Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 15(6): 864-872, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38894924

ABSTRACT

We were attracted to the therapeutic potential of inhibiting Casitas B-lineage lymphoma proto-oncogene-b (Cbl-b), a RING E3 ligase that plays a critical role in regulating the activation of T cells. However, given that only protein-protein interactions were involved, it was unclear whether inhibition by a small molecule would be a viable approach. After screening an ∼6 billion member DNA-encoded library (DEL) using activated Cbl-b, we identified compound 1 as a hit for which the cis-isomer (2) was confirmed by biochemical and surface plasmon resonance (SPR) assays. Our hit optimization effort was greatly accelerated when we obtained a cocrystal structure of 2 with Cbl-b, which demonstrated induced binding at the substrate binding site, namely, the Src homology-2 (SH2) domain. This was quite noteworthy given that there are few reports of small molecule inhibitors that bind to SH2 domains and block protein-protein interactions. Structure- and property-guided optimization led to compound 27, which demonstrated measurable cell activity, albeit only at high concentrations.

2.
Biochem Soc Trans ; 48(4): 1323-1336, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32794575

ABSTRACT

The proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates plasma LDL cholesterol levels by binding to the liver LDL receptor (LDLR) and promoting its degradation. Therefore, PCSK9 has become a compelling new therapeutic target for lipid lowering and the prevention of cardiovascular disease. PCSK9 contains two regions of conformational flexibility, the N-terminal regions of the prodomain and of the catalytic domain. The recognition that the latter region, the so-called P' helix, is able to transition from an α-helical to a disordered state gave rise to new strategies to develop small molecule inhibitors of PCSK9 for lipid lowering. In the ordered state the P' helix is buried in a groove of the PCSK9 catalytic domain located next to the main LDLR binding site. The transition to a disordered state leaves the groove site vacated and accessible for compounds to antagonize LDLR binding. By use of a groove-directed phage display strategy we were able to identify several groove-binding peptides. Based on structural information of PCSK9-peptide complexes, a minimized groove-binding peptide was generated and utilized as an anchor to extend towards the adjacent main LDLR binding site, either by use of a phage-displayed peptide extension library, or by appending organic moieties to yield organo-peptides. Both strategies led to antagonists with pharmacologic activities in cell-based assays. The intricate bipartite mechanism of the potent organo-peptide inhibitors was revealed by structural studies, showing that the core peptide occupies the N-terminal groove, while the organic moiety interacts with the LDLR binding site to create antagonism. These findings validate the PCSK9 groove as an attractive target site and should inspire the development of a new class of small molecule antagonists of PCSK9.


Subject(s)
Anticholesteremic Agents/chemistry , Cholesterol, LDL/blood , Drug Design , Proprotein Convertase 9/metabolism , Serine Proteinase Inhibitors/chemistry , Animals , Anticholesteremic Agents/pharmacology , Binding Sites , Humans , PCSK9 Inhibitors , Proprotein Convertase 9/chemistry , Receptors, LDL/metabolism , Serine Proteinase Inhibitors/pharmacology
3.
ACS Chem Biol ; 15(6): 1392-1400, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32302100

ABSTRACT

Ubiquitin specific protease 7 (USP7) regulates the protein stability of key cellular regulators in pathways ranging from apoptosis to neuronal development, making it a promising therapeutic target. Here we used an engineered, activated variant of the USP7 catalytic domain to perform structure-activity studies of electrophilic peptidomimetic inhibitors. Employing this USP7 variant, we found that inhibitors with a cyanopyrrolidine warhead unexpectedly promoted a ß-elimination reaction of the initial covalent adducts, thereby converting the active-site cysteine residue to dehydroalanine. We determined that this phenomenon is specific for the USP7 catalytic cysteine and that structural features of the inhibitor and protein microenvironment impact elimination rates. Using comprehensive docking studies, we propose that the characteristic conformational dynamics of USP7 allow access to conformations that promote the ligand-induced elimination. Unlike in conventional reversible-covalent inhibition, the compounds described here irreversibly destroy a catalytic residue while simultaneously converting the inhibitor to a nonelectrophilic byproduct. Accordingly, this unexpected finding expands the scope of covalent inhibitor modalities and offers intriguing insights into enzyme-inhibitor dynamics.


Subject(s)
Catalytic Domain/drug effects , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Cysteine/chemistry , Cysteine/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Molecular Docking Simulation , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Ubiquitin-Specific Peptidase 7/chemistry , Ubiquitin-Specific Peptidase 7/metabolism
4.
ACS Chem Biol ; 15(2): 425-436, 2020 02 21.
Article in English | MEDLINE | ID: mdl-31962046

ABSTRACT

Proprotein convertase subtilisin/kexin 9 (PCSK9) has become an important therapeutic target for lipid lowering, since it regulates low-density lipoprotein cholesterol (LDL-c) levels by binding to liver LDL receptors (LDLR) and effecting their intracellular degradation. However, the development of small molecule inhibitors is hampered by the lack of attractive PCSK9 target sites. We recently discovered helical peptides that are able to bind to a cryptic groove site on PCSK9, which is situated in proximity to the main LDLR binding site. Here, we designed potent bipartite PCSK9 inhibitors by appending organic moieties to a helical groove-binding peptide to reach a hydrophobic pocket in the proximal LDLR binding region. The ultimately designed 1-amino-4-phenylcyclohexane-1-carbonyl extension improved the peptide affinity by >100-fold, yielding organo-peptide antagonists that potently inhibited PCSK9 binding to LDLR and preserved cellular LDLR. These new bipartite antagonists have reduced mass and improved potency compared to the first-generation peptide antagonists, further validating the PCSK9 groove as a viable therapeutic target site.


Subject(s)
PCSK9 Inhibitors , Peptides/pharmacology , Serine Proteinase Inhibitors/pharmacology , Binding Sites , Crystallography, X-Ray , Drug Design , Hep G2 Cells , Humans , Molecular Structure , Peptides/chemistry , Peptides/metabolism , Proprotein Convertase 9/chemistry , Proprotein Convertase 9/metabolism , Protein Binding , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/metabolism
5.
Article in English | MEDLINE | ID: mdl-30104274

ABSTRACT

There is a critical need for new antibacterial strategies to counter the growing problem of antibiotic resistance. In Gram-negative bacteria, the outer membrane (OM) provides a protective barrier against antibiotics and other environmental insults. The outer leaflet of the outer membrane is primarily composed of lipopolysaccharide (LPS). Outer membrane biogenesis presents many potentially compelling drug targets as this pathway is absent in higher eukaryotes. Most proteins involved in LPS biosynthesis and transport are essential; however, few compounds have been identified that inhibit these proteins. The inner membrane ABC transporter MsbA carries out the first essential step in the trafficking of LPS to the outer membrane. We conducted a biochemical screen for inhibitors of MsbA and identified a series of quinoline compounds that kill Escherichia coli through inhibition of its ATPase and transport activity, with no loss of activity against clinical multidrug-resistant strains. Identification of these selective inhibitors indicates that MsbA is a viable target for new antibiotics, and the compounds we identified serve as useful tools to further probe the LPS transport pathway in Gram-negative bacteria.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Lipopolysaccharides/metabolism , Anti-Bacterial Agents/pharmacology , Biological Transport/drug effects , Biological Transport/physiology , Escherichia coli/drug effects
6.
Structure ; 26(1): 72-84.e7, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29249604

ABSTRACT

Ubiquitin-specific protease 7 (USP7) deubiquitinase activity is controlled by a number of regulatory factors, including stimulation by intramolecular accessory domains. Alone, the USP7 catalytic domain (USP7cd) shows limited activity and apo USP7cd crystal structures reveal a disrupted catalytic triad. By contrast, ubiquitin-conjugated USP7cd structures demonstrate the canonical cysteine protease active-site geometry; however, the structural features of the USP7cd that stabilize the inactive conformation and the mechanism of transition between inactive and active states remain unclear. Here we use comparative structural analyses, molecular dynamics simulations, and in silico sequence re-engineering via directed sampling by RosettaDesign to identify key molecular determinants of USP7cd activation and successfully engineer USP7cd for improved activity. Full kinetic analysis and multiple X-ray crystal structures of our designs indicate that electrostatic interactions in the distal "switching loop" region and local packing in the hydrophobic core mediate subtle but significant conformational changes that modulate USP7cd activation.


Subject(s)
Enzyme Inhibitors/chemistry , Mutation , Peptidomimetics/chemistry , Ubiquitin-Specific Peptidase 7/chemistry , Amino Acid Sequence , Amino Acid Substitution , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Enzyme Activation , Enzyme Inhibitors/chemical synthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Molecular Dynamics Simulation , Peptidomimetics/chemical synthesis , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Static Electricity , Substrate Specificity , Thermodynamics , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Ubiquitin-Specific Peptidase 7/genetics , Ubiquitin-Specific Peptidase 7/metabolism
7.
Sci Rep ; 7(1): 12524, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28970566

ABSTRACT

Fibroblast Activation Protein (FAP) is a membrane-bound serine protease whose expression is often elevated in activated fibroblasts associated with tissue remodeling in various common diseases such as cancer, arthritis and fibrosis. Like the closely related dipeptidyl peptidase DPPIV, the extracellular domain of FAP can be released into circulation as a functional enzyme, and limited studies suggest that the circulating level of FAP correlates with the degree of tissue fibrosis. Here we describe a novel homogeneous fluorescence intensity assay for circulating FAP activity based on a recently identified natural substrate, FGF21. This assay is unique in that it can effectively distinguish endopeptidase activity of FAP from that of other related enzymes such as prolyl endopeptidase (PREP) and was validated using Fap-deficient mice. Structural modeling was used to elucidate the mechanistic basis for the observed specificity in substrate recognition by FAP, but not by DPPIV or PREP. Finally, the assay was used to detect elevated FAP activity in human patients diagnosed with liver cirrhosis and to determine the effectiveness of a chemical inhibitor for FAP in mice. We propose that the assay presented here could thus be utilized for diagnosis of FAP-related pathologies and for the therapeutic development of FAP inhibitors.


Subject(s)
Fibroblast Growth Factors/genetics , Fibrosis/genetics , Gelatinases/genetics , Liver Cirrhosis/genetics , Membrane Proteins/genetics , Serine Endopeptidases/genetics , Animals , Endopeptidases/genetics , Fibroblasts/enzymology , Fibroblasts/metabolism , Fibrosis/enzymology , Fibrosis/pathology , Gene Expression Regulation, Enzymologic , Humans , Liver Cirrhosis/enzymology , Liver Cirrhosis/pathology , Mice , Prolyl Oligopeptidases , Substrate Specificity
8.
Nat Struct Mol Biol ; 24(10): 848-856, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28825733

ABSTRACT

Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates plasma LDL cholesterol (LDL-c) levels by promoting the degradation of liver LDL receptors (LDLRs). Antibodies that inhibit PCSK9 binding to the EGF(A) domain of the LDLR are effective in lowering LDL-c. However, the discovery of small-molecule therapeutics is hampered by difficulty in targeting the relatively flat EGF(A)-binding site on PCSK9. Here we demonstrate that it is possible to target this site, based on the finding that the PCSK9 P' helix displays conformational flexibility. As a consequence, the vacated N-terminal groove of PCSK9, which is adjacent to the EGF(A)-binding site, is in fact accessible to small peptides. In phage-display experiments, the EGF(A)-mimicking peptide Pep2-8 was used as an anchor peptide for the attachment of an extension peptide library directed toward the groove site. Guided by structural information, we further engineered the identified groove-binding peptides into antagonists, which encroach on the EGF(A)-binding site and inhibit LDLR binding.


Subject(s)
Enzyme Inhibitors/metabolism , PCSK9 Inhibitors , Peptides/metabolism , Proprotein Convertase 9/metabolism , Binding Sites , Enzyme Inhibitors/isolation & purification , Humans , Molecular Docking Simulation , Peptide Library , Peptides/isolation & purification
9.
Cancer Cell ; 29(4): 477-493, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-26996308

ABSTRACT

Activating mutations in protein kinases drive many cancers. While how recurring point mutations affect kinase activity has been described, the effect of in-frame deletions is not well understood. We show that oncogenic deletions within the ß3-αC loop of HER2 and BRAF are analogous to the recurrent EGFR exon 19 deletions. We identify pancreatic carcinomas with BRAF deletions mutually exclusive with KRAS mutations. Crystal structures of BRAF deletions reveal the truncated loop restrains αC in an active "in" conformation, imparting resistance to inhibitors like vemurafenib that bind the αC "out" conformation. Characterization of loop length explains the prevalence of five amino acid deletions in BRAF, EGFR, and HER2 and highlights the importance of this region for kinase activity and inhibitor efficacy.


Subject(s)
Genes, erbB-1 , Genes, erbB-2 , Mutation , Neoplasm Proteins/genetics , Neoplasms/genetics , Proto-Oncogene Proteins B-raf/genetics , Amino Acid Sequence , Amino Acid Substitution , Antineoplastic Agents/pharmacology , Base Pairing/genetics , Conserved Sequence , Dimerization , Drug Resistance, Neoplasm/genetics , Enzyme Activation/genetics , ErbB Receptors/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms/enzymology , Protein Conformation , Protein Interaction Mapping , Protein Kinase Inhibitors/pharmacology , Protein Structure, Secondary , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Structure-Activity Relationship
10.
J Biomol Screen ; 19(5): 758-70, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24518067

ABSTRACT

Several small-compound library subsets (14,000 to 56,000) have been established to complement screening of a larger Genentech corporate library (~1,300,000). Two validation sets (~1% of the total library) containing compounds representative of the main library were chosen by selection of plates or individual compounds. Use of these subsets guided selection of assay configuration, validated assay reproducibility, and provided estimates of hit rates expected from our full library. A larger diversity subset representing the scaffold diversity of the full library (3.4% of the total) was designed for screening more challenging targets with limited reagent availability or low-throughput assays. Retrospective analysis of this subset showed hit rates similar to those of the main library while recovering a higher proportion of hit scaffolds. Finally, a property-restricted diversity set called the "in-between library" was established to identify ligand-efficient compounds of molecular size between those typically found in fragment and high-throughput screening libraries. It was screened at fivefold higher concentrations than the main library to facilitate identification of less potent yet ligand-efficient compounds. Taken together, this work underscores the value of generating multiple purpose-focused, diversity-based library subsets that are designed using computational approaches coupled with internal screening data analyses to accelerate the lead discovery process.


Subject(s)
Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Small Molecule Libraries/chemistry , Chemistry, Pharmaceutical/methods , Dose-Response Relationship, Drug , Drug Discovery , Inhibitory Concentration 50 , Ligands , Reproducibility of Results
11.
Bioorg Med Chem Lett ; 24(3): 954-62, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24433859

ABSTRACT

The fragment-based identification of two novel and potent biochemical inhibitors of the nicotinamide phosphoribosyltransferase (NAMPT) enzyme is described. These compounds (51 and 63) incorporate an amide moiety derived from 3-aminopyridine, and are thus structurally distinct from other known anti-NAMPT agents. Each exhibits potent inhibition of NAMPT biochemical activity (IC50=19 and 15 nM, respectively) as well as robust antiproliferative properties in A2780 cell culture experiments (IC50=121 and 99 nM, respectively). However, additional biological studies indicate that only inhibitor 51 exerts its A2780 cell culture effects via a NAMPT-mediated mechanism. The crystal structures of both 51 and 63 in complex with NAMPT are also independently described.


Subject(s)
Amides/chemical synthesis , Amides/pharmacology , Aminopyridines/chemical synthesis , Cytokines/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Amides/chemistry , Aminopyridines/chemistry , Aminopyridines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Crystallography, X-Ray , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Humans , Inhibitory Concentration 50 , Models, Molecular , Structure-Activity Relationship
12.
J Med Chem ; 57(3): 770-92, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24405419

ABSTRACT

Potent, trans-2-(pyridin-3-yl)cyclopropanecarboxamide-containing inhibitors of the human nicotinamide phosphoribosyltransferase (NAMPT) enzyme were identified using fragment-based screening and structure-based design techniques. Multiple crystal structures were obtained of initial fragment leads, and this structural information was utilized to improve the biochemical and cell-based potency of the associated molecules. Many of the optimized compounds exhibited nanomolar antiproliferative activities against human tumor lines in in vitro cell culture experiments. In a key example, a fragment lead (13, KD = 51 µM) was elaborated into a potent NAMPT inhibitor (39, NAMPT IC50 = 0.0051 µM, A2780 cell culture IC50 = 0.000 49 µM) which demonstrated encouraging in vivo efficacy in an HT-1080 mouse xenograft tumor model.


Subject(s)
Amides/chemical synthesis , Antineoplastic Agents/chemical synthesis , Cyclopropanes/chemical synthesis , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Pyridines/chemical synthesis , Sulfones/chemical synthesis , Amides/chemistry , Amides/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Cyclopropanes/chemistry , Cyclopropanes/pharmacology , Drug Screening Assays, Antitumor , Heterografts , Humans , Mice , Mice, Nude , Models, Molecular , Neoplasm Transplantation , Protein Conformation , Pyridines/chemistry , Pyridines/pharmacology , Stereoisomerism , Structure-Activity Relationship , Sulfones/chemistry , Sulfones/pharmacology
13.
J Biol Chem ; 289(2): 942-55, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24225950

ABSTRACT

PCSK9 (proprotein convertase subtilisin/kexin type 9) is a negative regulator of the hepatic LDL receptor, and clinical studies with PCSK9-inhibiting antibodies have demonstrated strong LDL-c-lowering effects. Here we screened phage-displayed peptide libraries and identified the 13-amino acid linear peptide Pep2-8 as the smallest PCSK9 inhibitor with a clearly defined mechanism of inhibition that has been described. Pep2-8 bound to PCSK9 with a KD of 0.7 µm but did not bind to other proprotein convertases. It fully restored LDL receptor surface levels and LDL particle uptake in PCSK9-treated HepG2 cells. The crystal structure of Pep2-8 bound to C-terminally truncated PCSK9 at 1.85 Å resolution showed that the peptide adopted a strand-turn-helix conformation, which is remarkably similar to its solution structure determined by NMR. Consistent with the functional binding site identified by an Ala scan of PCSK9, the structural Pep2-8 contact region of about 400 Å(2) largely overlapped with that contacted by the EGF(A) domain of the LDL receptor, suggesting a competitive inhibition mechanism. Consistent with this, Pep2-8 inhibited LDL receptor and EGF(A) domain binding to PCSK9 with IC50 values of 0.8 and 0.4 µm, respectively. Remarkably, Pep2-8 mimicked secondary structural elements of the EGF(A) domain that interact with PCSK9, notably the ß-strand and a discontinuous short α-helix, and it engaged in the same ß-sheet hydrogen bonds as EGF(A) does. Although Pep2-8 itself may not be amenable to therapeutic applications, this study demonstrates the feasibility of developing peptidic inhibitors to functionally relevant sites on PCSK9.


Subject(s)
Oligopeptides/pharmacology , Proprotein Convertases/metabolism , Receptors, LDL/metabolism , Serine Endopeptidases/metabolism , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive/drug effects , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Hep G2 Cells , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Oligopeptides/chemistry , Oligopeptides/metabolism , Peptide Library , Proprotein Convertase 9 , Proprotein Convertases/chemistry , Proprotein Convertases/genetics , Protein Binding/drug effects , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, LDL/chemistry , Receptors, LDL/genetics , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics
14.
Clin Cancer Res ; 19(7): 1760-72, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23287563

ABSTRACT

PURPOSE: We describe the preclinical pharmacology and antitumor activity of GDC-0068, a novel highly selective ATP-competitive pan-Akt inhibitor currently in clinical trials for the treatment of human cancers. EXPERIMENTAL DESIGN: The effect of GDC-0068 on Akt signaling was characterized using specific biomarkers of the Akt pathway, and response to GDC-0068 was evaluated in human cancer cell lines and xenograft models with various genetic backgrounds, either as a single agent or in combination with chemotherapeutic agents. RESULTS: GDC-0068 blocked Akt signaling both in cultured human cancer cell lines and in tumor xenograft models as evidenced by dose-dependent decrease in phosphorylation of downstream targets. Inhibition of Akt activity by GDC-0068 resulted in blockade of cell-cycle progression and reduced viability of cancer cell lines. Markers of Akt activation, including high-basal phospho-Akt levels, PTEN loss, and PIK3CA kinase domain mutations, correlate with sensitivity to GDC-0068. Isogenic PTEN knockout also sensitized MCF10A cells to GDC-0068. In multiple tumor xenograft models, oral administration of GDC-0068 resulted in antitumor activity ranging from tumor growth delay to regression. Consistent with the role of Akt in a survival pathway, GDC-0068 also enhanced antitumor activity of classic chemotherapeutic agents. CONCLUSIONS: GDC-0068 is a highly selective, orally bioavailable Akt kinase inhibitor that shows pharmacodynamic inhibition of Akt signaling and robust antitumor activity in human cancer cells in vitro and in vivo. Our preclinical data provide a strong mechanistic rationale to evaluate GDC-0068 in cancers with activated Akt signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/metabolism , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrimidines/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Enzyme Activation/drug effects , Female , Humans , Male , Mice , Neoplasms/drug therapy , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
15.
Proc Natl Acad Sci U S A ; 109(47): 19368-73, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23134728

ABSTRACT

The protein kinase v-akt murine thymoma viral oncogene homolog (AKT), a key regulator of cell survival and proliferation, is frequently hyperactivated in human cancers. Intramolecular pleckstrin homology (PH) domain-kinase domain (KD) interactions are important in maintaining AKT in an inactive state. AKT activation proceeds after a conformational change that dislodges the PH from the KD. To understand these autoinhibitory interactions, we generated mutations at the PH-KD interface and found that most of them lead to constitutive activation of AKT. Such mutations are likely another mechanism by which activation may occur in human cancers and other diseases. In support of this likelihood, we found somatic mutations in AKT1 at the PH-KD interface that have not been previously described in human cancers. Furthermore, we show that the AKT1 somatic mutants are constitutively active, leading to oncogenic signaling. Additionally, our studies show that the AKT1 mutants are not effectively inhibited by allosteric AKT inhibitors, consistent with the requirement for an intact PH-KD interface for allosteric inhibition. These results have important implications for therapeutic intervention in patients with AKT mutations at the PH-KD interface.


Subject(s)
Neoplasms/enzymology , Neoplasms/genetics , Oncogenes/genetics , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/genetics , Allosteric Regulation/drug effects , Allosteric Regulation/genetics , Animals , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Enzyme Activation/drug effects , Humans , Mice , Models, Molecular , Mutant Proteins/metabolism , Mutation/genetics , NIH 3T3 Cells , Protein Binding/drug effects , Protein Binding/genetics , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
16.
J Med Chem ; 55(18): 8110-27, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22934575

ABSTRACT

The discovery and optimization of a series of 6,7-dihydro-5H-cyclopenta[d]pyrimidine compounds that are ATP-competitive, selective inhibitors of protein kinase B/Akt is reported. The initial design and optimization was guided by the use of X-ray structures of inhibitors in complex with Akt1 and the closely related protein kinase A. The resulting compounds demonstrate potent inhibition of all three Akt isoforms in biochemical assays and poor inhibition of other members of the cAMP-dependent protein kinase/protein kinase G/protein kinase C extended family and block the phosphorylation of multiple downstream targets of Akt in human cancer cell lines. Biological studies with one such compound, 28 (GDC-0068), demonstrate good oral exposure resulting in dose-dependent pharmacodynamic effects on downstream biomarkers and a robust antitumor response in xenograft models in which the phosphatidylinositol 3-kinase-Akt-mammalian target of rapamycin pathway is activated. 28 is currently being evaluated in human clinical trials for the treatment of cancer.


Subject(s)
Adenosine Triphosphate/metabolism , Binding, Competitive , Drug Discovery , Piperazines/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/metabolism , Pyrimidines/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Humans , Inhibitory Concentration 50 , Models, Molecular , Piperazines/chemistry , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/chemistry , Pyrimidines/chemistry , Substrate Specificity
17.
J Mol Biol ; 422(5): 685-696, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22728257

ABSTRACT

LDL (low-density lipoprotein) receptor (LDLR) binds to its negative regulator proprotein convertase subtilisin/kexin type 9 (PCSK9) through the first EGF (epidermal growth factor-like) domain [EGF(A)]. The isolated EGF(A) domain is a poor antagonist due to its low affinity for PCSK9. To improve binding affinity, we used a phage display approach by randomizing seven PCSK9 contact residues of EGF(A), including the Ca(2+)-coordinating Asp310. The library was panned in Ca(2+)-free solution, and 26 unique clones that bind to PCSK9 were identified. Four selected variants demonstrated improved inhibitory activities in a PCSK9-LDLR competition binding ELISA. The Fc fusion protein of variant EGF66 bound to PCSK9 with a K(d) value of 71 nM versus 935 nM of wild type [EGF(A)-Fc] and showed significantly improved potency in inhibiting LDLR degradation in vitro and in vivo. The five mutations in EGF66 could be modeled in the EGF(A) structure without perturbation of the EGF domain fold, and their contribution to affinity improvement could be rationalized. The most intriguing change was the substitution of the Ca(2+)-coordinating Asp310 by a Lys residue, whose side-chain amine may have functionally replaced Ca(2+). EGF66-Fc and other EGF variants having the Asp310Lys change bound to PCSK9 in a Ca(2+)-independent fashion. The findings indicate that randomization of an important Ca(2+)-chelating residue in conjunction with "selection pressure" applied by Ca(2+)-free phage selection conditions can yield variants with an alternatively stabilized Ca(2+) loop and with increased binding affinities. This approach may provide a new paradigm for the use of diversity libraries to improve affinities of members of the Ca(2+)-binding EGF domain subfamily.


Subject(s)
Calcium/metabolism , Proprotein Convertases/antagonists & inhibitors , Receptors, LDL/metabolism , Amino Acid Sequence , Amino Acid Substitution , Models, Molecular , Molecular Sequence Data , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Peptide Library , Proprotein Convertase 9 , Protein Binding , Protein Interaction Domains and Motifs , Receptors, LDL/genetics , Serine Endopeptidases
18.
Proc Natl Acad Sci U S A ; 109(14): 5299-304, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22431598

ABSTRACT

The Ras gene is frequently mutated in cancer, and mutant Ras drives tumorigenesis. Although Ras is a central oncogene, small molecules that bind to Ras in a well-defined manner and exert inhibitory effects have not been uncovered to date. Through an NMR-based fragment screen, we identified a group of small molecules that all bind to a common site on Ras. High-resolution cocrystal structures delineated a unique ligand-binding pocket on the Ras protein that is adjacent to the switch I/II regions and can be expanded upon compound binding. Structure analysis predicts that compound-binding interferes with the Ras/SOS interactions. Indeed, selected compounds inhibit SOS-mediated nucleotide exchange and prevent Ras activation by blocking the formation of intermediates of the exchange reaction. The discovery of a small-molecule binding pocket on Ras with functional significance provides a new direction in the search of therapeutically effective inhibitors of the Ras oncoprotein.


Subject(s)
Nucleotides/metabolism , Son of Sevenless Proteins/metabolism , ras Proteins/metabolism , Binding Sites , Cell Line , Humans , Ligands , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , ras Proteins/chemistry
19.
Bioorg Med Chem Lett ; 21(8): 2410-4, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21392984

ABSTRACT

A novel series of spirochromane pan-Akt inhibitors is reported. SAR optimization furnished compounds with improved enzyme potencies and excellent selectivity over the related AGC kinase PKA. Attempted replacement of the phenol hinge binder provided compounds with excellent Akt enzyme and cell activities but greatly diminished selectivity over PKA.


Subject(s)
Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Binding Sites , Crystallography, X-Ray , Drug Evaluation, Preclinical , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...