Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Transl Med ; 16(749): eabp8334, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38809966

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disease driven by gain-of-function variants in activin receptor-like kinase 2 (ALK2), the most common variant being ALK2R206H. In FOP, ALK2 variants display increased and dysregulated signaling through the bone morphogenetic protein (BMP) pathway resulting in progressive and permanent replacement of skeletal muscle and connective tissues with heterotopic bone, ultimately leading to severe debilitation and premature death. Here, we describe the discovery of BLU-782 (IPN60130), a small-molecule ALK2R206H inhibitor developed for the treatment of FOP. A small-molecule library was screened in a biochemical ALK2 binding assay to identify potent ALK2 binding compounds. Iterative rounds of structure-guided drug design were used to optimize compounds for ALK2R206H binding, ALK2 selectivity, and other desirable pharmacokinetic properties. BLU-782 preferentially bound to ALK2R206H with high affinity, inhibiting signaling from ALK2R206H and other rare FOP variants in cells in vitro without affecting signaling of closely related homologs ALK1, ALK3, and ALK6. In vivo efficacy of BLU-782 was demonstrated using a conditional knock-in ALK2R206H mouse model, where prophylactic oral dosing reduced edema and prevented cartilage and heterotopic ossification (HO) in both muscle and bone injury models. BLU-782 treatment preserved the normal muscle-healing response in ALK2R206H mice. Delayed dosing revealed a short 2-day window after injury when BLU-782 treatment prevented HO in ALK2R206H mice, but dosing delays of 4 days or longer abrogated HO prevention. Together, these data suggest that BLU-782 may be a candidate for prevention of HO in FOP.


Subject(s)
Disease Models, Animal , Myositis Ossificans , Ossification, Heterotopic , Animals , Myositis Ossificans/drug therapy , Myositis Ossificans/metabolism , Ossification, Heterotopic/drug therapy , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/prevention & control , Mice , Humans , Activin Receptors, Type II/metabolism , Activin Receptors, Type I/metabolism , Activin Receptors, Type I/antagonists & inhibitors , Signal Transduction/drug effects
2.
Cancer Discov ; 9(12): 1686-1695, 2019 12.
Article in English | MEDLINE | ID: mdl-31575540

ABSTRACT

Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality worldwide with no clinically confirmed oncogenic driver. Although preclinical studies implicate the FGF19 receptor FGFR4 in hepatocarcinogenesis, the dependence of human cancer on FGFR4 has not been demonstrated. Fisogatinib (BLU-554) is a potent and selective inhibitor of FGFR4 and demonstrates clinical benefit and tumor regression in patients with HCC with aberrant FGF19 expression. Mutations were identified in the gatekeeper and hinge-1 residues in the kinase domain of FGFR4 upon disease progression in 2 patients treated with fisogatinib, which were confirmed to mediate resistance in vitro and in vivo. A gatekeeper-agnostic, pan-FGFR inhibitor decreased HCC xenograft growth in the presence of these mutations, demonstrating continued FGF19-FGFR4 pathway dependence. These results validate FGFR4 as an oncogenic driver and warrant further therapeutic targeting of this kinase in the clinic. SIGNIFICANCE: Our study is the first to demonstrate on-target FGFR4 kinase domain mutations as a mechanism of acquired clinical resistance to targeted therapy. This further establishes FGF19-FGFR4 pathway activation as an oncogenic driver. These findings support further investigation of fisogatinib in HCC and inform the profile of potential next-generation inhibitors.See related commentary by Subbiah and Pal, p. 1646.This article is highlighted in the In This Issue feature, p. 1631.


Subject(s)
Carcinoma, Hepatocellular/diagnostic imaging , Drug Resistance, Neoplasm , Liver Neoplasms/diagnostic imaging , Pyrans/pharmacology , Quinazolines/pharmacology , Receptor, Fibroblast Growth Factor, Type 4/genetics , Aged, 80 and over , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Female , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Middle Aged , Models, Molecular , Mutation , Neoplasm Transplantation , Protein Domains , Receptor, Fibroblast Growth Factor, Type 4/chemistry , Receptor, Fibroblast Growth Factor, Type 4/metabolism
3.
Cancer Discov ; 8(7): 836-849, 2018 07.
Article in English | MEDLINE | ID: mdl-29657135

ABSTRACT

The receptor tyrosine kinase rearranged during transfection (RET) is an oncogenic driver activated in multiple cancers, including non-small cell lung cancer (NSCLC), medullary thyroid cancer (MTC), and papillary thyroid cancer. No approved therapies have been designed to target RET; treatment has been limited to multikinase inhibitors (MKI), which can have significant off-target toxicities and limited efficacy. BLU-667 is a highly potent and selective RET inhibitor designed to overcome these limitations. In vitro, BLU-667 demonstrated ≥10-fold increased potency over approved MKIs against oncogenic RET variants and resistance mutants. In vivo, BLU-667 potently inhibited growth of NSCLC and thyroid cancer xenografts driven by various RET mutations and fusions without inhibiting VEGFR2. In first-in-human testing, BLU-667 significantly inhibited RET signaling and induced durable clinical responses in patients with RET-altered NSCLC and MTC without notable off-target toxicity, providing clinical validation for selective RET targeting.Significance: Patients with RET-driven cancers derive limited benefit from available MKIs. BLU-667 is a potent and selective RET inhibitor that induces tumor regression in cancer models with RET mutations and fusions. BLU-667 attenuated RET signaling and produced durable clinical responses in patients with RET-altered tumors, clinically validating selective RET targeting. Cancer Discov; 8(7); 836-49. ©2018 AACR.See related commentary by Iams and Lovly, p. 797This article is highlighted in the In This Issue feature, p. 781.


Subject(s)
Antineoplastic Agents/therapeutic use , Mutation , Neoplasms/drug therapy , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Proto-Oncogene Proteins c-ret/genetics , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/genetics , Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-ret/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Xenograft Model Antitumor Assays
4.
J Med Chem ; 56(3): 712-21, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23360348

ABSTRACT

PI3Kα has been identified as an oncogene in human tumors. By use of rational drug design, a targeted covalent inhibitor 3 (CNX-1351) was created that potently and specifically inhibits PI3Kα. We demonstrate, using mass spectrometry and X-ray crystallography, that the selective inhibitor covalently modifies PI3Kα on cysteine 862 (C862), an amino acid unique to the α isoform, and that PI3Kß, -γ, and -δ are not covalently modified. 3 is able to potently (EC(50) < 100 nM) and specifically inhibit signaling in PI3Kα-dependent cancer cell lines, and this leads to a potent antiproliferative effect (GI(50) < 100 nM). A covalent probe, 8 (CNX-1220), which selectively bonds to PI3Kα, was used to investigate the duration of occupancy of 3 with PI3Kα in vivo. This is the first report of a PI3Kα-selective inhibitor, and these data demonstrate the biological impact of selectively targeting PI3Kα.


Subject(s)
Drug Discovery , Isoenzymes/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Nuclear Magnetic Resonance, Biomolecular , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/chemistry , Signal Transduction
5.
Nat Chem Biol ; 7(1): 22-4, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21113170

ABSTRACT

Designing selective inhibitors of proteases has proven problematic, in part because pharmacophores that confer potency exploit the conserved catalytic apparatus. We developed a fundamentally different approach by designing irreversible inhibitors that target noncatalytic cysteines that are structurally unique to a target in a protein family. We have successfully applied this approach to the important therapeutic target HCV protease, which has broad implications for the design of other selective protease inhibitors.


Subject(s)
Cysteine Proteinase Inhibitors/therapeutic use , Cysteine/antagonists & inhibitors , Drug Design , Oligopeptides/therapeutic use , Biocatalysis , Biochemistry/methods , Crystallography, X-Ray , Cysteine/metabolism , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Hepacivirus/drug effects , Hepacivirus/enzymology , Hepacivirus/growth & development , Oligopeptides/chemistry , Oligopeptides/pharmacology , Virology/methods
6.
Cell Immunol ; 220(2): 134-42, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12657248

ABSTRACT

In order to discover novel immunomodulators for application in treating autoimmune diseases, a stable Jurkat transfectant was constructed in which luciferase reporter gene is driven by a full-length IL-2 promotor. A chemical library was screened to identify compounds that inhibited luciferase expression in Jurkat transfectants stimulated with PMA and ionomycin. A class of compounds (bis-trifluoromethyl pyrazole, BTPs) was identified from this screen. BTPs were shown to inhibit anti-CD3 and anti-CD28 antibody-induced IL-2 secretion, mixed lymphocyte reaction, and Con A-induced T cell proliferation in normal human peripheral blood T cells. In addition, mRNA levels of IL-4, IL-5, IL-9, IL-10, IL-13, IL-15, and IFN-gamma were markedly inhibited by BTPs in peripheral blood mononuclear cells stimulated by Con A as determined by multi-probe RNA protection assay. Furthermore, IL-2, IL-4, IL-5, and IFN-gamma secretion by Hut 78 cells or CD3(+) T cells stimulated with PMA plus ionomycin or anti-CD3 antibody plus PMA were inhibited in a concentration-dependent manner by BTPs. Therefore, BTPs inhibit a wide spectrum of cytokine production including TH1 and TH2 type cytokines. Taken together, these compounds may be useful for treating autoimmune diseases and organ transplant rejection.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cytokines/antagonists & inhibitors , Pyrazoles/pharmacology , Th1 Cells/drug effects , Th2 Cells/drug effects , Calcium/immunology , Cell Division/immunology , Concanavalin A/immunology , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/immunology , Humans , Interleukin-2/immunology , Ionomycin/immunology , Ionophores/immunology , Jurkat Cells/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Promoter Regions, Genetic/immunology , RNA/genetics , RNA/immunology , Tetradecanoylphorbol Acetate/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL