drug Discovery, Pain drug therapy, Protein Kinase Inhibitors pharmacology, Receptor Protein-Tyrosine Kinases antagonists & inhibitors
- Abstract
Hormones of the neurotrophin family, nerve growth factor (NGF), brain derived neurotrophic factor (BDNF), neurotrophin 3 (NT3), and neurotrophin 4 (NT4), are known to activate the family of Tropomyosin receptor kinases (TrkA, TrkB, and TrkC). Moreover, inhibition of the TrkA kinase pathway in pain has been clinically validated by the NGF antibody tanezumab, leading to significant interest in the development of small molecule inhibitors of TrkA. Furthermore, Trk inhibitors having an acceptable safety profile will require minimal brain availability. Herein, we discuss the discovery of two potent, selective, peripherally restricted, efficacious, and well-tolerated series of pan-Trk inhibitors which successfully delivered three candidate quality compounds 10b, 13b, and 19. All three compounds are predicted to possess low metabolic clearance in human that does not proceed via aldehyde oxidase-catalyzed reactions, thus addressing the potential clearance prediction liability associated with our current pan-Trk development candidate PF-06273340.
- Published
- 2018
- Full Text
- View/download PDF
13. Evaluation and Characterization of Trk Kinase Inhibitors for the Treatment of Pain: Reliable Binding Affinity Predictions from Theory and Computation.
- Author
-
Wan S, Bhati AP, Skerratt S, Omoto K, Shanmugasundaram V, Bagal SK, and Coveney PV
- Subjects
- Molecular Docking Simulation, Molecular Dynamics Simulation, Pain enzymology, Protein Binding, Protein Kinase Inhibitors therapeutic use, Receptor, trkA chemistry, Thermodynamics, Drug Design, Pain drug therapy, Protein Kinase Inhibitors metabolism, Protein Kinase Inhibitors pharmacology, Receptor, trkA antagonists & inhibitors, Receptor, trkA metabolism
- Abstract
Optimization of ligand binding affinity to the target protein of interest is a primary objective in small-molecule drug discovery. Until now, the prediction of binding affinities by computational methods has not been widely applied in the drug discovery process, mainly because of its lack of accuracy and reproducibility as well as the long turnaround times required to obtain results. Herein we report on a collaborative study that compares tropomyosin receptor kinase A (TrkA) binding affinity predictions using two recently formulated fast computational approaches, namely, Enhanced Sampling of Molecular dynamics with Approximation of Continuum Solvent (ESMACS) and Thermodynamic Integration with Enhanced Sampling (TIES), to experimentally derived TrkA binding affinities for a set of Pfizer pan-Trk compounds. ESMACS gives precise and reproducible results and is applicable to highly diverse sets of compounds. It also provides detailed chemical insight into the nature of ligand-protein binding. TIES can predict and thus optimize more subtle changes in binding affinities between compounds of similar structure. Individual binding affinities were calculated in a few hours, exhibiting good correlations with the experimental data of 0.79 and 0.88 from the ESMACS and TIES approaches, respectively. The speed, level of accuracy, and precision of the calculations are such that the affinity predictions can be used to rapidly explain the effects of compound modifications on TrkA binding affinity. The methods could therefore be used as tools to guide lead optimization efforts across multiple prospective structurally enabled programs in the drug discovery setting for a wide range of compounds and targets.
- Published
- 2017
- Full Text
- View/download PDF
14. The Discovery of a Potent, Selective, and Peripherally Restricted Pan-Trk Inhibitor (PF-06273340) for the Treatment of Pain.
