Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Elife ; 122023 03 28.
Article in English | MEDLINE | ID: mdl-36975198

ABSTRACT

The voltage-gated sodium (NaV) channel NaV1.7 has been identified as a potential novel analgesic target due to its involvement in human pain syndromes. However, clinically available NaV channel-blocking drugs are not selective among the nine NaV channel subtypes, NaV1.1-NaV1.9. Moreover, the two currently known classes of NaV1.7 subtype-selective inhibitors (aryl- and acylsulfonamides) have undesirable characteristics that may limit their development. To this point understanding of the structure-activity relationships of the acylsulfonamide class of NaV1.7 inhibitors, exemplified by the clinical development candidate GDC-0310, has been based solely on a single co-crystal structure of an arylsulfonamide inhibitor bound to voltage-sensing domain 4 (VSD4). To advance inhibitor design targeting the NaV1.7 channel, we pursued high-resolution ligand-bound NaV1.7-VSD4 structures using cryogenic electron microscopy (cryo-EM). Here, we report that GDC-0310 engages the NaV1.7-VSD4 through an unexpected binding mode orthogonal to the arylsulfonamide inhibitor class binding pose, which identifies a previously unknown ligand binding site in NaV channels. This finding enabled the design of a novel hybrid inhibitor series that bridges the aryl- and acylsulfonamide binding pockets and allows for the generation of molecules with substantially differentiated structures and properties. Overall, our study highlights the power of cryo-EM methods to pursue challenging drug targets using iterative and high-resolution structure-guided inhibitor design. This work also underscores an important role of the membrane bilayer in the optimization of selective NaV channel modulators targeting VSD4.


Subject(s)
Cryoelectron Microscopy , Humans , Ligands , Protein Domains , Binding Sites , Structure-Activity Relationship
2.
Bioorg Med Chem Lett ; 50: 128335, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34425201

ABSTRACT

Fulvestrant is an FDA-approved drug with a dual mechanism of action (MOA), acting as a full antagonist and degrader of the estrogen receptor protein. A significant limitation of fulvestrant is the dosing regimen required for efficacy. Due to its high lipophilicity and poor pharmacokinetic profile, fulvestrant needs to be administered through intramuscular injections which leads to injection site soreness. This route of administration also limits the dose and target occupancy in patients. We envisioned a best-in-class molecule that would function with the same dual MOA as fulvestrant, but with improved physicochemical properties and would be orally bioavailable. Herein we report our progress toward that goal, resulting in a new lead GNE-502 which addressed some of the liabilities of our previously reported lead molecule GNE-149.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/drug therapy , Drug Discovery , Receptors, Estrogen/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , Female , Humans , MCF-7 Cells , Mice , Molecular Structure , Protein Conformation , Xenograft Model Antitumor Assays
3.
J Med Chem ; 64(16): 11841-11856, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34251202

ABSTRACT

Breast cancer remains a leading cause of cancer death in women, representing a significant unmet medical need. Here, we disclose our discovery efforts culminating in a clinical candidate, 35 (GDC-9545 or giredestrant). 35 is an efficient and potent selective estrogen receptor degrader (SERD) and a full antagonist, which translates into better antiproliferation activity than known SERDs (1, 6, 7, and 9) across multiple cell lines. Fine-tuning the physiochemical properties enabled once daily oral dosing of 35 in preclinical species and humans. 35 exhibits low drug-drug interaction liability and demonstrates excellent in vitro and in vivo safety profiles. At low doses, 35 induces tumor regressions either as a single agent or in combination with a CDK4/6 inhibitor in an ESR1Y537S mutant PDX or a wild-type ERα tumor model. Currently, 35 is being evaluated in Phase III clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Carbolines/therapeutic use , Estrogen Receptor Antagonists/therapeutic use , Estrogen Receptor alpha/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Carbolines/chemistry , Carbolines/pharmacokinetics , Dogs , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor Antagonists/pharmacokinetics , Female , Humans , MCF-7 Cells , Macaca fascicularis , Mice , Molecular Structure , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
4.
J Med Chem ; 64(6): 2953-2966, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33682420

ABSTRACT

Nav1.7 is an extensively investigated target for pain with a strong genetic link in humans, yet in spite of this effort, it remains challenging to identify efficacious, selective, and safe inhibitors. Here, we disclose the discovery and preclinical profile of GDC-0276 (1) and GDC-0310 (2), selective Nav1.7 inhibitors that have completed Phase 1 trials. Our initial search focused on close-in analogues to early compound 3. This resulted in the discovery of GDC-0276 (1), which possessed improved metabolic stability and an acceptable overall pharmacokinetics profile. To further derisk the predicted human pharmacokinetics and enable QD dosing, additional optimization of the scaffold was conducted, resulting in the discovery of a novel series of N-benzyl piperidine Nav1.7 inhibitors. Improvement of the metabolic stability by blocking the labile benzylic position led to the discovery of GDC-0310 (2), which possesses improved Nav selectivity and pharmacokinetic profile over 1.


Subject(s)
Azetidines/pharmacology , Benzamides/pharmacology , Drug Discovery , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Sulfonamides/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Animals , Azetidines/chemistry , Azetidines/pharmacokinetics , Benzamides/chemistry , Benzamides/pharmacokinetics , Cells, Cultured , HEK293 Cells , Humans , Piperidines/chemistry , Piperidines/pharmacokinetics , Piperidines/pharmacology , Rats, Sprague-Dawley , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/pharmacokinetics
5.
Org Biomol Chem ; 17(43): 9510-9513, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31657418

ABSTRACT

Nucleophilic addition of Grignard reagents to tetrahydro-ß-carboline (THC) N-sulfonyl N,S-acetal generates exclusively cis-1,3-disubstituted THCs with a unique 1,3-diaxial conformation. The stereochemical relationship of the 1,3-substituents was confirmed by 2-dimensional NMR spectroscopy and X-ray crystallography. The mechanism of the reaction is proposed based on crystal structures and molecular orbital calculations.

6.
Bioorg Med Chem Lett ; 29(16): 2090-2093, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31311734

ABSTRACT

Phenolic groups are responsible for the high clearance and low oral bioavailability of the estrogen receptor alpha (ERα) clinical candidate GDC-0927. An exhaustive search for a backup molecule with improved pharmacokinetic (PK) properties identified several metabolically stable analogs, although in general at the expense of the desired potency and degradation efficiency. C-8 hydroxychromene 30 is the first example of a phenol-containing chromene that not only maintained excellent potency but also exhibited 10-fold higher oral exposure in rats. The improved in vivo clearance in rat was hypothesized to be the result of C-8 hydroxy group being sterically protected from glucuronide conjugation. The excellent potency underscores the possibility of replacing the presumed indispensable phenolic group at C-6 or C-7 of the chromene core. Co-crystal structures were obtained to highlight the change in key interactions and rationalize the retained potency.


Subject(s)
Azetidines/pharmacology , Estrogen Receptor alpha/metabolism , Flavonoids/pharmacology , Administration, Oral , Animals , Azetidines/administration & dosage , Azetidines/metabolism , Azetidines/pharmacokinetics , Crystallography, X-Ray , Drug Discovery , Drug Stability , Flavonoids/administration & dosage , Flavonoids/metabolism , Flavonoids/pharmacokinetics , Humans , MCF-7 Cells , Microsomes, Liver/metabolism , Rats , Stereoisomerism , Structure-Activity Relationship
7.
J Med Chem ; 62(8): 4091-4109, 2019 04 25.
Article in English | MEDLINE | ID: mdl-30943032

ABSTRACT

Using structure- and ligand-based design principles, a novel series of piperidyl chromane arylsulfonamide Nav1.7 inhibitors was discovered. Early optimization focused on improvement of potency through refinement of the low energy ligand conformation and mitigation of high in vivo clearance. An in vitro hepatotoxicity hazard was identified and resolved through optimization of lipophilicity and lipophilic ligand efficiency to arrive at GNE-616 (24), a highly potent, metabolically stable, subtype selective inhibitor of Nav1.7. Compound 24 showed a robust PK/PD response in a Nav1.7-dependent mouse model, and site-directed mutagenesis was used to identify residues critical for the isoform selectivity profile of 24.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/chemistry , Sulfonamides/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Analgesics/chemistry , Analgesics/metabolism , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Binding Sites , Cell Line , Cell Survival/drug effects , Chronic Pain/drug therapy , Chronic Pain/pathology , Dogs , Half-Life , Humans , Ligands , Male , Mice , Molecular Docking Simulation , Mutagenesis, Site-Directed , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Rats , Structure-Activity Relationship , Sulfonamides/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Voltage-Gated Sodium Channel Blockers/metabolism , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/therapeutic use
8.
J Comput Aided Mol Des ; 33(3): 307-330, 2019 03.
Article in English | MEDLINE | ID: mdl-30756207

