Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cell ; 178(4): 949-963.e18, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31353221

ABSTRACT

Estrogen receptor-positive (ER+) breast cancers frequently remain dependent on ER signaling even after acquiring resistance to endocrine agents, prompting the development of optimized ER antagonists. Fulvestrant is unique among approved ER therapeutics due to its capacity for full ER antagonism, thought to be achieved through ER degradation. The clinical potential of fulvestrant is limited by poor physicochemical features, spurring attempts to generate ER degraders with improved drug-like properties. We show that optimization of ER degradation does not guarantee full ER antagonism in breast cancer cells; ER "degraders" exhibit a spectrum of transcriptional activities and anti-proliferative potential. Mechanistically, we find that fulvestrant-like antagonists suppress ER transcriptional activity not by ER elimination, but by markedly slowing the intra-nuclear mobility of ER. Increased ER turnover occurs as a consequence of ER immobilization. These findings provide proof-of-concept that small molecule perturbation of transcription factor mobility may enable therapeutic targeting of this challenging target class.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor Antagonists/pharmacology , Fulvestrant/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cinnamates/pharmacology , Drug Resistance, Neoplasm , Estrogen Receptor Antagonists/therapeutic use , Female , Fulvestrant/therapeutic use , HEK293 Cells , Heterografts , Humans , Indazoles/pharmacology , Ligands , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Polymorphism, Single Nucleotide , Proteolysis/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects
2.
Cell ; 179(2): 417-431.e19, 2019 Oct 03.
Article in English | MEDLINE | ID: mdl-31585081

ABSTRACT

Severe asthma patients with low type 2 inflammation derive less clinical benefit from therapies targeting type 2 cytokines and represent an unmet need. We show that mast cell tryptase is elevated in severe asthma patients independent of type 2 biomarker status. Active ß-tryptase allele count correlates with blood tryptase levels, and asthma patients carrying more active alleles benefit less from anti-IgE treatment. We generated a noncompetitive inhibitory antibody against human ß-tryptase, which dissociates active tetramers into inactive monomers. A 2.15 Å crystal structure of a ß-tryptase/antibody complex coupled with biochemical studies reveal the molecular basis for allosteric destabilization of small and large interfaces required for tetramerization. This anti-tryptase antibody potently blocks tryptase enzymatic activity in a humanized mouse model, reducing IgE-mediated systemic anaphylaxis, and inhibits airway tryptase in Ascaris-sensitized cynomolgus monkeys with favorable pharmacokinetics. These data provide a foundation for developing anti-tryptase as a clinical therapy for severe asthma.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Asthma/therapy , Mast Cells/enzymology , Mast Cells/immunology , Tryptases/antagonists & inhibitors , Tryptases/immunology , Adolescent , Allosteric Regulation/immunology , Animals , Cell Line , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Rabbits
4.
Drug Metab Dispos ; 48(5): 408-419, 2020 05.
Article in English | MEDLINE | ID: mdl-32132091

ABSTRACT

The objectives of the present study were to characterize GNE-947 for its phosphoinositide 3-kinase (PI3K) and mammalian target of rapamycin (mTOR) inhibitory activities, in vitro anti-cell migration activity in human umbilical vein endothelial cells (HUVECs), in vivo antineovascularization activity in laser-induced rat choroidal neovascular (CNV) eyes, pharmacokinetics in rabbit plasma and eyes, and ocular distribution using matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI-IMS) and autoradioluminography. Its PI3K and mTOR K i were 0.0005 and 0.045 µM, respectively, and its HUVEC IC50 was 0.093 µM. GNE-947 prevented neovascularization in the rat CNV model at 50 or 100 µg per eye with repeat dosing. After a single intravenous injection at 2.5 and 500 µg/kg in rabbits, its plasma terminal half-lives (t 1/2) were 9.11 and 9.59 hours, respectively. After a single intravitreal injection of a solution at 2.5 µg per eye in rabbits, its apparent t 1/2 values were 14.4, 16.3, and 23.2 hours in the plasma, vitreous humor, and aqueous humor, respectively. After a single intravitreal injection of a suspension at 33.5, 100, 200 µg per eye in rabbits, the t 1/2 were 29, 74, and 219 days in the plasma and 46, 143, and 191 days in the eyes, respectively. MALDI-IMS and autoradioluminography images show that GNE-947 did not homogenously distribute in the vitreous humor and aggregated at the injection sites after injection of the suspension, which was responsible for the long t 1/2 of the suspension because of the slow dissolution process. This hypothesis was supported by pharmacokinetic modeling analyses. In conclusion, the PI3K/mTOR inhibitor GNE-947 prevented neovascularization in a rat CNV model, with t 1/2 up to approximately 6 months after a single intravitreal injection of the suspension in rabbit eyes. SIGNIFICANCE STATEMENT: GNE-947 is a potent phosphoinositide 3-kinase/mammalian target of rapamycin inhibitor and exhibits anti-choroidal neovascular activity in rat eyes. The duration of GNE-947 in the rabbit eyes after intravitreal injection in a solution is short, with a half-life (t 1/2) of less than a day. However, the duration after intravitreal dose of a suspension is long, with t 1/2 up to 6 months due to low solubility and slow dissolution. These results indicate that intravitreal injection of a suspension for low-solubility drugs can be used to achieve long-term drug exposure.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Choroidal Neovascularization/drug therapy , Phosphoinositide-3 Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/therapeutic use , Animals , Choroidal Neovascularization/etiology , Choroidal Neovascularization/pathology , Disease Models, Animal , Half-Life , Human Umbilical Vein Endothelial Cells , Humans , Injections, Intravenous , Intravitreal Injections , Male , Models, Biological , Ophthalmic Solutions/pharmacology , Ophthalmic Solutions/therapeutic use , Phosphoinositide-3 Kinase Inhibitors/chemistry , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Rabbits , Rats , Solubility , TOR Serine-Threonine Kinases/metabolism , Tissue Distribution
5.
Pharmaceuticals (Basel) ; 17(2)2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38399394

