Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Sci Rep ; 14(1): 2038, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38263191

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), responsible for the COVID-19 pandemic, uses a surface expressed trimeric spike glycoprotein for cell entry. This trimer is the primary target for neutralizing antibodies making it a key candidate for vaccine development. During the global pandemic circulating variants of concern (VOC) caused several waves of infection, severe disease, and death. The reduced efficacy of the ancestral trimer-based vaccines against emerging VOC led to the need for booster vaccines. Here we present a detailed characterization of the Sanofi Beta trimer, utilizing cryo-EM for structural elucidation. We investigate the conformational dynamics and stabilizing features using orthogonal SPR, SEC, nanoDSF, and HDX-MS techniques to better understand how this antigen elicits superior broad neutralizing antibodies as a variant booster vaccine. This structural analysis confirms the Beta trimer preference for canonical quaternary structure with two RBD in the up position and the reversible equilibrium between the canonical spike and open trimer conformations. Moreover, this report provides a better understanding of structural differences between spike antigens contributing to differential vaccine efficacy.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Pandemias , Psicoterapia
2.
Nucleic Acids Res ; 51(21): 11732-11747, 2023 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-37870477

RESUMEN

The classical Non-Homologous End Joining (c-NHEJ) pathway is the predominant process in mammals for repairing endogenous, accidental or programmed DNA Double-Strand Breaks. c-NHEJ is regulated by several accessory factors, post-translational modifications, endogenous chemical agents and metabolites. The metabolite inositol-hexaphosphate (IP6) stimulates c-NHEJ by interacting with the Ku70-Ku80 heterodimer (Ku). We report cryo-EM structures of apo- and DNA-bound Ku in complex with IP6, at 3.5 Å and 2.74 Å resolutions respectively, and an X-ray crystallography structure of a Ku in complex with DNA and IP6 at 3.7 Å. The Ku-IP6 interaction is mediated predominantly via salt bridges at the interface of the Ku70 and Ku80 subunits. This interaction is distant from the DNA, DNA-PKcs, APLF and PAXX binding sites and in close proximity to XLF binding site. Biophysical experiments show that IP6 binding increases the thermal stability of Ku by 2°C in a DNA-dependent manner, stabilizes Ku on DNA and enhances XLF affinity for Ku. In cells, selected mutagenesis of the IP6 binding pocket reduces both Ku accrual at damaged sites and XLF enrolment in the NHEJ complex, which translate into a lower end-joining efficiency. Thus, this study defines the molecular bases of the IP6 metabolite stimulatory effect on the c-NHEJ repair activity.


Asunto(s)
Proteínas de Unión al ADN , Ácido Fítico , Animales , ADN/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/genética , Autoantígeno Ku/metabolismo , Mamíferos/genética , Humanos
3.
Cell ; 186(5): 1013-1025.e24, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36827973

RESUMEN

The emergence of drug-resistant tuberculosis has created an urgent need for new anti-tubercular agents. Here, we report the discovery of a series of macrolides called sequanamycins with outstanding in vitro and in vivo activity against Mycobacterium tuberculosis (Mtb). Sequanamycins are bacterial ribosome inhibitors that interact with the ribosome in a similar manner to classic macrolides like erythromycin and clarithromycin, but with binding characteristics that allow them to overcome the inherent macrolide resistance of Mtb. Structures of the ribosome with bound inhibitors were used to optimize sequanamycin to produce the advanced lead compound SEQ-9. SEQ-9 was efficacious in mouse models of acute and chronic TB as a single agent, and it demonstrated bactericidal activity in a murine TB infection model in combination with other TB drugs. These results support further investigation of this series as TB clinical candidates, with the potential for use in new regimens against drug-susceptible and drug-resistant TB.


Asunto(s)
Antituberculosos , Mycobacterium tuberculosis , Animales , Ratones , Antituberculosos/farmacología , Macrólidos , Farmacorresistencia Bacteriana , Claritromicina
4.
SLAS Discov ; 26(8): 1014-1019, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34238039

RESUMEN

The variety and complexity of drug targets are expanding rapidly. At the same time, there is significant interest in exploring a larger chemical space to identify new candidates. Fragment-based screening (FBS) has emerged as a popular alternative to traditional high-throughput screening campaigns to identify such drug candidates. FBS identifies hit fragments that exhibit weak interactions with the target of interest, thereby enabling the rational design of small-molecule compounds from the identified hit fragments, which serve as building blocks. This strategy reduces the number of molecules to screen while also allowing the exploration of a greater chemical space.Here we use temperature-related intensity change (TRIC) technology to perform FBS against the target MAPK/ERK kinase-1 (Mek1). TRIC describes the change in fluorescence intensity of a fluorescently labeled molecule upon a change in temperature. This intensity variation is dependent on the physicochemical environment in the vicinity of the dye and strongly affected by binding events. Thus, the detection of binding events is independent of mass, making TRIC an ideal tool for FBS.Using only 150 pmol of labeled Mek1, the authors screened 193 fragments from a prescreened library in less than 1 h of measurement time, leading to 66 hits. Among those hits, they identified more than 80% of the published top hits found using orthogonal techniques. Furthermore, TRIC allowed the identification of fragments that were of poor solubility but could be mistaken as false-positive hits in other methods.


Asunto(s)
Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Colorantes Fluorescentes , Humanos , Temperatura
5.
Commun Biol ; 3(1): 427, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32759959

RESUMEN

The mutant gdPT R9K/E129G is a genetically detoxified variant of the pertussis toxin (PTx) and represents an attractive candidate for the development of improved pertussis vaccines. The impact of the mutations on the overall protein structure and its immunogenicity has remained elusive. Here we present the crystal structure of gdPT and show that it is nearly identical to that of PTx. Hydrogen-deuterium exchange mass spectrometry revealed dynamic changes in the catalytic domain that directly impacted NAD+ binding which was confirmed by biolayer interferometry. Distal changes in dynamics were also detected in S2-S5 subunit interactions resulting in tighter packing of B-oligomer corresponding to increased thermal stability. Finally, antigen stimulation of human whole blood, analyzed by a previously unreported mass cytometry assay, indicated broader immunogenicity of gdPT compared to pertussis toxoid. These findings establish a direct link between the conserved structure of gdPT and its ability to generate a robust immune response.


Asunto(s)
Toxina del Pertussis/química , Vacuna contra la Tos Ferina/genética , Conformación Proteica , Toxoides/genética , Animales , Bordetella pertussis/genética , Bordetella pertussis/patogenicidad , Células CHO , Cricetinae , Cricetulus , Cristalografía por Rayos X , Medición de Intercambio de Deuterio , Humanos , Toxina del Pertussis/genética , Vacuna contra la Tos Ferina/química , Tos Ferina/microbiología , Tos Ferina/prevención & control
6.
J Med Chem ; 63(2): 512-528, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31721572

RESUMEN

More than 75% of breast cancers are estrogen receptor alpha (ERα) positive (ER+), and resistance to current hormone therapies occurs in one-third of ER+ patients. Tumor resistance is still ERα-dependent, but mutations usually confer constitutive activation to the hormone receptor, rendering ERα modulator drugs such as tamoxifen and aromatase inhibitors ineffective. Fulvestrant is a potent selective estrogen receptor degrader (SERD), which degrades the ERα receptor in drug-resistant tumors and has been approved for the treatment of hormone-receptor-positive metastatic breast cancer following antiestrogen therapy. However, fulvestrant shows poor pharmacokinetic properties in human, low solubility, weak permeation, and high metabolism, limiting its administration to inconvenient intramuscular injections. This Drug Annotation describes the identification and optimization of a new series of potent orally available SERDs, which led to the discovery of 6-(2,4-dichlorophenyl)-5-[4-[(3S)-1-(3-fluoropropyl)pyrrolidin-3-yl]oxyphenyl]-8,9-dihydro-7H-benzo[7]annulene-2-carboxylic acid (43d), showing promising antitumor activity in breast cancer mice xenograft models and whose properties warranted clinical evaluation.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Descubrimiento de Drogas/métodos , Pirrolidinas/síntesis química , Pirrolidinas/farmacología , Receptores de Estrógenos/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Animales , Neoplasias de la Mama/metabolismo , Cristalografía por Rayos X , Perros , Resistencia a Antineoplásicos , Femenino , Semivida , Ensayos Analíticos de Alto Rendimiento , Humanos , Ligandos , Ratones , Modelos Moleculares , Ratas , Receptores de Estrógenos/efectos de los fármacos , Moduladores Selectivos de los Receptores de Estrógeno/farmacocinética , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cells ; 8(12)2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31779273

RESUMEN

CD38 is a transmembrane glycoprotein with ectoenzymatic activity involved in regulation of migration, signal transduction, and receptor-mediated adhesion. CD38 is highly expressed on various malignant cells, including multiple myeloma (MM), and at relatively low levels in other tissues, making it a suitable target for therapeutic antibodies. Several anti-CD38 therapies have been, or are being, developed for the treatment of MM, including daratumumab and isatuximab (SAR650984), respectively. Studies have shown that anti-CD38 therapies are effective in the treatment of relapsed/refractory MM and are well tolerated, with infusion reactions being the most common side effects. They can be used as monotherapy or in combination with immunomodulatory agents, such as pomalidomide, or proteasome inhibitors to potentiate their activity. Here we examine isatuximab and several anti-CD38 agents in development that were generated using new antibody engineering techniques and that may lead to more effective CD38 targeting. We also summarize trials assessing these antibodies in MM, other malignancies, and solid organ transplantation. Finally, we propose that further research on the mechanisms of resistance to anti-CD38 therapy and the development of biomarkers and new backbone regimens with CD38 antibodies will be important steps in building more personalized treatment for patients with MM.


Asunto(s)
ADP-Ribosil Ciclasa 1/antagonistas & inhibidores , Anticuerpos Monoclonales/farmacología , Antineoplásicos Inmunológicos/farmacología , Glicoproteínas de Membrana/antagonistas & inhibidores , ADP-Ribosil Ciclasa 1/química , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Citotoxicidad Celular Dependiente de Anticuerpos , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/uso terapéutico , Humanos , Glicoproteínas de Membrana/química , Terapia Molecular Dirigida , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/inmunología , Mieloma Múltiple/metabolismo , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
8.
ChemMedChem ; 14(11): 1115-1127, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-30925009

RESUMEN

Ligand-based NMR screening represents a powerful method in fragment-based drug discovery for the identification of chemical matter interacting with the receptor of interest. The large dynamic range of these methods allows the detection of weakly binding ligands. However, the methodology has not been extensively used for quantifying the strength of these interactions. This knowledge is important for ranking fragments according to their binding strength and for prioritizing structure-based and medicinal chemistry activities. Rapid NMR methods for measuring the dissociation constant in direct and competition modes are presented here. The theory underpinning these methods are presented, along with their application to the measurement of the binding affinities of several ligands of the heat shock protein 90.


Asunto(s)
Proteínas HSP90 de Choque Térmico/química , Descubrimiento de Drogas , Ligandos , Espectroscopía de Resonancia Magnética , Estructura Molecular
9.
Med Sci (Paris) ; 35(12): 1171-1174, 2019 Dec.
Artículo en Francés | MEDLINE | ID: mdl-31903933

RESUMEN

Therapeutic antibodies generation has to be faster with less development costs. This requires combination of in silico predictions associated with cutting edge screening and characterization technologies. Here, non-exhaustive examples illustrate this simultaneity need.


TITLE: Exemples d'études de développabilité apportant un éclairage à la prise de décision. ABSTRACT: De nos jours, la génération d'anticorps thérapeutiques doit être plus rapide avec des coûts de développement moins importants. Pour cela, des prédictions in silico sont associées à des technologies de criblage et de caractérisation de pointe. Les exemples choisis ici sont non-exhaustifs mais illustrent ce besoin de travailler en parallèle.


Asunto(s)
Anticuerpos Monoclonales , Simulación por Computador , Toma de Decisiones , Diseño de Fármacos , Desarrollo de Medicamentos , Evaluación Preclínica de Medicamentos , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/economía , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales/uso terapéutico , Desarrollo de Medicamentos/economía , Desarrollo de Medicamentos/métodos , Desarrollo de Medicamentos/organización & administración , Desarrollo de Medicamentos/normas , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/normas , Industria Farmacéutica/economía , Industria Farmacéutica/métodos , Industria Farmacéutica/normas , Ensayos Analíticos de Alto Rendimiento/economía , Ensayos Analíticos de Alto Rendimiento/métodos , Ensayos Analíticos de Alto Rendimiento/normas , Humanos
10.
J Chem Theory Comput ; 14(7): 3859-3869, 2018 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-29768913

RESUMEN

Drug-target residence time (τ), one of the main determinants of drug efficacy, remains highly challenging to predict computationally and, therefore, is usually not considered in the early stages of drug design. Here, we present an efficient computational method, τ-random acceleration molecular dynamics (τRAMD), for the ranking of drug candidates by their residence time and obtaining insights into ligand-target dissociation mechanisms. We assessed τRAMD on a data set of 70 diverse drug-like ligands of the N-terminal domain of HSP90α, a pharmaceutically important target with a highly flexible binding site, obtaining computed relative residence times with an accuracy of about 2.3τ for 78% of the compounds and less than 2.0τ within congeneric series. Analysis of dissociation trajectories reveals features that affect ligand unbinding rates, including transient polar interactions and steric hindrance. These results suggest that τRAMD will be widely applicable as a computationally efficient aid to improving drug residence times during lead optimization.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Sitios de Unión , Descubrimiento de Drogas , Proteínas HSP90 de Choque Térmico/química , Humanos , Cinética , Ligandos , Simulación de Dinámica Molecular , Unión Proteica , Dominios Proteicos
11.
J Med Chem ; 59(15): 7066-74, 2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-27355974

RESUMEN

The HGF/MET pathway is frequently activated in a variety of cancer types. Several selective small molecule inhibitors of the MET kinase are currently in clinical evaluation, in particular for NSCLC, liver, and gastric cancer patients. We report herein the discovery of a series of triazolopyridazines that are selective inhibitors of wild-type (WT) MET kinase and several clinically relevant mutants. We provide insight into their mode of binding and report unprecedented crystal structures of the Y1230H variant. A multiparametric chemical optimization approach allowed the identification of compound 12 (SAR125844) as a development candidate. In this chemical series, absence of CYP3A4 inhibition was obtained at the expense of satisfactory oral absorption. Compound 12, a promising parenteral agent for the treatment of MET-dependent cancers, promoted sustained target engagement at tolerated doses in a human xenograft tumor model. Preclinical pharmacokinetics conducted in several species were predictive for the observed pharmacokinetic behavior of 12 in cancer patients.


Asunto(s)
Benzotiazoles/farmacología , Benzotiazoles/farmacocinética , Descubrimiento de Drogas , Neoplasias Experimentales/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Urea/análogos & derivados , Animales , Benzotiazoles/administración & dosificación , Benzotiazoles/química , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Modelos Moleculares , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-met/metabolismo , Relación Estructura-Actividad , Urea/administración & dosificación , Urea/química , Urea/farmacocinética , Urea/farmacología
12.
MAbs ; 8(5): 867-78, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26984268

RESUMEN

Bispecific immunoglobulins (Igs) typically contain at least two distinct variable domains (Fv) that bind to two different target proteins. They are conceived to facilitate clinical development of biotherapeutic agents for diseases where improved clinical outcome is obtained or expected by combination therapy compared to treatment by single agents. Almost all existing formats are linear in their concept and differ widely in drug-like and manufacture-related properties. To overcome their major limitations, we designed cross-over dual variable Ig-like proteins (CODV-Ig). Their design is akin to the design of circularly closed repeat architectures. Indeed, initial results showed that the traditional approach of utilizing (G4S)x linkers for biotherapeutics design does not identify functional CODV-Igs. Therefore, we applied an unprecedented molecular modeling strategy for linker design that consistently results in CODV-Igs with excellent biochemical and biophysical properties. CODV architecture results in a circular self-contained structure functioning as a self-supporting truss that maintains the parental antibody affinities for both antigens without positional effects. The format is universally suitable for therapeutic applications targeting both circulating and membrane-localized proteins. Due to the full functionality of the Fc domains, serum half-life extension as well as antibody- or complement-dependent cytotoxicity may support biological efficiency of CODV-Igs. We show that judicious choice in combination of epitopes and paratope orientations of bispecific biotherapeutics is anticipated to be critical for clinical outcome. Uniting the major advantages of alternative bispecific biotherapeutics, CODV-Igs are applicable in a wide range of disease areas for fast-track multi-parametric drug optimization.


Asunto(s)
Anticuerpos Biespecíficos/biosíntesis , Diseño de Fármacos , Modelos Moleculares , Humanos , Ingeniería de Proteínas/métodos
13.
J Biomol Screen ; 21(4): 414-21, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26637553

RESUMEN

Fragment-based lead discovery has proved to be an effective alternative to high-throughput screenings in identifying chemical matter that can be developed into robust lead compounds. The search for optimal combinations of biophysical techniques that can correctly and efficiently identify and quantify binding can be challenging due to the physicochemical properties of fragments. In order to minimize the time and costs of screening, optimal combinations of biophysical techniques with maximal information content, sensitivity, and robustness are needed. Here we describe an approach utilizing automated microscale thermophoresis (MST) affinity screening to identify fragments active against MEK1 kinase. MST identified multiple hits that were confirmed by X-ray crystallography but not detected by orthogonal methods. Furthermore, MST also provided information about ligand-induced aggregation and protein denaturation. The technique delivered a large number of binders while reducing experimentation time and sample consumption, demonstrating the potential of MST to execute and maximize the efficacy of fragment screening campaigns.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , MAP Quinasa Quinasa 1/química , Inhibidores de Proteínas Quinasas/química , Bibliotecas de Moléculas Pequeñas/química , Cristalografía por Rayos X , Difusión , Descubrimiento de Drogas , Expresión Génica , Ensayos Analíticos de Alto Rendimiento/instrumentación , Humanos , Ligandos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Modelos Moleculares , Unión Proteica , Desnaturalización Proteica , Resonancia por Plasmón de Superficie , Temperatura
14.
Sci Rep ; 5: 7642, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25560837

RESUMEN

Monoacylglycerol lipase (MAGL) represents a primary degradation enzyme of the endogenous cannabinoid (eCB), 2-arachidonoyglycerol (2-AG). This study reports a potent covalent MAGL inhibitor, SAR127303. The compound behaves as a selective and competitive inhibitor of mouse and human MAGL, which potently elevates hippocampal levels of 2-AG in mice. In vivo, SAR127303 produces antinociceptive effects in assays of inflammatory and visceral pain. In addition, the drug alters learning performance in several assays related to episodic, working and spatial memory. Moreover, long term potentiation (LTP) of CA1 synaptic transmission and acetylcholine release in the hippocampus, two hallmarks of memory function, are both decreased by SAR127303. Although inactive in acute seizure tests, repeated administration of SAR127303 delays the acquisition and decreases kindled seizures in mice, indicating that the drug slows down epileptogenesis, a finding deserving further investigation to evaluate the potential of MAGL inhibitors as antiepileptics. However, the observation that 2-AG hydrolysis blockade alters learning and memory performance, suggests that such drugs may have limited value as therapeutic agents.


