Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
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.
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
3.
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
4.
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
5.
ACS Med Chem Lett ; 7(6): 595-600, 2016 Jun 09.
Article in English | MEDLINE | ID: mdl-27326333

ABSTRACT

Using Sorafenib as a starting point, a series of potent and selective inhibitors of CDK8 was developed. When cocrystallized with CDK8 and cyclin C, these compounds exhibit a Type-II (DMG-out) binding mode.

6.
ACS Med Chem Lett ; 7(3): 223-8, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26985305

ABSTRACT

Beginning with promiscuous COT inhibitors, which were found to inhibit CDK8, a series of 6-aza-benzothiophene containing compounds were developed into potent, selective CDK8 inhibitors. When cocrystallized with CDK8 and cyclin C, these compounds exhibit an unusual binding mode, making a single hydrogen bond to the hinge residue A100, a second to K252, and a key cation-π interaction with R356. Structure-based drug design resulted in tool compounds 13 and 32, which are highly potent, kinase selective, permeable compounds with a free fraction >2% and no measurable efflux. Despite these attractive properties, these compounds exhibit weak antiproliferative activity in the HCT-116 colon cancer cell line. Further examination of the activity of 32 in this cell line revealed that the compound reduced phosphorylation of the known CDK8 substrate STAT1 in a manner identical to a CDK8 knockout clone, illustrating the complex effects of inhibition of CDK8 kinase activity in proliferation in these cells.

7.
J Comput Aided Mol Des ; 29(6): 511-23, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25921252

ABSTRACT

Structure- and property-based drug design is an integral part of modern drug discovery, enabling the design of compounds aimed at improving potency and selectivity. However, building molecules using desktop modeling tools can easily lead to poor designs that appear to form many favorable interactions with the protein's active site. Although a proposed molecule looks good on screen and appears to fit into the protein site X-ray crystal structure or pharmacophore model, doing so might require a high-energy small molecule conformation, which would likely be inactive. To help scientists make better design decisions, we have built integrated, easy-to-use, interactive software tools to perform docking experiments, de novo design, shape and pharmacophore based database searches, small molecule conformational analysis and molecular property calculations. Using a combination of these tools helps scientists in assessing the likelihood that a designed molecule will be active and have desirable drug metabolism and pharmacokinetic properties. Small molecule discovery success requires project teams to rapidly design and synthesize potent molecules with good ADME properties. Empowering scientists to evaluate ideas quickly and make better design decisions with easy-to-access and easy-to-understand software on their desktop is now a key part of our discovery process.


Subject(s)
Drug Design , Molecular Docking Simulation , Quantitative Structure-Activity Relationship , Software , Computer-Aided Design , Molecular Conformation , TYK2 Kinase/antagonists & inhibitors , TYK2 Kinase/chemistry
8.
ACS Med Chem Lett ; 5(6): 662-7, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24944740

ABSTRACT

Because of the promise of BCL-2 antagonists in combating chronic lymphocytic leukemia (CLL) and non-Hodgkin's lymphoma (NHL), interest in additional selective antagonists of antiapoptotic proteins has grown. Beginning with a series of selective, potent BCL-XL antagonists containing an undesirable hydrazone functionality, in silico design and X-ray crystallography were utilized to develop alternative scaffolds that retained the selectivity and potency of the starting compounds.

9.
Bioorg Med Chem Lett ; 23(18): 5097-104, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23932790

ABSTRACT

A series of N-7-methyl-imidazolopyrimidine inhibitors of the mTOR kinase have been designed and prepared, based on the hypothesis that the N-7-methyl substituent on imidazolopyrimidine would impart selectivity for mTOR over the related PI3Kα and δ kinases. The corresponding N-Me substituted pyrrolo[3,2-d]pyrimidines and pyrazolo[4,3-d]pyrimidines also show potent mTOR inhibition with selectivity toward both PI3α and δ kinases. The most potent compound synthesized is pyrazolo[4,3-d]pyrimidine 21c. Compound 21c shows a Ki of 2 nM against mTOR inhibition, remarkable selectivity (>2900×) over PI3 kinases, and excellent potency in cell-based assays.


Subject(s)
Drug Discovery , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship , TOR Serine-Threonine Kinases/metabolism
10.
J Med Chem ; 56(11): 4764-85, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-23659214

ABSTRACT

Herein we report on the structure-based discovery of a C-2 hydroxyethyl moiety which provided consistently high levels of selectivity for JAK1 over JAK2 to the imidazopyrrolopyridine series of JAK1 inhibitors. X-ray structures of a C-2 hydroxyethyl analogue in complex with both JAK1 and JAK2 revealed differential ligand/protein interactions between the two isoforms and offered an explanation for the observed selectivity. Analysis of historical data from related molecules was used to develop a set of physicochemical compound design parameters to impart desirable properties such as acceptable membrane permeability, potent whole blood activity, and a high degree of metabolic stability. This work culminated in the identification of a highly JAK1 selective compound (31) exhibiting favorable oral bioavailability across a range of preclinical species and robust efficacy in a rat CIA model.


Subject(s)
Antirheumatic Agents/chemical synthesis , Heterocyclic Compounds, 3-Ring/chemical synthesis , Imidazoles/chemical synthesis , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Pyridines/chemical synthesis , Pyrroles/chemical synthesis , Administration, Oral , Animals , Antirheumatic Agents/chemistry , Antirheumatic Agents/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/etiology , Biological Availability , Cell Membrane Permeability , Collagen , Crystallography, X-Ray , Dogs , Haplorhini , Heterocyclic Compounds, 3-Ring/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Janus Kinase 1/chemistry , Janus Kinase 2/chemistry , Madin Darby Canine Kidney Cells , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Pyridines/chemistry , Pyridines/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Rats , Stereoisomerism
11.
ACS Med Chem Lett ; 4(1): 103-7, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-24900569

ABSTRACT

Aberrant activation of the PI3K-Akt-mTOR signaling pathway has been observed in human tumors and tumor cell lines, indicating that these protein kinases may be attractive therapeutic targets for treating cancer. Optimization of advanced lead 1 culminated in the discovery of clinical development candidate 8h, GDC-0349, a potent and selective ATP-competitive inhibitor of mTOR. GDC-0349 demonstrates pathway modulation and dose-dependent efficacy in mouse xenograft cancer models.

12.
J Med Chem ; 55(24): 10958-71, 2012 Dec 27.
Article in English | MEDLINE | ID: mdl-23199076

ABSTRACT

Selective inhibitors of mammalian target of rapamycin (mTOR) kinase based upon saturated heterocycles fused to a pyrimidine core were designed and synthesized. Each series produced compounds with K(i) < 10 nM for the mTOR kinase and >500-fold selectivity over closely related PI3 kinases. This potency translated into strong pathway inhibition, as measured by phosphorylation of mTOR substrate proteins and antiproliferative activity in cell lines with a constitutively active PI3K pathway. Two compounds exhibiting suitable mouse PK were profiled in in vivo tumor models and were shown to suppress mTORC1 and mTORC2 signaling for over 12 h when dosed orally. Both compounds were additionally shown to suppress tumor growth in vivo in a PC3 prostate cancer model over a 14 day study.


Subject(s)
Antineoplastic Agents/chemical synthesis , Multiprotein Complexes/antagonists & inhibitors , Pyrimidines/chemical synthesis , Pyrroles/chemical synthesis , Quinazolines/chemical synthesis , TOR Serine-Threonine Kinases/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biological Availability , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Humans , Male , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Nude , Molecular Docking Simulation , Neoplasm Transplantation , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Prostatic Neoplasms , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Quinazolines/chemistry , Quinazolines/pharmacology , Structure-Activity Relationship , Transplantation, Heterologous
13.
Bioorg Med Chem Lett ; 22(24): 7627-33, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23107482

ABSTRACT

Herein we describe our successful efforts in obtaining C-2 substituted imidazo-pyrrolopyridines with improved JAK1 selectivity relative to JAK2 by targeting an amino acid residue that differs between the two isoforms (JAK1: E966; JAK2: D939). Efforts to improve cellular potency by reducing the polarity of the inhibitors are also detailed. The X-ray crystal structure of a representative inhibitor in complex with the JAK1 enzyme is also disclosed.


