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










Publication year range
1.
Nat Commun ; 13(1): 3372, 2022 06 11.
Article in English | MEDLINE | ID: mdl-35690592

ABSTRACT

Glycogen is the major glucose reserve in eukaryotes, and defects in glycogen metabolism and structure lead to disease. Glycogenesis involves interaction of glycogenin (GN) with glycogen synthase (GS), where GS is activated by glucose-6-phosphate (G6P) and inactivated by phosphorylation. We describe the 2.6 Å resolution cryo-EM structure of phosphorylated human GS revealing an autoinhibited GS tetramer flanked by two GN dimers. Phosphorylated N- and C-termini from two GS protomers converge near the G6P-binding pocket and buttress against GS regulatory helices. This keeps GS in an inactive conformation mediated by phospho-Ser641 interactions with a composite "arginine cradle". Structure-guided mutagenesis perturbing interactions with phosphorylated tails led to increased basal/unstimulated GS activity. We propose that multivalent phosphorylation supports GS autoinhibition through interactions from a dynamic "spike" region, allowing a tuneable rheostat for regulating GS activity. This work therefore provides insights into glycogen synthesis regulation and facilitates studies of glycogen-related diseases.


Subject(s)
Glucosyltransferases , Glycogen Synthase , Glucose-6-Phosphate/metabolism , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Glycogen/metabolism , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Glycoproteins/metabolism , Humans , Muscle, Skeletal/metabolism , Phosphorylation
2.
Bioorg Med Chem ; 23(10): 2387-407, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25882525

ABSTRACT

Compound 1, a hit from the screening of our chemical collection displaying activity against JAK2, was deconstructed for SAR analysis into three regions, which were explored. A series of compounds was synthesized leading to the identification of the potent and orally bioavailable JAK2 inhibitor 16 (NMS-P830), which showed an encouraging tumour growth inhibition in SET-2 xenograft tumour model, with evidence for JAK2 pathway suppression demonstrated by in vivo pharmacodynamic effects.


Subject(s)
Amides/chemical synthesis , Antineoplastic Agents/chemical synthesis , Janus Kinase 2/antagonists & inhibitors , Leukemia, Megakaryoblastic, Acute/drug therapy , Protein Kinase Inhibitors/chemical synthesis , Pyrroles/chemical synthesis , Amides/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression , High-Throughput Screening Assays , Humans , Janus Kinase 2/chemistry , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Leukemia, Megakaryoblastic, Acute/enzymology , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/pathology , Megakaryocyte Progenitor Cells/drug effects , Megakaryocyte Progenitor Cells/enzymology , Megakaryocyte Progenitor Cells/pathology , Mice , Mice, Nude , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Structure-Activity Relationship , Xenograft Model Antitumor Assays
3.
Bioorg Med Chem ; 22(17): 4998-5012, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25009002

ABSTRACT

We report herein the discovery, structure guided design, synthesis and biological evaluation of a novel class of JAK2 inhibitors. Optimization of the series led to the identification of the potent and orally bioavailable JAK2 inhibitor 28 (NMS-P953). Compound 28 displayed significant tumour growth inhibition in SET-2 xenograft tumour model, with a mechanism of action confirmed in vivo by typical modulation of known biomarkers, and with a favourable pharmacokinetic and safety profile.


Subject(s)
Antineoplastic Agents/pharmacology , Janus Kinase 2/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Janus Kinase 2/metabolism , Mice , Mice, SCID , Models, Molecular , Molecular Structure , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Structure-Activity Relationship , Substrate Specificity
4.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 5): 1465-75, 2014 May.
Article in English | MEDLINE | ID: mdl-24816114

ABSTRACT

Bloom's syndrome is an autosomal recessive genome-instability disorder associated with a predisposition to cancer, premature aging and developmental abnormalities. It is caused by mutations that inactivate the DNA helicase activity of the BLM protein or nullify protein expression. The BLM helicase has been implicated in the alternative lengthening of telomeres (ALT) pathway, which is essential for the limitless replication of some cancer cells. This pathway is used by 10-15% of cancers, where inhibitors of BLM are expected to facilitate telomere shortening, leading to apoptosis or senescence. Here, the crystal structure of the human BLM helicase in complex with ADP and a 3'-overhang DNA duplex is reported. In addition to the helicase core, the BLM construct used for crystallization (residues 640-1298) includes the RecQ C-terminal (RQC) and the helicase and ribonuclease D C-terminal (HRDC) domains. Analysis of the structure provides detailed information on the interactions of the protein with DNA and helps to explain the mechanism coupling ATP hydrolysis and DNA unwinding. In addition, mapping of the missense mutations onto the structure provides insights into the molecular basis of Bloom's syndrome.


Subject(s)
Adenosine Diphosphate/metabolism , RecQ Helicases/chemistry , RecQ Helicases/metabolism , Bloom Syndrome/genetics , Catalytic Domain , Crystallography, X-Ray , Humans , Hydrolysis , Models, Molecular , Mutation , Nucleic Acid Heteroduplexes , Protein Conformation , RecQ Helicases/genetics
5.
Biochemistry ; 52(37): 6380-7, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23914841

ABSTRACT

Maternal embryonic leucine zipper kinase (MELK) is upregulated in several types of tumor, including breast, prostate, and brain tumors. Its expression is generally associated with cell survival, cell proliferation, and resistance to apoptosis. Therefore, the potential of MELK inhibitors as therapeutic agents is recently attracting considerable interest. Here we report the first structures of MELK in complex with AMP-PNP and with nanomolar inhibitors. Our studies shed light on the role of the MELK UBA domain, provide a characterization of the kinase active site, and identify key residues for achieving high potency, laying the groundwork for structure-based drug design efforts.


Subject(s)
Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Adenylyl Imidodiphosphate/pharmacology , Antineoplastic Agents/pharmacology , Catalytic Domain , Cell Line, Tumor , Drug Design , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , Pyrazoles/chemistry , Pyrazoles/pharmacology
6.
J Med Chem ; 56(2): 437-50, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23245311

ABSTRACT

Valosine containing protein (VCP), also known as p97, is a member of AAA ATPase family that is involved in several biological processes and plays a central role in the ubiquitin-mediated degradation of misfolded proteins. VCP is an ubiquitously expressed, highly abundant protein and has been found overexpressed in many tumor types, sometimes associated with poor prognosis. In this respect, VCP has recently received a great deal of attention as a potential new target for cancer therapy. In this paper, the discovery and structure-activity relationships of alkylsulfanyl-1,2,4-triazoles, a new class of potent, allosteric VCP inhibitors, are described. Medicinal chemistry manipulation of compound 1, identified via HTS, led to the discovery of potent and selective inhibitors with submicromolar activity in cells and clear mechanism of action at consistent doses. This represents a first step toward a new class of potential anticancer agents.


Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , Cell Cycle Proteins/antagonists & inhibitors , Triazoles/pharmacology , Adenosine Triphosphatases/chemistry , Allosteric Regulation , Cell Cycle Proteins/chemistry , Humans , Neoplasms/pathology , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry , Valosin Containing Protein
7.
Bioorg Med Chem Lett ; 22(12): 3946-50, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22608961

ABSTRACT

A series of phenylacylsulfonamides has been prepared as antagonists of Bcl-2/Bcl-xL. In addition to potent binding affinities for both Bcl-2 and Bcl-xL, these compounds were shown to induce classical markers of apoptosis in isolated mitochondria. Overall weak cellular potency was improved by the incorporation of polar functionality resulting in compounds with moderate antiproliferative activity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Mitochondria/drug effects , Sulfonamides/chemical synthesis , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-X Protein/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Crystallography, X-Ray , Cytochromes c/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Mitochondria/metabolism , Models, Molecular , Sulfonamides/pharmacology , bcl-2-Associated X Protein/chemistry , bcl-X Protein/chemistry
8.
Bioorg Med Chem Lett ; 22(1): 96-101, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22154349

ABSTRACT

The discovery and characterization of two new chemical classes of potent and selective Polo-like kinase 1 (PLK1) inhibitors is reported. For the most interesting compounds, we discuss the biological activities, crystal structures and preliminary pharmacokinetic parameters. The more advanced compounds inhibit PLK1 in the enzymatic assay at the nM level and exhibit good activity in cell proliferation on A2780 cells. Furthermore, these compounds showed high levels of selectivity on a panel of unrelated kinases, as well as against PLK2 and PLK3 isoforms. Additionally, the compounds show acceptable oral bioavailability in mice making these inhibitors suitable candidates for further in vivo activity studies.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pyridones/chemistry , Pyrimidines/pharmacology , Pyrroles/chemistry , Administration, Oral , Algorithms , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry, Pharmaceutical/methods , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor/methods , Enzymes/chemistry , Humans , Mice , Models, Chemical , Protein Isoforms , Pyridones/pharmacology , Pyrimidines/chemical synthesis , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Tumor Suppressor Proteins , Polo-Like Kinase 1
9.
J Biol Chem ; 286(52): 44811-20, 2011 Dec 30.
Article in English | MEDLINE | ID: mdl-22006928

ABSTRACT

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are dually activated by hyperpolarization and binding of cAMP to their cyclic nucleotide binding domain (CNBD). HCN isoforms respond differently to cAMP; binding of cAMP shifts activation of HCN2 and HCN4 by 17 mV but shifts that of HCN1 by only 2-4 mV. To explain the peculiarity of HCN1, we solved the crystal structures and performed a biochemical-biophysical characterization of the C-terminal domain (C-linker plus CNBD) of the three isoforms. Our main finding is that tetramerization of the C-terminal domain of HCN1 occurs at basal cAMP concentrations, whereas those of HCN2 and HCN4 require cAMP saturating levels. Therefore, HCN1 responds less markedly than HCN2 and HCN4 to cAMP increase because its CNBD is already partly tetrameric. This is confirmed by voltage clamp experiments showing that the right-shifted position of V(½) in HCN1 is correlated with its propensity to tetramerize in vitro. These data underscore that ligand-induced CNBD tetramerization removes tonic inhibition from the pore of HCN channels.


Subject(s)
Cyclic AMP/metabolism , Ion Channel Gating/physiology , Ion Channels/chemistry , Ion Channels/metabolism , Protein Multimerization/physiology , Animals , Cyclic AMP/chemistry , Cyclic AMP/genetics , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/genetics , Oocytes , Potassium Channels , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Xenopus laevis
10.
Biochem J ; 436(2): 331-9, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21410432

ABSTRACT

CARM1 (co-activator-associated arginine methyltransferase 1) is a PRMT (protein arginine N-methyltransferase) family member that catalyses the transfer of methyl groups from SAM (S-adenosylmethionine) to the side chain of specific arginine residues of substrate proteins. This post-translational modification of proteins regulates a variety of transcriptional events and other cellular processes. Moreover, CARM1 is a potential oncological target due to its multiple roles in transcription activation by nuclear hormone receptors and other transcription factors such as p53. Here, we present crystal structures of the CARM1 catalytic domain in complex with cofactors [SAH (S-adenosyl-L-homocysteine) or SNF (sinefungin)] and indole or pyazole inhibitors. Analysis of the structures reveals that the inhibitors bind in the arginine-binding cavity and the surrounding pocket that exists at the interface between the N- and C-terminal domains. In addition, we show using ITC (isothermal titration calorimetry) that the inhibitors bind to the CARM1 catalytic domain only in the presence of the cofactor SAH. Furthermore, sequence differences for select residues that interact with the inhibitors may be responsible for the CARM1 selectivity against PRMT1 and PRMT3. Together, the structural and biophysical information should aid in the design of both potent and specific inhibitors of CARM1.


Subject(s)
Indoles/antagonists & inhibitors , Indoles/chemistry , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/chemistry , Pyrazoles/antagonists & inhibitors , Pyrazoles/chemistry , Amino Acid Sequence , Catalytic Domain/drug effects , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Indoles/metabolism , Molecular Sequence Data , Protein Binding/drug effects , Protein-Arginine N-Methyltransferases/metabolism , Pyrazoles/metabolism
11.
Cancer Res ; 70(24): 10255-64, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21159646

ABSTRACT

MPS1 kinase is a key regulator of the spindle assembly checkpoint (SAC), a mitotic mechanism specifically required for proper chromosomal alignment and segregation. It has been found aberrantly overexpressed in a wide range of human tumors and is necessary for tumoral cell proliferation. Here we report the identification and characterization of NMS-P715, a selective and orally bioavailable MPS1 small-molecule inhibitor, which selectively reduces cancer cell proliferation, leaving normal cells almost unaffected. NMS-P715 accelerates mitosis and affects kinetochore components localization causing massive aneuploidy and cell death in a variety of tumoral cell lines and inhibits tumor growth in preclinical cancer models. Inhibiting the SAC could represent a promising new approach to selectively target cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Mitosis/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Quinazolines/pharmacology , Spindle Apparatus/drug effects , Aneuploidy , Animals , Antineoplastic Agents/chemistry , Cell Cycle Proteins/chemistry , Cell Growth Processes/drug effects , HCT116 Cells , HeLa Cells , Humans , Mice , Mice, Nude , Models, Molecular , Molecular Targeted Therapy/methods , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/chemistry , Protein-Tyrosine Kinases , Xenograft Model Antitumor Assays
12.
Bioorg Med Chem ; 18(19): 7113-20, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20817473

ABSTRACT

A novel series of 3-amino-1H-thieno[3,2-c]pyrazole derivatives demonstrating high potency in inhibiting Aurora kinases was developed. Here we describe the synthesis and a preliminary structure-activity relationship, which led to the discovery of a representative compound (38), which showed low nanomolar inhibitory activity in the anti-proliferation assay and was able to block the cell cycle in HCT-116 cell line. This compound demonstrated favorable pharmacokinetic properties and good efficacy in the HL-60 xenograft tumor model.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Thiophenes/pharmacology , Animals , Antineoplastic Agents/chemistry , Aurora Kinases , Cell Cycle/drug effects , Cell Proliferation/drug effects , Computational Biology , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , HL-60 Cells , Humans , Male , Mice , Mice, SCID , Models, Molecular , Molecular Dynamics Simulation , Molecular Structure , Neoplasms, Experimental/drug therapy , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Stereoisomerism , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry , Transplantation, Heterologous
13.
Biochemistry ; 49(32): 6813-25, 2010 Aug 17.
Article in English | MEDLINE | ID: mdl-20695522

ABSTRACT

Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase involved in the development of several human cancers and, as a result, is a recognized target for the development of small-molecule inhibitors for the treatment of ALK-positive malignancies. Here, we present the crystal structures of the unphosphorylated human ALK kinase domain in complex with the ATP competitive ligands PHA-E429 and NVP-TAE684. Analysis of these structures provides valuable information concerning the specific characteristics of the ALK active site as well as giving indications about how to obtain selective ALK inhibitors. In addition, the ALK-KD-PHA-E429 structure led to the identification of a potential regulatory mechanism involving a link made between a short helical segment immediately following the DFG motif and an N-terminal two-stranded beta-sheet. Finally, mapping of the activating mutations associated with neuroblastoma onto our structures may explain the roles these residues have in the activation process.


Subject(s)
Models, Molecular , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/chemistry , Pyrimidines/chemistry , Anaplastic Lymphoma Kinase , Animals , Cell Line , Enzyme Inhibitors/chemistry , Humans , Protein Structure, Tertiary , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases , Spodoptera
14.
J Med Chem ; 53(9): 3532-51, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20397705

ABSTRACT

Polo-like kinase 1 (Plk1) is a fundamental regulator of mitotic progression whose overexpression is often associated with oncogenesis and therefore is recognized as an attractive therapeutic target in the treatment of proliferative diseases. Here we discuss the structure-activity relationship of the 4,5-dihydro-1H-pyrazolo[4,3-h]quinazoline class of compounds that emerged from a high throughput screening (HTS) campaign as potent inhibitors of Plk1 kinase. Furthermore, we describe the discovery of 49, 8-{[2-methoxy-5-(4-methylpiperazin-1-yl)phenyl]amino}-1-methyl-4,5-dihydro-1H-pyrazolo[4,3-h]quinazoline-3-carboxamide, as a highly potent and specific ATP mimetic inhibitor of Plk1 (IC(50) = 0.007 microM) as well as its crystal structure in complex with the methylated Plk1(36-345) construct. Compound 49 was active in cell proliferation against different tumor cell lines with IC(50) values in the submicromolar range and active in vivo in the HCT116 xenograft model where it showed 82% tumor growth inhibition after repeated oral administration.


Subject(s)
Antineoplastic Agents/chemistry , Cell Cycle Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Quinazolines/pharmacology , Adenosine Triphosphate , Administration, Oral , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Humans , Molecular Mimicry , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Quinazolines/chemistry , Quinazolines/therapeutic use , Structure-Activity Relationship , Tumor Burden , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
15.
J Med Chem ; 52(2): 293-307, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-19115845

ABSTRACT

Cdc7 kinase is a key regulator of the S-phase of the cell cycle, known to promote the activation of DNA replication origins in eukaryotic organisms. Cdc7 inhibition can cause tumor-cell death in a p53-independent manner, supporting the rationale for developing Cdc7 inhibitors for the treatment of cancer. In this paper, we conclude the structure-activity relationships study of the 2-heteroaryl-pyrrolopyridinone class of compounds that display potent inhibitory activity against Cdc7 kinase. Furthermore, we also describe the discovery of 89S, [(S)-2-(2-aminopyrimidin-4-yl)-7-(2-fluoro-ethyl)-1,5,6,7-tetrahydropyrrolo[3,2-c]pyridin-4-one], as a potent ATP mimetic inhibitor of Cdc7. Compound 89S has a Ki value of 0.5 nM, inhibits cell proliferation of different tumor cell lines with an IC50 in the submicromolar range, and exhibits in vivo tumor growth inhibition of 68% in the A2780 xenograft model.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridones/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Chromatography, High Pressure Liquid , Dogs , Drug Discovery , Humans , Magnetic Resonance Spectroscopy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Pyridones/chemistry , Pyridones/pharmacokinetics , Rats , Rats, Wistar , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Ultraviolet , Structure-Activity Relationship
16.
J Med Chem ; 49(5): 1562-75, 2006 Mar 09.
Article in English | MEDLINE | ID: mdl-16509574

ABSTRACT

A novel class of highly selective inhibitors of p38 MAP kinase was discovered from high throughput screening. The synthesis and optimization of a series of 5-amino-N-phenyl-1H-pyrazol-4-yl-3-phenylmethanones is described. An X-ray crystal structure of this series bound in the ATP binding pocket of unphosphorylated p38alpha established the presence of a unique hydrogen bond between the exocyclic amine of the inhibitor and threonine 106 which likely contributes to the selectivity for p38. The crystallographic information was used to optimize the potency and physicochemical properties of the series. The incorporation of the 2,3-dihydroxypropoxy moiety on the pyrazole scaffold resulted in a compound with excellent drug-like properties including high oral bioavailability. These efforts identified 63 (RO3201195) as an orally bioavailable and highly selective inhibitor of p38 which was selected for advancement into Phase I clinical trials.


Subject(s)
Anti-Inflammatory Agents/chemical synthesis , Pyrazoles/chemical synthesis , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Adenosine Triphosphate/chemistry , Administration, Oral , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/drug therapy , Binding Sites , Biological Availability , Cell Line , Crystallography, X-Ray , Dogs , Female , Haplorhini , Humans , Interleukin-1/antagonists & inhibitors , Interleukin-1/biosynthesis , Interleukin-6/antagonists & inhibitors , Interleukin-6/biosynthesis , Models, Molecular , Pyrazoles/chemistry , Pyrazoles/pharmacology , Rats , Rats, Inbred Lew , Stereoisomerism , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/chemistry
17.
J Chem Inf Model ; 45(5): 1282-90, 2005.
Article in English | MEDLINE | ID: mdl-16180905

ABSTRACT

An evaluation and comparison of two different approaches, GRID/CPCA and GRIND/CPCA (CPCA = consensus principal component analysis; GRIND = GRid-INdependent Descriptors), suitable for visualizing the structural differences between related proteins is presented. Ten crystal structures of CDK2/cyclin A and GSK3beta solved in-house with different inhibitors were compared with the aim of highlighting regions that could be potential sites for gaining selectivity for CDK2 versus GSK3beta. The analyses pointed out remarkable differences in the backs of the CDK2-GSK3beta ATP binding pockets that guided the optimization toward a selective benzodipyrazole CDK2 inhibitor. The gain in selectivity can be associated with the two main differences in the ATP pocket between the enzymes. Phe80 of CDK2, the so-called gatekeeper residue often exploited for the design of kinase selective ligands, is replaced by a leucine in GSK3beta, and Ala144 is replaced by a cysteine. As a consequence of these mutations, CDK2 has a less elongated and less flat buried region at the back of the ATP pocket.


Subject(s)
Computational Biology/methods , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/metabolism , Glycogen Synthase Kinase 3/chemistry , Glycogen Synthase Kinase 3/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Crystallization , Crystallography, X-Ray , Cyclin A/chemistry , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/genetics , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Models, Molecular , Principal Component Analysis , Protein Binding , Protein Conformation , Sensitivity and Specificity , Software , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...