Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
J Biol Chem ; 297(2): 100928, 2021 08.
Article in English | MEDLINE | ID: mdl-34274316

ABSTRACT

B-cell lymphoma 6 (BCL6) is a zinc finger transcriptional repressor possessing a BTB-POZ (BR-C, ttk, and bab for BTB; pox virus and zinc finger for POZ) domain, which is required for homodimerization and association with corepressors. BCL6 has multiple roles in normal immunity, autoimmunity, and some types of lymphoma. Mice bearing disrupted BCL6 loci demonstrate suppressed high-affinity antibody responses to T-dependent antigens. The corepressor binding groove in the BTB-POZ domain is a potential target for small compound-mediated therapy. Several inhibitors targeting this binding groove have been described, but these compounds have limited or absent in vivo activity. Biophysical studies of a novel compound, GSK137, showed an in vitro pIC50 of 8 and a cellular pIC50 of 7.3 for blocking binding of a peptide derived from the corepressor silencing mediator for retinoid or thyroid hormone receptors to the BCL6 BTB-POZ domain. The compound has good solubility (128 µg/ml) and permeability (86 nM/s). GSK137 caused little change in cell viability or proliferation in four BCL6-expressing B-cell lymphoma lines, although there was modest dose-dependent accumulation of G1 phase cells. Pharmacokinetic studies in mice showed a profile compatible with achieving good levels of target engagement. GSK137, administered orally, suppressed immunoglobulin G responses and reduced numbers of germinal centers and germinal center B cells following immunization of mice with the hapten trinitrophenol. Overall, we report a novel small-molecule BCL6 inhibitor with in vivo activity that inhibits the T-dependent antigen immune response.


Subject(s)
Proto-Oncogene Proteins c-bcl-6 , Animals , B-Lymphocytes/metabolism , Humans , Mice , Transcription, Genetic , Zinc Fingers
2.
J Invest Dermatol ; 141(9): 2272-2279, 2021 09.
Article in English | MEDLINE | ID: mdl-33744298

ABSTRACT

Regulation of proteolytic activity in the skin plays a pivotal role in epidermal homeostasis. This is best exemplified in Netherton syndrome, a severe genetic skin condition caused by loss-of-function mutations in the gene serine protease inhibitor Kazal-type 5 encoding lympho-epithelial Kazal-type-related inhibitor, a serine protease inhibitor that regulates kallikrein (KLK)-related peptidase 5, 7, and 14 activities. KLK5 plays a central role in stratum corneum shedding and inflammatory cell signaling, activates KLK7 and KLK14, and is therefore an optimal therapeutic target. We aimed to identify a potent and selective small-molecule inhibitor of KLK5 amenable to epidermal delivery. GSK951 was identified using a structure-based design strategy and showed a half maximal inhibitory concentration of 250 pM for KLK5 and greater than 100-fold selectivity over KLK7 and KLK14. Cocrystal structure analysis identified the critical catalytic site interactions to a surrogate for KLK5. Topical application of GSK951-containing cream inhibited KLK5 activity in TgKLK5 mouse skin, reduced transepidermal water loss, and decreased proinflammatory cytokine expression. GSK951 achieved high concentrations in healthy human epidermis following topical application in a cream formulation. Finally, KLK5 protease activity was increased in stratum corneum of patients with Netherton syndrome and significantly inhibited by GSK951. These findings unveil a KLK5-specific small-molecule inhibitor with a high therapeutic potential for patients with Netherton syndrome.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Boron Compounds/therapeutic use , Inflammation/drug therapy , Kallikreins/antagonists & inhibitors , Netherton Syndrome/drug therapy , Skin/pathology , Administration, Topical , Animals , Disease Models, Animal , Humans , Kallikreins/genetics , Mice , Mice, Transgenic , Signal Transduction , Skin/drug effects , Skin Cream
3.
Bioorg Med Chem Lett ; 41: 127973, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33753261

ABSTRACT

α1-antitrypsin deficiency is characterised by the misfolding and intracellular polymerisation of mutant α1-antitrypsin protein within the endoplasmic reticulum (ER) of hepatocytes. Small molecules that bind and stabilise Z α1-antitrypsin were identified via a DNA-encoded library screen. A subsequent structure based optimisation led to a series of highly potent, selective and cellular active α1-antitrypsin correctors.


