Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
J Med Chem ; 67(5): 3287-3306, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38431835

ABSTRACT

Transient receptor potential ankyrin 1 (TRPA1) is a nonselective calcium ion channel highly expressed in the primary sensory neurons, functioning as a polymodal sensor for exogenous and endogenous stimuli, and has been implicated in neuropathic pain and respiratory disease. Herein, we describe the optimization of potent, selective, and orally bioavailable TRPA1 small molecule antagonists with strong in vivo target engagement in rodent models. Several lead molecules in preclinical single- and short-term repeat-dose toxicity studies exhibited profound prolongation of coagulation parameters. Based on a thorough investigative toxicology and clinical pathology analysis, anticoagulation effects in vivo are hypothesized to be manifested by a metabolite─generated by aldehyde oxidase (AO)─possessing a similar pharmacophore to known anticoagulants (i.e., coumarins, indandiones). Further optimization to block AO-mediated metabolism yielded compounds that ameliorated coagulation effects in vivo, resulting in the discovery and advancement of clinical candidate GDC-6599, currently in Phase II clinical trials for respiratory indications.


Subject(s)
Respiratory Tract Diseases , Transient Receptor Potential Channels , Humans , Transient Receptor Potential Channels/metabolism , TRPA1 Cation Channel , Aldehyde Oxidase/metabolism , Oxidoreductases/metabolism , Cytoskeletal Proteins/metabolism
2.
Anal Chem ; 95(11): 4834-4839, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36876898

ABSTRACT

The growing opportunities recognized for covalent drug inhibitors, like KRAS G12C inhibitors, are driving the need for mass spectrometry methods that can quickly and robustly measure therapeutic drug activity in vivo for drug discovery research and development. Effective front-end sample preparation is critical for proteins extracted from tumors but is generally labor intensive and impractical for large sample numbers typical in pharmacodynamic (PD) studies. Herein, we describe an automated and integrated sample preparation method for the measurement of activity levels of KRAS G12C drug inhibitor alkylation from complex tumor samples involving high throughput detergent removal and preconcentration followed by quantitation using mass spectrometry. We introduce a robust assay with an average intra-assay coefficient of variation (CV) of 4% and an interassay CV of 6% obtained from seven studies, enabling us to understand the relationship between KRAS G12C target occupancy and the therapeutic PD effect from mouse tumor samples. Further, the data demonstrated that the drug candidate GDC-6036, a KRAS G12C covalent inhibitor, shows dose-dependent target inhibition (KRAS G12C alkylation) and MAPK pathway inhibition, which correlate with high antitumor potency in the MIA PaCa-2 pancreatic xenograft model.


Subject(s)
Antineoplastic Agents , Proto-Oncogene Proteins p21(ras) , Humans , Animals , Mice , Proto-Oncogene Proteins p21(ras)/genetics , Cell Line, Tumor , Mutation , Antineoplastic Agents/pharmacology , Disease Models, Animal
3.
ACS Med Chem Lett ; 12(5): 791-797, 2021 May 13.
Article in English | MEDLINE | ID: mdl-34055227

ABSTRACT

Structure-based optimization of a set of aryl urea RAF inhibitors has led to the identification of Type II pan-RAF inhibitor GNE-9815 (7), which features a unique pyrido[2,3-d]pyridazin-8(7H)-one hinge-binding motif. With minimal polar hinge contacts, the pyridopyridazinone hinge binder moiety affords exquisite kinase selectivity in a lipophilic efficient manner. The improved physicochemical properties of GNE-9815 provided a path for oral dosing without enabling formulations. In vivo evaluation of GNE-9815 in combination with the MEK inhibitor cobimetinib demonstrated synergistic MAPK pathway modulation in an HCT116 xenograft mouse model. To the best of our knowledge, GNE-9815 is among the most highly kinase-selective RAF inhibitors reported to date.