Asunto(s)
Analgésicos/farmacología , Ácidos Araquidónicos/metabolismo , Carbamatos/farmacología , Endocannabinoides/metabolismo , Glicéridos/metabolismo , Aprendizaje/efectos de los fármacos , Memoria a Corto Plazo/efectos de los fármacos , Monoacilglicerol Lipasas/metabolismo , Sulfonamidas/farmacología , Acetilcolina/metabolismo , Administración Oral , Analgésicos/química , Analgésicos/uso terapéutico , Animales , Ácidos Araquidónicos/química , Sitios de Unión , Encéfalo/metabolismo , Antagonistas de Receptores de Cannabinoides/farmacología , Carbamatos/química , Carbamatos/uso terapéutico , Cromatografía Líquida de Alta Presión , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Estimulación Eléctrica , Endocannabinoides/química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Glicéridos/química , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Hidrólisis , Técnicas In Vitro , Potenciación a Largo Plazo/efectos de los fármacos , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Monoacilglicerol Lipasas/antagonistas & inhibidores , Dolor/tratamiento farmacológico , Dolor/patología , Piperidinas/farmacología , Estructura Terciaria de Proteína , Pirazoles/farmacología , Rimonabant , Convulsiones/tratamiento farmacológico , Convulsiones/patología , Sulfonamidas/química , Sulfonamidas/uso terapéutico
15.
J Biol Chem ; 290(2): 762-74, 2015 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-25391653

RESUMEN

Cancer-associated point mutations in isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) confer a neomorphic enzymatic activity: the reduction of α-ketoglutarate to d-2-hydroxyglutaric acid, which is proposed to act as an oncogenic metabolite by inducing hypermethylation of histones and DNA. Although selective inhibitors of mutant IDH1 and IDH2 have been identified and are currently under investigation as potential cancer therapeutics, the mechanistic basis for their selectivity is not yet well understood. A high throughput screen for selective inhibitors of IDH1 bearing the oncogenic mutation R132H identified compound 1, a bis-imidazole phenol that inhibits d-2-hydroxyglutaric acid production in cells. We investigated the mode of inhibition of compound 1 and a previously published IDH1 mutant inhibitor with a different chemical scaffold. Steady-state kinetics and biophysical studies show that both of these compounds selectively inhibit mutant IDH1 by binding to an allosteric site and that inhibition is competitive with respect to Mg(2+). A crystal structure of compound 1 complexed with R132H IDH1 indicates that the inhibitor binds at the dimer interface and makes direct contact with a residue involved in binding of the catalytically essential divalent cation. These results show that targeting a divalent cation binding residue can enable selective inhibition of mutant IDH1 and suggest that differences in magnesium binding between wild-type and mutant enzymes may contribute to the inhibitors' selectivity for the mutant enzyme.


Asunto(s)
Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Isocitrato Deshidrogenasa/química , Neoplasias/tratamiento farmacológico , Sitio Alostérico , Cristalografía por Rayos X , Metilación de ADN/genética , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/uso terapéutico , Escherichia coli , Regulación Neoplásica de la Expresión Génica , Humanos , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Isocitrato Deshidrogenasa/biosíntesis , Isocitrato Deshidrogenasa/genética , Magnesio/química , Proteínas Mutantes/química , Proteínas Mutantes/genética , Neoplasias/genética , Neoplasias/patología , Conformación Proteica
16.
Protein Sci ; 23(12): 1698-707, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25209176

RESUMEN

Various important biological pathways are modulated by TGFß isoforms; as such they are potential targets for therapeutic intervention. Fresolimumab, also known as GC1008, is a pan-TGFß neutralizing antibody that has been tested clinically for several indications including an ongoing trial for focal segmental glomerulosclerosis. The structure of the antigen-binding fragment of fresolimumab (GC1008 Fab) in complex with TGFß3 has been reported previously, but the structural capacity of fresolimumab to accommodate tight interactions with TGFß1 and TGFß2 was insufficiently understood. We report the crystal structure of the single-chain variable fragment of fresolimumab (GC1008 scFv) in complex with target TGFß1 to a resolution of 3.00 Å and the crystal structure of GC1008 Fab in complex with TGFß2 to 2.83 Å. The structures provide further insight into the details of TGFß recognition by fresolimumab, give a clear indication of the determinants of fresolimumab pan-specificity and provide potential starting points for the development of isoform-specific antibodies using a fresolimumab scaffold.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos/inmunología , Reacciones Antígeno-Anticuerpo/inmunología , Factor de Crecimiento Transformador beta/química , Factor de Crecimiento Transformador beta/inmunología , Anticuerpos Monoclonales Humanizados , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/inmunología , Cristalografía por Rayos X , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/inmunología , Modelos Moleculares , Conformación Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/inmunología , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/inmunología
17.
J Med Chem ; 57(3): 903-20, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24387221

RESUMEN

Compelling molecular biology publications have reported the implication of phosphoinositide kinase PI3Kß in PTEN-deficient cell line growth and proliferation. These findings supported a scientific rationale for the development of PI3Kß-specific inhibitors for the treatment of PTEN-deficient cancers. This paper describes the discovery of 2-[2-(2,3-dihydro-indol-1-yl)-2-oxo-ethyl]-6-morpholin-4-yl-3H-pyrimidin-4-one (7) and the optimization of this new series of active and selective pyrimidone indoline amide PI3Kß inhibitors. 2-[2-(2-Methyl-2,3-dihydro-indol-1-yl)-2-oxo-ethyl]-6-morpholin-4-yl-3H-pyrimidin-4-one (28), identified following a carefully designed methyl scan, displayed improved physicochemical and in vitro pharmacokinetic properties. Structural biology efforts enabled the acquisition of the first X-ray cocrystal structure of p110ß with the selective inhibitor compound 28 bound to the ATP site. The nonplanar binding mode described herein is consistent with observed structure-activity relationship for the series. Compound 28 demonstrated significant in vivo activity in a UACC-62 xenograft model in mice, warranting further preclinical investigation. Following successful development, compound 28 entered phase I/Ib clinical trial in patients with advanced cancer.


Asunto(s)
Antineoplásicos/química , Indoles/química , Neoplasias/tratamiento farmacológico , Fosfohidrolasa PTEN/deficiencia , Inhibidores de las Quinasa Fosfoinosítidos-3 , Pirimidinonas/química , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Disponibilidad Biológica , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Cristalografía por Rayos X , Perros , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Xenoinjertos , Humanos , Indoles/farmacocinética , Indoles/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Microsomas Hepáticos/metabolismo , Conformación Molecular , Simulación del Acoplamiento Molecular , Trasplante de Neoplasias , Neoplasias/enzimología , Fosfohidrolasa PTEN/genética , Unión Proteica , Pirimidinonas/farmacocinética , Pirimidinonas/farmacología , Ratas , Ratas Desnudas , Solubilidad , Estereoisomerismo , Relación Estructura-Actividad
18.
Bioorg Med Chem Lett ; 23(12): 3620-6, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23648182

RESUMEN

We report the analysis of an in-house fragment screening campaign for the oncology target MEK1. The application of virtual screening (VS) as a primary fragment screening approach, followed by biophysical validation using differential screening fluorimetry (DSF), with resultant binding mode determination by X-ray crystallography (X-ray), is presented as the most time and cost-effective combination of in silico and in vitro methods to identify fragments. We demonstrate the effectiveness of the VS-DSF workflow for the early identification of fragments to both 'jump-start' the drug discovery project and to complement biochemical screening data.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Fluorometría/métodos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/química , Humanos , MAP Quinasa Quinasa 1/química , MAP Quinasa Quinasa 1/metabolismo , Modelos Moleculares , Fosforilación , Relación Estructura-Actividad
19.
J Med Chem ; 55(10): 4788-805, 2012 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-22524426

RESUMEN

Most of the phosphoinositide-3 kinase (PI3K) kinase inhibitors currently in clinical trials for cancer treatment exhibit pan PI3K isoform profiles. Single PI3K isoforms differentially control tumorigenesis, and PI3Kß has emerged as the isoform involved in the tumorigenicity of PTEN-deficient tumors. Herein we describe the discovery and optimization of a new series of benzimidazole- and benzoxazole-pyrimidones as small molecular mass PI3Kß-selective inhibitors. Starting with compound 5 obtained from a one-pot reaction via a novel intermediate 1, medicinal chemistry optimization led to the discovery of compound 8, which showed a significant activity and selectivity for PI3Kß and adequate in vitro pharmacokinetic properties. The X-ray costructure of compound 8 in PI3Kδ showed key interactions and structural features supporting the observed PI3Kß isoform selectivity. Compound 8 achieved sustained target modulation and tumor growth delay at well tolerated doses when administered orally to SCID mice implanted with PTEN-deficient human tumor xenografts.


Asunto(s)
Antineoplásicos/síntesis química , Bencimidazoles/síntesis química , Benzoxazoles/síntesis química , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Neoplasias Experimentales/tratamiento farmacológico , Fosfohidrolasa PTEN/deficiencia , Pirimidinonas/síntesis química , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Bencimidazoles/farmacocinética , Bencimidazoles/farmacología , Benzoxazoles/farmacocinética , Benzoxazoles/farmacología , Línea Celular Tumoral , Cristalografía por Rayos X , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Humanos , Isoenzimas/antagonistas & inhibidores , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Ratones , Ratones SCID , Modelos Moleculares , Estructura Molecular , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/patología , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinonas/farmacocinética , Pirimidinonas/farmacología , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Curr Biol ; 20(3): 198-208, 2010 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-20116244

RESUMEN

BACKGROUND: Cell-cell adhesion and intracellular trafficking are regulated by signaling pathways from small GTPases of the Rho, Arf, and Rab subfamilies. How signaling from distinct small GTPases are integrated in a given process is poorly understood. RESULTS: We find that a TBC/RabGAP protein, Armus, integrates signaling between Arf6, Rac1, and Rab7 during junction disassembly. Armus binds specifically to activated Rac1 and its C-terminal TBC/RabGAP domain inactivates Rab7. Thus, Armus is a novel Rac1 effector and a bona fide GAP for Rab7 in vitro and in vivo, a unique and previously unreported combination. Arf6 activation efficiently disrupts cell-cell contacts and is known to activate Rac1 and Rab7. Arf6-induced E-cadherin degradation is efficiently blocked by expression of Armus C-terminal domain or after Armus RNAi. Coexpression of Arf6 with dominant-negative Rab7 or Rac1 also inhibits junction disassembly. Importantly, Armus RabGAP expression also prevents EGF-induced scattering in keratinocytes, a process shown here to require Arf6, Rac1, and Rab7 function. To our knowledge, this is the first report to demonstrate a molecular and functional link between Rac1 and Rab7. CONCLUSIONS: Our data indicate that active Rac1 recruits Armus to locally inactivate Rab7 and facilitate E-cadherin degradation in lysosomes. Thus, the integration of Rac1 and Rab7 activities by Armus provides an important regulatory node for E-cadherin turnover and stability of cell-cell contacts.


Asunto(s)
Cadherinas/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Proteínas de Unión al GTP rab/antagonistas & inhibidores , Proteína de Unión al GTP rac1/metabolismo , Animales , Células COS , Proteínas Portadoras/genética , Comunicación Celular , Células Cultivadas , Chlorocebus aethiops , Proteínas Activadoras de GTPasa/genética , Humanos , Uniones Intercelulares/metabolismo , Queratinocitos/metabolismo , Modelos Biológicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Proteínas de Unión a GTP rab7
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...