Subject(s)
Drug Design , Protein Folding , alpha 1-Antitrypsin/metabolism , Crystallization , Drug Development/methods , Drug Evaluation, Preclinical , Endoplasmic Reticulum/metabolism , Gene Library , Hepatocytes/metabolism , Humans , Models, Molecular , Protein Conformation , alpha 1-Antitrypsin/genetics
4.
EMBO Mol Med ; 13(3): e13167, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33512066

ABSTRACT

Severe α1 -antitrypsin deficiency results from the Z allele (Glu342Lys) that causes the accumulation of homopolymers of mutant α1 -antitrypsin within the endoplasmic reticulum of hepatocytes in association with liver disease. We have used a DNA-encoded chemical library to undertake a high-throughput screen to identify small molecules that bind to, and stabilise Z α1 -antitrypsin. The lead compound blocks Z α1 -antitrypsin polymerisation in vitro, reduces intracellular polymerisation and increases the secretion of Z α1 -antitrypsin threefold in an iPSC model of disease. Crystallographic and biophysical analyses demonstrate that GSK716 and related molecules bind to a cryptic binding pocket, negate the local effects of the Z mutation and stabilise the bound state against progression along the polymerisation pathway. Oral dosing of transgenic mice at 100 mg/kg three times a day for 20 days increased the secretion of Z α1 -antitrypsin into the plasma by sevenfold. There was no observable clearance of hepatic inclusions with respect to controls over the same time period. This study provides proof of principle that "mutation ameliorating" small molecules can block the aberrant polymerisation that underlies Z α1 -antitrypsin deficiency.


Subject(s)
alpha 1-Antitrypsin Deficiency , alpha 1-Antitrypsin , Animals , Endoplasmic Reticulum , Hepatocytes , Mice , alpha 1-Antitrypsin/genetics
5.
Bioorg Med Chem Lett ; 30(21): 127533, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32919012

ABSTRACT

Structure-based led optimisation of orally active reversible Methionine Aminopeptidase-2 (MetAP-2) inhibitors utilising a 'molecular budget' medicinal chemistry strategy is described. The key physicochemical parameters of target molecules (cLogP, molecular size and H-bond donor count) were monitored through straightforward and intuitive use of atom count and distribution. The balance between structure-based design and an awareness of the physicochemical properties of the compounds synthesised enabled the rapid identification of a potent molecule with good oral pharmacokinetic (PK) characteristics by making fewer, higher quality compounds. The resulting candidate quality molecule was validated in a mechanistic cellular assay and a rodent secondary immunisation model.


Subject(s)
Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Methionyl Aminopeptidases/antagonists & inhibitors , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Indoles/chemical synthesis , Indoles/chemistry , Methionyl Aminopeptidases/metabolism , Molecular Structure , Structure-Activity Relationship
6.
J Med Chem ; 63(6): 3348-3358, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32109056

ABSTRACT

ER aminopeptidase 1 (ERAP1) is an intracellular enzyme that generates antigenic peptides and is an emerging target for cancer immunotherapy and the control of autoimmunity. ERAP1 inhibitors described previously target the active site and are limited in selectivity, minimizing their clinical potential. To address this, we targeted the regulatory site of ERAP1 using a high-throughput screen and discovered a small molecule hit that is highly selective for ERAP1. (4aR,5S,6R,8S,8aR)-5-(2-(Furan-3-yl)ethyl)-8-hydroxy-5,6,8a-trimethyl-3,4,4a,5,6,7,8,8a-octahydronaphthalene-1-carboxylic acid is a natural product found in Dodonaea viscosa that constitutes a submicromolar, highly selective, and cell-active modulator of ERAP1. Although the compound activates hydrolysis of small model substrates, it is a competitive inhibitor for physiologically relevant longer peptides. Crystallographic analysis confirmed that the compound targets the regulatory site of the enzyme that normally binds the C-terminus of the peptide substrate. Our findings constitute a novel starting point for the development of selective ERAP1 modulators that have potential for further clinical development.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Antigen Presentation/drug effects , Diterpenes, Clerodane/pharmacology , Epitopes/metabolism , Peptides/metabolism , Protease Inhibitors/pharmacology , Allosteric Site , Aminopeptidases/chemistry , Aminopeptidases/metabolism , Animals , Catalytic Domain , Crystallography, X-Ray , Diterpenes, Clerodane/chemistry , Diterpenes, Clerodane/metabolism , Enzyme Activators/chemistry , Enzyme Activators/metabolism , Enzyme Activators/pharmacology , Epitopes/chemistry , HeLa Cells , Humans , Mice , Minor Histocompatibility Antigens/chemistry , Minor Histocompatibility Antigens/metabolism , Peptides/chemistry , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Protein Binding , Proteolysis/drug effects
8.
Br J Pharmacol ; 177(8): 1853-1864, 2020 04.
Article in English | MEDLINE | ID: mdl-31877231

ABSTRACT

BACKGROUND AND PURPOSE: Miridesap, a depleter of serum amyloid P component (SAP), forms an essential component of a novel approach to remove systemic amyloid deposits; low oral bioavailability necessitates that it is given parenterally. We sought to identify and clinically characterise a pro-drug that preserves the pharmacological properties of miridesap while having adequate oral bioavailability and physical stability. EXPERIMENTAL APPROACH: We utilised a preclinical screening cascade focused on appropriate physicochemical properties, physical and gut stability, and conversion to miridesap in liver microsomes and blood. GSK3039294 (GSK294) had the desired in vitro profile and progressed to preclinical in vivo pharmacokinetic and safety assessments. Based on a favourable profile, it was tested in healthy participants after single and repeat dosing. KEY RESULTS: GSK294 was highly soluble and stable in simulated gastric and intestinal fluids, stable in intestinal microsomes, and permeable in Madine Darby Canine Kidney type II cells. GSK294 was rapidly hydrolysed to miridesap and its mono pro-drug ester in blood and liver microsomes. GSK294 showed good oral bioavailability of miridesap in rats and dogs. Following administration of GSK294 600 mg QD for 7 days in humans, pharmacodynamically active concentrations of miridesap were achieved with substantial and sustained depletion of plasma SAP. The study was terminated due to observations of arrhythmia, the relation of which to GSK294 remains unclear. CONCLUSION AND IMPLICATIONS: Using a preclinical screening cascade, we identified a pro-drug for a palindromic molecule with unique pharmacology (miridesap). The pro-drug depleted circulating SAP with a time course and extent similar to that of parenterally administered miridesap.


Subject(s)
Prodrugs , Administration, Oral , Animals , Biological Availability , Carboxylic Acids , Dogs , Microsomes, Liver/metabolism , Pyrrolidines , Rats , Serum Amyloid P-Component/metabolism
9.
Bioorg Med Chem Lett ; 29(20): 126675, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31521475

ABSTRACT

The connection between Netherton syndrome and overactivation of epidermal/dermal proteases, particularly Kallikrein 5 (KLK5) has been well established and it is expected that a KLK5 inhibitor would improve the dermal barrier and also reduce the pain and itch that afflict Netherton syndrome patients. One of the challenges of covalent protease inhibitors has been achieving selectivity over closely related targets. In this paper we describe the use of structural insight to design and develop a selective and highly potent reversibly covalent KLK5 inhibitor from an initial weakly binding fragment.


Subject(s)
Benzamidines/chemistry , Kallikreins/antagonists & inhibitors , Netherton Syndrome/drug therapy , Serine Proteinase Inhibitors/chemistry , Amino Acid Sequence , Benzamidines/pharmacology , Binding Sites , Drug Evaluation, Preclinical , Humans , Isomerism , Models, Molecular , Molecular Structure , Mutation , Protein Binding , Serine Peptidase Inhibitor Kazal-Type 5/genetics , Serine Proteinase Inhibitors/pharmacology , Structure-Activity Relationship
10.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 5): 385-391, 2019 May 01.
Article in English | MEDLINE | ID: mdl-31045568

ABSTRACT

The inhibition of kallikrein 5 (KLK5) has been identified as a potential strategy for treatment of the genetic skin disorder Netherton syndrome, in which loss-of-function mutations in the SPINK5 gene lead to down-regulation of the endogenous inhibitor LEKTI-1 and profound skin-barrier defects with severe allergic manifestations. To aid in the development of a medicine for this target, an X-ray crystallographic system was developed to facilitate fragment-guided chemistry and knowledge-based drug-discovery approaches. Here, the development of a surrogate crystallographic system in place of KLK5, which proved to be challenging to crystallize, is described. The biochemical robustness of the crystallographic surrogate and the suitability of the system for the study of small nonpeptidic fragments and lead-like molecules are demonstrated.