ABSTRACT

Targeting the interaction with or displacement of the 'right' water molecule can significantly increase inhibitor potency in structure-guided drug design. Multiple computational approaches exist to predict which waters should be targeted for displacement to achieve the largest gain in potency. However, the relative success of different methods remains underexplored. Here, we present a comparison of the ability of five water prediction programs (3D-RISM, SZMAP, WaterFLAP, WaterRank, and WaterMap) to predict crystallographic water locations, calculate their binding free energies, and to relate differences in these energies to observed changes in potency. The structural cohort included nine Bruton's Tyrosine Kinase (BTK) structures, and nine bromodomain structures. Each program accurately predicted the locations of most crystallographic water molecules. However, the predicted binding free energies correlated poorly with the observed changes in inhibitor potency when solvent atoms were displaced by chemical changes in closely related compounds.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/chemistry , Computer Simulation , Models, Molecular , Protein Kinase Inhibitors/chemistry , Water/chemistry , Crystallography, X-Ray , Ligands , Protein Binding , Protein Domains , Software , Solvents/chemistry , Structure-Activity Relationship , Thermodynamics
9.
Bioorg Med Chem Lett ; 29(7): 905-911, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30732944

ABSTRACT

Despite tremendous progress made in the understanding of the ERα signaling pathway and the approval of many therapeutic agents, ER+ breast cancer continues to be a leading cause of cancer death in women. We set out to discover compounds with a dual mechanism of action in which they not only compete with estradiol for binding with ERα, but also can induce the degradation of the ERα protein itself. We were attracted to the constrained chromenes containing a tetracyclic benzopyranobenzoxepine scaffold, which were reported as potent selective estrogen receptor modulators (SERMs). Incorporation of a fluoromethyl azetidine side chain yielded highly potent and efficacious selective estrogen receptor degraders (SERDs), such as 16aa and surprisingly, also its enantiomeric pair 16ab. Co-crystal structures of the enantiomeric pair 16aa and 16ab in complex with ERα revealed default (mimics the A-D rings of endogenous ligand estradiol) and core-flipped binding modes, rationalizing the equivalent potency observed for these enantiomers in the ERα degradation and MCF-7 anti-proliferation assays.


Subject(s)
Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Estrogen Receptor alpha/chemistry , Antineoplastic Agents/chemistry , Benzopyrans/chemistry , Crystallization , Humans , MCF-7 Cells , Models, Molecular , Molecular Structure , Protein Conformation , Signal Transduction , Structure-Activity Relationship
10.
J Med Chem ; 62(2): 908-927, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30499663

ABSTRACT

Herein, we report the discovery and optimization of a series of orally bioavailable acyl sulfonamide NaV1.7 inhibitors that are selective for NaV1.7 over NaV1.5 and highly efficacious in in vivo models of pain and hNaV1.7 target engagement. An analysis of the physicochemical properties of literature NaV1.7 inhibitors suggested that acyl sulfonamides with high fsp3 could overcome some of the pharmacokinetic (PK) and efficacy challenges seen with existing series. Parallel library syntheses lead to the identification of analogue 7, which exhibited moderate potency against NaV1.7 and an acceptable PK profile in rodents, but relatively poor stability in human liver microsomes. Further, design strategy then focused on the optimization of potency against hNaV1.7 and improvement of human metabolic stability, utilizing induced fit docking in our previously disclosed X-ray cocrystal of the NaV1.7 voltage sensing domain. These investigations culminated in the discovery of tool compound 33, one of the most potent and efficacious NaV1.7 inhibitors reported to date.