Subject(s)
Drug Discovery , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Animals , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Imidazoles/chemistry , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Male , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Pyridines/administration & dosage , Pyridines/chemistry , Pyrroles/administration & dosage , Pyrroles/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
14.
J Med Chem ; 55(13): 6176-93, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22698084

ABSTRACT

Herein we report the discovery of the C-2 methyl substituted imidazopyrrolopyridine series and its optimization to provide potent and orally bioavailable JAK1 inhibitors with selectivity over JAK2. The C-2 methyl substituted inhibitor 4 exhibited not only improved JAK1 potency relative to unsubstituted compound 3 but also notable JAK1 vs JAK2 selectivity (20-fold and >33-fold in biochemical and cell-based assays, respectively). Features of the X-ray structures of 4 in complex with both JAK1 and JAK2 are delineated. Efforts to improve the in vitro and in vivo ADME properties of 4 while maintaining JAK1 selectivity are described, culminating in the discovery of a highly optimized and balanced inhibitor (20). Details of the biological characterization of 20 are disclosed including JAK1 vs JAK2 selectivity levels, preclinical in vivo PK profiles, performance in an in vivo JAK1-mediated PK/PD model, and attributes of an X-ray structure in complex with JAK1.


Subject(s)
Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/chemistry , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Animals , Biological Assay , Biological Availability , Cell Line , Crystallography, X-Ray , Dogs , Hepatocytes/cytology , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Humans , Janus Kinase 1/chemistry , Janus Kinase 2/chemistry , Mice , Models, Molecular , Rats , Structure-Activity Relationship
15.
J Med Chem ; 54(9): 3426-35, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21495671

ABSTRACT

A series of inhibitors of mTOR kinase based on a quaternary-substituted dihydrofuropyrimidine was designed and synthesized. The most potent compounds in this series inhibited mTOR kinase with K(i) < 1.0 nM and were highly (>100×) selective for mTOR over the closely related PI3 kinases. Compounds in this series showed inhibition of the pathway and antiproliferative activity in cell-based assays. Furthermore, these compounds had excellent mouse PK, and showed a robust PK-PD relationship in a mouse model of cancer.


Subject(s)
Antineoplastic Agents/chemical synthesis , Furans/chemical synthesis , Pyrimidines/chemical synthesis , TOR Serine-Threonine Kinases/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biological Availability , Cell Line, Tumor , Drug Screening Assays, Antitumor , Drug Stability , Furans/pharmacokinetics , Furans/pharmacology , Humans , Mice , Mice, Nude , Models, Molecular , Neoplasm Transplantation , Phosphoinositide-3 Kinase Inhibitors , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Species Specificity , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous
16.
Bioorg Med Chem Lett ; 20(7): 2229-33, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20189383

ABSTRACT

A series of IAP antagonists based on thiazole or benzothiazole amide isosteres was designed and synthesized. These compounds were tested for binding to the XIAP-BIR3 and ML-IAP BIR using a fluorescence polarization assay. The most potent of these compounds, 19a and 33b, were found to have K(i)'s of 20-30 nM against ML-IAP and 50-60 nM against XIAP-BIR3.


Subject(s)
Amides/chemistry , Amides/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Peptides/chemistry , Thiazoles/chemistry , Thiazoles/pharmacology , Binding Sites , Biomimetics , Crystallography, X-Ray , Humans , Inhibitor of Apoptosis Proteins/metabolism , Models, Molecular , Peptides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL