Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Med Chem ; 67(2): 864-884, 2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38197367

RESUMO

The DNA-encoded library (DEL) discovery platform has emerged as a powerful technology for hit identification in recent years. It has become one of the major parallel workstreams for small molecule drug discovery along with other strategies such as HTS and data mining. For many researchers working in the DEL field, it has become increasingly evident that many hits and leads discovered via DEL screening bind to target proteins with unique and unprecedented binding modes. This Perspective is our attempt to analyze reports of DEL screening with the purpose of providing a rigorous and useful account of the binding modes observed for DEL-derived ligands with a focus on binding mode novelty.


Assuntos
DNA , Bibliotecas de Moléculas Pequenas , Bibliotecas de Moléculas Pequenas/química , Ligantes , DNA/química , Descoberta de Drogas , Técnicas de Química Combinatória
2.
J Med Chem ; 64(23): 17146-17183, 2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34807608

RESUMO

Aberrant activity of the histone methyltransferase polycomb repressive complex 2 (PRC2) has been linked to several cancers, with small-molecule inhibitors of the catalytic subunit of the PRC2 enhancer of zeste homologue 2 (EZH2) being recently approved for the treatment of epithelioid sarcoma (ES) and follicular lymphoma (FL). Compounds binding to the EED subunit of PRC2 have recently emerged as allosteric inhibitors of PRC2 methyltransferase activity. In contrast to orthosteric inhibitors that target EZH2, small molecules that bind to EED retain their efficacy in EZH2 inhibitor-resistant cell lines. In this paper we disclose the discovery of potent and orally bioavailable EED ligands with good solubilities. The solubility of the EED ligands was optimized through a variety of design tactics, with the resulting compounds exhibiting in vivo efficacy in EZH2-driven tumors.


Assuntos
Inibidores Enzimáticos/farmacologia , Complexo Repressor Polycomb 2/antagonistas & inibidores , Regulação Alostérica , Animais , Domínio Catalítico , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Proteína Potenciadora do Homólogo 2 de Zeste/química , Proteína Potenciadora do Homólogo 2 de Zeste/efeitos dos fármacos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacocinética , Compostos Heterocíclicos/química , Humanos , Ligantes , Complexo Repressor Polycomb 2/química , Ratos , Relação Estrutura-Atividade
3.
J Med Chem ; 63(9): 4517-4527, 2020 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-32297743

RESUMO

JAK1, JAK2, JAK3, and TYK2 belong to the JAK (Janus kinase) family. They play critical roles in cytokine signaling. Constitutive activation of JAK/STAT pathways is associated with a wide variety of diseases. Particularly, pSTAT3 is observed in response to the treatment with inhibitors of oncogenic signaling pathways such as EGFR, MAPK, and AKT and is associated with resistance or poorer response to agents targeting these pathways. Among the JAK family kinases, JAK1 has been shown to be the primary driver of STAT3 phosphorylation and signaling; therefore, selective JAK1 inhibition can be a viable means to overcome such treatment resistances. Herein, an account of the medicinal chemistry optimization from the promiscuous kinase screening hit 3 to the candidate drug 21 (AZD4205), a highly selective JAK1 kinase inhibitor, is reported. Compound 21 has good preclinical pharmacokinetics. Compound 21 displayed an enhanced antitumor activity in combination with an approved EGFR inhibitor, osimertinib, in a preclinical non-small-cell lung cancer (NSCLC) xenograft NCI-H1975 model.


Assuntos
Indóis/uso terapêutico , Janus Quinase 1/antagonistas & inibidores , Inibidores de Proteínas Quinases/uso terapêutico , Acrilamidas/farmacologia , Compostos de Anilina/farmacologia , Animais , Linhagem Celular Tumoral , Desenho de Fármacos , Descoberta de Drogas , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Receptores ErbB/antagonistas & inibidores , Feminino , Humanos , Indóis/síntese química , Indóis/farmacocinética , Camundongos Nus , Estrutura Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacocinética , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
4.
ACS Chem Biol ; 14(10): 2134-2140, 2019 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-31525019

RESUMO

Enhancer of zeste homologue 2 (EZH2), the catalytic subunit of polycomb repressive complex 2 (PRC2), regulates chromatin state and gene expression by methylating histone H3 lysine 27. EZH2 is overexpressed or mutated in various hematological malignancies and solid cancers. Our previous efforts to identify inhibitors of PRC2 methyltransferase activity by high-throughput screening (HTS) resulted in large numbers of false positives and thus a significant hit deconvolution challenge. More recently, others have reported compounds that bind to another PRC2 core subunit, EED, and allosterically inhibit EZH2 activity. This mechanism is particularly appealing as it appears to retain potency in cell lines that have acquired resistance to orthosteric EZH2 inhibition. By designing a fluorescence polarization probe based on the reported EED binding compounds, we were able to quickly and cleanly re-triage our previously challenging HTS hit list and identify novel allosteric PRC2 inhibitors.