ABSTRACT

Early stage chemical development presents numerous challenges, and achieving a functional balance is a major hurdle, with many early compounds not meeting the clinical requirements for advancement benchmarks due to issues like poor oral bioavailability. There is a need to develop strategies for achieving the desired systemic concentration for these compounds. This will enable further evaluation of the biological response upon a compound-target interaction, providing deeper insight into the postulated biological pathways. Our study elucidates alternative drug delivery paradigms by comparing formulation strategies across oral (PO), intraperitoneal (IP), subcutaneous (SC), and intravenous (IV) routes. While each modality boasts its own set of merits and constraints, it is the drug's formulation that crucially influences its pharmacokinetic (PK) trajectory and the maintenance of its therapeutic levels. Our examination of model compounds G7883 and G6893 highlighted their distinct physio-chemical attributes. By harnessing varied formulation methods, we sought to fine-tune their PK profiles. PK studies showcased G7883's extended half-life using an SC oil formulation, resulting in a 4.5-fold and 2.5-fold enhancement compared with the IP and PO routes, respectively. In contrast, with G6893, we achieved a prolonged systemic coverage time above the desired target concentration through a different approach using an IV infusion pump. These outcomes underscore the need for tailored formulation strategies, which are dictated by the compound's innate properties, to reach the optimal in vivo systemic concentrations. Prioritizing formulation and delivery optimization early on is pivotal for effective systemic uptake, thereby facilitating a deeper understanding of biological pathways and expediting the overall clinical drug development timeline.

6.
Bioorg Med Chem Lett ; 23(21): 5923-30, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24042009

ABSTRACT

A highly ligand efficient, novel 8-oxo-pyridopyrimidine containing inhibitor of Jak1 and Jak2 isoforms with a pyridone moiety as the hinge-binding motif was discovered. Structure-based design strategies were applied to significantly improve enzyme potency and the polarity of the molecule was adjusted to gain cellular activity. The crystal structures of two representative inhibitors bound to Jak1 were obtained to enable SAR exploration.


Subject(s)
Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Humans , Janus Kinase 1/chemistry , Janus Kinase 1/metabolism , Janus Kinase 2/chemistry , Janus Kinase 2/metabolism , Molecular Docking Simulation , Structure-Activity Relationship
7.
Bioorg Med Chem Lett ; 23(12): 3592-8, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23642482

ABSTRACT

The identification of a novel fused triazolo-pyrrolopyridine scaffold, optimized derivatives of which display nanomolar inhibition of Janus kinase 1, is described. Prototypical example 3 demonstrated lower cell potency shift, better permeability in cells and higher oral exposure in rat than the corresponding, previously reported, imidazo-pyrrolopyridine analogue 2. Examples 6, 7 and 18 were subsequently identified from an optimization campaign and demonstrated modest selectivity over JAK2, moderate to good oral bioavailability in rat with overall pharmacokinetic profiles comparable to that reported for an approved pan-JAK inhibitor (tofacitinib).


Subject(s)
Janus Kinase 1/antagonists & inhibitors , Pyridines/pharmacology , Animals , Crystallography, X-Ray , Janus Kinase 1/chemistry , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/chemistry , Kinetics , Models, Molecular , Pyridines/chemistry , Pyrroles/chemistry , Pyrroles/pharmacology , Rats
8.
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
9.
Sci Rep ; 11(1): 6079, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33727605

ABSTRACT

Mutations in the GBA1 gene encoding glucocerebrosidase (GCase) are linked to Gaucher (GD) and Parkinson's Disease (PD). Since some GD and PD patients develop ocular phenotypes, we determined whether ocular phenotypes might result from impaired GCase activity and the corresponding accumulation of glucosylceramide (GluCer) and glucosylsphingosine (GluSph) in the Gba1D409V/D409V knock-in (Gba KI/KI; "KI") mouse. Gba KI mice developed age-dependent pupil dilation deficits to an anti-muscarinic agent; histologically, the iris covered the anterior part of the lens with adhesions between the iris and the anterior surface of the lens (posterior synechia). This may prevent pupil dilation in general, beyond an un-responsiveness of the iris to anti-muscarinics. Gba KI mice displayed atrophy and pigment dispersion of the iris, and occlusion of the iridocorneal angle by pigment-laden cells, reminiscent of secondary open angle glaucoma. Gba KI mice showed progressive thinning of the retina consistent with retinal degeneration. GluSph levels were increased in the anterior and posterior segments of the eye, suggesting that accumulation of lipids in the eye may contribute to degeneration in this compartment. We conclude that the Gba KI model provides robust and reproducible eye phenotypes which may be used to test for efficacy and establish biomarkers for GBA1-related therapies.


Subject(s)
Gaucher Disease , Glaucoma, Open-Angle , Glucosylceramidase , Mutation, Missense , Parkinson Disease , Amino Acid Substitution , Animals , Disease Models, Animal , Gaucher Disease/enzymology , Gaucher Disease/genetics , Gaucher Disease/pathology , Gene Knock-In Techniques , Glaucoma, Open-Angle/enzymology , Glaucoma, Open-Angle/genetics , Glaucoma, Open-Angle/pathology , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Humans , Mice , Mice, Transgenic , Parkinson Disease/enzymology , Parkinson Disease/genetics , Parkinson Disease/pathology
10.
Sci Transl Med ; 10(468)2018 11 21.
Article in English | MEDLINE | ID: mdl-30463918

ABSTRACT

Preclinical and clinical evidence indicates that a subset of asthma is driven by type 2 cytokines such as interleukin-4 (IL-4), IL-5, IL-9, and IL-13. Additional evidence predicts pathogenic roles for IL-6 and type I and type II interferons. Because each of these cytokines depends on Janus kinase 1 (JAK1) for signal transduction, and because many of the asthma-related effects of these cytokines manifest in the lung, we hypothesized that lung-restricted JAK1 inhibition may confer therapeutic benefit. To test this idea, we synthesized iJak-381, an inhalable small molecule specifically designed for local JAK1 inhibition in the lung. In pharmacodynamic models, iJak-381 suppressed signal transducer and activator of transcription 6 activation by IL-13. Furthermore, iJak-381 suppressed ovalbumin-induced lung inflammation in both murine and guinea pig asthma models and improved allergen-induced airway hyperresponsiveness in mice. In a model driven by human allergens, iJak-381 had a more potent suppressive effect on neutrophil-driven inflammation compared to systemic corticosteroid administration. The inhibitor iJak-381 reduced lung pathology, without affecting systemic Jak1 activity in rodents. Our data show that local inhibition of Jak1 in the lung can suppress lung inflammation without systemic Jak inhibition in rodents, suggesting that this strategy might be effective for treating asthma.


Subject(s)
Asthma/drug therapy , Asthma/enzymology , Janus Kinase 1/antagonists & inhibitors , Lung/enzymology , Protein Kinase Inhibitors/therapeutic use , Administration, Inhalation , Allergens , Animals , Asthma/pathology , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Disease Models, Animal , Eosinophils/drug effects , Eosinophils/metabolism , Eosinophils/pathology , Guinea Pigs , Inflammation/pathology , Janus Kinase 1/metabolism , Lung/drug effects , Lung/pathology , Ovalbumin , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Signal Transduction , Treatment Outcome
12.
J Pharm Sci ; 102(2): 750-60, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23225118

ABSTRACT

A quantitative bioanalytical method for pharmacokinetic studies using diluted whole blood from serially bled mice was developed. Oral exposure profiles in mice for five model anticancer compounds dacarbazine, gefitinib, gemcitabine, imatinib, and topotecan were determined following discrete and cassette (five-in-one) dosing. Six micro blood samples per animal were collected and added to a fixed amount of water. This dilution served several purposes: the red blood cells were lysed; an anticoagulant was unnecessary and the fluid volume of diluted sample was sufficient for bioanalytical assays. AUC values obtained from blood concentrations were within twofold for discrete and cassette dosing except for imatinib (2.1-fold difference) and in agreement with those obtained from plasma concentrations after discrete dosing. All compounds were stable in plasma and diluted blood samples for at least 2 weeks at approximately -80°C. Matrix and intermatrix effects were evaluated to ensure robustness and integrity of the bioanalytical assays. This method provides significant process improvement by enhancing efficiency for sample collection and processing and reducing resources (e.g., reduced compound, cost, and animal requirement) compared with conventional methods. Our study demonstrates the applicability of using diluted micro blood samples for small molecule quantitative bioanalysis to support mouse studies in drug discovery.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Biological Assay/methods , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Female , Mice , Random Allocation
13.
Cancer Discov ; 3(2): 168-81, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23229345

ABSTRACT

UNLABELLED: Approximately half of EGFR-mutant non-small cell lung cancer (NSCLC) patients treated with small-molecule EGFR kinase inhibitors develop drug resistance associated with the EGF receptor (EGFR) T790M "gatekeeper" substitution, prompting efforts to develop covalent EGFR inhibitors, which can effectively suppress EGFR T790M in preclinical models. However, these inhibitors have yet to prove clinically efficacious, and their toxicity in skin, reflecting activity against wild-type EGFR, may limit dosing required to effectively suppress EGFR T790M in vivo. While profiling sensitivity to various kinase inhibitors across a large cancer cell line panel, we identified indolocarbazole compounds, including a clinically well-tolerated FLT3 inhibitor, as potent and reversible inhibitors of EGFR T790M that spare wild-type EGFR. These findings show the use of broad cancer cell profiling of kinase inhibitor efficacy to identify unanticipated novel applications, and they identify indolocarbazole compounds as potentially effective EGFR inhibitors in the context of T790M-mediated drug resistance in NSCLC. SIGNIFICANCE: EGFR-mutant lung cancer patients who respond to currently used EGFR kinase inhibitors invariably develop drug resistance, which is associated with the EGFR T790M resistance mutation in about half these cases. We unexpectedly identified a class of reversible potent inhibitors of EGFR T790M that do not inhibit wild-type EGFR, revealing a promising therapeutic strategy to overcome T790M-associated drug-resistant lung cancers.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Acrylamides/pharmacology , Amino Acid Substitution , Animals , Area Under Curve , Base Sequence , Carbazoles/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Erlotinib Hydrochloride , Humans , Immunoblotting , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacology , Quinazolines/pharmacology , RNA Interference , Staurosporine/analogs & derivatives , Staurosporine/pharmacokinetics , Staurosporine/pharmacology , Xenograft Model Antitumor Assays
14.
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
15.
J Med Chem ; 55(12): 5901-21, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22591402

ABSTRACT

A therapeutic rationale is proposed for the treatment of inflammatory diseases, such as rheumatoid arthritis (RA), by specific targeting of the JAK1 pathway. Examination of the preferred binding conformation of clinically effective, pan-JAK inhibitor 1 led to identification of a novel, tricyclic hinge binding scaffold 3. Exploration of SAR through a series of cycloamino and cycloalkylamino analogues demonstrated this template to be highly tolerant of substitution, with a predisposition to moderate selectivity for the JAK1 isoform over JAK2. This study culminated in the identification of subnanomolar JAK1 inhibitors such as 22 and 49, having excellent cell potency, good rat pharmacokinetic characteristics, and excellent kinase selectivity. Determination of the binding modes of the series in JAK1 and JAK2 by X-ray crystallography supported the design of analogues to enhance affinity and selectivity.


Subject(s)
Imidazoles/chemistry , Janus Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Animals , Cell Line , Janus Kinase 1/chemistry , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/chemistry , Models, Molecular , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Protein Structure, Tertiary , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Rats , Substrate Specificity
16.
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
17.
Bioanalysis ; 3(17): 1923-33, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21899502

ABSTRACT

BACKGROUND: In early drug-discovery research, understanding the tissue distribution of drug at the site of action can help to predict the toxicity, efficacy and exposure level of the drug. The bottleneck of tissue analysis by LC-MS/MS is the time-consuming homogenization step. RESULTS: Both mechanical and enzymatic techniques for mouse tissue homogenization were evaluated, which included bead beater, polytron and enzymatic digestion. Brain, bone marrow, kidney, spleen and liver tissues can be homogenized effectively using the bead beater alone. Lung and heart tissues were best treated with collagenase first and then homogenized by the bead beater. CONCLUSION: Homogenization conditions for seven mouse tissues have been evaluated and optimized. These findings will expedite the preparation of tissue samples for analysis.


Subject(s)
Chromatography, Liquid/instrumentation , Chromatography, Liquid/methods , Drug Discovery , Tandem Mass Spectrometry/instrumentation , Tandem Mass Spectrometry/methods , Tissue Extracts/analysis , Animals , Female , Mice , Molecular Structure , Particle Size , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Pyridazines/chemistry , Pyridazines/pharmacokinetics , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL