Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 15(683): eabj3289, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36791209

ABSTRACT

Innate immunity not only shapes the way epithelial barriers interpret environmental cues but also drives adaptive responses. Therefore, modulators of innate immune responses are expected to have high therapeutic potential across immune-mediated inflammatory diseases. IRAK4 is a kinase that integrates signaling downstream of receptors acting at the interface between innate and adaptive immune responses, such as Toll-like receptors (TLRs), interleukin-1R (IL-1R), and IL-18R. Because effects of IRAK4 inhibition are stimulus, cell type, and species dependent, the evaluation of the therapeutic potential of IRAK4 inhibitors requires a highly translational approach. Here, we profiled a selective IRAK4 inhibitor, GLPG2534, in an extensive panel of models of inflammatory skin diseases, translationally expanding evidence from in vitro to in vivo and from mouse to human. In vitro, IRAK4 inhibition resulted in substantial inhibition of TLR and IL-1 responses in dendritic cells, keratinocytes, granulocytes, and T cells but only weakly affected dermal fibroblast responses. Furthermore, disease activity in murine models of skin inflammation (IL-23-, IL-33-, imiquimod-, and MC903-induced) was markedly dampened by IRAK4 inhibition. Last, inhibiting IRAK4 reversed pathogenic molecular signatures in human lesional psoriasis and atopic dermatitis biopsies. Over the variety of models used, IRAK4 inhibition consistently affected central mediators of psoriasis (IL-17A) and atopic dermatitis (IL-4 and IL-13). Overall, our data highlight IRAK4 as a central player in skin inflammatory processes and demonstrate the potential of IRAK4 inhibition as a therapeutic strategy in chronic inflammatory skin diseases.


Subject(s)
Dermatitis, Atopic , Psoriasis , Humans , Mice , Animals , Interleukin-1 Receptor-Associated Kinases/metabolism , Dermatitis, Atopic/pathology , Signal Transduction , Toll-Like Receptors/therapeutic use , Skin/pathology , Psoriasis/drug therapy
2.
J Med Chem ; 64(19): 14557-14586, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34581584

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive lung disease. Current treatments only slow down disease progression, making new therapeutic strategies compelling. Increasing evidence suggests that S1P2 antagonists could be effective agents against fibrotic diseases. Our compound collection was mined for molecules possessing substructure features associated with S1P2 activity. The weakly potent indole hit 6 evolved into a potent phthalazone series, bearing a carboxylic acid, with the aid of a homology model. Suboptimal pharmacokinetics of a benzimidazole subseries were improved by modifications targeting potential interactions with transporters, based on concepts deriving from the extended clearance classification system (ECCS). Scaffold hopping, as a part of a chemical enablement strategy, permitted the rapid exploration of the position adjacent to the carboxylic acid. Compound 38, with good pharmacokinetics and in vitro potency, was efficacious at 10 mg/kg BID in three different in vivo mouse models of fibrotic diseases in a therapeutic setting.


Subject(s)
Carboxylic Acids/pharmacology , Drug Discovery , Idiopathic Pulmonary Fibrosis/drug therapy , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Carboxylic Acids/administration & dosage , Disease Models, Animal , Humans , Mice
3.
J Med Chem ; 64(9): 6037-6058, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33939425

ABSTRACT

Mounting evidence from the literature suggests that blocking S1P2 receptor (S1PR2) signaling could be effective for the treatment of idiopathic pulmonary fibrosis (IPF). However, only a few antagonists have been so far disclosed. A chemical enablement strategy led to the discovery of a pyridine series with good antagonist activity. A pyridazine series with improved lipophilic efficiency and with no CYP inhibition liability was identified by scaffold hopping. Further optimization led to the discovery of 40 (GLPG2938), a compound with exquisite potency on a phenotypic IL8 release assay, good pharmacokinetics, and good activity in a bleomycin-induced model of pulmonary fibrosis.


Subject(s)
Drug Design , Idiopathic Pulmonary Fibrosis/drug therapy , Pyridazines/chemistry , Pyridazines/pharmacology , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Animals , CHO Cells , Cricetulus , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Interleukin-8/metabolism , Male , Mice , Pyridazines/pharmacokinetics , Pyridazines/therapeutic use , Structure-Activity Relationship , Tissue Distribution
4.
J Med Chem ; 64(6): 2937-2952, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33719441

ABSTRACT

There are currently no approved disease-modifying osteoarthritis (OA) drugs (DMOADs). The aggrecanase ADAMTS-5 is key in the degradation of human aggrecan (AGC), a component of cartilage. Therefore, ADAMTS-5 is a promising target for the identification of DMOADs. We describe the discovery of GLPG1972/S201086, a potent and selective ADAMTS-5 inhibitor obtained by optimization of a promising hydantoin series following an HTS. Biochemical activity against rat and human ADAMTS-5 was assessed via a fluorescence-based assay. ADAMTS-5 inhibitory activity was confirmed with human aggrecan using an AGC ELISA. The most promising compounds were selected based on reduction of glycosaminoglycan release after interleukin-1 stimulation in mouse cartilage explants and led to the discovery of GLPG1972/S201086. The anticatabolic activity was confirmed in mouse cartilage explants (IC50 < 1.5 µM). The cocrystal structure of GLPG1972/S201086 with human recombinant ADAMTS-5 is discussed. GLPG1972/S201086 has been investigated in a phase 2 clinical study in patients with knee OA (NCT03595618).


Subject(s)
ADAMTS5 Protein/antagonists & inhibitors , Osteoarthritis/drug therapy , ADAMTS5 Protein/metabolism , Animals , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Dogs , Glycosaminoglycans/metabolism , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Osteoarthritis/metabolism , Rats
5.
J Med Chem ; 63(22): 13526-13545, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32902984

ABSTRACT

GPR84 is a medium chain free fatty acid-binding G-protein-coupled receptor associated with inflammatory and fibrotic diseases. As the only reported antagonist of GPR84 (PBI-4050) that displays relatively low potency and selectivity, a clear need exists for an improved modulator. Structural optimization of GPR84 antagonist hit 1, identified through high-throughput screening, led to the identification of potent and selective GPR84 inhibitor GLPG1205 (36). Compared with the initial hit, 36 showed improved potency in a guanosine 5'-O-[γ-thio]triphosphate assay, exhibited metabolic stability, and lacked activity against phosphodiesterase-4. This novel pharmacological tool allowed investigation of the therapeutic potential of GPR84 inhibition. At once-daily doses of 3 and 10 mg/kg, GLPG1205 reduced disease activity index score and neutrophil infiltration in a mouse dextran sodium sulfate-induced chronic inflammatory bowel disease model, with efficacy similar to positive-control compound sulfasalazine. The drug discovery steps leading to GLPG1205 identification, currently under phase II clinical investigation, are described herein.


Subject(s)
Drug Discovery/methods , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Acetates/chemistry , Acetates/pharmacology , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Caco-2 Cells , Cells, Cultured , Dogs , Drug Evaluation, Preclinical/methods , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley
6.
Front Pharmacol ; 10: 514, 2019.
Article in English | MEDLINE | ID: mdl-31143125

ABSTRACT

The deletion of phenylalanine at position 508 (F508del) in cystic fibrosis transmembrane conductance regulator (CFTR) causes a severe defect in folding and trafficking of the chloride channel resulting in its absence at the plasma membrane of epithelial cells leading to cystic fibrosis. Progress in the understanding of the disease increased over the past decades and led to the awareness that combinations of mechanistically different CFTR modulators are required to obtain meaningful clinical benefit. Today, there remains an unmet need for identification and development of more effective CFTR modulator combinations to improve existing therapies for patients carrying the F508del mutation. Here, we describe the identification of a novel F508del corrector using functional assays. We provide experimental evidence that the clinical candidate GLPG/ABBV-2737 represents a novel class of corrector exerting activity both on its own and in combination with VX809 or GLPG/ABBV-2222.

8.
J Med Chem ; 61(4): 1425-1435, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29148763

ABSTRACT

Cystic fibrosis (CF) is caused by mutations in the gene for the cystic fibrosis transmembrane conductance regulator (CFTR). With the discovery of Ivacaftor and Orkambi, it has been shown that CFTR function can be partially restored by administering one or more small molecules. These molecules aim at either enhancing the amount of CFTR on the cell surface (correctors) or at improving the gating function of the CFTR channel (potentiators). Here we describe the discovery of a novel potentiator GLPG1837, which shows enhanced efficacy on CFTR mutants harboring class III mutations compared to Ivacaftor, the first marketed potentiator. The optimization of potency, efficacy, and pharmacokinetic profile will be described.


Subject(s)
Chloride Channel Agonists/chemistry , Cystic Fibrosis/drug therapy , Drug Discovery , Mutant Proteins/drug effects , Aminophenols/pharmacokinetics , Animals , Chloride Channel Agonists/pharmacokinetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Humans , Mutation , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Quinolones/pharmacokinetics , Rats , Structure-Activity Relationship
9.
J Med Chem ; 60(17): 7371-7392, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28731719

ABSTRACT

Autotaxin (ATX) is a secreted enzyme playing a major role in the production of lysophosphatidic acid (LPA) in blood through hydrolysis of lysophosphatidyl choline (LPC). The ATX-LPA signaling axis arouses a high interest in the drug discovery industry as it has been implicated in several diseases including cancer, fibrotic diseases, and inflammation, among others. An imidazo[1,2-a]pyridine series of ATX inhibitors was identified out of a high-throughput screening (HTS). A cocrystal structure with one of these compounds and ATX revealed a novel binding mode with occupancy of the hydrophobic pocket and channel of ATX but no interaction with zinc ions of the catalytic site. Exploration of the structure-activity relationship led to compounds displaying high activity in biochemical and plasma assays, exemplified by compound 40. Compound 40 was also able to decrease the plasma LPA levels upon oral administration to rats.


Subject(s)
Imidazoles/chemistry , Imidazoles/pharmacology , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Pyridines/chemistry , Pyridines/pharmacology , Animals , Humans , Imidazoles/pharmacokinetics , Lysophosphatidylcholines/metabolism , Lysophospholipids/metabolism , Male , Mice , Molecular Docking Simulation , Phosphodiesterase Inhibitors/pharmacokinetics , Phosphoric Diester Hydrolases/chemistry , Pyridines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
10.
J Med Chem ; 60(9): 3580-3590, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28414242

ABSTRACT

Autotaxin is a circulating enzyme with a major role in the production of lysophosphatic acid (LPA) species in blood. A role for the autotaxin/LPA axis has been suggested in many disease areas including pulmonary fibrosis. Structural modifications of the known autotaxin inhibitor lead compound 1, to attenuate hERG inhibition, remove CYP3A4 time-dependent inhibition, and improve pharmacokinetic properties, led to the identification of clinical candidate GLPG1690 (11). Compound 11 was able to cause a sustained reduction of LPA levels in plasma in vivo and was shown to be efficacious in a bleomycin-induced pulmonary fibrosis model in mice and in reducing extracellular matrix deposition in the lung while also reducing LPA 18:2 content in bronchoalveolar lavage fluid. Compound 11 is currently being evaluated in an exploratory phase 2a study in idiopathic pulmonary fibrosis patients.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , Imidazoles/therapeutic use , Phosphoric Diester Hydrolases/drug effects , Pyrimidines/therapeutic use , Animals , Humans , Imidazoles/pharmacology , Mice , Mice, Knockout , Phosphoric Diester Hydrolases/genetics , Pyrimidines/pharmacology , Rats
11.
J Med Chem ; 57(23): 10044-57, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25380412

ABSTRACT

FFA2, also called GPR43, is a G-protein coupled receptor for short chain fatty acids which is involved in the mediation of inflammatory responses. A class of azetidines was developed as potent FFA2 antagonists. Multiparametric optimization of early hits with moderate potency and suboptimal ADME properties led to the identification of several compounds with nanomolar potency on the receptor combined with excellent pharmacokinetic (PK) parameters. The most advanced compound, 4-[[(R)-1-(benzo[b]thiophene-3-carbonyl)-2-methyl-azetidine-2-carbonyl]-(3-chloro-benzyl)-amino]-butyric acid 99 (GLPG0974), is able to inhibit acetate-induced neutrophil migration strongly in vitro and demonstrated ability to inhibit a neutrophil-based pharmacodynamic (PD) marker, CD11b activation-specific epitope [AE], in a human whole blood assay. All together, these data supported the progression of 99 toward next phases, becoming the first FFA2 antagonist to reach the clinic.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Azetidines/metabolism , Butyrates/chemical synthesis , Receptors, Cell Surface/antagonists & inhibitors , Thiophenes/chemical synthesis , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Azetidines/chemical synthesis , Azetidines/pharmacokinetics , Azetidines/pharmacology , Butyrates/pharmacokinetics , Butyrates/pharmacology , Humans , Immune System Diseases , Inhibitory Concentration 50 , Leukocyte Disorders , Mice , Microsomes, Liver/metabolism , Rats, Sprague-Dawley , Structure-Activity Relationship , Thiophenes/pharmacokinetics , Thiophenes/pharmacology
12.
J Med Chem ; 57(22): 9323-42, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25369270

ABSTRACT

Janus kinases (JAK1, JAK2, JAK3, and TYK2) are involved in the signaling of multiple cytokines important in cellular function. Blockade of the JAK-STAT pathway with a small molecule has been shown to provide therapeutic immunomodulation. Having identified JAK1 as a possible new target for arthritis at Galapagos, the compound library was screened against JAK1, resulting in the identification of a triazolopyridine-based series of inhibitors represented by 3. Optimization within this chemical series led to identification of GLPG0634 (65, filgotinib), a selective JAK1 inhibitor currently in phase 2B development for RA and phase 2A development for Crohn's disease (CD).


Subject(s)
Chemistry, Pharmaceutical/methods , Janus Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Triazoles/chemistry , Adenosine Triphosphate/chemistry , Animals , Arthritis/drug therapy , Collagen/chemistry , Crohn Disease/drug therapy , Crystallography, X-Ray , Cytokines/metabolism , Dimerization , Disease Models, Animal , Drug Design , Drug Evaluation, Preclinical , Humans , Inhibitory Concentration 50 , Kinetics , Phosphorylation , Rats , Recombinant Proteins/chemistry , Structure-Activity Relationship
13.
Cell ; 157(7): 1565-76, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24949969

ABSTRACT

Mycobacterium ulcerans, the etiological agent of Buruli ulcer, causes extensive skin lesions, which despite their severity are not accompanied by pain. It was previously thought that this remarkable analgesia is ensured by direct nerve cell destruction. We demonstrate here that M. ulcerans-induced hypoesthesia is instead achieved through a specific neurological pathway triggered by the secreted mycobacterial polyketide mycolactone. We decipher this pathway at the molecular level, showing that mycolactone elicits signaling through type 2 angiotensin II receptors (AT2Rs), leading to potassium-dependent hyperpolarization of neurons. We further validate the physiological relevance of this mechanism with in vivo studies of pain sensitivity in mice infected with M. ulcerans, following the disruption of the identified pathway. Our findings shed new light on molecular mechanisms evolved by natural systems for the induction of very effective analgesia, opening up the prospect of new families of analgesics derived from such systems.


Subject(s)
Angiotensins/metabolism , Buruli Ulcer/pathology , Macrolides/isolation & purification , Mycobacterium ulcerans , Analgesics/isolation & purification , Animals , Buruli Ulcer/metabolism , Buruli Ulcer/microbiology , Disease Models, Animal , Edema/microbiology , Humans , Hypesthesia/chemically induced , Macrolides/chemistry , Macrolides/metabolism , Mice , Neurons/metabolism , Potassium Channels/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Receptor, Angiotensin, Type 2/metabolism , Signal Transduction/drug effects
14.
J Immunol ; 191(7): 3568-77, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24006460

ABSTRACT

The JAKs receive continued interest as therapeutic targets for autoimmune, inflammatory, and oncological diseases. JAKs play critical roles in the development and biology of the hematopoietic system, as evidenced by mouse and human genetics. JAK1 is critical for the signal transduction of many type I and type II inflammatory cytokine receptors. In a search for JAK small molecule inhibitors, GLPG0634 was identified as a lead compound belonging to a novel class of JAK inhibitors. It displayed a JAK1/JAK2 inhibitor profile in biochemical assays, but subsequent studies in cellular and whole blood assays revealed a selectivity of ∼30-fold for JAK1- over JAK2-dependent signaling. GLPG0634 dose-dependently inhibited Th1 and Th2 differentiation and to a lesser extent the differentiation of Th17 cells in vitro. GLPG0634 was well exposed in rodents upon oral dosing, and exposure levels correlated with repression of Mx2 expression in leukocytes. Oral dosing of GLPG0634 in a therapeutic set-up in a collagen-induced arthritis model in rodents resulted in a significant dose-dependent reduction of the disease progression. Paw swelling, bone and cartilage degradation, and levels of inflammatory cytokines were reduced by GLPG0634 treatment. Efficacy of GLPG0634 in the collagen-induced arthritis models was comparable to the results obtained with etanercept. In conclusion, the JAK1 selective inhibitor GLPG0634 is a promising novel therapeutic with potential for oral treatment of rheumatoid arthritis and possibly other immune-inflammatory diseases.


Subject(s)
Inflammation/metabolism , Janus Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Triazoles/pharmacology , Animals , Cell Differentiation/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Gene Silencing , Humans , Inflammation/drug therapy , Inhibitory Concentration 50 , Interleukin-6/pharmacology , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Male , Mice , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , Rats , STAT1 Transcription Factor/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Triazoles/administration & dosage
15.
Nat Med ; 19(9): 1157-60, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23913123

ABSTRACT

New therapeutic strategies are needed to combat the tuberculosis pandemic and the spread of multidrug-resistant (MDR) and extensively drug-resistant (XDR) forms of the disease, which remain a serious public health challenge worldwide. The most urgent clinical need is to discover potent agents capable of reducing the duration of MDR and XDR tuberculosis therapy with a success rate comparable to that of current therapies for drug-susceptible tuberculosis. The last decade has seen the discovery of new agent classes for the management of tuberculosis, several of which are currently in clinical trials. However, given the high attrition rate of drug candidates during clinical development and the emergence of drug resistance, the discovery of additional clinical candidates is clearly needed. Here, we report on a promising class of imidazopyridine amide (IPA) compounds that block Mycobacterium tuberculosis growth by targeting the respiratory cytochrome bc1 complex. The optimized IPA compound Q203 inhibited the growth of MDR and XDR M. tuberculosis clinical isolates in culture broth medium in the low nanomolar range and was efficacious in a mouse model of tuberculosis at a dose less than 1 mg per kg body weight, which highlights the potency of this compound. In addition, Q203 displays pharmacokinetic and safety profiles compatible with once-daily dosing. Together, our data indicate that Q203 is a promising new clinical candidate for the treatment of tuberculosis.


Subject(s)
Adenosine Triphosphate/biosynthesis , Electron Transport Complex III/antagonists & inhibitors , Extensively Drug-Resistant Tuberculosis/drug therapy , Imidazoles/pharmacology , Mycobacterium tuberculosis/drug effects , Piperidines/pharmacology , Pyridines/pharmacology , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Electron Transport Complex III/genetics , Imidazoles/pharmacokinetics , Mice , Mice, Inbred BALB C , Piperidines/pharmacokinetics , Pyridines/pharmacokinetics , Rats , Rats, Sprague-Dawley
16.
PLoS One ; 8(7): e68767, 2013.
Article in English | MEDLINE | ID: mdl-23874756

ABSTRACT

Classical target-based, high-throughput screening has been useful for the identification of inhibitors for known molecular mechanisms involved in the HIV life cycle. In this study, the development of a cell-based assay that uses a phenotypic drug discovery approach based on automated high-content screening is described. Using this screening approach, the antiviral activity of 26,500 small molecules from a relevant chemical scaffold library was evaluated. Among the selected hits, one sulfonamide compound showed strong anti-HIV activity against wild-type and clinically relevant multidrug resistant HIV strains. The biochemical inhibition, point resistance mutations and the activity of structural analogs allowed us to understand the mode of action and propose a binding model for this compound with HIV-1 reverse transcriptase.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , HIV-1/drug effects , Sulfonamides/pharmacology , Virus Replication/drug effects , Antiviral Agents/metabolism , Cell Line , Cell Survival , Enzyme-Linked Immunosorbent Assay , HIV-1/enzymology , High-Throughput Screening Assays , Humans , Models, Biological , Protein Binding , RNA-Directed DNA Polymerase/metabolism , Small Molecule Libraries , Sulfonamides/metabolism
17.
J Med Chem ; 55(14): 6391-402, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22738293

ABSTRACT

In this paper, we describe the screening of a 14640-compound library using a novel whole mycobacteria phenotypic assay to discover inhibitors of EthR, a transcriptional repressor implicated in the innate resistance of Mycobacterium tuberculosis to the second-line antituberculosis drug ethionamide. From this screening a new chemical family of EthR inhibitors bearing an N-phenylphenoxyacetamide motif was identified. The X-ray structure of the most potent compound crystallized with EthR inspired the synthesis of a 960-member focused library. These compounds were tested in vitro using a rapid thermal shift assay on EthR to accelerate the optimization. The best compounds were synthesized on a larger scale and confirmed as potent ethionamide boosters on M. tuberculosis -infected macrophages. Finally, the cocrystallization of the best optimized analogue with EthR revealed an unexpected reorientation of the ligand in the binding pocket.


Subject(s)
Acetamides/chemical synthesis , Acetamides/pharmacology , Antitubercular Agents/pharmacology , Drug Discovery , Ethionamide/pharmacology , High-Throughput Screening Assays , Repressor Proteins/antagonists & inhibitors , Acetamides/chemistry , Animals , Cell Line , Chemistry Techniques, Synthetic , Drug Synergism , Ligands , Macrophages/drug effects , Macrophages/microbiology , Mice , Models, Molecular , Mycobacterium tuberculosis/drug effects , Protein Conformation , Repressor Proteins/chemistry
18.
J Med Chem ; 55(1): 68-83, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22098589

ABSTRACT

Mycobacterial transcriptional repressor EthR controls the expression of EthA, the bacterial monooxygenase activating ethionamide, and is thus largely responsible for the low sensitivity of the human pathogen Mycobacterium tuberculosis to this antibiotic. We recently reported structure-activity relationships of a series of 1,2,4-oxadiazole EthR inhibitors leading to the discovery of potent ethionamide boosters. Despite high metabolic stability, pharmacokinetic evaluation revealed poor mice exposure; therefore, a second phase of optimization was required. Herein a structure-property relationship study is reported according to the replacement of the two aromatic heterocycles: 2-thienyl and 1,2,4-oxadiazolyl moieties. This work was done using a combination of structure-based drug design and in vitro/ex vivo evaluations of ethionamide boosters on the targeted protein EthR and on the human pathogen Mycobacterium tuberculosis. Thanks to this process, we identified compound 42 (BDM41906), which displays improved efficacy in addition to high exposure to mice after oral administration.


