Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Cell Chem Biol ; 30(10): 1191-1210.e20, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37557181

ABSTRACT

KAT6A, and its paralog KAT6B, are histone lysine acetyltransferases (HAT) that acetylate histone H3K23 and exert an oncogenic role in several tumor types including breast cancer where KAT6A is frequently amplified/overexpressed. However, pharmacologic targeting of KAT6A to achieve therapeutic benefit has been a challenge. Here we describe identification of a highly potent, selective, and orally bioavailable KAT6A/KAT6B inhibitor CTx-648 (PF-9363), derived from a benzisoxazole series, which demonstrates anti-tumor activity in correlation with H3K23Ac inhibition in KAT6A over-expressing breast cancer. Transcriptional and epigenetic profiling studies show reduced RNA Pol II binding and downregulation of genes involved in estrogen signaling, cell cycle, Myc and stem cell pathways associated with CTx-648 anti-tumor activity in ER-positive (ER+) breast cancer. CTx-648 treatment leads to potent tumor growth inhibition in ER+ breast cancer in vivo models, including models refractory to endocrine therapy, highlighting the potential for targeting KAT6A in ER+ breast cancer.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/genetics , Histones/metabolism , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Signal Transduction , Cell Line, Tumor
2.
Mol Cancer Ther ; 21(1): 3-15, 2022 01.
Article in English | MEDLINE | ID: mdl-34737197

ABSTRACT

Protein arginine methyltransferase 5 (PRMT5) overexpression in hematologic and solid tumors methylates arginine residues on cellular proteins involved in important cancer functions including cell-cycle regulation, mRNA splicing, cell differentiation, cell signaling, and apoptosis. PRMT5 methyltransferase function has been linked with high rates of tumor cell proliferation and decreased overall survival, and PRMT5 inhibitors are currently being explored as an approach for targeting cancer-specific dependencies due to PRMT5 catalytic function. Here, we describe the discovery of potent and selective S-adenosylmethionine (SAM) competitive PRMT5 inhibitors, with in vitro and in vivo characterization of clinical candidate PF-06939999. Acquired resistance mechanisms were explored through the development of drug resistant cell lines. Our data highlight compound-specific resistance mutations in the PRMT5 enzyme that demonstrate structural constraints in the cofactor binding site that prevent emergence of complete resistance to SAM site inhibitors. PRMT5 inhibition by PF-06939999 treatment reduced proliferation of non-small cell lung cancer (NSCLC) cells, with dose-dependent decreases in symmetric dimethyl arginine (SDMA) levels and changes in alternative splicing of numerous pre-mRNAs. Drug sensitivity to PF-06939999 in NSCLC cells associates with cancer pathways including MYC, cell cycle and spliceosome, and with mutations in splicing factors such as RBM10. Translation of efficacy in mouse tumor xenograft models with splicing mutations provides rationale for therapeutic use of PF-06939999 in the treatment of splicing dysregulated NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , S-Adenosylmethionine/metabolism , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Drug Resistance , Female , Humans , Lung Neoplasms/pathology , Mice
3.
ACS Med Chem Lett ; 11(6): 1175-1184, 2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32550998

ABSTRACT

Two novel compounds were identified as Naa50 binders/inhibitors using DNA-encoded technology screening. Biophysical and biochemical data as well as cocrystal structures were obtained for both compounds (3a and 4a) to understand their mechanism of action. These data were also used to rationalize the binding affinity differences observed between the two compounds and a MLGP peptide-containing substrate. Cellular target engagement experiments further confirm the Naa50 binding of 4a and demonstrate its selectivity toward related enzymes (Naa10 and Naa60). Additional analogs of inhibitor 4a were also evaluated to study the binding mode observed in the cocrystal structures.

