Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
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
2.
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
3.
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
4.
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.

5.
Bioorg Med Chem Lett ; 18(23): 6273-8, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18929486

ABSTRACT

Information from X-ray crystal structures were used to optimize the potency of a HTS hit in a Hsp90 competitive binding assay. A class of novel and potent small molecule Hsp90 inhibitors were thereby identified. Enantio-pure compounds 31 and 33 were potent in PGA-based competitive binding assay and inhibited proliferation of various human cancer cell lines in vitro, with IC(50) values averaging 20 nM.


Subject(s)
Amides/chemical synthesis , Amides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Amides/chemistry , Amino Acids/chemistry , Antineoplastic Agents/chemistry , Combinatorial Chemistry Techniques , Crystallography, X-Ray , Drug Design , Drug Screening Assays, Antitumor , Humans , Molecular Chaperones/metabolism , Molecular Conformation , Molecular Structure , Structure-Activity Relationship
6.
Mol Cancer Ther ; 6(3): 945-56, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17363489

ABSTRACT

Poly(ADP-ribose) polymerase (PARP)-1 (EC 2.4.2.30) is a nuclear enzyme that promotes the base excision repair of DNA breaks. Inhibition of PARP-1 enhances the efficacy of DNA alkylating agents, topoisomerase I poisons, and ionizing radiation. Our aim was to identify a PARP inhibitor for clinical trial from a panel of 42 potent PARP inhibitors (K(i), 1.4-15.1 nmol/L) based on the quinazolinone, benzimidazole, tricyclic benzimidazole, tricyclic indole, and tricyclic indole-1-one core structures. We evaluated chemosensitization of temozolomide and topotecan using LoVo and SW620 human colorectal cells; in vitro radiosensitization was measured using LoVo cells, and the enhancement of antitumor activity of temozolomide was evaluated in mice bearing SW620 xenografts. Excellent chemopotentiation and radiopotentiation were observed in vitro, with 17 of the compounds causing a greater temozolomide and topotecan sensitization than the benchmark inhibitor AG14361 and 10 compounds were more potent radiosensitizers than AG14361. In tumor-bearing mice, none of the compounds were toxic when given alone, and the antitumor activity of the PARP inhibitor-temozolomide combinations was unrelated to toxicity. Compounds that were more potent chemosensitizers in vivo than AG14361 were also more potent in vitro, validating in vitro assays as a prescreen. These studies have identified a compound, AG14447, as a PARP inhibitor with outstanding in vivo chemosensitization potency at tolerable doses, which is at least 10 times more potent than the initial lead, AG14361. The phosphate salt of AG14447 (AG014699), which has improved aqueous solubility, has been selected for clinical trial.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Cell Line, Tumor/drug effects , Cell Line, Tumor/radiation effects , Colorectal Neoplasms/radiotherapy , DNA Repair/drug effects , DNA Repair/radiation effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Gamma Rays , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Heterocyclic Compounds, 3-Ring/chemistry , Humans , Maximum Tolerated Dose , Mice , Mice, Nude , Poly(ADP-ribose) Polymerases/metabolism , Structure-Activity Relationship , Temozolomide , Topoisomerase I Inhibitors , Topotecan/pharmacology
7.
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
8.
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
9.
Structure ; 12(8): 1533-44, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15296746

ABSTRACT

Human rhinoviruses (HRV), the predominant members of the Picornaviridae family of positive-strand RNA viruses, are the major causative agents of the common cold. Given the lack of effective treatments for rhinoviral infections, virally encoded proteins have become attractive therapeutic targets. The HRV genome encodes an RNA-dependent RNA polymerase (RdRp) denoted 3Dpol, which is responsible for replicating the viral genome and for synthesizing a protein primer used in the replication. Here the crystal structures for three viral serotypes (1B, 14, and 16) of HRV 3Dpol have been determined. The three structures are very similar to one another, and to the closely related poliovirus (PV) 3Dpol enzyme. Because the reported PV crystal structure shows significant disorder, HRV 3Dpol provides the first complete view of a picornaviral RdRp. The folding topology of HRV 3Dpol also resembles that of RdRps from hepatitis C virus (HCV) and rabbit hemorrhagic disease virus (RHDV) despite very low sequence homology.


Subject(s)
Models, Molecular , Protein Folding , RNA-Dependent RNA Polymerase/chemistry , Rhinovirus/enzymology , Amino Acid Sequence , Cloning, Molecular , Crystallography, X-Ray , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid
10.
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
11.
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
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.
J Med Chem ; 46(2): 210-3, 2003 Jan 16.
Article in English | MEDLINE | ID: mdl-12519059
14.
J Med Chem ; 47(22): 5467-81, 2004 Oct 21.
Article in English | MEDLINE | ID: mdl-15481984

ABSTRACT

The design, synthesis, and biological evaluation of potent inhibitors of poly(ADP-ribose) polymerase-1 (PARP-1) are reported. A novel series of 3,4-dihydro-2H-[1,4]diazepino[6,7,1-hi]indol-1-ones were designed using a combination of protein structure-based drug design, molecular modeling, and structure-activity relationships (SAR). These novel submicromolar inhibitors possess a tricyclic ring system conformationally restricting the benzamide in the preferred cis orientation. The compounds were designed to optimize space-filling and atomic interactions within the NAD+ binding site of PARP-1. Previously described and newly adapted methods were applied to syntheses of these tricyclic inhibitors. Various modifications were made to the diazepinoindolones at the 6- and 7-positions in order to study this region of the active site and optimize noncovalent interactions. The electron density of derivative 28 bound to chicken PARP-1 revealed that the oxime makes a tight hydrogen bond with the catalytic gamma-carboxylate of glutamic acid (Glu) 988 in accordance with our original designs and models. Most of the compounds have been evaluated for inhibition of human PARP-1. Selected inhibitors were also tested for the ability to potentiate the cytotoxic effect of the DNA-damaging agent Topotecan.


Subject(s)
Antineoplastic Agents/chemical synthesis , Azepines/chemical synthesis , Indoles/chemical synthesis , Poly(ADP-ribose) Polymerase Inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Azepines/chemistry , Azepines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Design , Drug Resistance, Neoplasm , Drug Synergism , Humans , Indoles/chemistry , Indoles/pharmacology , Models, Molecular , Structure-Activity Relationship , Topoisomerase I Inhibitors
15.
J Med Chem ; 45(23): 4961-74, 2002 Nov 07.
Article in English | MEDLINE | ID: mdl-12408707

ABSTRACT

A series of novel compounds have been designed that are potent inhibitors of poly(ADP-ribose) polymerase-1 (PARP-1), and the activity and physical properties have been characterized. The new structural classes, 3,4,5,6-tetrahydro-1H-azepino[5,4,3-cd]indol-6-ones and 3,4-dihydropyrrolo[4,3,2-de]isoquinolin-5-(1H)-ones, have conformationally locked benzamide cores that specifically interact with the PARP-1 protein. The compounds have been evaluated with in vitro cellular assays that measure the ability of the PARP-1 inhibitors to enhance the effect of cytotoxic agents against cancer cell lines.


Subject(s)
Antineoplastic Agents/chemical synthesis , Dacarbazine/analogs & derivatives , Enzyme Inhibitors/chemical synthesis , Indoles/chemical synthesis , Isoquinolines/chemical synthesis , Poly(ADP-ribose) Polymerase Inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Crystallography, X-Ray , Dacarbazine/pharmacology , Drug Design , Drug Screening Assays, Antitumor , Drug Synergism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Indoles/chemistry , Indoles/pharmacology , Isoquinolines/chemistry , Isoquinolines/pharmacology , NAD/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Structure-Activity Relationship , Temozolomide , Topotecan/pharmacology , Tumor Cells, Cultured
17.
Structure ; 17(12): 1625-1635, 2009 Dec 09.
Article in English | MEDLINE | ID: mdl-20004166

ABSTRACT

Novel inhibitors are needed to counteract the rapid emergence of drug-resistant HIV variants. HIV-1 reverse transcriptase (RT) has both DNA polymerase and RNase H (RNH) enzymatic activities, but approved drugs that inhibit RT target the polymerase. Inhibitors that act against new targets, such as RNH, should be effective against all of the current drug-resistant variants. Here, we present 2.80 A and 2.04 A resolution crystal structures of an RNH inhibitor, beta-thujaplicinol, bound at the RNH active site of both HIV-1 RT and an isolated RNH domain. beta-thujaplicinol chelates two divalent metal ions at the RNH active site. We provide biochemical evidence that beta-thujaplicinol is a slow-binding RNH inhibitor with noncompetitive kinetics and suggest that it forms a tropylium ion that interacts favorably with RT and the RNA:DNA substrate.


Subject(s)
HIV Reverse Transcriptase/chemistry , Reverse Transcriptase Inhibitors/chemistry , Ribonucleases/metabolism , Tropolone/analogs & derivatives , Catalytic Domain , Crystallography, X-Ray , HIV Reverse Transcriptase/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Reverse Transcriptase Inhibitors/metabolism , Tropolone/chemistry , Tropolone/metabolism
18.
Anal Biochem ; 351(2): 260-5, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16527235

ABSTRACT

A fluorescence polarization (FP) microplate assay suitable for screening compounds against the ribonuclease H (RNase H) activity of HIV-1 reverse transcriptase has been developed. This homogeneous assay uses a hybrid 18-mer DNA/RNA duplex substrate composed of an RNA oligonucleotide labeled with 6-carboxytetramethyl rhodamine at the 3' end that is annealed to a complementary unlabeled DNA strand. The labeled RNA/DNA duplex demonstrated Michaelis-Menten kinetics with a Km value of 9.6+/-2.8 nM. Substrate cleavage by RNase H to produce small RNA fragments (1-4 mer) resulted in a large change in the measured FP value. This FP assay was amenable to kinetics protocols as well as stopped endpoint measurements. When using the latter for conducting robotics runs, Z' values greater than 0.8 typically were observed. The stopped endpoint FP assay was used successfully in a high-throughput screening campaign to screen 1.8 million compounds for RNase H inhibition.


Subject(s)
Fluorescence Polarization/methods , HIV Reverse Transcriptase/antagonists & inhibitors , Ribonuclease H/antagonists & inhibitors , Fluorescence Resonance Energy Transfer , Inhibitory Concentration 50 , Kinetics , Rhodamines
19.
Bioorg Med Chem Lett ; 15(11): 2829-33, 2005 Jun 02.
Article in English | MEDLINE | ID: mdl-15911263

ABSTRACT

The structure-based design, chemical synthesis, and biological evaluation of novel MTAP substrates are described. These compounds incorporate various C5'-moieties and are shown to have different k(cat)/K(m) values compared with the natural MTAP substrate (MTA).


Subject(s)
Purine-Nucleoside Phosphorylase/metabolism , Drug Design , Humans , Molecular Structure , Substrate Specificity
20.
Bioorg Med Chem Lett ; 14(10): 2433-7, 2004 May 17.
Article in English | MEDLINE | ID: mdl-15109627

ABSTRACT

The synthesis and biological evaluation of a new series of amine-substituted 2-arylbenzimidazole-4-carboxamide inhibitors of the DNA-repair enzyme poly(ADP-ribose) polymerase-1 (PARP-1) is reported. The introduction of an amine substituent at the 2-aryl position is not detrimental to activity, with most inhibitors exhibiting K(i) values for PARP-1 inhibition in the low nanomolar range. Two compounds in this series were found to potentiate the cytotoxicity of the DNA-methylating agent temozolomide by 4-5-fold in a human colorectal cancer cell line.


Subject(s)
Amides/pharmacology , Antineoplastic Agents/chemical synthesis , Benzimidazoles/pharmacology , Dacarbazine/analogs & derivatives , Poly(ADP-ribose) Polymerase Inhibitors , Amides/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Benzimidazoles/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Dacarbazine/pharmacology , Drug Synergism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Structure-Activity Relationship , Temozolomide
SELECTION OF CITATIONS
SEARCH DETAIL