Subject(s)
Antitubercular Agents/chemical synthesis , Ethionamide/pharmacokinetics , Oxadiazoles/chemical synthesis , Piperidines/chemical synthesis , Prodrugs/pharmacokinetics , Repressor Proteins/antagonists & inhibitors , Administration, Oral , Animals , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacokinetics , Cell Line , Crystallography, X-Ray , Drug Design , Drug Synergism , In Vitro Techniques , Macrophages/drug effects , Macrophages/microbiology , Mice , Microsomes, Liver/metabolism , Models, Molecular , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Oxadiazoles/chemistry , Oxadiazoles/pharmacokinetics , Piperidines/chemistry , Piperidines/pharmacokinetics , Repressor Proteins/chemistry , Stereoisomerism , Structure-Activity Relationship
19.
Curr Opin Chem Biol ; 15(4): 534-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21684803

ABSTRACT

The last decade has seen the development of automated microscopy and its adaptation for various areas of research, particularly infectious disease. Most of the high-content screening (HCS) platforms now integrate all of the following necessary steps: automated pipettes for assay miniaturization in 384-well plates, automated image acquisition and data storage and analysis. HCS was initially associated with RNA interference genetic screens for identifying host factors involved in host-pathogen interactions. More recently, both in academia and in industry, HCS has been adapted for drug discovery purposes. High-content analysis enables intracellular tracking of viral particles to profile the antiviral mechanisms of each compound. Adaptation to high-throughput screening in bacteriology and parasitology has already led to the discovery of new types of host-specific inhibitors that differ from those inhibitors that act directly on microbes.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antiparasitic Agents/pharmacology , Antiviral Agents/pharmacology , Communicable Diseases/drug therapy , Genetic Testing/methods , High-Throughput Screening Assays/methods , Biological Assay , Drug Discovery , Host-Pathogen Interactions , Humans , Miniaturization , RNA Interference , Structure-Activity Relationship
20.
J Med Chem ; 54(8): 2994-3010, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21417236

ABSTRACT

We report in this article an extensive structure-activity relationships (SAR) study with 58 thiophen-2-yl-1,2,4-oxadiazoles as inhibitors of EthR, a transcriptional regulator controling ethionamide bioactivation in Mycobacterium tuberculosis. We explored the replacement of two key fragments of the starting lead BDM31343. We investigated the potency of all analogues to boost subactive doses of ethionamide on a phenotypic assay involving M. tuberculosis infected macrophages and then ascertained the mode of action of the most active compounds using a functional target-based surface plasmon resonance assay. This process revealed that introduction of 4,4,4-trifluorobutyryl chain instead of cyanoacetyl group was crucial for intracellular activity. Replacement of 1,4-piperidyl by (R)-1,3-pyrrolidyl scaffold did not enhance activity but led to improved pharmacokinetic properties. Furthermore, the crystal structures of ligand-EthR complexes were consistent with the observed SAR. In conclusion, we identified EthR inhibitors that boost antibacterial activity of ethionamide with nanomolar potency while improving solubility and metabolic stability.


Subject(s)
Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Ethionamide/chemistry , Ethionamide/pharmacology , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Repressor Proteins/antagonists & inhibitors , Animals , Antitubercular Agents/chemical synthesis , Base Sequence , Cell Line , Chromatography, High Pressure Liquid , Crystallography, X-Ray , DNA Primers , Dose-Response Relationship, Drug , Ethionamide/chemical synthesis , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Models, Molecular , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Structure-Activity Relationship , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL
...