Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
JCI Insight ; 6(8)2021 04 22.
Article in English | MEDLINE | ID: mdl-33705358

ABSTRACT

Currently, no effective therapies exist for fibrodysplasia ossificans progressiva (FOP), a rare congenital syndrome in which heterotopic bone is formed in soft tissues owing to dysregulated activity of the bone morphogenetic protein (BMP) receptor kinase ALK2 (also known as ACVR1). From a screen of known biologically active compounds, we identified saracatinib as a potent ALK2 kinase inhibitor. In enzymatic and cell-based assays, saracatinib preferentially inhibited ALK2, compared with other receptors of the BMP/TGF-ß signaling pathway, and induced dorsalization in zebrafish embryos consistent with BMP antagonism. We further tested the efficacy of saracatinib using an inducible ACVR1Q207D-transgenic mouse line, which provides a model of heterotopic ossification (HO), as well as an inducible ACVR1R206H-knockin mouse, which serves as a genetically and physiologically faithful FOP model. In both models, saracatinib was well tolerated and potently inhibited the development of HO, even when administered transiently following soft tissue injury. Together, these data suggest that saracatinib is an efficacious clinical candidate for repositioning in FOP treatment, offering an accelerated path to clinical proof-of-efficacy studies and potentially significant benefits to individuals with this devastating condition.


Subject(s)
Activin Receptors, Type I/genetics , Benzodioxoles/pharmacology , Bone Morphogenetic Proteins/drug effects , Muscles/drug effects , Myositis Ossificans/genetics , Quinazolines/pharmacology , Activin Receptors, Type I/antagonists & inhibitors , Animals , Benzodioxoles/therapeutic use , Bone Morphogenetic Proteins/metabolism , Drug Evaluation, Preclinical , Gene Knock-In Techniques , Mice , Mice, Transgenic , Muscles/metabolism , Myositis Ossificans/metabolism , Myositis Ossificans/pathology , Ossification, Heterotopic/genetics , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/pathology , Quinazolines/therapeutic use , Zebrafish
2.
Sci Transl Med ; 8(366): 366ra163, 2016 11 23.
Article in English | MEDLINE | ID: mdl-27881824

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP), a congenital heterotopic ossification (HO) syndrome caused by gain-of-function mutations of bone morphogenetic protein (BMP) type I receptor ACVR1, manifests with progressive ossification of skeletal muscles, tendons, ligaments, and joints. In this disease, HO can occur in discrete flares, often triggered by injury or inflammation, or may progress incrementally without identified triggers. Mice harboring an Acvr1R206H knock-in allele recapitulate the phenotypic spectrum of FOP, including injury-responsive intramuscular HO and spontaneous articular, tendon, and ligament ossification. The cells that drive HO in these diverse tissues can be compartmentalized into two lineages: an Scx+ tendon-derived progenitor that mediates endochondral HO of ligaments and joints without exogenous injury, and a muscle-resident interstitial Mx1+ population that mediates intramuscular, injury-dependent endochondral HO. Expression of Acvr1R206H in either lineage confers aberrant gain of BMP signaling and chondrogenic differentiation in response to activin A and gives rise to mutation-expressing hypertrophic chondrocytes in HO lesions. Compared to Acvr1R206H, expression of the man-made, ligand-independent ACVR1Q207D mutation accelerates and increases the penetrance of all observed phenotypes, but does not abrogate the need for antecedent injury in muscle HO, demonstrating the need for an injury factor in addition to enhanced BMP signaling. Both injury-dependent intramuscular and spontaneous ligament HO in Acvr1R206H knock-in mice were effectively controlled by the selective ACVR1 inhibitor LDN-212854. Thus, diverse phenotypes of HO found in FOP are rooted in cell-autonomous effects of dysregulated ACVR1 signaling in nonoverlapping tissue-resident progenitor pools that may be addressed by systemic therapy or by modulating injury-mediated factors involved in their local recruitment.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Myxovirus Resistance Proteins/metabolism , Ossification, Heterotopic/metabolism , Stem Cells/cytology , Activin Receptors, Type I/genetics , Alleles , Animals , Bone Morphogenetic Proteins/metabolism , Cell Lineage , Disease Models, Animal , Female , Gene Knock-In Techniques , Genotype , Humans , Joints/metabolism , Ligaments/metabolism , Ligands , Male , Mice , Mice, Transgenic , Mutation , Phenotype
3.
J Med Chem ; 57(19): 7900-15, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25101911

ABSTRACT

There are currently no effective therapies for fibrodysplasia ossificans progressiva (FOP), a debilitating and progressive heterotopic ossification disease caused by activating mutations of ACVR1 encoding the BMP type I receptor kinase ALK2. Recently, a subset of these same mutations of ACVR1 have been identified in diffuse intrinsic pontine glioma (DIPG) tumors. Here we describe the structure-activity relationship for a series of novel ALK2 inhibitors based on the 2-aminopyridine compound K02288. Several modifications increased potency in kinase, thermal shift, or cell-based assays of BMP signaling and transcription, as well as selectivity for ALK2 versus closely related BMP and TGF-ß type I receptor kinases. Compounds in this series exhibited a wide range of in vitro cytotoxicity that was not correlated with potency or selectivity, suggesting mechanisms independent of BMP or TGF-ß inhibition. The study also highlights a potent 2-methylpyridine derivative 10 (LDN-214117) with a high degree of selectivity for ALK2 and low cytotoxicity that could provide a template for preclinical development. Contrary to the notion that activating mutations of ALK2 might alter inhibitor efficacy due to potential conformational changes in the ATP-binding site, the compounds demonstrated consistent binding to a panel of mutant and wild-type ALK2 proteins. Thus, BMP inhibitors identified via activity against wild-type ALK2 signaling are likely to be of clinical relevance for the diverse ALK2 mutant proteins associated with FOP and DIPG.


Subject(s)
Activin Receptors, Type I/antagonists & inhibitors , Aminopyridines/pharmacology , Mutation , Myositis Ossificans/drug therapy , Protein Kinase Inhibitors/pharmacology , Activin Receptors, Type I/genetics , Aminopyridines/chemical synthesis , Aminopyridines/metabolism , Humans , Myositis Ossificans/genetics , Phenols/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Structure-Activity Relationship
4.
PLoS One ; 8(4): e62721, 2013.
Article in English | MEDLINE | ID: mdl-23646137

ABSTRACT

Growth factor signaling pathways are tightly regulated by phosphorylation and include many important kinase targets of interest for drug discovery. Small molecule inhibitors of the bone morphogenetic protein (BMP) receptor kinase ALK2 (ACVR1) are needed urgently to treat the progressively debilitating musculoskeletal disease fibrodysplasia ossificans progressiva (FOP). Dorsomorphin analogues, first identified in zebrafish, remain the only BMP inhibitor chemotype reported to date. By screening an assay panel of 250 recombinant human kinases we identified a highly selective 2-aminopyridine-based inhibitor K02288 with in vitro activity against ALK2 at low nanomolar concentrations similar to the current lead compound LDN-193189. K02288 specifically inhibited the BMP-induced Smad pathway without affecting TGF-ß signaling and induced dorsalization of zebrafish embryos. Comparison of the crystal structures of ALK2 with K02288 and LDN-193189 revealed additional contacts in the K02288 complex affording improved shape complementarity and identified the exposed phenol group for further optimization of pharmacokinetics. The discovery of a new chemical series provides an independent pharmacological tool to investigate BMP signaling and offers multiple opportunities for pre-clinical development.


Subject(s)
Aminopyridines/pharmacology , Bone Morphogenetic Proteins/antagonists & inhibitors , Phenols/pharmacology , Signal Transduction/drug effects , Activin Receptors, Type I/antagonists & inhibitors , Activin Receptors, Type I/chemistry , Aminopyridines/chemistry , Animals , Body Patterning/drug effects , Bone Morphogenetic Proteins/chemistry , Bone Morphogenetic Proteins/metabolism , Humans , Models, Molecular , Molecular Conformation , Neovascularization, Physiologic/drug effects , Phenols/chemistry , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Smad Proteins/metabolism , Zebrafish
5.
ACS Chem Biol ; 8(6): 1291-302, 2013.
Article in English | MEDLINE | ID: mdl-23547776

ABSTRACT

The bone morphogenetic protein (BMP) signaling pathway has essential functions in development, homeostasis, and the normal and pathophysiologic remodeling of tissues. Small molecule inhibitors of the BMP receptor kinase family have been useful for probing physiologic functions of BMP signaling in vitro and in vivo and may have roles in the treatment of BMP-mediated diseases. Here we describe the development of a selective and potent inhibitor of the BMP type I receptor kinases, LDN-212854, which in contrast to previously described BMP receptor kinase inhibitors exhibits nearly 4 orders of selectivity for BMP versus the closely related TGF-ß and Activin type I receptors. In vitro, LDN-212854 exhibits some selectivity for ALK2 in preference to other BMP type I receptors, ALK1 and ALK3, which may permit the interrogation of ALK2-mediated signaling, transcriptional activity, and function. LDN-212854 potently inhibits heterotopic ossification in an inducible transgenic mutant ALK2 mouse model of fibrodysplasia ossificans progressiva. These findings represent a significant step toward developing selective inhibitors targeting individual members of the highly homologous BMP type I receptor family. Such inhibitors would provide greater resolution as probes of physiologic function and improved selectivity against therapeutic targets.


Subject(s)
Activin Receptors, Type I/antagonists & inhibitors , Bone Morphogenetic Protein Receptors, Type I/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Quinolines/chemistry , Quinolines/pharmacology , Activin Receptors, Type I/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cell Line , Humans , Mice , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...