- Author
-
Skerratt SE, Andrews M, Bagal SK, Bilsland J, Brown D, Bungay PJ, Cole S, Gibson KR, Jones R, Morao I, Nedderman A, Omoto K, Robinson C, Ryckmans T, Skinner K, Stupple P, and Waldron G
- Subjects
- Dose-Response Relationship, Drug, Humans, Models, Molecular, Molecular Structure, Pain metabolism, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors chemistry, Pyrimidines chemical synthesis, Pyrimidines chemistry, Pyrroles chemical synthesis, Pyrroles chemistry, Quantitative Structure-Activity Relationship, Drug Discovery, Pain drug therapy, Protein Kinase Inhibitors pharmacology, Protein Kinases metabolism, Pyrimidines pharmacology, Pyrroles pharmacology
- Abstract
The neurotrophin family of growth factors, comprised of nerve growth factor (NGF), brain derived neurotrophic factor (BDNF), neurotrophin 3 (NT3), and neurotrophin 4 (NT4), is implicated in the physiology of chronic pain. Given the clinical efficacy of anti-NGF monoclonal antibody (mAb) therapies, there is significant interest in the development of small molecule modulators of neurotrophin activity. Neurotrophins signal through the tropomyosin related kinase (Trk) family of tyrosine kinase receptors, hence Trk kinase inhibition represents a potentially "druggable" point of intervention. To deliver the safety profile required for chronic, nonlife threatening pain indications, highly kinase-selective Trk inhibitors with minimal brain availability are sought. Herein we describe how the use of SBDD, 2D QSAR models, and matched molecular pair data in compound design enabled the delivery of the highly potent, kinase-selective, and peripherally restricted clinical candidate PF-06273340.
- Published
- 2016
- Full Text
- View/download PDF
15. Discovery and Optimization of Selective Nav1.8 Modulator Series That Demonstrate Efficacy in Preclinical Models of Pain.
- Author
-
Bagal SK, Bungay PJ, Denton SM, Gibson KR, Glossop MS, Hay TL, Kemp MI, Lane CA, Lewis ML, Maw GN, Million WA, Payne CE, Poinsard C, Rawson DJ, Stammen BL, Stevens EB, and Thompson LR
- Abstract
Voltage-gated sodium channels, in particular Nav1.8, can be targeted for the treatment of neuropathic and inflammatory pain. Herein, we described the optimization of Nav1.8 modulator series to deliver subtype selective, state, and use-dependent chemical matter that is efficacious in preclinical models of neuropathic and inflammatory pain.
- Published
- 2015
- Full Text
- View/download PDF
16. Voltage gated sodium channels as drug discovery targets.
- Author
-
Bagal SK, Marron BE, Owen RM, Storer RI, and Swain NA
- Subjects
- Amino Acid Sequence, Analgesics chemistry, Animals, Anti-Arrhythmia Agents chemistry, Anticonvulsants chemistry, Humans, Molecular Sequence Data, Sodium Channel Blockers chemistry, Analgesics pharmacology, Anti-Arrhythmia Agents pharmacology, Anticonvulsants pharmacology, Drug Discovery methods, Sodium Channel Blockers pharmacology, Voltage-Gated Sodium Channels metabolism
- Abstract
Voltage-gated sodium (NaV) channels are a family of transmembrane ion channel proteins. They function by forming a gated, water-filled pore to help establish and control cell membrane potential via control of the flow of ions between the intracellular and the extracellular environments. Blockade of NaVs has been successfully accomplished in the clinic to enable control of pathological firing patterns that occur in a diverse range of conditions such as chronic pain, epilepsy, and cardiac arrhythmias. First generation sodium channel modulator drugs, despite low inherent subtype selectivity, preferentially act on over-excited cells which reduces undesirable side effects in the clinic. However, the limited therapeutic indices observed with the first generation demanded a new generation of sodium channel inhibitors. The structure, function and the state of the art in sodium channel modulator drug discovery are discussed in this chapter.
- Published
- 2015
- Full Text
- View/download PDF
17. Recent progress in sodium channel modulators for pain.
- Author
-
Bagal SK, Chapman ML, Marron BE, Prime R, Storer RI, and Swain NA
- Subjects
- Animals, Drug Discovery, Humans, Sodium Channel Blockers chemistry, Voltage-Gated Sodium Channels chemistry, Pain drug therapy, Sodium Channel Blockers pharmacology, Sodium Channel Blockers therapeutic use, Voltage-Gated Sodium Channels metabolism
- Abstract
Voltage-gated sodium channels (Navs) are an important family of transmembrane ion channel proteins and Nav drug discovery is an exciting field. Pharmaceutical investment in Navs for pain therapeutics has expanded exponentially due to genetic data such as SCN10A mutations and an improved ability to establish an effective screen sequence for example IonWorks Barracuda®, Synchropatch® and Qube®. Moreover, emerging clinical data (AZD-3161, XEN402, CNV1014802, PF-05089771, PF-04531083) combined with recent breakthroughs in Nav structural biology pave the way for a future of fruitful prospective Nav drug discovery., (Copyright © 2014 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
18. Ion channels as therapeutic targets: a drug discovery perspective.
- Author
-
Bagal SK, Brown AD, Cox PJ, Omoto K, Owen RM, Pryde DC, Sidders B, Skerratt SE, Stevens EB, Storer RI, and Swain NA
- Subjects
- Humans, Ion Channels chemistry, Models, Molecular, Phylogeny, Drug Discovery, Ion Channels drug effects
- Abstract
Ion channels are membrane proteins expressed in almost all living cells. The sequencing of the human genome has identified more than 400 putative ion channels, but only a fraction of these have been cloned and functionally tested. The widespread tissue distribution of ion channels, coupled with the plethora of physiological consequences of their opening and closing, makes ion-channel-targeted drug discovery highly compelling. However, despite some important drugs in clinical use today, as a class, ion channels remain underexploited in drug discovery and many existing drugs are poorly selective with significant toxicities or suboptimal efficacy. This Perspective seeks to review the ion channel family, its structural and functional features, and the diseases that are known to be modulated by members of the family. In particular, we will explore the structure and properties of known ligands and consider the future prospects for drug discovery in this challenging but high potential area.
- Published
- 2013
- Full Text
- View/download PDF
19. Minimizing Drug Exposure in the CNS while Maintaining Good Oral Absorption.
- Author
-
Bagal SK and Bungay PJ
- Abstract
In some drug discovery approaches, it is advantageous to restrict the access of compounds to the CNS to minimize the risk of side effects. By choosing appropriate physicochemical properties and building in the ability to act as substrates for active efflux transporters, it is possible to achieve CNS restriction and still retain sufficient absorption through the intestinal epithelium to retain good oral bioavailability. Potential risks in employing this approach are considered.
- Published
- 2012
- Full Text
- View/download PDF
20. Syntheses of the enantiomers of 1-deoxynojirimycin and 1-deoxyaltronojirimycin via chemo- and diastereoselective olefinic oxidation of unsaturated amines.
- Author
-
Bagal SK, Davies SG, Lee JA, Roberts PM, Scott PM, and Thomson JE
- Subjects
- Magnetic Resonance Spectroscopy, Molecular Structure, Oxidation-Reduction, Stereoisomerism, 1-Deoxynojirimycin chemical synthesis, Alkenes chemistry, Amines chemistry, Epoxy Compounds chemistry
- Abstract
Oxidation of enantiomerically pure (R)-N(1)-1'-(1''-naphthyl)ethyl-2,7-dihydro-1H-azepine with m-CPBA in the presence of HBF(4) and BnOH gave (3S,4R,5S,6S,1'R)-N(1)-1'-(1''-naphthyl)ethyl-3-hydroxy-4-benzyloxy-5,6-epoxyazepane as the major product and as a single diastereoisomer after chromatography. Elaboration of this highly functionalized intermediate via ring contraction to (2S,3R,4S,5S,1'R)-N(1)-benzyl-2-chloromethyl-3-benzyloxy-4,5-epoxypiperidine followed by regioselective epoxide ring opening, functional group manipulation, and deprotection gave (+)-1-deoxyaltronojirimycin. Alternatively, resolution of (RS,RS)-N(1)-benzyl-3-hydroxy-4-benzyloxy-2,3,4,7-tetrahydro-1H-azepine or (3RS,4SR,5RS,6RS)-N(1)-benzyl-3-hydroxy-4-benzyloxy-5,6-epoxyazepane by preparative chiral HPLC and subsequent elaboration allows access to the enantiomers of 1-deoxynojirimycin and 1-deoxyaltronojirimycin, respectively.
- Published
- 2010
- Full Text
- View/download PDF
21. An oxidation and ring contraction approach to the synthesis of (+/-)-1-deoxynojirimycin and (+/-)-1-deoxyaltronojirimycin.
- Author
-
Bagal SK, Davies SG, Lee JA, Roberts PM, Russell AJ, Scott PM, and Thomson JE
- Subjects
- 1-Deoxynojirimycin chemistry, Catalysis, Crystallography, X-Ray, Molecular Conformation, Molecular Structure, Oxidation-Reduction, Stereoisomerism, 1-Deoxynojirimycin chemical synthesis, Azepines chemistry
- Abstract
A reaction sequence involving the chemoselective olefinic oxidation of N(1)-benzyl-2,7-dihydro-1H-azepine with m-CPBA in the presence of HBF(4) and BnOH followed by ring contraction facilitates the stereoselective preparation of either of the epoxide diastereoisomers of (2RS,3SR)-N(1)-benzyl-2-chloromethyl-3-benzyloxy-4,5-epoxypiperidine by simple modification of the reaction conditions. Epoxide ring opening, functional group interconversion, and deprotection allow the synthesis of (+/-)-1-deoxynojirimycin and (+/-)-1-deoxyaltronojirimycin.
- Published
- 2010
- Full Text
- View/download PDF
22. Recent advances in biomimetic natural product synthesis.
- Author
-
Bulger PG, Bagal SK, and Marquez R
- Subjects
- Biological Products chemistry, Molecular Structure, Biological Products chemical synthesis, Molecular Mimicry
- Abstract
This review highlights some of the most elegant and instructive biomimetic syntheses of natural products over the last few years, providing an updated overview of this area of research.
- Published
- 2008
- Full Text
- View/download PDF
23. Cyano(ethoxycarbonothioylthio)methyl benzoate: a novel one-carbon radical equivalent.
- Author
-
Bagal SK, de Greef M, and Zard SZ
- Abstract
[reaction: see text] Cyano(ethoxycarbonothioylthio)methyl benzoate 3 has been prepared and shown to be an excellent one-carbon radical equivalent that can be applied for the introduction of an acyl unit via xanthate transfer radical addition to olefins. The corresponding adducts can be further elaborated. A rare 1,5-nitrile translocation was also observed during the study.
- Published
- 2006
- Full Text
- View/download PDF
24. Dimerization of butenolide structures. A biomimetic approach to the dimeric sesquiterpene lactones (+/-)-biatractylolide and (+/-)-biepiasterolide.
- Author
-
Bagal SK, Adlington RM, Baldwin JE, and Marquez R
- Subjects
- Dimerization, Models, Molecular, Molecular Mimicry, Molecular Structure, Spectrum Analysis, X-Ray Diffraction, 4-Butyrolactone analogs & derivatives, 4-Butyrolactone chemistry, Furans chemistry, Sesquiterpenes chemistry
- Abstract
The biomimetic synthesis of the bisesquiterpene lactones (+/-)-biatractylolide 1 and (+/-)-biepiasterolide 2 via dimerization of the captodative stabilized radical 8 is reported. Atractylon 7 has also been shown to be a possible intermediate during the biosynthesis of biatractylolide 1, biepiasterolide 2, atractylolide 3, and hydroxyatractylolide 4.
- Published
- 2004
- Full Text
- View/download PDF
25. Biomimetic synthesis of biatractylolide and biepiasterolide.
- Author
-
Bagal SK, Adlington RM, Baldwin JE, Marquez R, and Cowley A
- Subjects
- Atractylodes chemistry, Biomimetics, Dimerization, Models, Molecular, Furans chemical synthesis, Sesquiterpenes chemical synthesis, Sesquiterpenes chemistry
- Abstract
[reaction: see text] The biomimetic synthesis of the bisesquiterpenoids biatractylolide 1 and biepiasterolide 2 is reported.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.