Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
ChemMedChem ; 17(19): e202200303, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35950546

ABSTRACT

Inhibition of the YAP-TEAD protein-protein interaction is an attractive therapeutic concept under intense investigation with the objective to treat cancers associated with a dysregulation of the Hippo pathway. However, owing to the very extended surface of interaction of the two proteins, the identification of small drug-like molecules able to efficiently prevent YAP from binding to TEAD by direct competition has been elusive so far. We disclose here the discovery of the first class of small molecules potently inhibiting the YAP-TEAD interaction by binding at one of the main interaction sites of YAP at the surface of TEAD. These inhibitors, providing a path forward to pharmacological intervention in the Hippo pathway, evolved from a weakly active virtual screening hit advanced to high potency by structure-based design.


Subject(s)
Neoplasms , Transcription Factors , Adaptor Proteins, Signal Transducing/chemistry , Humans , Transcription Factors/metabolism , YAP-Signaling Proteins
2.
Nat Chem Biol ; 17(3): 280-290, 2021 03.
Article in English | MEDLINE | ID: mdl-33462494

ABSTRACT

Although most acute skin wounds heal rapidly, non-healing skin ulcers represent an increasing and substantial unmet medical need that urgently requires effective therapeutics. Keratinocytes resurface wounds to re-establish the epidermal barrier by transitioning to an activated, migratory state, but this ability is lost in dysfunctional chronic wounds. Small-molecule regulators of keratinocyte plasticity with the potential to reverse keratinocyte malfunction in situ could offer a novel therapeutic approach in skin wound healing. Utilizing high-throughput phenotypic screening of primary keratinocytes, we identify such small molecules, including bromodomain and extra-terminal domain (BET) protein family inhibitors (BETi). BETi induce a sustained activated, migratory state in keratinocytes in vitro, increase activation markers in human epidermis ex vivo and enhance skin wound healing in vivo. Our findings suggest potential clinical utility of BETi in promoting keratinocyte re-epithelialization of skin wounds. Importantly, this novel property of BETi is exclusively observed after transient low-dose exposure, revealing new potential for this compound class.


Subject(s)
Cell Cycle Proteins/genetics , Epidermis/drug effects , Re-Epithelialization/drug effects , Skin Ulcer/drug therapy , Small Molecule Libraries/pharmacology , Transcription Factors/genetics , Wounds, Nonpenetrating/drug therapy , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Disease Models, Animal , Epidermis/metabolism , Epidermis/pathology , Fluorescence Resonance Energy Transfer , Gene Expression Regulation , High-Throughput Screening Assays , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Mice , Mice, Inbred C57BL , Primary Cell Culture , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Precursors/antagonists & inhibitors , Protein Precursors/genetics , Protein Precursors/metabolism , Re-Epithelialization/genetics , Skin Ulcer/genetics , Skin Ulcer/metabolism , Skin Ulcer/pathology , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcription, Genetic , Wounds, Nonpenetrating/genetics , Wounds, Nonpenetrating/metabolism , Wounds, Nonpenetrating/pathology
3.
ChemMedChem ; 14(14): 1305-1314, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31066983

ABSTRACT

Hdm2 (human MDM2, human double minute 2 homologue) counteracts p53 function by direct binding to p53 and by ubiquitin-dependent p53 protein degradation. Activation of p53 by inhibitors of the p53-Hdm2 interaction is being pursued as a therapeutic strategy in p53 wild-type cancers. In addition, HdmX (human MDMX, human MDM4) was also identified as an important therapeutic target to efficiently reactivate p53, and it is likely that dual inhibition of Hdm2 and HdmX is beneficial. Herein we report four new X-ray structures for Hdm2 and five new X-ray structures for HdmX complexes, involving different classes of synthetic compounds (including the worldwide highest resolutions for Hdm2 and HdmX, at 1.13 and 1.20 Å, respectively). We also reveal the key additive 18-crown-ether, which we discovered to enable HdmX crystallization and show its stabilization of various Lys residues. In addition, we report the previously unpublished details of X-ray structure determinations for eight further Hdm2 complexes, including the clinical trial compounds NVP-CGM097 and NVP-HDM201. An analysis of all compound binding modes reveals new and deepened insight into the possible adaptations and structural states of Hdm2 (e.g., flip of F55, flip of Y67, reorientation of H96) and HdmX (e.g., flip of H55, dimer induction), enabling key binding interactions for different compound classes. To facilitate comparisons, we used the same numbering for Hdm2 (as in Q00987) and HdmX (as in O15151, but minus 1). Taken together, these structural insights should prove useful for the design and optimization of further selective and/or dual Hdm2/HdmX inhibitors.


Subject(s)
Cell Cycle Proteins/metabolism , Heterocyclic Compounds/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins/metabolism , Binding Sites , Cell Cycle Proteins/chemistry , Crystallography, X-Ray , Heterocyclic Compounds/chemistry , Humans , Protein Binding , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-mdm2/chemistry
4.
Bioorg Med Chem Lett ; 28(20): 3404-3408, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30217415

ABSTRACT

Small molecule inhibitors of the p53-MDM2 protein complex are under intense investigation in clinical trials as anti-cancer agents, including our first generation inhibitor NVP-CGM097. We recently described the rational design of a novel pyrazolopyrrolidinone core as a new lead structure and now we report on the synthesis and optimization of this to provide a highly potent lead compound. This new compound displayed excellent oral efficacy in our preclinical mechanistic in vivo model and marked a significant milestone towards the identification of our second generation clinical candidate NVP-HDM201.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrrolidinones/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Dogs , Haplorhini , Humans , Male , Mice , Microsomes, Liver/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Pyrrolidinones/chemical synthesis , Pyrrolidinones/chemistry , Pyrrolidinones/pharmacokinetics , Rats, Sprague-Dawley , Stereoisomerism
5.
Bioorg Med Chem Lett ; 26(19): 4837-4841, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27542305

ABSTRACT

The p53-MDM2 interaction is an anticancer drug target under investigation in the clinic. Our compound NVP-CGM097 is one of the small molecule inhibitors of this protein-protein interaction currently evaluated in cancer patients. As part of our effort to identify new classes of p53-MDM2 inhibitors that could lead to additional clinical candidates, we report here the design of highly potent inhibitors having a pyrazolopyrrolidinone core structure. The conception of these new inhibitors originated in a consideration on the MDM2 bound conformation of the dihydroisoquinolinone class of inhibitors to which NVP-CGM097 belongs. This work forms the foundation of the discovery of HDM201, a second generation p53-MDM2 inhibitor that recently entered phase I clinical trial.


Subject(s)
Drug Discovery , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/antagonists & inhibitors , Crystallography, X-Ray , Fluorescence Resonance Energy Transfer , Molecular Conformation , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism
6.
Bioorg Med Chem Lett ; 26(19): 4729-4734, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27575470

ABSTRACT

In vitro metabolic identification studies with a PI3K-α inhibitor lead molecule 1 identified a single predominant site of oxidative metabolism to be occurring within a tert.butyl moiety. Modification of the tert.butyl group within the lead molecule 1, to the corresponding d9-tert.butyl analogue 2, led to an increase in both the in vitro and in vivo metabolic stability. This increase in metabolic stability resulted in a 2-fold increase in the oral bioavailability measured in the rat, and a 3-fold increase in potency in a chronic in vivo study in the mouse, for 2 when compared to 1.