4.
J Med Chem ; 64(7): 3843-3869, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33749283

ABSTRACT

Transient receptor potential ankyrin 1 (TRPA1) is a nonselective calcium-permeable ion channel highly expressed in the primary sensory neurons functioning as a polymodal sensor for exogenous and endogenous stimuli and has generated widespread interest as a target for inhibition due to its implication in neuropathic pain and respiratory disease. Herein, we describe the optimization of a series of potent, selective, and orally bioavailable TRPA1 small molecule antagonists, leading to the discovery of a novel tetrahydrofuran-based linker. Given the balance of physicochemical properties and strong in vivo target engagement in a rat AITC-induced pain assay, compound 20 was progressed into a guinea pig ovalbumin asthma model where it exhibited significant dose-dependent reduction of inflammatory response. Furthermore, the structure of the TRPA1 channel bound to compound 21 was determined via cryogenic electron microscopy to a resolution of 3 Å, revealing the binding site and mechanism of action for this class of antagonists.


Subject(s)
Asthma/drug therapy , Furans/therapeutic use , Purines/therapeutic use , TRPA1 Cation Channel/antagonists & inhibitors , Animals , Asthma/chemically induced , Asthma/complications , CHO Cells , Cricetulus , Furans/chemical synthesis , Furans/metabolism , Guinea Pigs , Humans , Inflammation/drug therapy , Inflammation/etiology , Ligands , Male , Molecular Structure , Ovalbumin , Oxadiazoles/chemical synthesis , Oxadiazoles/metabolism , Oxadiazoles/therapeutic use , Protein Binding , Purines/chemical synthesis , Purines/metabolism , Rats, Sprague-Dawley , Structure-Activity Relationship , TRPA1 Cation Channel/metabolism
5.
J Med Chem ; 64(7): 3940-3955, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33780623

ABSTRACT

Optimization of a series of aryl urea RAF inhibitors led to the identification of type II pan-RAF inhibitor GNE-0749 (7), which features a fluoroquinazolinone hinge-binding motif. By minimizing reliance on common polar hinge contacts, this hinge binder allows for a greater contribution of RAF-specific residue interactions, resulting in exquisite kinase selectivity. Strategic substitution of fluorine at the C5 position efficiently masked the adjacent polar NH functionality and increased solubility by impeding a solid-state conformation associated with stronger crystal packing of the molecule. The resulting improvements in permeability and solubility enabled oral dosing of 7. In vivo evaluation of 7 in combination with the MEK inhibitor cobimetinib demonstrated synergistic pathway inhibition and significant tumor growth inhibition in a KRAS mutant xenograft mouse model.


Subject(s)
Neoplasms/drug therapy , Phenylurea Compounds/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Quinazolinones/therapeutic use , raf Kinases/antagonists & inhibitors , Animals , Azetidines/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Dogs , Drug Combinations , Drug Synergism , Female , Humans , Madin Darby Canine Kidney Cells , Mice, Nude , Molecular Structure , Mutation , Phenylurea Compounds/chemistry , Phenylurea Compounds/metabolism , Piperidines/therapeutic use , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Quinazolinones/chemistry , Quinazolinones/metabolism , Structure-Activity Relationship , Xenograft Model Antitumor Assays , raf Kinases/genetics , raf Kinases/metabolism
6.
ACS Med Chem Lett ; 11(4): 541-549, 2020 Apr 09.
Article in English | MEDLINE | ID: mdl-32292562

ABSTRACT

A class of imidazoisoindole (III) heme-binding indoleamine-2,3-dioxygenase (IDO1) inhibitors were optimized via structure-based drug design into a series of tryptophan-2,3-dioxygenase (TDO)-selective inhibitors. Kynurenine pathway modulation was demonstrated in vivo, which enabled evaluation of TDO as a potential cancer immunotherapy target. As means of mitigating the risk of drug-drug interactions arising from cytochrome P450 inhibition, a novel property-based drug design parameter, herein referred to as the CYP Index, was implemented for the design of inhibitors with appreciable selectivity for TDO over CYP3A4. We anticipate the CYP Index will be a valuable design parameter for optimizing CYP inhibition of any small molecule inhibitor containing a Lewis basic motif capable of binding heme.

7.
J Pharm Sci ; 108(6): 1934-1943, 2019 06.
Article in English | MEDLINE | ID: mdl-30639736

ABSTRACT

Solubilization of new chemical entities for toxicity assessment must use excipients that do not negatively impact drug pharmacokinetics and toxicology. In this study, we investigated the tolerability of a model freebase compound, GDC-0152, solubilized by pH adjustment with succinic acid and complexation with hydroxypropyl-ß-cyclodextrin (HP-ß-CD) to enable intravenous use. Solubility, critical micelle concentration, and association constant with HP-ß-CD were determined. Blood compatibility and potential for hemolysis were assessed in vitro. Local tolerability was assessed after intravenous and subcutaneous injections in rats. A pharmacokinetic study was conducted in rats after intravenous bolus administration. GDC-0152 exhibited pH-dependent solubility that was influenced by self-association. The presence of succinic acid increased solubility in a concentration-dependent manner. HP-ß-CD alone also increased solubility, but the extent of solubility enhancement was significantly lower than succinic acid alone. Inclusion of HP-ß-CD in the solution of GDC-0152 improved blood compatibility, reduced hemolytic potential by ∼20-fold in vitro, and increased the maximum tolerated dose to 80 mg/kg.


Subject(s)
2-Hydroxypropyl-beta-cyclodextrin/pharmacokinetics , Cyclohexanes/toxicity , Drug Evaluation, Preclinical/methods , Excipients/pharmacokinetics , Pyrroles/toxicity , Toxicity Tests, Acute/methods , 2-Hydroxypropyl-beta-cyclodextrin/administration & dosage , Animals , Cyclohexanes/administration & dosage , Cyclohexanes/pharmacokinetics , Dose-Response Relationship, Drug , Drug Interactions , Excipients/administration & dosage , Hemolysis/drug effects , Injections, Intravenous , Injections, Subcutaneous , Male , Maximum Tolerated Dose , Models, Animal , Pyrroles/administration & dosage , Pyrroles/pharmacokinetics , Rats , Solubility
8.
Cancer Cell ; 34(4): 611-625.e7, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30300582

ABSTRACT

Targeting KRAS mutant tumors through inhibition of individual downstream pathways has had limited clinical success. Here we report that RAF inhibitors exhibit little efficacy in KRAS mutant tumors. In combination drug screens, MEK and PI3K inhibitors synergized with pan-RAF inhibitors through an RAS-GTP-dependent mechanism. Broad cell line profiling with RAF/MEK inhibitor combinations revealed synergistic efficacy in KRAS mutant and wild-type tumors, with KRASG13D mutants exhibiting greater synergy versus KRASG12 mutant tumors. Mechanistic studies demonstrate that MEK inhibition induced RAS-GTP levels, RAF dimerization and RAF kinase activity resulting in MEK phosphorylation in synergistic tumor lines regardless of KRAS status. Taken together, our studies uncover a strategy to rewire KRAS mutant tumors to confer sensitivity to RAF kinase inhibition.


Subject(s)
Phosphatidylinositol 3-Kinases/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/drug effects , Cell Line, Tumor , Guanosine Triphosphate/metabolism , Humans , Mutation/drug effects , Mutation/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins B-raf/drug effects , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , ras Proteins/drug effects , ras Proteins/genetics
9.
Bioorg Med Chem Lett ; 26(18): 4455-4461, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27524313

ABSTRACT

Modification of the δ-sultam ring of RORc inverse agonist 2 led to the discovery of more polar oxa-sultam 65. The less lipophilic inverse agonist (65) displayed high potency in a biochemical assay, which translated into inhibition of IL-17 production in human peripheral blood mononuclear cells. The successful reduction of lipophilicity of this new analog gave rise to additional improvements in ROR selectivity and aqueous kinetic solubility, as well as reduction in plasma protein binding, while maintaining high cellular permeability.