4.
Mol Cancer Ther ; 17(12): 2530-2542, 2018 12.
Article in English | MEDLINE | ID: mdl-30232146

ABSTRACT

Tumors use indoleamine 2,3-dioxygenase-1 (IDO1) as a major mechanism to induce an immunosuppressive microenvironment. IDO1 expression is upregulated in many cancers and considered to be a resistance mechanism to immune checkpoint therapies. IDO1 is induced in response to inflammatory stimuli such as IFNγ and promotes immune tolerance by depleting tryptophan and producing tryptophan catabolites, including kynurenine, in the tumor microenvironment. This leads to effector T-cell anergy and enhanced Treg function through upregulation of FoxP3. As a nexus for the induction of key immunosuppressive mechanisms, IDO1 represents an important immunotherapeutic target in oncology. Here, we report the identification and characterization of the novel selective, orally bioavailable IDO1 inhibitor EOS200271/PF-06840003. It reversed IDO1-induced T-cell anergy in vitro In mice carrying syngeneic tumor grafts, PF-06840003 reduced intratumoral kynurenine levels by over 80% and inhibited tumor growth both in monotherapy and, with an increased efficacy, in combination with antibodies blocking the immune checkpoint ligand PD-L1. We demonstrate that anti-PD-L1 therapy results in increased IDO1 metabolic activity thereby providing additional mechanistic rationale for combining PD-(L)1 blockade with IDO1 inhibition in cancer immunotherapies. Supported by these preclinical data and favorable predicted human pharmacokinetic properties of PF-06840003, a phase I open-label, multicenter clinical study (NCT02764151) has been initiated.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Biocatalysis , Enzyme Inhibitors/pharmacology , Immunotherapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoles/pharmacology , Succinimides/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , B7-H1 Antigen/metabolism , CTLA-4 Antigen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/metabolism , Kynurenine/blood , Lymphocytes, Tumor-Infiltrating/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Stereoisomerism , Substrate Specificity/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
5.
J Med Chem ; 61(3): 650-665, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29211475

ABSTRACT

A new series of lactam-derived EZH2 inhibitors was designed via ligand-based and physicochemical-property-based strategies to address metabolic stability and thermodynamic solubility issues associated with previous lead compound 1. The new inhibitors incorporated an sp3 hybridized carbon atom at the 7-position of the lactam moiety present in lead compound 1 as a replacement for a dimethylisoxazole group. This transformation enabled optimization of the physicochemical properties and potency compared to compound 1. Analysis of relationships between calculated log D (clogD) values and in vitro metabolic stability and permeability parameters identified a clogD range that afforded an increased probability of achieving favorable ADME data in a single molecule. Compound 23a exhibited the best overlap of potency and pharmaceutical properties as well as robust tumor growth inhibition in vivo and was therefore advanced as a development candidate (PF-06821497). A crystal structure of 23a in complex with the three-protein PRC2 complex enabled understanding of the key structural features required for optimal binding.


Subject(s)
Drug Design , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Isoquinolines/pharmacology , Isoquinolines/pharmacokinetics , Administration, Oral , Biological Availability , Cell Line, Tumor , Humans , Isoquinolines/administration & dosage , Isoquinolines/chemistry , Models, Molecular , Molecular Conformation
6.
J Med Chem ; 60(23): 9617-9629, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29111717

ABSTRACT

Tumors use tryptophan-catabolizing enzymes such as indoleamine 2,3-dioxygenase (IDO-1) to induce an immunosuppressive environment. IDO-1 is induced in response to inflammatory stimuli and promotes immune tolerance through effector T-cell anergy and enhanced Treg function. As such, IDO-1 is a nexus for the induction of a key immunosuppressive mechanism and represents an important immunotherapeutic target in oncology. Starting from HTS hit 5, IDO-1 inhibitor 6 (EOS200271/PF-06840003) has been developed. The structure-activity relationship around 6 is described and rationalized using the X-ray crystal structure of 6 bound to human IDO-1, which shows that 6, differently from most of the IDO-1 inhibitors described so far, does not bind to the heme iron atom and has a novel binding mode. Clinical candidate 6 shows good potency in an IDO-1 human whole blood assay and also shows a very favorable ADME profile leading to favorable predicted human pharmacokinetic properties, including a predicted half-life of 16-19 h.


Subject(s)
Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoles/pharmacology , Succinimides/pharmacology , Animals , Cell Line , Crystallography, X-Ray , Dogs , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Indoles/chemistry , Indoles/pharmacokinetics , Macaca fascicularis , Male , Mice , Molecular Docking Simulation , Rats , Structure-Activity Relationship , Succinimides/chemistry , Succinimides/pharmacokinetics
7.
Elife ; 62017 11 29.
Article in English | MEDLINE | ID: mdl-29185984

ABSTRACT

Polycomb repressive complex 2 (PRC2) is a key chromatin modifier responsible for methylation of lysine 27 in histone H3. PRC2 has been shown to interact with thousands of RNA species in vivo, but understanding the physiological function of RNA binding has been hampered by the lack of separation-of-function mutants. Here, we use comprehensive mutagenesis and hydrogen deuterium exchange mass spectrometry (HDX-MS) to identify critical residues for RNA interaction in PRC2 core complexes from Homo sapiens and Chaetomium thermophilum, for which crystal structures are known. Preferential binding of G-quadruplex RNA is conserved, surprisingly using different protein elements. Key RNA-binding residues are spread out along the surface of EZH2, with other subunits including EED also contributing, and missense mutations of some of these residues have been found in cancer patients. The unusual nature of this protein-RNA interaction provides a paradigm for other epigenetic modifiers that bind RNA without canonical RNA-binding motifs.


Subject(s)
Amino Acids/genetics , Amino Acids/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , G-Quadruplexes , RNA/metabolism , Chaetomium/enzymology , DNA Mutational Analysis , Enhancer of Zeste Homolog 2 Protein/chemistry , Humans , Mass Spectrometry , Protein Binding
8.
Methods Mol Biol ; 1608: 271-297, 2017.
Article in English | MEDLINE | ID: mdl-28695516

ABSTRACT

This chapter describes the approaches taken in the development of the first PARP inhibitor to enter clinical trial, rucaparib (now called Rubraca), in 2003. We describe the general principles of crystal-based drug design, the purification and crystallization of the PARP-1 catalytic domain and how this was used to develop highly potent PARP inhibitors, based on the nicotinamide pharmacophore. Several methods have been used to determine the inhibitory potency in cell-free and whole cell assays, each described with reference to its advantages and disadvantages.


Subject(s)
Enzyme Inhibitors/chemistry , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Animals , Catalytic Domain , Drug Design , Enzyme Inhibitors/pharmacology , Humans , Indoles/chemistry , Indoles/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
10.
J Med Chem ; 59(18): 8306-25, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27512831

ABSTRACT

A new enhancer of zeste homolog 2 (EZH2) inhibitor series comprising a substituted phenyl ring joined to a dimethylpyridone moiety via an amide linkage has been designed. A preferential amide torsion that improved the binding properties of the compounds was identified for this series via computational analysis. Cyclization of the amide linker resulted in a six-membered lactam analogue, compound 18. This transformation significantly improved the ligand efficiency/potency of the cyclized compound relative to its acyclic analogue. Additional optimization of the lactam-containing EZH2 inhibitors focused on lipophilic efficiency (LipE) improvement, which provided compound 31. Compound 31 displayed improved LipE and on-target potency in both biochemical and cellular readouts relative to compound 18. Inhibitor 31 also displayed robust in vivo antitumor growth activity and dose-dependent de-repression of EZH2 target genes.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Design , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Pyridones/chemistry , Pyridones/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cyclization , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Humans , Isoquinolines/chemistry , Isoquinolines/pharmacology , Isoquinolines/therapeutic use , Lactams/chemistry , Lactams/pharmacology , Mice , Mice, SCID , Models, Molecular , Neoplasms/drug therapy , Neoplasms/metabolism , Pyridones/therapeutic use
11.
Nat Commun ; 7: 11384, 2016 04 28.
Article in English | MEDLINE | ID: mdl-27122193

ABSTRACT

Polycomb repressive complex 2 (PRC2) mediates gene silencing through chromatin reorganization by methylation of histone H3 lysine 27 (H3K27). Overexpression of the complex and point mutations in the individual subunits of PRC2 have been shown to contribute to tumorigenesis. Several inhibitors of the PRC2 activity have shown efficacy in EZH2-mutated lymphomas and are currently in clinical development, although the molecular basis of inhibitor recognition remains unknown. Here we report the crystal structures of the inhibitor-bound wild-type and Y641N PRC2. The structures illuminate an important role played by a stretch of 17 residues in the N-terminal region of EZH2, we call the activation loop, in the stimulation of the enzyme activity, inhibitor recognition and the potential development of the mutation-mediated drug resistance. The work presented here provides new avenues for the design and development of next-generation PRC2 inhibitors through establishment of a structure-based drug design platform.


Subject(s)
Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/chemistry , Drug Resistance, Neoplasm , Enhancer of Zeste Homolog 2 Protein/chemistry , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Humans , Models, Molecular , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism
12.
Assay Drug Dev Technol ; 13(4): 235-40, 2015 May.
Article in English | MEDLINE | ID: mdl-26065559

ABSTRACT

Conformational remodeling of chromatin in cells is known to alter gene expression. The histone code hypothesis postulates that multiple modifications present on histone tails can regulate gene expression both through direct effects on chromatin compaction as well as through recruitment of unique complexes that signal specific downstream functions. Histone methylation is an important component of the histone code, and the dysregulation of histone methylation in disease makes methyltransferases and demethylases viable targets for drug discovery. We developed a biochemical assay platform, which takes advantage of the fact that protein methyltransferases (PMTs) all utilize the cofactor S-Adenosyl-L-methionine (SAM) as the methyl donor. The platform utilizes the High-throughput Mass Spectrometry (MS) technology to measure SAM and the S-Adenosyl-L-homocysteine product in a label-free manner. The platform has all the advantages of a label-free system coupled with the benefit of substrate agnostic measurements making it an ideal setup for PMT biochemical studies and drug discovery. In addition, MS is ideally suited for detecting multiple modification events within the same substrate. The ability to adjust the detection to monitor the methyl acceptor product allows for real-time measurements of multiple product species simultaneously, a distinct advantage over other commonly used assay formats.


Subject(s)
Enzyme Assays/methods , High-Throughput Screening Assays , Mass Spectrometry , Protein-Arginine N-Methyltransferases/analysis , Radiometry/methods , Binding Sites , Humans , Protein-Arginine N-Methyltransferases/metabolism
13.
Bioorg Med Chem Lett ; 25(7): 1532-7, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25746813

ABSTRACT

A series of novel enhancer of zeste homolog 2 (EZH2) inhibitors was designed based on the chemical structure of the histone methyltransferase (HMT) inhibitor SAH (S-adenosyl-l-homocysteine). These nucleoside-based EZH2 inhibitors blocked the methylation of nucleosomes at H3K27 in biochemical assays employing both WT PRC2 complex as well as a Y641N mutant PRC2 complex. The most potent compound, 27, displayed IC50's against both complexes of 270 nM and 70 nM, respectively. To our knowledge, compound 27 is the most potent SAH-derived inhibitor of the EZH2 PRC2 complex yet identified. This compound also displayed improved potency, lipophilic efficiency (LipE), and selectivity profile against other lysine methyltransferases compared with SAH.


Subject(s)
Polycomb Repressive Complex 2/antagonists & inhibitors , S-Adenosylhomocysteine/pharmacology , Dose-Response Relationship, Drug , Drug Design , Enhancer of Zeste Homolog 2 Protein , Humans , Models, Molecular , Molecular Structure , S-Adenosylhomocysteine/chemical synthesis , S-Adenosylhomocysteine/chemistry , Structure-Activity Relationship
15.
Bioorg Med Chem Lett ; 23(11): 3358-63, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23622982

ABSTRACT

The M2 isoform of pyruvate kinase is an emerging target for antitumor therapy. In this letter, we describe the discovery of 2-((1H-benzo[d]imidazol-1-yl)methyl)-4H-pyrido[1,2-a]pyrimidin-4-ones as potent and selective PKM2 activators which were found to have a novel binding mode. The original lead identified from high throughput screening was optimized into an efficient series via computer-aided structure-based drug design. Both a representative compound from this series and an activator described in the literature were used as molecular tools to probe the biological effects of PKM2 activation on cancer cells. Our results suggested that PKM2 activation alone is not sufficient to alter cancer cell metabolism.


Subject(s)
Benzimidazoles/chemistry , Carrier Proteins/agonists , Membrane Proteins/agonists , Pyrimidinones/chemistry , Thyroid Hormones/agonists , Binding Sites , Carrier Proteins/metabolism , Cell Line , Computer-Aided Design , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Humans , Kinetics , Membrane Proteins/metabolism , Molecular Docking Simulation , Protein Binding , Protein Structure, Tertiary , Pyrimidinones/chemical synthesis , Pyrimidinones/metabolism , Structure-Activity Relationship , Thyroid Hormones/metabolism , Thyroid Hormone-Binding Proteins
16.
PLoS One ; 8(1): e53994, 2013.
Article in English | MEDLINE | ID: mdl-23342057

ABSTRACT

The non-receptor tyrosine kinase Ack1 belongs to a unique multi-domain protein kinase family, Ack. Ack is the only family of SH3 domain containing kinases to have an SH3 domain following the kinase domain; others have their SH3 domains preceding the kinase domain. Previous reports have suggested that Ack1 does not require phosphorylation for activation and the enzyme activity of the isolated kinase domain is low relative to other kinases. It has been shown to dimerize in the cellular environment, which augments its enzyme activity. The molecular mechanism of activation, however, remains unknown. Here we present structural and biochemical data on Ack1 kinase domain, and kinase domain+SH3 domain that suggest that Ack1 in its monomeric state is autoinhibited, like EGFR and CDK. The activation of the kinase domain may require N-lobe mediated symmetric dimerization, which may be facilitated by the N-terminal SAM domain. Results presented here show that SH3 domain, unlike in Src family tyrosine kinases, does not directly control the activation state of the enzyme. Instead we speculate that the SH3 domain may play a regulatory role by facilitating binding of the MIG6 homologous region to the kinase domain. We postulate that features of Ack1 activation and regulation parallel those of receptor tyrosine kinase EGFR with some interesting differences.


Subject(s)
Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , Allosteric Regulation , Animals , Crystallography, X-Ray , Enzyme Activation , Humans , Mice , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Multimerization , Sf9 Cells , Spodoptera
17.
Methods Mol Biol ; 780: 463-89, 2011.
Article in English | MEDLINE | ID: mdl-21870278

ABSTRACT

This chapter describes the approaches taken in the development of the first PARP inhibitor to enter into clinical trial, AG-014699. We describe the general principles of crystal-based drug design, the purification, and crystallization of the PARP-1 catalytic domain, and how this approach was used to develop highly potent PARP inhibitors based on the nicotinamide pharmacophore. Several methods have been used to determine the inhibitory potency of designed inhibitors in cell-free and whole cell assays; each is described with reference to its advantages and disadvantages.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Crystallography , Drug Design , Enzyme Inhibitors/pharmacology , Humans
18.
Bioorg Med Chem Lett ; 21(12): 3557-62, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21612924

ABSTRACT

A series of novel and potent small molecule Hsp90 inhibitors was optimized using X-ray crystal structures. These compounds bind in a deep pocket of the Hsp90 enzyme that is partially comprised by residues Asn51 and Ser52. Displacement of several water molecules observed crystallographically in this pocket using rule-based strategies led to significant improvements in inhibitor potency. An optimized inhibitor (compound 17) exhibited potent Hsp90 inhibition in ITC, biochemical, and cell-based assays (K(d)=1.3 nM, K(i)=15 nM, and cellular IC(50)=0.5 µM).


Subject(s)
Drug Design , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Small Molecule Libraries/chemistry , Binding Sites/drug effects , Crystallography, X-Ray , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/chemistry , Pyrroles/pharmacology , Small Molecule Libraries/pharmacology
19.
Antiviral Res ; 91(2): 102-11, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21620899

ABSTRACT

Several potent inhibitors of hepatitis C virus (HCV) NS3/4A protease have been identified that show great clinical potential against genotype 1. Due to the tremendous genetic diversity that exists among HCV isolates, development of broad spectrum inhibitors is challenging. With a limited number of lab strains available for preclinical testing, new tools are required for assessing protease inhibitor activity. We developed a chimeric replicon system for evaluating NS3 protease inhibitor activity against naturally occurring isolates. NS3/4A genes were cloned from the plasma of HCV-infected individuals and inserted into lab strain replicons, replacing the native sequences. The chimeric reporter replicons were transfected into Huh 7.5 cells, their replication monitored by luciferase assays, and their susceptibilities to inhibitors determined. Viable chimeras expressing heterologous genotypes 1, 2, 3, and 4 protease domains were identified that exhibited varying susceptibilities to inhibitors. Protease inhibitor spectrums observed against the chimeric replicon panel strongly correlated with published enzymatic and clinical results. This cell-based chimeric replicon system can be used to characterize the activities of protease inhibitors against diverse natural isolates and may improve the ability to predict dose and clinical efficacy.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/genetics , Protease Inhibitors/chemistry , Replicon , Viral Nonstructural Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemistry , Base Sequence , Binding Sites , Carbamates/chemistry , Carbamates/pharmacology , Cell Line , Cloning, Molecular , Fluorescence Resonance Energy Transfer/methods , Genetic Variation , Genetic Vectors/genetics , Genotype , Hepacivirus/drug effects , Hepacivirus/physiology , Humans , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Microbial Sensitivity Tests , Oligopeptides/chemistry , Oligopeptides/pharmacology , Phylogeny , Protease Inhibitors/pharmacology , Quinolines/chemistry , Quinolines/pharmacology , Sequence Analysis, Protein/methods , Thiazoles/chemistry , Thiazoles/pharmacology , Viral Nonstructural Proteins/chemistry , Virus Replication
20.
J Med Chem ; 54(9): 3368-85, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21438541

ABSTRACT

A novel class of heat shock protein 90 (Hsp90) inhibitors was discovered by high-throughput screening and was subsequently optimized using a combination of structure-based design, parallel synthesis, and the application of medicinal chemistry principles. Through this process, the biochemical and cell-based potency of the original HTS lead were substantially improved along with the corresponding metabolic stability properties. These efforts culminated with the identification of a development candidate (compound 42) which displayed desired PK/PD relationships, significant efficacy in a melanoma A2058 xenograft tumor model, and attractive DMPK profiles.


Subject(s)
Antineoplastic Agents/chemical synthesis , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Pyrazoles/chemical synthesis , Pyrimidines/chemical synthesis , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Binding, Competitive , Biological Availability , Blood Proteins/metabolism , Cell Line, Tumor , Cell Membrane Permeability , Drug Screening Assays, Antitumor , Drug Stability , Female , Humans , Hydrophobic and Hydrophilic Interactions , In Vitro Techniques , Male , Melanoma/drug therapy , Melanoma/pathology , Mice , Mice, Nude , Microsomes, Liver/metabolism , Models, Molecular , Neoplasm Transplantation , Protein Binding , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Structure-Activity Relationship , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...