Subject(s)
Deuterium/metabolism , Enzyme Inhibitors/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Amides/chemistry , Animals , Biological Availability , Class I Phosphatidylinositol 3-Kinases , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Kinetics , Phosphoinositide-3 Kinase Inhibitors , Proline/chemistry , Rats , Thiazoles/chemistry , Urea/chemistry
7.
Bioorg Med Chem Lett ; 26(8): 2057-64, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26951753

ABSTRACT

Taking the pyrrolopyrimidine derived IGF-1R inhibitor NVP-AEW541 as the starting point, the benzyl ether back-pocket binding moiety was replaced with a series of 2-cyclic ether methyl ethers leading to the identification of novel achiral [2.2.1]-bicyclic ether methyl ether containing analogues with improved IGF-1R activities and kinase selectivities. Further exploration of the series, including a fluorine scan of the 5-phenyl substituent, and optimisation of the sugar-pocket binding moiety identified compound 33 containing (S)-2-tetrahydrofuran methyl ether 6-fluorophenyl ether back-pocket, and cis-N-Ac-Pip sugar-pocket binding groups. Compound 33 showed improved selectivity and pharmacokinetics compared to NVP-AEW541, and produced comparable in vivo efficacy to linsitinib in inhibiting the growth of an IGF-1R dependent tumour xenograft model in the mouse.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Mice , Mice, Nude , Molecular Structure , NIH 3T3 Cells , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrazines/chemical synthesis , Pyrazines/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Receptor, IGF Type 1/metabolism , Structure-Activity Relationship , Xenograft Model Antitumor Assays
8.
PLoS One ; 8(10): e77652, 2013.
Article in English | MEDLINE | ID: mdl-24204904

ABSTRACT

Malignant rhabdoid tumors (MRTs) are aggressive pediatric cancers arising in brain, kidney and soft tissues, which are characterized by loss of the tumor suppressor SNF5/SMARCB1. MRTs are poorly responsive to chemotherapy and thus a high unmet clinical need exists for novel therapies for MRT patients. SNF5 is a core subunit of the SWI/SNF chromatin remodeling complex which affects gene expression by nucleosome remodeling. Here, we report that loss of SNF5 function correlates with increased expression of fibroblast growth factor receptors (FGFRs) in MRT cell lines and primary tumors and that re-expression of SNF5 in MRT cells causes a marked repression of FGFR expression. Conversely, siRNA-mediated impairment of SWI/SNF function leads to elevated levels of FGFR2 in human fibroblasts. In vivo, treatment with NVP-BGJ398, a selective FGFR inhibitor, blocks progression of a murine MRT model. Hence, we identify FGFR signaling as an aberrantly activated oncogenic pathway in MRTs and propose pharmacological inhibition of FGFRs as a potential novel clinical therapy for MRTs.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Rhabdoid Tumor/metabolism , Transcription Factors/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Proliferation , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Fibroblasts/metabolism , HEK293 Cells , Humans , Mice , Promoter Regions, Genetic/genetics , Receptors, Fibroblast Growth Factor/genetics , Rhabdoid Tumor/genetics , SMARCB1 Protein , Signal Transduction/genetics , Transcription Factors/genetics
10.
J Bone Miner Res ; 28(4): 899-911, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23129509

ABSTRACT

Fibroblast growth factor 23 (FGF23) is a circulating factor secreted by osteocytes that is essential for phosphate homeostasis. In kidney proximal tubular cells FGF23 inhibits phosphate reabsorption and leads to decreased synthesis and enhanced catabolism of 1,25-dihydroxyvitamin D3 (1,25[OH]2 D3 ). Excess levels of FGF23 cause renal phosphate wasting and suppression of circulating 1,25(OH)2 D3 levels and are associated with several hereditary hypophosphatemic disorders with skeletal abnormalities, including X-linked hypophosphatemic rickets (XLH) and autosomal recessive hypophosphatemic rickets (ARHR). Currently, therapeutic approaches to these diseases are limited to treatment with activated vitamin D analogues and phosphate supplementation, often merely resulting in partial correction of the skeletal aberrations. In this study, we evaluate the use of FGFR inhibitors for the treatment of FGF23-mediated hypophosphatemic disorders using NVP-BGJ398, a novel selective, pan-specific FGFR inhibitor currently in Phase I clinical trials for cancer therapy. In two different hypophosphatemic mouse models, Hyp and Dmp1-null mice, resembling the human diseases XLH and ARHR, we find that pharmacological inhibition of FGFRs efficiently abrogates aberrant FGF23 signaling and normalizes the hypophosphatemic and hypocalcemic conditions of these mice. Correspondingly, long-term FGFR inhibition in Hyp mice leads to enhanced bone growth, increased mineralization, and reorganization of the disturbed growth plate structure. We therefore propose NVP-BGJ398 treatment as a novel approach for the therapy of FGF23-mediated hypophosphatemic diseases.


Subject(s)
Fibroblast Growth Factors/metabolism , Phenylurea Compounds/pharmacology , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Rickets, Hypophosphatemic/metabolism , Signal Transduction/drug effects , Animals , Body Weight/drug effects , Bone Development/drug effects , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/metabolism , Femur/drug effects , Femur/pathology , Fibroblast Growth Factor-23 , Growth Plate/drug effects , Growth Plate/pathology , Homeostasis/drug effects , Ions , Kidney/drug effects , Kidney/metabolism , Mice, Inbred C57BL , Minerals/metabolism , Phenylurea Compounds/therapeutic use , Pyrimidines/therapeutic use , Receptors, Fibroblast Growth Factor/metabolism , Rickets, Hypophosphatemic/drug therapy , Rickets, Hypophosphatemic/pathology , Tail/anatomy & histology , Vitamin D/analogs & derivatives , Vitamin D/biosynthesis
11.
Cancer Discov ; 2(12): 1118-33, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23002168

ABSTRACT

UNLABELLED: Patient stratification biomarkers that enable the translation of cancer genetic knowledge into clinical use are essential for the successful and rapid development of emerging targeted anticancer therapeutics. Here, we describe the identification of patient stratification biomarkers for NVP-BGJ398, a novel and selective fibroblast growth factor receptor (FGFR) inhibitor. By intersecting genome-wide gene expression and genomic alteration data with cell line-sensitivity data across an annotated collection of cancer cell lines called the Cancer Cell Line Encyclopedia, we show that genetic alterations for FGFR family members predict for sensitivity to NVP-BGJ398. For the first time, we report oncogenic FGFR1 amplification in osteosarcoma as a potential patient selection biomarker. Furthermore, we show that cancer cell lines harboring FGF19 copy number gain at the 11q13 amplicon are sensitive to NVP-BGJ398 only when concomitant expression of ß-klotho occurs. Thus, our findings provide the rationale for the clinical development of FGFR inhibitors in selected patients with cancer harboring tumors with the identified predictors of sensitivity. SIGNIFICANCE: The success of a personalized medicine approach using targeted therapies ultimately depends on being able to identify the patients who will benefit the most from any given drug. To this end, we have integrated the molecular profiles for more than 500 cancer cell lines with sensitivity data for the novel anticancer drug NVP-BGJ398 and showed that FGFR genetic alterations are the most significant predictors for sensitivity. This work has ultimately endorsed the incorporation of specific patient selection biomakers in the clinical trials for NVP-BGJ398.


Subject(s)
Neoplasms/drug therapy , Neoplasms/enzymology , Phenylurea Compounds/pharmacology , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/genetics , Animals , Cell Line, Tumor , Gene Amplification/drug effects , HEK293 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Mice , Models, Molecular , Neoplasms/genetics , Neoplasms/pathology , Phenylurea Compounds/chemistry , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemistry , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , Xenograft Model Antitumor Assays
12.
Cancer Discov ; 2(10): 948-59, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22874768

ABSTRACT

The overall power of kinase inhibitors is substantially overshadowed by the acquisition of drug resistance. To address this issue, we systematically assessed the potential of secreted proteins to induce resistance to kinase inhibitors. To this end, we developed a high-throughput platform for screening a cDNA library encoding 3,432 secreted proteins in cellular assays. Using cancer cells originally dependent on either MET, FGFR2, or FGFR3, we observed a bypass of dependence through ligand-mediated activation of alternative receptor tyrosine kinases (RTK). Our findings indicate a broad and versatile potential for RTKs from the HER and FGFR families as well as MET to compensate for loss of each other. We further provide evidence that combined inhibition of simultaneously active RTKs can lead to an added anticancer effect.


Subject(s)
Neoplasms/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-2/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , High-Throughput Screening Assays , Humans , Mice , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/genetics , Signal Transduction/drug effects , Transplantation, Heterologous
13.
J Med Chem ; 54(20): 7066-83, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21936542

ABSTRACT

A novel series of N-aryl-N'-pyrimidin-4-yl ureas has been optimized to afford potent and selective inhibitors of the fibroblast growth factor receptor tyrosine kinases 1, 2, and 3 by rationally designing the substitution pattern of the aryl ring. On the basis of its in vitro profile, compound 1h (NVP-BGJ398) was selected for in vivo evaluation and showed significant antitumor activity in RT112 bladder cancer xenografts models overexpressing wild-type FGFR3. These results support the potential therapeutic use of 1h as a new anticancer agent.


Subject(s)
Antineoplastic Agents/chemical synthesis , Phenylurea Compounds/chemical synthesis , Pyrimidines/chemical synthesis , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Female , Humans , Mice , Mice, Nude , Models, Molecular , Neoplasm Transplantation , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/pharmacology , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Rats, Wistar , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Structure-Activity Relationship , Transplantation, Heterologous , Urinary Bladder Neoplasms
14.
Bioorg Med Chem Lett ; 18(3): 897-900, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18248988

ABSTRACT

A pyrimidin-4-yl-urea motif forming a pseudo ring by intramolecular hydrogen bonding has been designed to mimic the pyrido[2,3-d]pyrimidin-7-one core structure of a well-established class of protein kinase inhibitors. Potent inhibition of a number of protein kinases was obtained with the first prototype compound synthesized to probe the design concept.


Subject(s)
Drug Design , Models, Molecular , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Pyridones/chemistry , Pyridones/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Urea/analogs & derivatives , Urea/chemical synthesis , Urea/pharmacology , Amino Acids/chemistry , Amino Acids/pharmacology , Molecular Conformation , Molecular Mimicry , Molecular Structure , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemistry , Structure-Activity Relationship , Urea/chemistry
15.
Bioorg Med Chem Lett ; 17(2): 358-62, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17095212

ABSTRACT

A series of beta-lactam derivatives has been designed and synthesized to inhibit the chymotrypsin-like activity of the human 20S proteasome. The most potent compounds of this new structural class of beta-subunit selective 20S proteasome inhibitors exhibit IC50 values in the low-nanomolar range and show good selectivity over the trypsin-like and post-glutamyl-peptide hydrolytic activities of the enzyme.


Subject(s)
Chymotrypsin/antagonists & inhibitors , Proteasome Inhibitors , Trypsin Inhibitors/chemical synthesis , Trypsin Inhibitors/pharmacology , beta-Lactams/chemical synthesis , beta-Lactams/pharmacology , Crystallography, X-Ray , Drug Design , Humans , Models, Molecular , Peptides/chemistry , Structure-Activity Relationship
16.
J Med Chem ; 49(15): 4451-4, 2006 Jul 27.
Article in English | MEDLINE | ID: mdl-16854049

ABSTRACT

FLT3 kinase inhibitors are currently under investigation as a new treatment for acute myeloid leukemia. We report here a molecular concept invoking interactions between an aromatic ring and the side chains of Phe691 and Cys828, two residues of the ATP pocket, to obtain potent and specific inhibitors of this kinase. The hypothesis has been validated by the successful design of a new inhibitor prototype showing promising antiproliferative activity in cellular assays.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cysteine/chemistry , Phenylalanine/chemistry , Thiazoles/chemical synthesis , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/chemistry , Acute Disease , Adenosine Triphosphate/chemistry , Antineoplastic Agents/chemistry , Binding Sites , Cell Line, Tumor , Humans , Leukemia, Myeloid , Models, Molecular , Mutation , Protein Structure, Tertiary , Thiazoles/chemistry , Thiazoles/pharmacology , fms-Like Tyrosine Kinase 3/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...