Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 16(730): eadf1691, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38232139

ABSTRACT

Glycogen synthase 1 (GYS1), the rate-limiting enzyme in muscle glycogen synthesis, plays a central role in energy homeostasis and has been proposed as a therapeutic target in multiple glycogen storage diseases. Despite decades of investigation, there are no known potent, selective small-molecule inhibitors of this enzyme. Here, we report the preclinical characterization of MZ-101, a small molecule that potently inhibits GYS1 in vitro and in vivo without inhibiting GYS2, a related isoform essential for synthesizing liver glycogen. Chronic treatment with MZ-101 depleted muscle glycogen and was well tolerated in mice. Pompe disease, a glycogen storage disease caused by mutations in acid α glucosidase (GAA), results in pathological accumulation of glycogen and consequent autophagolysosomal abnormalities, metabolic dysregulation, and muscle atrophy. Enzyme replacement therapy (ERT) with recombinant GAA is the only approved treatment for Pompe disease, but it requires frequent infusions, and efficacy is limited by suboptimal skeletal muscle distribution. In a mouse model of Pompe disease, chronic oral administration of MZ-101 alone reduced glycogen buildup in skeletal muscle with comparable efficacy to ERT. In addition, treatment with MZ-101 in combination with ERT had an additive effect and could normalize muscle glycogen concentrations. Biochemical, metabolomic, and transcriptomic analyses of muscle tissue demonstrated that lowering of glycogen concentrations with MZ-101, alone or in combination with ERT, corrected the cellular pathology in this mouse model. These data suggest that substrate reduction therapy with GYS1 inhibition may be a promising therapeutic approach for Pompe disease and other glycogen storage diseases.


Subject(s)
Glycogen Storage Disease Type II , Mice , Animals , Glycogen Storage Disease Type II/drug therapy , Glycogen Synthase/metabolism , Glycogen Synthase/pharmacology , Mice, Knockout , Glycogen/metabolism , Muscle, Skeletal/metabolism , Enzyme Replacement Therapy/methods
2.
Cell Rep ; 40(1): 111041, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35793618

ABSTRACT

Glycogen is the primary energy reserve in mammals, and dysregulation of glycogen metabolism can result in glycogen storage diseases (GSDs). In muscle, glycogen synthesis is initiated by the enzymes glycogenin-1 (GYG1), which seeds the molecule by autoglucosylation, and glycogen synthase-1 (GYS1), which extends the glycogen chain. Although both enzymes are required for proper glycogen production, the nature of their interaction has been enigmatic. Here, we present the human GYS1:GYG1 complex in multiple conformations representing different functional states. We observe an asymmetric conformation of GYS1 that exposes an interface for close GYG1 association, and propose this state facilitates handoff of the GYG1-associated glycogen chain to a GYS1 subunit for elongation. Full activation of GYS1 widens the GYG1-binding groove, enabling GYG1 release concomitant with glycogen chain growth. This structural mechanism connecting chain nucleation and extension explains the apparent stepwise nature of glycogen synthesis and suggests distinct states to target for GSD-modifying therapeutics.


Subject(s)
Glycogen Synthase , Glycogenolysis , Glycoproteins , Glucosyltransferases/metabolism , Glycogen/metabolism , Glycogen Synthase/metabolism , Glycoproteins/metabolism , Humans
3.
J Am Chem Soc ; 142(9): 4445-4455, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32064871

ABSTRACT

The lipopolysaccharide biosynthesis pathway is considered an attractive drug target against the rising threat of multi-drug-resistant Gram-negative bacteria. Here, we report two novel small-molecule inhibitors (compounds 1 and 2) of the acyltransferase LpxA, the first enzyme in the lipopolysaccharide biosynthesis pathway. We show genetically that the antibacterial activities of the compounds against efflux-deficient Escherichia coli are mediated by LpxA inhibition. Consistently, the compounds inhibited the LpxA enzymatic reaction in vitro. Intriguingly, using biochemical, biophysical, and structural characterization, we reveal two distinct mechanisms of LpxA inhibition; compound 1 is a substrate-competitive inhibitor targeting apo LpxA, and compound 2 is an uncompetitive inhibitor targeting the LpxA/product complex. Compound 2 exhibited more favorable biological and physicochemical properties than compound 1 and was optimized using structural information to achieve improved antibacterial activity against wild-type E. coli. These results show that LpxA is a promising antibacterial target and imply the advantages of targeting enzyme/product complexes in drug discovery.


Subject(s)
Acyltransferases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Pyrazoles/pharmacology , Acyltransferases/metabolism , Anti-Bacterial Agents/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Escherichia coli/drug effects , Escherichia coli/enzymology , Imidazoles/metabolism , Microbial Sensitivity Tests , Protein Binding , Pyrazoles/metabolism
4.
J Med Chem ; 63(5): 2013-2027, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31059256

ABSTRACT

Direct pharmacological inhibition of RAS has remained elusive, and efforts to target CRAF have been challenging due to the complex nature of RAF signaling, downstream of activated RAS, and the poor overall kinase selectivity of putative RAF inhibitors. Herein, we describe 15 (LXH254, Aversa, R.; et al. Int. Patent WO2014151616A1, 2014), a selective B/C RAF inhibitor, which was developed by focusing on drug-like properties and selectivity. Our previous tool compound, 3 (RAF709; Nishiguchi, G. A.; et al. J. Med. Chem. 2017, 60, 4969), was potent, selective, efficacious, and well tolerated in preclinical models, but the high human intrinsic clearance precluded further development and prompted further investigation of close analogues. A structure-based approach led to a pyridine series with an alcohol side chain that could interact with the DFG loop and significantly improved cell potency. Further mitigation of human intrinsic clearance and time-dependent inhibition led to the discovery of 15. Due to its excellent properties, it was progressed through toxicology studies and is being tested in phase 1 clinical trials.


Subject(s)
Antineoplastic Agents/chemistry , Drug Discovery/methods , Mutation/genetics , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Animals , Antineoplastic Agents/pharmacology , Drug Design , Drug Discovery/trends , Humans , Molecular Docking Simulation/methods , Molecular Docking Simulation/trends , Mutation/drug effects , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays/methods
6.
J Med Chem ; 60(12): 4869-4881, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28557458

ABSTRACT

RAS oncogenes have been implicated in >30% of human cancers, all representing high unmet medical need. The exquisite dependency on CRAF kinase in KRAS mutant tumors has been established in genetically engineered mouse models and human tumor cells. To date, many small molecule approaches are under investigation to target CRAF, yet kinase-selective and cellular potent inhibitors remain challenging to identify. Herein, we describe 14 (RAF709) [ Aversa , Biaryl amide compounds as kinase inhibitors and their preparation . WO 2014151616, 2014 ], a selective B/C RAF inhibitor, which was developed through a hypothesis-driven approach focusing on drug-like properties. A key challenge encountered in the medicinal chemistry campaign was maintaining a balance between good solubility and potent cellular activity (suppression of pMEK and proliferation) in KRAS mutant tumor cell lines. We investigated the small molecule crystal structure of lead molecule 7 and hypothesized that disruption of the crystal packing would improve solubility, which led to a change from N-methylpyridone to a tetrahydropyranyl oxy-pyridine derivative. 14 proved to be soluble, kinase selective, and efficacious in a KRAS mutant xenograft model.


Subject(s)
2,2'-Dipyridyl/analogs & derivatives , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , raf Kinases/antagonists & inhibitors , ras Proteins/genetics , 2,2'-Dipyridyl/chemistry , 2,2'-Dipyridyl/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Benzamides/chemistry , Crystallography, X-Ray , Dogs , Drug Design , Drug Discovery , Drug Stability , Humans , Inhibitory Concentration 50 , Mice , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
7.
Bioorg Med Chem Lett ; 26(3): 742-746, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26774655

ABSTRACT

Alterations in PI3K/AKT signaling are known to be implicated with tumorigenesis. The PI3 kinases family of lipid kinases has been an attractive therapeutic target for cancer treatment. Imidazopyridine compound 1, a potent, selective, and orally available pan-PI3K inhibitor, identified by scaffold morphing of a benzothiazole hit, was further optimized in order to achieve efficacy in a PTEN-deleted A2780 ovarian cancer mouse xenograft model. With a hypothesis that a planar conformation between the core and the 6-heteroaryl ring will allow for the accommodation of larger 5'-substituents in a hydrophobic area under P-loop, SAR efforts focused on 5'-alkoxy heteroaryl rings at the 6-position of imidazopyridine and imidazopyridazine cores that have the same dihedral angle of zero degrees. 6'-Alkoxy 5'-aminopyrazines in the imidazopyridine series were identified as the most potent compounds in the A2780 cell line. Compound 14 with 1,1,1-trifluoroisopropoxy group at 6'-position demonstrated excellent potency and selectivity, good oral exposure in rats and in vivo efficacy in A2780 tumor-bearing mouse. Also, we disclose the X-ray co-crystal structure of one enantiomer of compound 14 in PI3Kα, confirming that the trifluoromethyl group fits nicely in the hydrophobic hot spot under P-loop.


Subject(s)
Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Disease Models, Animal , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Female , Half-Life , Heterografts , Humans , Mice , Molecular Docking Simulation , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Protein Structure, Tertiary , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Rats , Stereoisomerism , Structure-Activity Relationship
8.
Protein Expr Purif ; 110: 172-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25818999

ABSTRACT

Extracellular signal-regulated kinase 2 (ERK2) is a serine/threonine protein kinase involved in many cellular programs, such as cell proliferation, differentiation, motility and programed cell-death. It is therefore considered an important target in the treatment of cancer. In an effort to support biochemical screening and small molecule drug discovery, we established a robust system to generate both inactive and active forms of ERK2 using insect expression system. We report here, for the first time, that inactive ERK2 can be expressed and purified with 100% homogeneity in the unphosphorylated form using insect system. This resulted in a significant 20-fold yield improvement compared to that previously reported using bacterial expression system. We also report a newly developed system to generate active ERK2 in insect cells through in vivo co-expression with a constitutively active MEK1 (S218D S222D). Isolated active ERK2 was confirmed to be doubly phosphorylated at the correct sites, T185 and Y187, in the activation loop of ERK2. Both ERK2 forms, inactive and active, were well characterized by biochemical activity assay for their kinase function. Inactive and active ERK2 were the two key reagents that enabled successful high through-put biochemical assay screen and structural drug discovery studies.


Subject(s)
Baculoviridae/genetics , Cloning, Molecular/methods , Mitogen-Activated Protein Kinase 1/genetics , Plasmids/metabolism , Recombinant Fusion Proteins/genetics , Animals , Baculoviridae/metabolism , Enzyme Activation , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Engineering , Histidine/genetics , Histidine/metabolism , Humans , Kinetics , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 1/isolation & purification , Oligopeptides/genetics , Oligopeptides/metabolism , Phosphorylation , Plasmids/chemistry , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Sf9 Cells , Spodoptera
9.
Bioorg Med Chem Lett ; 23(16): 4652-6, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23820386

ABSTRACT

PI3 kinases are a family of lipid kinases mediating numerous cell processes such as proliferation, migration and differentiation. The PI3 Kinase pathway is often de-regulated in cancer through PI3Kα overexpression, gene amplification, mutations and PTEN phosphatase deletion. PI3K inhibitors represent therefore an attractive therapeutic modality for cancer treatment. Herein we describe how the potency of a benzothiazole fragment hit was quickly improved based on structural information and how this early chemotype was further optimized through scaffold hopping. This effort led to the identification of a series of 2-acetamido-5-heteroaryl imidazopyridines showing potent in vitro activity against all class I PI3Ks and attractive pharmacokinetic properties.


Subject(s)
Azo Compounds/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors , Pyridines/chemical synthesis , Pyridines/pharmacology , Azo Compounds/chemistry , Azo Compounds/pharmacology , Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Cell Line, Tumor , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Female , Humans , Imides/chemical synthesis , Imides/chemistry , Imides/pharmacology , Inhibitory Concentration 50 , Models, Molecular , Pyridines/chemistry , Solubility , Structure-Activity Relationship
10.
Cancer Cell ; 23(5): 594-602, 2013 May 13.
Article in English | MEDLINE | ID: mdl-23680146

ABSTRACT

ATP competitive inhibitors of the BRAF(V600E) oncogene paradoxically activate downstream signaling in cells bearing wild-type BRAF (BRAF(WT)). In this study, we investigate the biochemical mechanism of wild-type RAF (RAF(WT)) activation by multiple catalytic inhibitors using kinetic analysis of purified BRAF(V600E) and RAF(WT) enzymes. We show that activation of RAF(WT) is ATP dependent and directly linked to RAF kinase activity. These data support a mechanism involving inhibitory autophosphorylation of RAF's phosphate-binding loop that, when disrupted either through pharmacologic or genetic alterations, results in activation of RAF and the mitogen-activated protein kinase (MAPK) pathway. This mechanism accounts not only for compound-mediated activation of the MAPK pathway in BRAF(WT) cells but also offers a biochemical mechanism for BRAF oncogenesis.


Subject(s)
MAP Kinase Signaling System/drug effects , raf Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/physiology , Cell Line, Tumor , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins B-raf/physiology , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins c-raf/physiology , raf Kinases/genetics , raf Kinases/metabolism
11.
ACS Med Chem Lett ; 2(10): 774-9, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-24900266

ABSTRACT

Phosphoinositide-3-kinases (PI3Ks) are important oncology targets due to the deregulation of this signaling pathway in a wide variety of human cancers. Herein we describe the structure guided optimization of a series of 2-morpholino, 4-substituted, 6-heterocyclic pyrimidines where the pharmacokinetic properties were improved by modulating the electronics of the 6-position heterocycle, and the overall druglike properties were fine-tuned further by modification of the 4-position substituent. The resulting 2,4-bismorpholino 6-heterocyclic pyrimidines are potent class I PI3K inhibitors showing mechanism modulation in PI3K dependent cell lines and in vivo efficacy in tumor xenograft models with PI3K pathway deregulation (A2780 ovarian and U87MG glioma). These efforts culminated in the discovery of 15 (NVP-BKM120), currently in Phase II clinical trials for the treatment of cancer.

12.
Bioorg Med Chem Lett ; 20(23): 6895-8, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21035331

ABSTRACT

PI3 Kinases are a family of lipid kinases mediating numerous cell processes such as proliferation, migration, and differentiation. The PI3 kinase pathway is often de-regulated in cancer through PI3Kα overexpression, gene amplification, mutations, and PTEN phosphatase deletion. PI3K inhibitors represent therefore an attractive therapeutic modality for cancer treatment. Herein we describe a novel series of PI3K inhibitors sharing a pyrimidine core and showing significant potency against class I PI3 kinases in the biochemical assay and in cells. The discovery, synthesis and SAR of this chemotype are described.


Subject(s)
Antineoplastic Agents/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Morpholines/chemistry , Morpholines/pharmacology , Phosphorylation/drug effects , Pyrimidines/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...