Subject(s)
Analgesics/chemistry , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Sulfonamides/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Analgesics/metabolism , Analgesics/therapeutic use , Animals , Binding Sites , Drug Design , Half-Life , Humans , Male , Mice , Mice, Transgenic , Microsomes, Liver/metabolism , Molecular Docking Simulation , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pain/chemically induced , Pain/drug therapy , Pain/pathology , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Sulfonamides/metabolism , Sulfonamides/therapeutic use , Voltage-Gated Sodium Channel Blockers/metabolism , Voltage-Gated Sodium Channel Blockers/therapeutic use
11.
J Med Chem ; 61(11): 4810-4831, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29737846

ABSTRACT

The sodium channel NaV1.7 has emerged as a promising target for the treatment of pain based on strong genetic validation of its role in nociception. In recent years, a number of aryl and acyl sulfonamides have been reported as potent inhibitors of NaV1.7, with high selectivity over the cardiac isoform NaV1.5. Herein, we report on the discovery of a novel series of N-([1,2,4]triazolo[4,3- a]pyridin-3-yl)methanesulfonamides as selective NaV1.7 inhibitors. Starting with the crystal structure of an acyl sulfonamide, we rationalized that cyclization to form a fused heterocycle would improve physicochemical properties, in particular lipophilicity. Our design strategy focused on optimization of potency for block of NaV1.7 and human metabolic stability. Lead compounds 10, 13 (GNE-131), and 25 showed excellent potency, good in vitro metabolic stability, and low in vivo clearance in mouse, rat, and dog. Compound 13 also displayed excellent efficacy in a transgenic mouse model of induced pain.


Subject(s)
Drug Design , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pain/drug therapy , Sulfonamides/chemistry , Sulfonamides/pharmacology , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/pharmacology , Amino Acid Sequence , Animals , Dogs , Drug Stability , Humans , Kinetics , Mice , Molecular Conformation , Pain/metabolism , Rats , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/therapeutic use
12.
J Med Chem ; 61(6): 2227-2245, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29457982

ABSTRACT

Bruton's tyrosine kinase (Btk) is a nonreceptor cytoplasmic tyrosine kinase involved in B-cell and myeloid cell activation, downstream of B-cell and Fcγ receptors, respectively. Preclinical studies have indicated that inhibition of Btk activity might offer a potential therapy in autoimmune diseases such as rheumatoid arthritis and systemic lupus erythematosus. Here we disclose the discovery and preclinical characterization of a potent, selective, and noncovalent Btk inhibitor currently in clinical development. GDC-0853 (29) suppresses B cell- and myeloid cell-mediated components of disease and demonstrates dose-dependent activity in an in vivo rat model of inflammatory arthritis. It demonstrates highly favorable safety, pharmacokinetic (PK), and pharmacodynamic (PD) profiles in preclinical and Phase 2 studies ongoing in patients with rheumatoid arthritis, lupus, and chronic spontaneous urticaria. On the basis of its potency, selectivity, long target residence time, and noncovalent mode of inhibition, 29 has the potential to be a best-in-class Btk inhibitor for a wide range of immunological indications.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridones/pharmacology , Agammaglobulinaemia Tyrosine Kinase/drug effects , Agammaglobulinaemia Tyrosine Kinase/genetics , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/toxicity , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Dogs , Drug Discovery , Humans , Lupus Erythematosus, Systemic/drug therapy , Madin Darby Canine Kidney Cells , Models, Molecular , Molecular Structure , Piperazines/pharmacokinetics , Piperazines/toxicity , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/toxicity , Pyridones/pharmacokinetics , Pyridones/toxicity , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley
13.
Proc Natl Acad Sci U S A ; 115(4): E792-E801, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29311306

ABSTRACT

Many ion channels, including Nav1.7, Cav1.3, and Kv1.3, are linked to human pathologies and are important therapeutic targets. To develop efficacious and safe drugs, subtype-selective modulation is essential, but has been extremely difficult to achieve. We postulate that this challenge is caused by the poor assay design, and investigate the Nav1.7 membrane potential assay, one of the most extensively employed screening assays in modern drug discovery. The assay uses veratridine to activate channels, and compounds are identified based on the inhibition of veratridine-evoked activities. We show that this assay is biased toward nonselective pore blockers and fails to detect the most potent, selective voltage-sensing domain 4 (VSD4) blockers, including PF-05089771 (PF-771) and GX-936. By eliminating a key binding site for pore blockers and replacing veratridine with a VSD-4 binding activator, we directed the assay toward non-pore-blocking mechanisms and discovered Nav1.7-selective chemical scaffolds. Hence, we address a major hurdle in Nav1.7 drug discovery, and this mechanistic approach to assay design is applicable to Cav3.1, Kv1.3, and many other ion channels to facilitate drug discovery.