Subject(s)
Lipids/chemistry , Nuclear Receptor Subfamily 1, Group F, Member 1/agonists , Drug Discovery , Drug Inverse Agonism , Naphthalenesulfonates/chemistry
10.
J Med Chem ; 59(19): 9080-9093, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27564586

ABSTRACT

Inhibitors targeting the activating mutants of the epidermal growth factor receptor (EGFR) have found success in the treatment of EGFR mutant positive non-small-cell lung cancer. A secondary point mutation (T790M) in the inhibitor binding site has been linked to the acquired resistance against those first generation therapeutics. Herein, we describe the lead optimization of a series of reversible, pan-mutant (L858R, del746-750, T790M/L858R, and T790M/del746-750) EGFR inhibitors. By use of a noncovalent double mutant (T790M/L858R and T790M/del746-750) selective EGFR inhibitor (2) as a starting point, activities against the single mutants (L858R and del746-750) were introduced through a series of structure-guided modifications. The in vitro ADME-PK properties of the lead molecules were further optimized through a number of rational structural changes. The resulting inhibitor (21) exhibited excellent cellular activity against both the single and double mutants of EGFR, demonstrating target engagement in vivo and ADME-PK properties that are suitable for further evaluation. The reversible, noncovalent inhibitors described complement the covalent pan-mutant EGFR inhibitors that have shown encouraging results in recent clinical trials.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Resistance, Neoplasm , ErbB Receptors/genetics , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Models, Molecular , Mutation , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology
11.
Bioorg Med Chem Lett ; 26(15): 3518-24, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27346791

ABSTRACT

A series of 4-azaindole-containing p21-activated kinase-1 (PAK1) inhibitors was prepared with the goal of improving physicochemical properties relative to an indole starting point. Indole 1 represented an attractive, non-basic scaffold with good PAK1 affinity and cellular potency but was compromised by high lipophilicity (clogD=4.4). Azaindole 5 was designed as an indole surrogate with the goal of lowering logD and resulted in equipotent PAK1 inhibition with a 2-fold improvement in cellular potency over 1. Structure-activity relationship studies around 5 identified additional 4-azaindole analogs with superior PAK1 biochemical activity (Ki <10nM) and up to 24-fold selectivity for group I over group II PAKs. Compounds from this series showed enhanced permeability, improved aqueous solubility, and lower plasma protein binding over indole 1. The improvement in physicochemical properties translated to a 20-fold decrease in unbound clearance in mouse PK studies for azaindole 5 relative to indole 1.


Subject(s)
Indoles/pharmacology , Protein Kinase Inhibitors/pharmacology , p21-Activated Kinases/antagonists & inhibitors , Animals , Dogs , Dose-Response Relationship, Drug , Humans , Indoles/chemical synthesis , Indoles/chemistry , Madin Darby Canine Kidney Cells , Mice , Mice, Nude , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , p21-Activated Kinases/metabolism
12.
J Med Chem ; 59(11): 5520-41, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27167326

ABSTRACT

p21-activated kinase 1 (PAK1) has an important role in transducing signals in several oncogenic pathways. The concept of inhibiting this kinase has garnered significant interest over the past decade, particularly for targeting cancers associated with PAK1 amplification. Animal studies with the selective group I PAK (pan-PAK1, 2, 3) inhibitor G-5555 from the pyrido[2,3-d]pyrimidin-7-one class uncovered acute toxicity with a narrow therapeutic window. To attempt mitigating the toxicity, we introduced significant structural changes, culminating in the discovery of the potent pyridone side chain analogue G-9791. Mouse tolerability studies with this compound, other members of this series, and compounds from two structurally distinct classes revealed persistent toxicity and a correlation of minimum toxic concentrations and PAK1/2 mediated cellular potencies. Broad screening of selected PAK inhibitors revealed PAK1, 2, and 3 as the only overlapping targets. Our data suggest acute cardiovascular toxicity resulting from the inhibition of PAK2, which may be enhanced by PAK1 inhibition, and cautions against continued pursuit of pan-group I PAK inhibitors in drug discovery.


Subject(s)
Cardiovascular Diseases/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , p21-Activated Kinases/antagonists & inhibitors , Acute Disease , Animals , Dose-Response Relationship, Drug , Female , Mice , Mice, Inbred BALB C , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyridines/chemical synthesis , Pyridines/chemistry , Pyridones , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship , p21-Activated Kinases/metabolism
13.
Drug Metab Lett ; 10(2): 91-100, 2016.
Article in English | MEDLINE | ID: mdl-27063863

ABSTRACT

BACKGROUND: Significant under-prediction of in vivo clearance in rat was observed for a potent p21-activated kinase (PAK1) inhibitor, GNE1. OBJECTIVE: Rate-determining (rapid uptake) and rate-limiting (slow excretion) steps in systemic clearance and elimination of GNE1, respectively, were evaluated to better understand the cause of the in vitro-in vivo (IVIV) disconnect. METHODS: A series of in vivo, ex vivo, and in vitro experiments were carried out: 1) the role of organic cation transporters (Oct or Slc22a) was investigated in transporter knock-out and wild-type animals with or without 1-aminobenzotriazole (ABT) pretreatment; 2) the concentration-dependent hepatic extraction ratio was determined in isolated perfused rat liver; and 3) excreta were collected from both bile duct cannulated and non-cannulated rats after intravenous injection. RESULTS: After intravenous dosing, the rate-determining step in clearance was found to be mediated by the active uptake transporter, Oct1. In cannulated rats, biliary and renal clearance of GNE1 accounted for only approximately 14 and 16% of the total clearance, respectively. N-acetylation, an important metabolic pathway, accounted for only about 10% of the total dose. In non-cannulated rats, the majority of the dose was recovered in feces as unchanged parent (up to 91%) overnight following intravenous administration. CONCLUSION: Because the clearance of GNE1 is mediated through uptake transporters rather than metabolism, the extrahepatic expression of Oct1 in kidney and intestine in rat likely plays an important role in the IVIV disconnect in hepatic clearance prediction. The slow process of intestinal secretion is the rate-limiting step for in vivo clearance of GNE1.


Subject(s)
Catecholamine Plasma Membrane Transport Proteins/metabolism , Liver/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , p21-Activated Kinases/antagonists & inhibitors , Animals , Catecholamine Plasma Membrane Transport Proteins/genetics , Female , Male , Mice , Mice, Knockout , Organic Cation Transport Proteins/metabolism , Rats , Rats, Sprague-Dawley , Triazoles/pharmacology
15.
ACS Med Chem Lett ; 7(1): 100-4, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26819674

ABSTRACT

The rapid advancement of a series of noncovalent inhibitors of T790M mutants of EGFR is discussed. The optimization of pyridone 1, a nonselective high-throughput screening hit, to potent molecules with high levels of selectivity over wtEGFR and the broader kinome is described herein.

16.
Bioorg Med Chem Lett ; 26(2): 534-539, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26639762

ABSTRACT

The treatment of epidermal growth factor receptor (EGFR)-driven non-small cell lung cancers with the T790M resistance mutation remains a significant unmet medical need. We report the identification of 4-aminoindazolyl-dihydrofuro[3,4-d]pyrimidines as non-covalent inhibitors of EGFR, with excellent activity against the T790M resistance double mutants and initial single activating mutants. Using an optimization strategy focused on structure-based design and improving PK properties through metabolite identification, we obtained advanced leads with high oral exposure.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Furans/pharmacology , Indazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Acrylamides/pharmacology , Aniline Compounds/pharmacology , Animals , Binding Sites , Crystallography, X-Ray , Dogs , ErbB Receptors/chemistry , Erlotinib Hydrochloride/pharmacology , Furans/chemical synthesis , Furans/chemistry , Furans/pharmacokinetics , Hepatocytes/metabolism , High-Throughput Screening Assays , Humans , Hydrogen Bonding , Indazoles/chemical synthesis , Indazoles/chemistry , Indazoles/pharmacokinetics , Mice , Microsomes, Liver/metabolism , Point Mutation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Rats
17.
ACS Med Chem Lett ; 6(12): 1241-6, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26713112

ABSTRACT

Signaling pathways intersecting with the p21-activated kinases (PAKs) play important roles in tumorigenesis and cancer progression. By recognizing that the limitations of FRAX1036 (1) were chiefly associated with the highly basic amine it contained, we devised a mitigation strategy to address several issues such as hERG activity. The 5-amino-1,3-dioxanyl moiety was identified as an effective means of reducing pK a and logP simultaneously. When positioned properly within the scaffold, this group conferred several benefits including potency, pharmacokinetics, and selectivity. Mouse xenograft PK/PD studies were carried out using an advanced compound, G-5555 (12), derived from this approach. These studies concluded that dose-dependent pathway modulation was achievable and paves the way for further in vivo investigations of PAK1 function in cancer and other diseases.

18.
J Med Chem ; 58(22): 8877-95, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26455919

ABSTRACT

Because of their increased activity against activating mutants, first-generation epidermal growth factor receptor (EGFR) kinase inhibitors have had remarkable success in treating non-small-cell lung cancer (NSCLC) patients, but acquired resistance, through a secondary mutation of the gatekeeper residue, means that clinical responses only last for 8-14 months. Addressing this unmet medical need requires agents that can target both of the most common double mutants: T790M/L858R (TMLR) and T790M/del(746-750) (TMdel). Herein we describe how a noncovalent double mutant selective lead compound was optimized using a strategy focused on the structure-guided increase in potency without added lipophilicity or reduction of three-dimensional character. Following successive rounds of design and synthesis it was discovered that cis-fluoro substitution on 4-hydroxy- and 4-methoxypiperidinyl groups provided synergistic, substantial, and specific potency gain through direct interaction with the enzyme and/or effects on the proximal ligand oxygen atom. Further development of the fluorohydroxypiperidine series resulted in the identification of a pair of diastereomers that showed 50-fold enzyme and cell based selectivity for T790M mutants over wild-type EGFR (wtEGFR) in vitro and pathway knock-down in an in vivo xenograft model.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Genes, erbB-1/drug effects , Animals , Antineoplastic Agents/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Dogs , Drug Design , Gene Knockdown Techniques , Humans , In Vitro Techniques , Lipids/chemistry , Lung Neoplasms/drug therapy , Macaca fascicularis , Microsomes, Liver/metabolism , Models, Molecular , Mutation , Rats , Stereoisomerism , Structure-Activity Relationship , Substrate Specificity , Xenograft Model Antitumor Assays
19.
Bioorg Med Chem Lett ; 25(19): 4109-13, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26321361

ABSTRACT

A high-throughput screen of the Genentech/Roche compound collection using a retinoic acid receptor-related orphan receptor C (RORc, RORγ, or NR1F3) biochemical assay revealed a N-sulfonyl-tetrahydroquinoline hit. Herein, we describe the hit-to-lead optimization and structure-activity relationships of these tetrahydroquinoline RORc inverse agonists. Through iterative synthesis and analog design, we identified compounds with improved biochemical RORc inverse agonist activity and RORc cellular potencies. These improved N-sulfonyl-tetrahydroquinoline compounds also exhibited selectivity for RORc over other nuclear receptors.


Subject(s)
Drug Inverse Agonism , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Quinolines/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Quinolines/chemical synthesis , Quinolines/chemistry , Structure-Activity Relationship
20.
J Med Chem ; 58(12): 5121-36, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26030457

ABSTRACT

The p21-activated kinases (PAKs) play important roles in cytoskeletal organization, cellular morphogenesis, and survival and have generated significant attention as potential therapeutic targets for cancer. Following a high-throughput screen, we identified an aminopyrazole scaffold-based series that was optimized to yield group I selective PAK inhibitors. A structure-based design effort aimed at targeting the ribose pocket for both potency and selectivity led to much-improved group I vs II selectivity. Early lead compounds contained a basic primary amine, which was found to be a major metabolic soft spot with in vivo clearance proceeding predominantly via N-acetylation. We succeeded in identifying replacements with improved metabolic stability, leading to compounds with lower in vivo rodent clearance and excellent group I PAK selectivity.


Subject(s)
Drug Design , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazoles/chemistry , Pyrazoles/pharmacology , p21-Activated Kinases/antagonists & inhibitors , Animals , Humans , Mice , Molecular Docking Simulation , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/pharmacokinetics , Rats , p21-Activated Kinases/chemistry , p21-Activated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...