Assuntos
Benzofuranos/química , Inibidores Enzimáticos/química , Complexo Repressor Polycomb 2/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/química , Regulação Alostérica/efeitos dos fármacos , Benzofuranos/metabolismo , Carbocianinas/química , Linhagem Celular Tumoral , Inibidores Enzimáticos/metabolismo , Corantes Fluorescentes/química , Ensaios de Triagem em Larga Escala , Humanos , Ligantes , Complexo Repressor Polycomb 2/isolamento & purificação , Complexo Repressor Polycomb 2/metabolismo , Ligação Proteica , Bibliotecas de Moléculas Pequenas/metabolismo
6.
J Med Chem ; 61(12): 5235-5244, 2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29856615

RESUMO

Janus kinases (JAKs) have been demonstrated to be critical in cytokine signaling and have thus been implicated in both cancer and inflammatory diseases. The JAK family consists of four highly homologous members: JAK1-3 and TYK2. The development of small-molecule inhibitors that are selective for a specific family member would represent highly desirable tools for deconvoluting the intricacies of JAK family biology. Herein, we report the discovery of a potent JAK1 inhibitor, 24, which displays ∼1000-fold selectivity over the other highly homologous JAK family members (determined by biochemical assays), while also possessing good selectivity over other kinases (determined by panel screening). Moreover, this compound was demonstrated to be orally bioavailable and possesses acceptable pharmacokinetic parameters. In an in vivo study, the compound was observed to dose dependently modulate the phosphorylation of STAT3 (a downstream marker of JAK1 inhibition).


Assuntos
Janus Quinase 1/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Disponibilidade Biológica , Linhagem Celular , Cristalografia por Raios X , Humanos , Janus Quinase 1/química , Janus Quinase 1/metabolismo , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/metabolismo , Janus Quinase 3/metabolismo , Camundongos , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Med Chem ; 61(3): 1061-1073, 2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29301085

RESUMO

Checkpoint kinase 1 (CHK1) inhibitors are potential cancer therapeutics that can be utilized for enhancing the efficacy of DNA damaging agents. Multiple small molecule CHK1 inhibitors from different chemical scaffolds have been developed and evaluated in clinical trials in combination with chemotherapeutics and radiation treatment. Scaffold morphing of thiophene carboxamide ureas (TCUs), such as AZD7762 (1) and a related series of triazoloquinolines (TZQs), led to the identification of fused-ring bicyclic CHK1 inhibitors, 7-carboxamide thienopyridines (7-CTPs), and 7-carboxamide indoles. X-ray crystal structures reveal a key intramolecular noncovalent sulfur-oxygen interaction in aligning the hinge-binding carboxamide group to the thienopyridine core in a coplanar fashion. An intramolecular hydrogen bond to an indole NH was also effective in locking the carboxamide in the preferred bound conformation to CHK1. Optimization on the 7-CTP series resulted in the identification of lead compound 44, which displayed respectable drug-like properties and good in vitro and in vivo potency.


Assuntos
Quinase 1 do Ponto de Checagem/antagonistas & inibidores , Descoberta de Drogas , Compostos Heterocíclicos/química , Compostos Heterocíclicos/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quinase 1 do Ponto de Checagem/química , Dano ao DNA , Humanos , Indóis/química , Modelos Moleculares , Domínios Proteicos , Piridinas/química
8.
J Med Chem ; 60(22): 9299-9319, 2017 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-29116812

RESUMO

Excessive activity of striatal-enriched protein tyrosine phosphatase (STEP) in the brain has been detected in numerous neuropsychiatric disorders including Alzheimer's disease. Notably, knockdown of STEP in an Alzheimer mouse model effected an increase in the phosphorylation levels of downstream STEP substrates and a significant reversal in the observed cognitive and memory deficits. These data point to the promising potential of STEP as a target for drug discovery in Alzheimer's treatment. We previously reported a substrate-based approach to the development of low molecular weight STEP inhibitors with Ki values as low as 7.8 µM. Herein, we disclose the first X-ray crystal structures of inhibitors bound to STEP and the surprising finding that they occupy noncoincident binding sites. Moreover, we utilize this structural information to optimize the inhibitor structure to achieve a Ki of 110 nM, with 15-60-fold selectivity across a series of phosphatases.


Assuntos
Organofosfonatos/química , Proteínas Tirosina Fosfatases não Receptoras/antagonistas & inibidores , Sulfonamidas/química , Doença de Alzheimer/tratamento farmacológico , Animais , Domínio Catalítico , Cristalografia por Raios X , Descoberta de Drogas , Estabilidade de Medicamentos , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Microssomos Hepáticos/metabolismo , Organofosfonatos/síntese química , Organofosfonatos/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/química , Ratos , Sulfonamidas/síntese química , Sulfonamidas/metabolismo
9.
Nat Commun ; 8(1): 1610, 2017 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-29151576

RESUMO

The CRISPR-Cas9 RNA-guided endonuclease system allows precise and efficient modification of complex genomes and is continuously developed to enhance specificity, alter targeting and add new functional moieties. However, one area yet to be explored is the base chemistry of the associated RNA molecules. Here we show the design and optimisation of hybrid DNA-RNA CRISPR and tracr molecules based on structure-guided approaches. Through careful mapping of the ribose requirements of Cas9, we develop hybrid versions possessing minimal RNA residues, which are sufficient to direct specific nuclease activity in vitro and in vivo with reduced off-target activity. We identify critical regions within these molecules that require ribose nucleotides and show a direct correlation between binding affinity/stability and cellular activity. This is the first demonstration of a non-RNA-guided Cas9 endonuclease and first step towards eliminating the ribose dependency of Cas9 to develop a XNA-programmable endonuclease.


Assuntos
Proteínas de Bactérias/química , DNA/química , Endonucleases/química , RNA Guia de Cinetoplastídeos/química , RNA/química , Proteínas de Bactérias/metabolismo , Biocatálise , Proteína 9 Associada à CRISPR , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , DNA/genética , Endonucleases/metabolismo , Conformação de Ácido Nucleico , RNA/genética , RNA Guia de Cinetoplastídeos/genética
10.
Proc Natl Acad Sci U S A ; 113(49): E7880-E7889, 2016 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-27864515

RESUMO

Millions of individuals are infected with and die from tuberculosis (TB) each year, and multidrug-resistant (MDR) strains of TB are increasingly prevalent. As such, there is an urgent need to identify novel drugs to treat TB infections. Current frontline therapies include the drug isoniazid, which inhibits the essential NADH-dependent enoyl-acyl-carrier protein (ACP) reductase, InhA. To inhibit InhA, isoniazid must be activated by the catalase-peroxidase KatG. Isoniazid resistance is linked primarily to mutations in the katG gene. Discovery of InhA inhibitors that do not require KatG activation is crucial to combat MDR TB. Multiple discovery efforts have been made against InhA in recent years. Until recently, despite achieving high potency against the enzyme, these efforts have been thwarted by lack of cellular activity. We describe here the use of DNA-encoded X-Chem (DEX) screening, combined with selection of appropriate physical properties, to identify multiple classes of InhA inhibitors with cell-based activity. The utilization of DEX screening allowed the interrogation of very large compound libraries (1011 unique small molecules) against multiple forms of the InhA enzyme in a multiplexed format. Comparison of the enriched library members across various screening conditions allowed the identification of cofactor-specific inhibitors of InhA that do not require activation by KatG, many of which had bactericidal activity in cell-based assays.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Mycobacterium tuberculosis/enzimologia , Oxirredutases/antagonistas & inibidores , Testes de Sensibilidade Microbiana , Bibliotecas de Moléculas Pequenas
11.
Cancer Res ; 76(3): 724-35, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26637668

RESUMO

MLK4 is a member of the mixed-lineage family of kinases that regulate the JNK, p38, and ERK kinase signaling pathways. MLK4 mutations have been identified in various human cancers, including frequently in colorectal cancer, where their function and pathobiological importance have been uncertain. In this study, we assessed the functional consequences of MLK4 mutations in colon tumorigenesis. Biochemical data indicated that a majority of MLK4 mutations are loss-of-function (LOF) mutations that can exert dominant-negative effects. In seeking to understand the abrogated activity of these mutants, we elucidated a new MLK4 catalytic domain structure. To determine whether MLK4 is required to maintain tumorigenic phenotypes, we reconstituted its signaling axis in colon cancer cells harboring MLK4-inactivating mutations. We found that restoring MLK4 activity reduced cell viability, proliferation, and colony formation in vitro and delayed tumor growth in vivo. Mechanistic investigations established that restoring the function of MLK4 selectively induced the JNK pathway and its downstream targets, cJUN, ATF3, and the cyclin-dependent kinase inhibitors CDKN1A and CDKN2B. Our work indicates that MLK4 is a novel tumor-suppressing kinase harboring frequent LOF mutations that lead to diminished signaling in the JNK pathway and enhanced proliferation in colon cancer.


Assuntos
Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Animais , Carcinogênese , Neoplasias do Colo/patologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Bioorg Med Chem Lett ; 25(24): 5743-7, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26546219

RESUMO

The propensity for cancer cells to accumulate additional centrosomes relative to normal cells could be exploited for therapeutic benefit in oncology. Following literature reports that suggested TNKS1 (tankyrase 1) and PARP16 may be involved with spindle structure and function and may play a role in suppressing multi-polar spindle formation in cells with supernumerary centrosomes, we initiated a phenotypic screen to look for small molecule poly (ADP-ribose) polymerase (PARP) enzyme family inhibitors that could produce a multi-polar spindle phenotype via declustering of centrosomes. Screening of AstraZeneca's collection of phthalazinone PARP inhibitors in HeLa cells using high-content screening techniques identified several compounds that produced a multi-polar spindle phenotype at low nanomolar concentrations. Characterization of these compounds across a broad panel of PARP family enzyme assays indicated that they had activity against several PARP family enzymes, including PARP1, 2, 3, 5a, 5b, and 6. Further optimization of these initial hits for improved declustering potency, solubility, permeability, and oral bioavailability resulted in AZ0108, a PARP1, 2, 6 inhibitor that potently inhibits centrosome clustering and is suitable for in vivo efficacy and tolerability studies.


Assuntos
Centrossomo/metabolismo , Ftalazinas/química , Inibidores de Poli(ADP-Ribose) Polimerases/química , Administração Oral , Animais , Sítios de Ligação , Células CACO-2 , Centrossomo/efeitos dos fármacos , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos , Células HeLa , Humanos , Microssomos/metabolismo , Conformação Molecular , Simulação de Dinâmica Molecular , Ftalazinas/administração & dosagem , Ftalazinas/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/administração & dosagem , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Estrutura Terciária de Proteína , Ratos , Tanquirases/antagonistas & inibidores , Tanquirases/metabolismo
13.
J Med Chem ; 57(1): 144-58, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24359159

RESUMO

Structure based design, synthesis, and biological evaluation of a novel series of 1-methyl-1H-imidazole, as potent Jak2 inhibitors to modulate the Jak/STAT pathway, are described. Using the C-ring fragment from our first clinical candidate AZD1480 (24), optimization of the series led to the discovery of compound 19a, a potent, orally bioavailable Jak2 inhibitor. Compound 19a displayed a high level of cellular activity in hematopoietic cell lines harboring the V617F mutation and in murine BaF3 TEL-Jak2 cells. Compound 19a demonstrated significant tumor growth inhibition in a UKE-1 xenograft model within a well-tolerated dose range.


Assuntos
Antineoplásicos/síntese química , Imidazóis/síntese química , Janus Quinase 2/antagonistas & inibidores , Inibidores de Proteínas Quinases/síntese química , Animais , Antineoplásicos/farmacologia , Cães , Descoberta de Drogas , Humanos , Imidazóis/farmacologia , Camundongos , Inibidores de Proteínas Quinases/farmacologia , Ratos , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Nat Chem Biol ; 10(2): 96-8, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24292073

RESUMO

Pyridomycin, a natural product with potent antituberculosis activity, inhibits a major drug target, the InhA enoyl reductase. Here, we unveil the co-crystal structure and unique ability of pyridomycin to block both the NADH cofactor- and lipid substrate-binding pockets of InhA. This is to our knowledge a first-of-a-kind binding mode that discloses a new means of InhA inhibition. Proof-of-principle studies show how structure-assisted drug design can improve the activity of new pyridomycin derivatives.


Assuntos
Antituberculosos/química , Proteínas de Bactérias/química , NAD/química , Oligopeptídeos/química , Oxirredutases/química , Sítios de Ligação , Cristalografia por Raios X , Modelos Moleculares , Especificidade por Substrato
15.
J Med Chem ; 56(21): 8533-42, 2013 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-24107081

RESUMO

InhA is a well validated Mycobacterium tuberculosis (Mtb) target as evidenced by the clinical success of isoniazid. Translating enzyme inhibition to bacterial cidality by targeting the fatty acid substrate site of InhA remains a daunting challenge. The recent disclosure of a methyl-thiazole series demonstrates that bacterial cidality can be achieved with potent enzyme inhibition and appropriate physicochemical properties. In this study, we report the molecular mode of action of a lead methyl-thiazole, along with analogues with improved CYP inhibition profile. We have identified a novel mechanism of InhA inhibition characterized by a hitherto unreported "Y158-out" inhibitor-bound conformation of the protein that accommodates a neutrally charged "warhead". An additional novel hydrophilic interaction with protein residue M98 allows the incorporation of favorable physicochemical properties for cellular activity. Notably, the methyl-thiazole prefers the NADH-bound form of the enzyme with a Kd of ~13.7 nM, as against the NAD(+)-bound form of the enzyme.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Mycobacterium tuberculosis/enzimologia , Oxirredutases/antagonistas & inibidores , Tiazóis/farmacologia , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Modelos Moleculares , Estrutura Molecular , Oxirredutases/metabolismo , Relação Estrutura-Atividade , Tiazóis/síntese química , Tiazóis/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...