Subject(s)
Drug Discovery/methods , Molecular Targeted Therapy , Voltage-Gated Sodium Channel Blockers/analysis , Animals , High-Throughput Screening Assays , Humans , Insect Proteins , Membrane Potentials , NAV1.7 Voltage-Gated Sodium Channel/drug effects , NAV1.7 Voltage-Gated Sodium Channel/genetics , Rats , Veratridine , Wasp Venoms
14.
Methods Mol Biol ; 1705: 345-350, 2018.
Article in English | MEDLINE | ID: mdl-29188571

ABSTRACT

The practice of computational chemistry in an industrial setting poses unique opportunities and challenges. Industrial computational chemists must manage large amounts of data, master modeling software, write scripts to perform custom calculations, and stay abreast of scientific advances in the field. Just as importantly, because computational chemists are full partners in the drug discovery effort at companies, in order to influence and streamline the drug discovery process, they must communicate effectively with medicinal chemists and other scientists to deliver results of their calculations in a timely fashion. The skills necessary to play this role require education that emphasizes a combination of chemistry, programming, and communication skills. Professors are encouraged to incorporate such training in their curriculum.


Subject(s)
Chemistry, Pharmaceutical , Computational Biology , Drug Discovery , Biotechnology , Chemistry, Pharmaceutical/methods , Computational Biology/methods , Drug Discovery/methods , Drug Industry , Humans
15.
ACS Med Chem Lett ; 8(6): 608-613, 2017 Jun 08.
Article in English | MEDLINE | ID: mdl-28626519

ABSTRACT

In our continued effort to discover and develop best-in-class Bruton's tyrosine kinase (Btk) inhibitors for the treatment of B-cell lymphomas, rheumatoid arthritis, and systemic lupus erythematosus, we devised a series of novel tricyclic compounds that improved upon the druglike properties of our previous chemical matter. Compounds exemplified by G-744 are highly potent, selective for Btk, metabolically stable, well tolerated, and efficacious in an animal model of arthritis.

16.
Bioorg Med Chem Lett ; 27(13): 2974-2981, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28512031

ABSTRACT

A high-throughput screening (HTS) of the Genentech/Roche library identified a novel, uncharged scaffold as a KDM5A inhibitor. Lacking insight into the binding mode, initial attempts to improve inhibitor potency failed to improve potency, and synthesis of analogs was further hampered by the presence of a C-C bond between the pyrrolidine and pyridine. Replacing this with a C-N bond significantly simplified synthesis, yielding pyrazole analog 35, of which we obtained a co-crystal structure with KDM5A. Using structure-based design approach, we identified 50 with improved biochemical, cell potency and reduced MW and lower lipophilicity (LogD) compared with the original hit. Furthermore, 50 showed lower clearance than 9 in mice. In combination with its remarkably low plasma protein binding (PPB) in mice (40%), oral dosing of 50 at 5mg/kg resulted in unbound Cmax ∼2-fold of its cell potency (PC9 H3K4Me3 0.96µM), meeting our criteria for an in vivo tool compound from a new scaffold.


Subject(s)
Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Pyrazoles/pharmacology , Retinoblastoma-Binding Protein 2/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Humans , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Docking Simulation , Molecular Structure , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Rats , Retinoblastoma-Binding Protein 2/metabolism , Structure-Activity Relationship
17.
J Comput Aided Mol Des ; 31(3): 287-291, 2017 03.
Article in English | MEDLINE | ID: mdl-27796615

ABSTRACT

Computational chemistry/informatics scientists and software engineers in Genentech Small Molecule Drug Discovery collaborate with experimental scientists in a therapeutic project-centric environment. Our mission is to enable and improve pre-clinical drug discovery design and decisions. Our goal is to deliver timely data, analysis, and modeling to our therapeutic project teams using best-in-class software tools. We describe our strategy, the organization of our group, and our approaches to reach this goal. We conclude with a summary of the interdisciplinary skills required for computational scientists and recommendations for their training.


