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.
ACS Chem Biol ; 18(3): 643-651, 2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36825662

RESUMO

The TEAD transcription factors are the most distal elements of the Hippo pathway, and their transcriptional activity is regulated by several proteins, including YAP. In some cancers, the Hippo pathway is deregulated and inhibitors of the YAP:TEAD interaction are foreseen as new anticancer drugs. The binding of YAP to TEAD is driven by the interaction of an α-helix and an Ω-loop present in its TEAD-binding domain with two distinct pockets at the TEAD surface. Using the mRNA-based display technique to screen a library of in vitro-translated cyclic peptides, we identified a peptide that binds with a nanomolar affinity to TEAD. The X-ray structure of this peptide in complex with TEAD reveals that it interacts with the α-helix pocket. Under our experimental conditions, this peptide can form a ternary complex with TEAD and YAP. Furthermore, combining it with a peptide binding to the Ω-loop pocket gives an additive inhibitory effect on the YAP:TEAD interaction. Overall, our results show that it is possible to identify nanomolar inhibitors of the YAP:TEAD interaction that bind to the α-helix pocket, suggesting that developing such compounds might be a strategy to treat cancers where the Hippo pathway is deregulated.


Assuntos
Neoplasias , Fatores de Transcrição , Humanos , Fatores de Transcrição/metabolismo , Conformação Proteica em alfa-Hélice , Fatores de Transcrição de Domínio TEA , Peptídeos/química
2.
Cell Chem Biol ; 29(2): 249-258.e5, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-34547225

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates plasma low-density lipoprotein cholesterol (LDL-C) levels by promoting hepatic LDL receptor (LDLR) degradation. Therapeutic antibodies that disrupt PCSK9-LDLR binding reduce LDL-C concentrations and cardiovascular disease risk. The epidermal growth factor precursor homology domain A (EGF-A) of the LDLR serves as a primary contact with PCSK9 via a flat interface, presenting a challenge for identifying small molecule PCSK9-LDLR disruptors. We employ an affinity-based screen of 1013in vitro-translated macrocyclic peptides to identify high-affinity PCSK9 ligands that utilize a unique, induced-fit pocket and partially disrupt the PCSK9-LDLR interaction. Structure-based design led to molecules with enhanced function and pharmacokinetic properties (e.g., 13PCSK9i). In mice, 13PCSK9i reduces plasma cholesterol levels and increases hepatic LDLR density in a dose-dependent manner. 13PCSK9i functions by a unique, allosteric mechanism and is the smallest molecule identified to date with in vivo PCSK9-LDLR disruptor function.


Assuntos
Peptídeos/farmacologia , Pró-Proteína Convertase 9/metabolismo , Receptores de LDL/antagonistas & inibidores , Animais , Relação Dose-Resposta a Droga , Células Hep G2 , Humanos , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/síntese química , Peptídeos/química , Conformação Proteica , Receptores de LDL/metabolismo
3.
RSC Chem Biol ; 2(6): 1661-1668, 2021 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-34977581

RESUMO

Mouse double minute 2 homolog (MDM2, Hdm2) is an important negative regulator of the tumor suppressor p53. Using a mRNA based display technique to screen a library of >1012 in vitro-translated cyclic peptides, we have identified a macrocyclic ligand that shows picomolar potency on MDM2. X-Ray crystallography reveals a novel binding mode utilizing a unique pharmacophore to occupy the Phe/Trp/Leu pockets on MDM2. Conjugation of a cyclic cell-penetrating peptide (cCPP) to the initially non cell-permeable ligand enables cellular uptake and a pharmacodynamic response in SJSA-1 cells. The demonstrated enhanced intracellular availability of cyclic peptides that are identified by a display technology exemplifies a process for the application of intracellular tools for drug discovery projects.