Subject(s)
Benzamidines/chemistry , Kallikreins/chemistry , Protease Inhibitors/chemistry , Amino Acid Sequence , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Benzamidines/pharmacology , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Drug Discovery , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Kallikreins/antagonists & inhibitors , Kallikreins/genetics , Kallikreins/metabolism , Kinetics , Models, Molecular , Mutation , Netherton Syndrome/drug therapy , Netherton Syndrome/enzymology , Protease Inhibitors/pharmacology , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sf9 Cells , Spodoptera , Static Electricity , Substrate Specificity
11.
Bioorg Med Chem Lett ; 29(12): 1454-1458, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31005442

ABSTRACT

The connection between Netherton syndrome and overactivation of epidermal/dermal proteases particularly KLK5 has been well established. To treat sufferers of this severe condition we wished to develop a topical KLK5 inhibitor in order to normalise epidermal shedding and reduce the associated inflammation and itching. In this paper we describe structure-based optimisation of a series of brightly coloured weak KLK5 inhibitors into colourless, non-irritant molecules with good KLK5 activity and selectivity over a range of serine proteases.


Subject(s)
Drug Design , Kallikreins/antagonists & inhibitors , Netherton Syndrome/drug therapy , Humans
12.
Bioorg Med Chem Lett ; 29(6): 821-825, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30691925

ABSTRACT

Netherton syndrome (NS) is a rare and debilitating severe autosomal recessive genetic skin disease with high mortality rates particularly in neonates. NS is caused by loss-of-function SPINK5 mutations leading to unregulated kallikrein 5 (KLK5) and kallikrein 7 (KLK7) activity. Furthermore, KLK5 inhibition has been proposed as a potential therapeutic treatment for NS. Identification of potent and selective KLK5 inhibitors would enable further exploration of the disease biology and could ultimately lead to a treatment for NS. This publication describes how fragmentation of known trypsin-like serine protease (TLSP) inhibitors resulted in the identification of a series of phenolic amidine-based KLK5 inhibitors 1. X-ray crystallography was used to find alternatives to the phenol interaction leading to identification of carbonyl analogues such as lactam 13 and benzimidazole 15. These reversible inhibitors, with selectivity over KLK1 (10-100 fold), provided novel starting points for the guided growth towards suitable tool molecules for the exploration of KLK5 biology.


Subject(s)
Benzamidines/chemistry , Kallikreins/antagonists & inhibitors , Serine Proteinase Inhibitors/chemistry , Animals , Benzamidines/chemical synthesis , Benzamidines/metabolism , Catalytic Domain , Drug Design , Kallikreins/metabolism , Netherton Syndrome/drug therapy , Protein Binding , Salicylamides/chemical synthesis , Salicylamides/chemistry , Salicylamides/metabolism , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/metabolism , Spodoptera/genetics
13.
Bioorg Med Chem Lett ; 27(21): 4914-4919, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28958623

ABSTRACT

A series of potent, selective and long-acting quinoline-based sulfonamide human H1 histamine receptor antagonists, designed for once-daily intranasal administration for the treatment of rhinitis were developed. Sulfonamide 33b had a slightly lower affinity for the H1 receptor than azelastine, had low oral bioavailability in the rat and dog, and was turned over to five major metabolites. Furthermore, 33b had longer duration of action than azelastine in guinea pigs, lower rat brain-penetration, and did not cause time dependent inhibition of CYP2D6 or CYP3A4. The clinical dose in humans is expected to be low (approximately 0.5mg per day) based on the clinical dose used for azelastine and a comparison of efficacy data from animal models for 33b and azelastine.


Subject(s)
Histamine H1 Antagonists/chemistry , Quinolines/chemistry , Receptors, Histamine H1/metabolism , Rhinitis, Allergic/drug therapy , Sulfanilamides/chemistry , Sulfonamides/chemistry , Sulfones/chemistry , Administration, Intranasal , Animals , Brain/metabolism , Dogs , Guinea Pigs , Half-Life , Histamine H1 Antagonists/pharmacokinetics , Histamine H1 Antagonists/therapeutic use , Inhibitory Concentration 50 , Quinolines/pharmacokinetics , Quinolines/therapeutic use , Rats , Receptors, Histamine H1/chemistry , Rhinitis, Allergic/metabolism , Rhinitis, Allergic/pathology , Structure-Activity Relationship , Sulfanilamide , Sulfanilamides/pharmacokinetics , Sulfanilamides/therapeutic use , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use , Sulfones/pharmacokinetics , Sulfones/therapeutic use
14.
Sci Rep ; 7(1): 10806, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28883402

ABSTRACT

The peptide hormone human relaxin-2 (H2-RLX) has emerged as a potential therapy for cardiovascular and fibrotic diseases, but its short in vivo half-life is an obstacle to long-term administration. The discovery of ML290 demonstrated that it is possible to identify small molecule agonists of the cognate G-protein coupled receptor for H2-RLX (relaxin family peptide receptor-1 (RXFP1)). In our efforts to generate a new medicine for liver fibrosis, we sought to identify improved small molecule functional mimetics of H2-RLX with selective, full agonist or positive allosteric modulator activity against RXFP1. First, we confirmed expression of RXFP1 in human diseased liver. We developed a robust cellular cAMP reporter assay of RXFP1 signaling in HEK293 cells transiently expressing RXFP1. A high-throughput screen did not identify further specific agonists or positive allosteric modulators of RXFP1, affirming the low druggability of this receptor. As an alternative approach, we generated novel ML290 analogues and tested their activity in the HEK293-RXFP1 cAMP assay and the human hepatic cell line LX-2. Differences in activity of compounds on cAMP activation compared with changes in expression of fibrotic markers indicate the need to better understand cell- and tissue-specific signaling mechanisms and their disease-relevant phenotypes in order to enable drug discovery.


Subject(s)
Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Enzyme Activators/isolation & purification , Liver Cirrhosis/drug therapy , Receptors, G-Protein-Coupled/agonists , Receptors, Peptide/agonists , Biopsy , Cells, Cultured , Enzyme Activators/chemical synthesis , Enzyme Activators/pharmacology , High-Throughput Screening Assays , Humans , Liver Cirrhosis/pathology
15.
Nat Commun ; 8: 15827, 2017 06 12.
Article in English | MEDLINE | ID: mdl-28604669

ABSTRACT

Kynurenine-3-monooxygenase (KMO) is a key FAD-dependent enzyme of tryptophan metabolism. In animal models, KMO inhibition has shown benefit in neurodegenerative diseases such as Huntington's and Alzheimer's. Most recently it has been identified as a target for acute pancreatitis multiple organ dysfunction syndrome (AP-MODS); a devastating inflammatory condition with a mortality rate in excess of 20%. Here we report and dissect the molecular mechanism of action of three classes of KMO inhibitors with differentiated binding modes and kinetics. Two novel inhibitor classes trap the catalytic flavin in a previously unobserved tilting conformation. This correlates with picomolar affinities, increased residence times and an absence of the peroxide production seen with previous substrate site inhibitors. These structural and mechanistic insights culminated in GSK065(C1) and GSK366(C2), molecules suitable for preclinical evaluation. Moreover, revising the repertoire of flavin dynamics in this enzyme class offers exciting new opportunities for inhibitor design.


Subject(s)
Enzyme Inhibitors/pharmacology , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Multiple Organ Failure/metabolism , Pancreatitis/metabolism , Animals , Enzyme Inhibitors/chemistry , Escherichia coli/genetics , Humans , Hydrogen Peroxide/metabolism , Kynurenine 3-Monooxygenase/chemistry , Kynurenine 3-Monooxygenase/metabolism , Models, Molecular , Protein Domains , Sf9 Cells
16.
ACS Med Chem Lett ; 8(5): 577-581, 2017 May 11.
Article in English | MEDLINE | ID: mdl-28523114

ABSTRACT

The synthesis of potent amide-containing phthalazinone H1 histamine receptor antagonists is described. Three analogues 3e, 3g, and 9g were equipotent with azelastine and were longer-acting in vitro. Amide 3g had low oral bioavailability, low brain-penetration, high metabolic clearance, and long duration of action in vivo, and it was suitable for once-daily dosing intranasally, with a predicted dose for humans of approximately 0.5 mg per day.

17.
J Med Chem ; 60(8): 3383-3404, 2017 04 27.
Article in English | MEDLINE | ID: mdl-28398044

ABSTRACT

Recently, we reported a novel role for KMO in the pathogenesis of acute pancreatitis (AP). A number of inhibitors of kynurenine 3-monooxygenase (KMO) have previously been described as potential treatments for neurodegenerative conditions and particularly for Huntington's disease. However, the inhibitors reported to date have insufficient aqueous solubility relative to their cellular potency to be compatible with the intravenous (iv) dosing route required in AP. We have identified and optimized a novel series of high affinity KMO inhibitors with favorable physicochemical properties. The leading example is exquisitely selective, has low clearance in two species, prevents lung and kidney damage in a rat model of acute pancreatitis, and is progressing into preclinical development.


Subject(s)
Enzyme Inhibitors/pharmacology , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Pancreatitis/drug therapy , Acute Disease , Animals , Enzyme Inhibitors/therapeutic use , Humans , Rats
19.
Bioorg Med Chem Lett ; 26(24): 5855-5859, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27866818

ABSTRACT

A novel series of potent quinoline-based human H1 and H3 bivalent histamine receptor antagonists, suitable for intranasal administration for the potential treatment of allergic rhinitis associated nasal congestion, were identified. Compound 18b had slightly lower H1 potency (pA2 8.8 vs 9.7 for the clinical goldstandard azelastine), and H3 potency (pKi 9.1vs 6.8 for azelastine), better selectivity over α1A, α1B and hERG, similar duration of action, making 18b a good back-up compound to our previous candidate, but with a more desirable profile.


Subject(s)
Drug Discovery , Histamine H1 Antagonists/pharmacology , Histamine H3 Antagonists/pharmacology , Quinolines/pharmacology , Receptors, Histamine H1/metabolism , Receptors, Histamine H3/metabolism , Dose-Response Relationship, Drug , Histamine H1 Antagonists/chemical synthesis , Histamine H1 Antagonists/chemistry , Histamine H3 Antagonists/chemical synthesis , Histamine H3 Antagonists/chemistry , Humans , Ligands , Molecular Structure , Quinolines/chemical synthesis , Quinolines/chemistry , Structure-Activity Relationship
20.
Nat Med ; 22(2): 202-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26752518

ABSTRACT

Acute pancreatitis (AP) is a common and devastating inflammatory condition of the pancreas that is considered to be a paradigm of sterile inflammation leading to systemic multiple organ dysfunction syndrome (MODS) and death. Acute mortality from AP-MODS exceeds 20% (ref. 3), and the lifespans of those who survive the initial episode are typically shorter than those of the general population. There are no specific therapies available to protect individuals from AP-MODS. Here we show that kynurenine-3-monooxygenase (KMO), a key enzyme of tryptophan metabolism, is central to the pathogenesis of AP-MODS. We created a mouse strain that is deficient for Kmo (encoding KMO) and that has a robust biochemical phenotype that protects against extrapancreatic tissue injury to the lung, kidney and liver in experimental AP-MODS. A medicinal chemistry strategy based on modifications of the kynurenine substrate led to the discovery of the oxazolidinone GSK180 as a potent and specific inhibitor of KMO. The binding mode of the inhibitor in the active site was confirmed by X-ray co-crystallography at 3.2 Å resolution. Treatment with GSK180 resulted in rapid changes in the levels of kynurenine pathway metabolites in vivo, and it afforded therapeutic protection against MODS in a rat model of AP. Our findings establish KMO inhibition as a novel therapeutic strategy in the treatment of AP-MODS, and they open up a new area for drug discovery in critical illness.


Subject(s)
Benzoxazoles/pharmacology , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Multiple Organ Failure/genetics , Oxazolidinones/pharmacology , Pancreatitis/genetics , Propionates/pharmacology , RNA, Messenger/metabolism , Acute Disease , Animals , Chromatography, Liquid , Crystallography, X-Ray , Disease Models, Animal , HEK293 Cells , Hepatocytes/metabolism , Humans , In Vitro Techniques , Kidney/metabolism , Kidney/pathology , Kynurenine 3-Monooxygenase/genetics , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Multiple Organ Failure/etiology , Multiple Organ Failure/pathology , Pancreas/metabolism , Pancreas/pathology , Pancreatitis/complications , Pancreatitis/pathology , Rats , Tandem Mass Spectrometry , Tryptophan/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...