Subject(s)
Computer-Aided Design , Drug Design , Models, Molecular , Chemistry, Pharmaceutical , Computational Biology , Drug Discovery , Molecular Dynamics Simulation , Software
18.
J Comput Aided Mol Des ; 30(11): 945-958, 2016 11.
Article in English | MEDLINE | ID: mdl-27718028

ABSTRACT

Small molecule distribution coefficients between immiscible nonaqueuous and aqueous phases-such as cyclohexane and water-measure the degree to which small molecules prefer one phase over another at a given pH. As distribution coefficients capture both thermodynamic effects (the free energy of transfer between phases) and chemical effects (protonation state and tautomer effects in aqueous solution), they provide an exacting test of the thermodynamic and chemical accuracy of physical models without the long correlation times inherent to the prediction of more complex properties of relevance to drug discovery, such as protein-ligand binding affinities. For the SAMPL5 challenge, we carried out a blind prediction exercise in which participants were tasked with the prediction of distribution coefficients to assess its potential as a new route for the evaluation and systematic improvement of predictive physical models. These measurements are typically performed for octanol-water, but we opted to utilize cyclohexane for the nonpolar phase. Cyclohexane was suggested to avoid issues with the high water content and persistent heterogeneous structure of water-saturated octanol phases, since it has greatly reduced water content and a homogeneous liquid structure. Using a modified shake-flask LC-MS/MS protocol, we collected cyclohexane/water distribution coefficients for a set of 53 druglike compounds at pH 7.4. These measurements were used as the basis for the SAMPL5 Distribution Coefficient Challenge, where 18 research groups predicted these measurements before the experimental values reported here were released. In this work, we describe the experimental protocol we utilized for measurement of cyclohexane-water distribution coefficients, report the measured data, propose a new bootstrap-based data analysis procedure to incorporate multiple sources of experimental error, and provide insights to help guide future iterations of this valuable exercise in predictive modeling.


Subject(s)
Cyclohexanes/chemistry , Pharmaceutical Preparations/chemistry , Solvents/chemistry , Water/chemistry , Chromatography, Liquid , Computer Simulation , Hydrogen-Ion Concentration , Models, Chemical , Solubility , Tandem Mass Spectrometry , Thermodynamics
19.
Bioorg Med Chem Lett ; 26(18): 4492-4496, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27499454

ABSTRACT

Features from a high throughput screening (HTS) hit and a previously reported scaffold were combined to generate 1,7-naphthyridones as novel KDM5 enzyme inhibitors with nanomolar potencies. These molecules exhibited high selectivity over the related KDM4C and KDM2B isoforms. An X-ray co-crystal structure of a representative molecule bound to KDM5A showed that these inhibitors are competitive with the co-substrate (2-oxoglutarate or 2-OG).


Subject(s)
Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Naphthyridines/pharmacology , Nuclear Proteins/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Retinoblastoma-Binding Protein 2/antagonists & inhibitors , Animals , Crystallography, X-Ray , Dogs , Drug Design , Humans , Madin Darby Canine Kidney Cells , Naphthyridines/chemistry , Structure-Activity Relationship
20.
ACS Chem Biol ; 11(10): 2897-2907, 2016 10 21.
Article in English | MEDLINE | ID: mdl-27571029

ABSTRACT

The Bruton's tyrosine kinase (Btk) inhibitor ibrutinib has shown impressive clinical efficacy in a range of B-cell malignancies. However, acquired resistance has emerged, and second generation therapies are now being sought. Ibrutinib is a covalent, irreversible inhibitor that modifies Cys481 in the ATP binding site of Btk and renders the enzyme inactive, thereby blocking B-cell receptor signal transduction. Not surprisingly, Cys481 is the most commonly mutated Btk residue in cases of acquired resistance to ibrutinib. Mutations at other sites, including Thr474, a gatekeeper residue, have also been detected. Herein, we describe noncovalent Btk inhibitors that differ from covalent inhibitors like ibrutinib in that they do not interact with Cys481, they potently inhibit the ibrutinib-resistant Btk C481S mutant in vitro and in cells, and they are exquisitely selective for Btk. Noncovalent inhibitors such as GNE-431 also show excellent potency against the C481R, T474I, and T474M mutants. X-ray crystallographic analysis of Btk provides insight into the unique mode of binding of these inhibitors that explains their high selectivity for Btk and their retained activity against mutant forms of Btk. This class of noncovalent Btk inhibitors may provide a treatment option to patients, especially those who have acquired resistance to ibrutinib by mutation of Cys481 or Thr474.


Subject(s)
Cysteine/genetics , Mutation , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Threonine/genetics , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Kinetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Piperidines , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...