4.
ACS Med Chem Lett ; 10(12): 1674-1679, 2019 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-31857845

RESUMO

Targeted antimitotic agents are a promising class of anticancer therapies. Herein, we describe the development of a potent and selective antimitotic Eg5 inhibitor based antibody-drug conjugate (ADC). Preliminary studies were performed using proprietary Eg5 inhibitors which were conjugated onto a HER2-targeting antibody using maleimido caproyl valine-citrulline para-amino benzocarbamate, or MC-VC-PABC cleavable linker. However, the resulting ADCs lacked antigen-specificity in vivo, probably from premature release of the payload. Second-generation ADCs were then developed, using noncleavable linkers, and the resulting conjugates (ADC-4 and ADC-10) led to in vivo efficacy in an HER-2 expressing (SK-OV-3ip) mouse xenograft model while ADC-11 led to in vivo efficacy in an anti-c-KIT (NCI-H526) mouse xenograft model in a target-dependent manner.

5.
J Med Chem ; 61(18): 8120-8135, 2018 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-30137981

RESUMO

Chronic myelogenous leukemia (CML) arises from the constitutive activity of the BCR-ABL1 oncoprotein. Tyrosine kinase inhibitors (TKIs) that target the ATP-binding site have transformed CML into a chronic manageable disease. However, some patients develop drug resistance due to ATP-site mutations impeding drug binding. We describe the discovery of asciminib (ABL001), the first allosteric BCR-ABL1 inhibitor to reach the clinic. Asciminib binds to the myristate pocket of BCR-ABL1 and maintains activity against TKI-resistant ATP-site mutations. Although resistance can emerge due to myristate-site mutations, these are sensitive to ATP-competitive inhibitors so that combinations of asciminib with ATP-competitive TKIs suppress the emergence of resistance. Fragment-based screening using NMR and X-ray yielded ligands for the myristate pocket. An NMR-based conformational assay guided the transformation of these inactive ligands into ABL1 inhibitors. Further structure-based optimization for potency, physicochemical, pharmacokinetic, and drug-like properties, culminated in asciminib, which is currently undergoing clinical studies in CML patients.


Assuntos
Descoberta de Drogas , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Niacinamida/análogos & derivados , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Regulação Alostérica , Animais , Cães , Proteínas de Fusão bcr-abl/genética , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Masculino , Camundongos , Modelos Moleculares , Estrutura Molecular , Mutação , Niacinamida/química , Niacinamida/farmacologia , Fosforilação , Conformação Proteica , Inibidores de Proteínas Quinases/química , Pirazóis/química , Ratos , Ratos Sprague-Dawley , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Nature ; 543(7647): 733-737, 2017 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-28329763

RESUMO

Chronic myeloid leukaemia (CML) is driven by the activity of the BCR-ABL1 fusion oncoprotein. ABL1 kinase inhibitors have improved the clinical outcomes for patients with CML, with over 80% of patients treated with imatinib surviving for more than 10 years. Second-generation ABL1 kinase inhibitors induce more potent molecular responses in both previously untreated and imatinib-resistant patients with CML. Studies in patients with chronic-phase CML have shown that around 50% of patients who achieve and maintain undetectable BCR-ABL1 transcript levels for at least 2 years remain disease-free after the withdrawal of treatment. Here we characterize ABL001 (asciminib), a potent and selective allosteric ABL1 inhibitor that is undergoing clinical development testing in patients with CML and Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukaemia. In contrast to catalytic-site ABL1 kinase inhibitors, ABL001 binds to the myristoyl pocket of ABL1 and induces the formation of an inactive kinase conformation. ABL001 and second-generation catalytic inhibitors have similar cellular potencies but distinct patterns of resistance mutations, with genetic barcoding studies revealing pre-existing clonal populations with no shared resistance between ABL001 and the catalytic inhibitor nilotinib. Consistent with this profile, acquired resistance was observed with single-agent therapy in mice; however, the combination of ABL001 and nilotinib led to complete disease control and eradicated CML xenograft tumours without recurrence after the cessation of treatment.


Assuntos
Sítio Alostérico/efeitos dos fármacos , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Niacinamida/análogos & derivados , Pirazóis/farmacologia , Regulação Alostérica/efeitos dos fármacos , Animais , Domínio Catalítico/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dasatinibe/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Quimioterapia Combinada , Proteínas de Fusão bcr-abl/química , Proteínas de Fusão bcr-abl/genética , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Camundongos , Mutação , Niacinamida/farmacologia , Niacinamida/uso terapêutico , Pirazóis/uso terapêutico , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Angew Chem Int Ed Engl ; 55(52): 16026-16030, 2016 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-27874234

RESUMO

To study the behavior of MDM2-p53 inhibitors in a disease-relevant cellular model, we have developed and validated a set of bioorthogonal probes that can be fluorescently labeled in cells and used in high-content screening assays. By using automated image analysis with single-cell resolution, we could visualize the intracellular target binding of compounds by co-localization and quantify target upregulation upon MDM2-p53 inhibition in an osteosarcoma model. Additionally, we developed a high-throughput assay to quantify target occupancy of non-tagged MDM2-p53 inhibitors by competition and to identify novel chemical matter. This approach could be expanded to other targets for lead discovery applications.


Assuntos
Antineoplásicos/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Corantes Fluorescentes/análise , Indóis/farmacologia , Osteossarcoma/tratamento farmacológico , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteína Supressora de Tumor p53/antagonistas & inibidores , Antineoplásicos/química , Técnicas Biossensoriais , Linhagem Celular Tumoral , Corantes Fluorescentes/química , Humanos , Indóis/química , Modelos Moleculares , Estrutura Molecular , Osteossarcoma/patologia , Análise de Célula Única
8.
ChemMedChem ; 10(11): 1884-91, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26381451

RESUMO

Farnesyl pyrophosphate synthase (FPPS) is an established target for the treatment of bone diseases, but also shows promise as an anticancer and anti-infective drug target. Currently available anti-FPPS drugs are active-site-directed bisphosphonate inhibitors, the peculiar pharmacological profile of which is inadequate for therapeutic indications beyond bone diseases. The recent discovery of an allosteric binding site has paved the way toward the development of novel non-bisphosphonate FPPS inhibitors with broader therapeutic potential, notably as immunomodulators in oncology. Herein we report the discovery, by an integrated lead finding approach, of two new chemical classes of allosteric FPPS inhibitors that belong to the salicylic acid and quinoline chemotypes. We present their synthesis, biochemical and cellular activities, structure-activity relationships, and provide X-ray structures of several representative FPPS complexes. These novel allosteric FPPS inhibitors are devoid of any affinity for bone mineral and could serve as leads to evaluate their potential in none-bone diseases.


Assuntos
Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Geraniltranstransferase/antagonistas & inibidores , Quinolinas/farmacologia , Ácido Salicílico/farmacologia , Regulação Alostérica/efeitos dos fármacos , Sítio Alostérico/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Geraniltranstransferase/metabolismo , Humanos , Estrutura Molecular , Quinolinas/síntese química , Quinolinas/química , Ácido Salicílico/síntese química , Ácido Salicílico/química , Relação Estrutura-Atividade
9.
Chimia (Aarau) ; 67(12-13): 899-904, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24594335

RESUMO

In view of our more comprehensive understanding with respect to intracellular pathways and their regulation, a vast number of interesting drug targets appear to be in a space that can be addressed neither by small molecules nor by biologics. Especially, interference with intracellular protein-protein interactions, if successful, seems to offer considerable opportunities to expand the application of peptides as therapeutics, in particular in the field of oncology. This review focuses on requirements for the development of peptide therapeutics aiming at intracellular targets. In addition, an outlook for developments in this field based on recent examples for peptides active in cellular assays, highlighting key requirement to assess permeability, is provided.


Assuntos
Peptídeos/administração & dosagem , Sequência de Aminoácidos , Dados de Sequência Molecular , Peptídeos/química
10.
Eur J Med Chem ; 57: 1-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23041456

RESUMO

Protein kinases are widely recognized as important therapeutic targets due to their involvement in signal transduction pathways. These pathways are tightly controlled and regulated, notably by the ability of kinases to selectively phosphorylate a defined set of substrates. A wide variety of disorders can arise as a consequence of abnormal kinase-mediated phosphorylation and numerous kinase inhibitors have earned their place as key components of the modern pharmacopeia. Although "traditional" kinase inhibitors typically act by preventing the interaction between the kinase and ATP, thus stopping substrate phosphorylation, an alternative approach consists in disrupting the protein-protein interaction between the kinase and its downstream partners. In order to facilitate the identification of potential chemical starting points for substrate-site inhibition approaches, we desired to investigate the application of Substrate Activity Screening to kinases. We herein report a proof-of-concept study demonstrating, on a model tyrosine kinase, that the key requirements of this methodology can be met. Namely, using peptides as model substrates, we show that a simple ADP-accumulation assay can be used to monitor substrate efficiency and that efficiency can be optimized in a modular manner. More importantly, we demonstrate that structure-efficiency relationships translate into structure-activity relationships upon conversion of the substrates into inhibitors.


Assuntos
Peptídeos/química , Inibidores de Proteínas Quinases/química , Receptores Proteína Tirosina Quinases/química , Trifosfato de Adenosina/química , Ensaios de Triagem em Larga Escala , Humanos , Cinética , Peptídeos/antagonistas & inibidores , Ligação Proteica , Inibidores de Proteínas Quinases/síntese química , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/química , Relação Estrutura-Atividade , Especificidade por Substrato
11.
Bioorg Med Chem Lett ; 21(23): 7030-3, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22004721

RESUMO

Protein kinases are widely recognized as important therapeutic targets due to their involvement in signal transduction pathways. These pathways are tightly controlled and regulated, notably by the ability of kinases to selectively phosphorylate a defined set of substrates. As part of a study on the substrate requirements of Insulin-like Growth Factor 1 Receptor (IGF-1R) and Insulin Receptor (InsR), we evaluated and applied a universal assay system able to monitor the phosphorylation of unlabelled peptides of any length in real time. In contrast to already reported profiling methodologies, we were able to assess the k(cat)/K(M) ratio of peptides as short as tetramers. Notably, we were able to identify an efficient pentamer substrate that exhibited kinetic properties close to those of a 250-amino acid protein derived from IRS-1, a natural substrate of IGF-1R and InsR.


Assuntos
Sondas Moleculares/química , Peptídeos/química , Receptor IGF Tipo 1/química , Sequência de Aminoácidos , Cristalografia por Raios X , Modelos Moleculares , Sondas Moleculares/genética , Dados de Sequência Molecular , Peptídeos/genética , Fosforilação , Ligação Proteica , Receptor de Insulina/química , Receptor de Insulina/genética , Especificidade por Substrato
12.
Org Lett ; 13(2): 320-3, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21162548

RESUMO

A flow method for the synthesis of aliphatic and aromatic diazoketones from acyl chloride precursors has been developed and used to prepare quinoxalines in a multistep sequence without isolation of the potentially explosive diazoketone. The protocol showcases an efficient in-line purification using supported scavengers with time-saving and safety benefits and in particular a reduction in the operator's exposure to carcinogenic phenylenediamines.


Assuntos
Química Orgânica/instrumentação , Compostos de Diazônio/síntese química , Cetonas/síntese química , Quinoxalinas/síntese química , Química Orgânica/métodos , Compostos de Diazônio/química , Cetonas/química , Estrutura Molecular , Fenilenodiaminas/toxicidade , Quinoxalinas/química
13.
Nat Chem Biol ; 6(9): 660-6, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20711197

RESUMO

Bisphosphonates are potent inhibitors of farnesyl pyrophosphate synthase (FPPS) and are highly efficacious in the treatment of bone diseases such as osteoporosis, Paget's disease and tumor-induced osteolysis. In addition, the potential for direct antitumor effects has been postulated on the basis of in vitro and in vivo studies and has recently been demonstrated clinically in early breast cancer patients treated with the potent bisphosphonate zoledronic acid. However, the high affinity of bisphosphonates for bone mineral seems suboptimal for the direct treatment of soft-tissue tumors. Here we report the discovery of the first potent non-bisphosphonate FPPS inhibitors. These new inhibitors bind to a previously unknown allosteric site on FPPS, which was identified by fragment-based approaches using NMR and X-ray crystallography. This allosteric and druggable pocket allows the development of a new generation of FPPS inhibitors that are optimized for direct antitumor effects in soft tissue.


Assuntos
Difosfonatos , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/farmacologia , Geraniltranstransferase/antagonistas & inibidores , Regulação Alostérica , Sítio Alostérico , Osso e Ossos/química , Osso e Ossos/metabolismo , Cristalografia por Raios X , Difosfonatos/análise , Difosfonatos/química , Difosfonatos/metabolismo , Difosfonatos/farmacologia , Inibidores Enzimáticos/química , Geraniltranstransferase/metabolismo , Humanos , Imidazóis/análise , Imidazóis/química , Imidazóis/farmacologia , Espectroscopia de Ressonância Magnética , Neoplasias de Tecidos Moles/tratamento farmacológico , Ácido Zoledrônico
14.
J Am Chem Soc ; 132(20): 7043-8, 2010 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-20450175

RESUMO

Allosteric inhibitors of Bcr-Abl have emerged as a novel therapeutic option for the treatment of CML. Using fragment-based screening, a search for novel Abl inhibitors that bind to the myristate pocket was carried out. Here we show that not all myristate ligands are functional inhibitors, but that the conformational state of C-terminal helix_I is a structural determinant for functional activity. We present an NMR-based conformational assay to monitor the conformation of this crucial helix_I and show that myristate ligands that bend helix_I are functional antagonists, whereas ligands that bind to the myristate pocket but do not induce this conformational change are kinase agonists. Activation of c-Abl by allosteric agonists has been confirmed in a biochemical assay.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-abl/agonistas , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Regulação Alostérica , Animais , Ativação Enzimática/efeitos dos fármacos , Humanos , Ligantes , Espectroscopia de Ressonância Magnética , Camundongos , Modelos Moleculares , Ácido Mirístico/metabolismo , Ligação Proteica , Inibidores de Proteínas Quinases/química , Estrutura Secundária de Proteína , Proteínas Proto-Oncogênicas c-abl/química , Proteínas Proto-Oncogênicas c-abl/metabolismo
15.
Biochim Biophys Acta ; 1804(3): 454-62, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20152788

RESUMO

The ATP-competitive inhibitors dasatinib and nilotinib, which bind to catalytically different conformations of the Abl kinase domain, have recently been approved for the treatment of imatinib-resistant CML. These two new drugs, albeit very efficient against most of the imatinib-resistant mutants of Bcr-Abl, fail to effectively suppress the Bcr-Abl activity of the T315I (or gatekeeper) mutation. Generating new ATP site-binding drugs that target the T315I in Abl has been hampered, amongst others, by target selectivity, which is frequently an issue when developing ATP-competitive inhibitors. Recently, using an unbiased cellular screening approach, GNF-2, a non-ATP-competitive inhibitor, has been identified that demonstrates cellular activity against Bcr-Abl transformed cells. The exquisite selectivity of GNF-2 is due to the finding that it targets the myristate binding site located near the C-terminus of the Abl kinase domain, as demonstrated by genetic approaches, solution NMR and X-ray crystallography. GNF-2, like myristate, is able to induce and/or stabilize the clamped inactive conformation of Abl analogous to the SH2-Y527 interaction of Src. The molecular mechanism for allosteric inhibition by the GNF-2 inhibitor class, and the combined effects with ATP-competitive inhibitors such as nilotinib and imatinib on wild-type Abl and imatinib-resistant mutants, in particular the T315I gatekeeper mutant, are reviewed.


Assuntos
Trifosfato de Adenosina/química , Ácido Mirístico/química , Inibidores de Proteínas Quinases/química , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-abl/química , Trifosfato de Adenosina/metabolismo , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/genética , Benzamidas , Cristalografia por Raios X , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Mesilato de Imatinib , Mutação de Sentido Incorreto , Ácido Mirístico/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Neoplasias/genética , Ressonância Magnética Nuclear Biomolecular , Piperazinas/química , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Estrutura Terciária de Proteína/genética , Proteínas Proto-Oncogênicas c-abl/genética , Proteínas Proto-Oncogênicas c-abl/metabolismo , Pirimidinas/química , Pirimidinas/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA