Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Phys Rev E ; 109(1-2): 015002, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38366536

RESUMO

Theoretical analysis of Richtmyer-Meshkov instability (RMI) experiments for solid strength shows that the strain rate for a given shock should be inversely proportional to the length scale of the sine wave perturbations when η_{0}k, the nondimensional amplitude to wavelength ratio, is held fixed. To isolate the effect of strain rate on strength, free-surface RMI specimens of annealed copper were prepared with three perturbation regions with the same η_{0}k but different length scales, characterized by the wavelength λ varying by a factor of 4.9 from 65 to 130 to 320µm. Three such targets with different fixed η_{0}k^{'}s were impacted to a shock pressure of 25 GPa, and the instability evolution was measured with photon Doppler velocimetry. Strengths estimated by comparing hydrocode simulation to the data increased from 700 to 1200 MPa as λ decreased. The different η_{0}k targets exercised increasing amounts of plastic strain yet showed no evidence of strain hardening. Physical regime sensitivity analysis determined that for 320-65µm wavelength perturbations, the effective strain rates increased from 8.7×10^{6} to 3.3×10^{7}s^{-1}, a factor of 3.8. Thus, the predicted strain rate scaling was mostly achieved but slightly suppressed by increased strength at higher rates. The RMI strength estimates were plotted against constitutive testing data on copper from the literature to show striking evidence of the strength upturn at higher strain rates.

2.
J Med Chem ; 66(1): 641-656, 2023 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-36548390

RESUMO

Therapeutic interventions are being developed for Huntington's disease (HD), a hallmark of which is mutant huntingtin protein (mHTT) aggregates. Following the advancement to human testing of two [11C]-PET ligands for aggregated mHTT, attributes for further optimization were identified. We replaced the pyridazinone ring of CHDI-180 with a pyrimidine ring and minimized off-target binding using brain homogenate derived from Alzheimer's disease patients. The major in vivo metabolic pathway via aldehyde oxidase was blocked with a 2-methyl group on the pyrimidine ring. A strategically placed ring-nitrogen on the benzoxazole core ensured high free fraction in the brain without introducing efflux. Replacing a methoxy pendant with a fluoro-ethoxy group and introducing deuterium atoms suppressed oxidative defluorination and accumulation of [18F]-signal in bones. The resulting PET ligand, CHDI-650, shows a rapid brain uptake and washout profile in non-human primates and is now being advanced to human testing.


Assuntos
Doença de Huntington , Tomografia por Emissão de Pósitrons , Animais , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Ligantes , Tomografia por Emissão de Pósitrons/métodos , Doença de Huntington/diagnóstico por imagem , Doença de Huntington/tratamento farmacológico , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo
3.
Drug Metab Dispos ; 40(12): 2297-306, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22942319

RESUMO

Understanding whether regulation of tryptophan metabolites can ameliorate neurodegeneration is of high interest to investigators. A recent publication describes 3,4-dimethoxy-N-(4-(3-nitrophenyl)-5-(piperidin-1-ylmethyl)thiazol-2-yl)benzenesulfonamide (JM6) as a novel prodrug for the kynurenine 3-monooxygenase (KMO) inhibitor 3,4-dimethoxy-N-(4-(3-nitrophenyl)thiazol-2-yl)benzenesulfonamide (Ro-61-8048) that elicits therapeutic effects in mouse models of Huntington's and Alzheimer's diseases (Cell 145:863-874, 2011). Our evaluation of the metabolism and pharmacokinetics of JM6 and Ro-61-8048 indicate instead that Ro-61-8048 concentrations in mouse plasma after JM6 administration originate from a Ro-61-8048 impurity (<0.1%) in JM6. After a 0.05 mg/kg Ro-61-8048 oral dose alone or coadministered with 10 mg/kg JM6 to mice, the Ro-61-8048 areas under the concentration-time curves (AUCs) from 0 to infinity were similar (4300 and 4900 nM × h, respectively), indicating no detectable contributions of JM6 metabolism to the Ro-61-8048 AUCs. JM6 was stable in incubations under acidic conditions and Ro-61-8048 was not a product of JM6 metabolism in vitro (plasma, blood, or hepatic models). Species differences in the quantitative rate of oxidative metabolism indicate that major circulating JM6 metabolite(s) in mice are unlikely to be major in humans: JM6 is rapidly metabolized via the piperidyl moiety in mouse (forming an iminium ion reactive intermediate) but is slowly metabolized in human (in vitro), primarily via O-dealkylation at the phenyl ring. Our data indicate that JM6 is not a prodrug for Ro-61-8048 and is not a potent KMO inhibitor.


Assuntos
Pró-Fármacos/farmacocinética , Sulfonamidas/farmacocinética , Tiazóis/farmacocinética , Animais , Área Sob a Curva , Linhagem Celular , Cães , Hepatócitos/enzimologia , Hepatócitos/metabolismo , Humanos , Células Madin Darby de Rim Canino , Masculino , Desintoxicação Metabólica Fase I , Camundongos , Camundongos Endogâmicos C57BL , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Ratos , Sulfonamidas/administração & dosagem , Tiazóis/administração & dosagem
4.
Sci Transl Med ; 14(630): eabm3682, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-35108063

RESUMO

Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder caused by a CAG trinucleotide expansion in the huntingtin (HTT) gene that encodes the pathologic mutant HTT (mHTT) protein with an expanded polyglutamine (polyQ) tract. Whereas several therapeutic programs targeting mHTT expression have advanced to clinical evaluation, methods to visualize mHTT protein species in the living brain are lacking. Here, we demonstrate the development and characterization of a positron emission tomography (PET) imaging radioligand with high affinity and selectivity for mHTT aggregates. This small molecule radiolabeled with 11C ([11C]CHDI-180R) allowed noninvasive monitoring of mHTT pathology in the brain and could track region- and time-dependent suppression of mHTT in response to therapeutic interventions targeting mHTT expression in a rodent model. We further showed that in these animals, therapeutic agents that lowered mHTT in the striatum had a functional restorative effect that could be measured by preservation of striatal imaging markers, enabling a translational path to assess the functional effect of mHTT lowering.


Assuntos
Doença de Huntington , Doenças Neurodegenerativas , Animais , Corpo Estriado/diagnóstico por imagem , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/diagnóstico por imagem , Doença de Huntington/genética , Doença de Huntington/metabolismo , Ligantes , Doenças Neurodegenerativas/patologia
5.
Sci Rep ; 11(1): 17977, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34504195

RESUMO

Huntington's disease (HD) is caused by a CAG trinucleotide repeat expansion in the first exon of the huntingtin (HTT) gene coding for the huntingtin (HTT) protein. The misfolding and consequential aggregation of CAG-expanded mutant HTT (mHTT) underpin HD pathology. Our interest in the life cycle of HTT led us to consider the development of high-affinity small-molecule binders of HTT oligomerized/amyloid-containing species that could serve as either cellular and in vivo imaging tools or potential therapeutic agents. We recently reported the development of PET tracers CHDI-180 and CHDI-626 as suitable for imaging mHTT aggregates, and here we present an in-depth pharmacological investigation of their binding characteristics. We have implemented an array of in vitro and ex vivo radiometric binding assays using recombinant HTT, brain homogenate-derived HTT aggregates, and brain sections from mouse HD models and humans post-mortem to investigate binding affinities and selectivity against other pathological proteins from indications such as Alzheimer's disease and spinocerebellar ataxia 1. Radioligand binding assays and autoradiography studies using brain homogenates and tissue sections from HD mouse models showed that CHDI-180 and CHDI-626 specifically bind mHTT aggregates that accumulate with age and disease progression. Finally, we characterized CHDI-180 and CHDI-626 regarding their off-target selectivity and binding affinity to beta amyloid plaques in brain sections and homogenates from Alzheimer's disease patients.


Assuntos
Proteína Huntingtina/metabolismo , Doença de Huntington/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Agregados Proteicos/genética , Agregação Patológica de Proteínas/diagnóstico por imagem , Compostos Radiofarmacêuticos/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Autorradiografia/métodos , Encéfalo/metabolismo , Modelos Animais de Doenças , Humanos , Proteína Huntingtina/genética , Doença de Huntington/patologia , Imuno-Histoquímica/métodos , Camundongos , Camundongos Transgênicos , Radioisótopos de Nitrogênio/metabolismo , Traçadores Radioativos , Ensaio Radioligante/métodos , Proteínas Recombinantes/metabolismo
6.
J Med Chem ; 64(16): 12003-12021, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34351166

RESUMO

The expanded polyglutamine-containing mutant huntingtin (mHTT) protein is implicated in neuronal degeneration of medium spiny neurons in Huntington's disease (HD) for which multiple therapeutic approaches are currently being evaluated to eliminate or reduce mHTT. Development of effective and orthogonal biomarkers will ensure accurate assessment of the safety and efficacy of pharmacologic interventions. We have identified and optimized a class of ligands that bind to oligomerized/aggregated mHTT, which is a hallmark in the HD postmortem brain. These ligands are potentially useful imaging biomarkers for HD therapeutic development in both preclinical and clinical settings. We describe here the optimization of the benzo[4,5]imidazo[1,2-a]pyrimidine series that show selective binding to mHTT aggregates over Aß- and/or tau-aggregates associated with Alzheimer's disease pathology. Compound [11C]-2 was selected as a clinical candidate based on its high free fraction in the brain, specific binding in the HD mouse model, and rapid brain uptake/washout in nonhuman primate positron emission tomography imaging studies.


Assuntos
Encéfalo/diagnóstico por imagem , Compostos Heterocíclicos com 3 Anéis/química , Proteína Huntingtina/metabolismo , Agregados Proteicos/fisiologia , Piridinas/química , Compostos Radiofarmacêuticos/química , Doença de Alzheimer , Animais , Biomarcadores/metabolismo , Encéfalo/metabolismo , Radioisótopos de Carbono/química , Feminino , Compostos Heterocíclicos com 3 Anéis/síntese química , Compostos Heterocíclicos com 3 Anéis/farmacocinética , Humanos , Macaca fascicularis , Masculino , Camundongos Endogâmicos C57BL , Estrutura Molecular , Tomografia por Emissão de Pósitrons , Piridinas/síntese química , Piridinas/farmacocinética , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/farmacocinética , Ratos Sprague-Dawley , Relação Estrutura-Atividade
7.
J Med Chem ; 63(15): 8608-8633, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32662649

RESUMO

Mutant huntingtin (mHTT) protein carrying the elongated N-terminal polyglutamine (polyQ) tract misfolds and forms protein aggregates characteristic of Huntington's disease (HD) pathology. A high-affinity ligand specific for mHTT aggregates could serve as a positron emission tomography (PET) imaging biomarker for HD therapeutic development and disease progression. To identify such compounds with binding affinity for polyQ aggregates, we embarked on systematic structural activity studies; lead optimization of aggregate-binding affinity, unbound fractions in brain, permeability, and low efflux culminated in the discovery of compound 1, which exhibited target engagement in autoradiography (ARG) studies in brain slices from HD mouse models and postmortem human HD samples. PET imaging studies with 11C-labeled 1 in both HD mice and WT nonhuman primates (NHPs) demonstrated that the right-hand-side labeled ligand [11C]-1R (CHDI-180R) is a suitable PET tracer for imaging of mHTT aggregates. [11C]-1R is now being advanced to human trials as a first-in-class HD PET radiotracer.


Assuntos
Proteína Huntingtina/análise , Doença de Huntington/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Agregação Patológica de Proteínas/diagnóstico por imagem , Animais , Modelos Animais de Doenças , Cães , Feminino , Humanos , Proteína Huntingtina/genética , Doença de Huntington/genética , Ligantes , Células Madin Darby de Rim Canino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Peptídeos/genética , Agregação Patológica de Proteínas/genética , Compostos Radiofarmacêuticos/análise , Ratos Sprague-Dawley
8.
Phys Rev E ; 100(5-1): 053002, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31869927

RESUMO

Recently, Richtmyer-Meshkov instability (RMI) experiments driven by high explosives and fielded with perturbations on a free surface have been used to study strength at extreme strain rates and near zero pressure. The RMI experiments reported here used impact loading, which is experimentally simpler, more accurate to analyze, and which also allows the exploration of a wider range of conditions. Three experiments were performed on tantalum at shock stresses from 20 to 34 GPa, with six different perturbation sizes at each shock level, making this the most comprehensive set of strength-focused RMI experiments reported to date on any material. The resulting estimated average strengths of 1200-1400 MPa at strain rates of 10^{7}/s exceeded, by 40% or more, a common power law extrapolation from data at strain rates below 10^{4}/s. Taken together with other data in the literature that show much higher strength at simultaneous high rates and high pressure, these RMI data isolated effects and indicated that, in the range of conditions examined, the pressure effects are more significant than rate effects.

9.
J Med Chem ; 58(3): 1159-83, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25590515

RESUMO

We report on the development of a series of pyrimidine carboxylic acids that are potent and selective inhibitors of kynurenine monooxygenase and competitive for kynurenine. We describe the SAR for this novel series and report on their inhibition of KMO activity in biochemical and cellular assays and their selectivity against other kynurenine pathway enzymes. We describe the optimization process that led to the identification of a program lead compound with a suitable ADME/PK profile for therapeutic development. We demonstrate that systemic inhibition of KMO in vivo with this lead compound provides pharmacodynamic evidence for modulation of kynurenine pathway metabolites both in the periphery and in the central nervous system.


Assuntos
Inibidores Enzimáticos/farmacologia , Doença de Huntington/tratamento farmacológico , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Pirimidinas/farmacologia , Animais , Células CHO , Proliferação de Células/efeitos dos fármacos , Cricetulus , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Doença de Huntington/metabolismo , Cinurenina/metabolismo , Quinurenina 3-Mono-Oxigenase/metabolismo , Camundongos , Modelos Moleculares , Estrutura Molecular , Pirimidinas/síntese química , Pirimidinas/química , Ratos , Relação Estrutura-Atividade
10.
J Biomol Screen ; 18(8): 879-89, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23690293

RESUMO

Kynurenine monooxygenase (KMO) catalyzes the conversion of kynurenine to 3-hydroxykynurenine. Modulation of KMO activity has been implicated in several neurodegenerative diseases, including Huntington disease. Our goal is to develop potent and selective small-molecule KMO inhibitors with suitable pharmacokinetic characteristics for in vivo proof-of-concept studies and subsequent clinical development. We developed a comprehensive panel of biochemical and cell-based assays that use liquid chromatography/tandem mass spectrometry to quantify unlabeled kynurenine and 3-hydroxykynurenine. We describe assays to measure KMO inhibition in cell and tissue extracts, as well as cellular assays including heterologous cell lines and primary rat microglia and human peripheral blood mononuclear cells.


Assuntos
Ensaios Enzimáticos/métodos , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Quinurenina 3-Mono-Oxigenase/metabolismo , Animais , Células CHO , Linhagem Celular , Cromatografia Líquida/métodos , Cricetulus , Cães , Descoberta de Drogas , Células HEK293 , Humanos , Doença de Huntington/tratamento farmacológico , Doença de Huntington/metabolismo , Cinurenina/análogos & derivados , Cinurenina/biossíntese , Cinurenina/metabolismo , Quinurenina 3-Mono-Oxigenase/química , Leucócitos Mononucleares/efeitos dos fármacos , Macaca fascicularis , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Wistar , Espectrometria de Massas em Tandem/métodos
11.
ACS Med Chem Lett ; 3(12): 1024-8, 2012 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-24900424

RESUMO

We report a series of irreversible transglutaminase 2 inhibitors starting from a known lysine dipeptide bearing an acrylamide warhead. We established new SARs resulting in compounds demonstrating improved potency and better physical and calculated properties. Transglutaminase selectivity profiling and in vitro ADME properties of selected compounds are also reported.

12.
ACS Med Chem Lett ; 3(9): 731-5, 2012 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-24900540

RESUMO

A new series of potent TG2 inhibitors are reported that employ a 4-aminopiperidine core bearing an acrylamide warhead. We establish the structure-activity relationship of this new series and report on the transglutaminase selectivity and in vitro ADME properties of selected compounds. We demonstrate that the compounds do not conjugate glutathione in an in vitro setting and have superior plasma stability over our previous series.

13.
J Med Chem ; 55(3): 1021-46, 2012 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-22224594

RESUMO

Tissue transglutaminase 2 (TG2) is a multifunctional protein primarily known for its calcium-dependent enzymatic protein cross-linking activity via isopeptide bond formation between glutamine and lysine residues. TG2 overexpression and activity have been found to be associated with Huntington's disease (HD); specifically, TG2 is up-regulated in the brains of HD patients and in animal models of the disease. Interestingly, genetic deletion of TG2 in two different HD mouse models, R6/1 and R6/2, results in improved phenotypes including a reduction in neuronal death and prolonged survival. Starting with phenylacrylamide screening hit 7d, we describe the SAR of this series leading to potent and selective TG2 inhibitors. The suitability of the compounds as in vitro tools to elucidate the biology of TG2 was demonstrated through mode of inhibition studies, characterization of druglike properties, and inhibition profiles in a cell lysate assay.


Assuntos
Acrilamidas/síntese química , Proteínas de Ligação ao GTP/antagonistas & inibidores , Doença de Huntington/tratamento farmacológico , Sulfonamidas/síntese química , Transglutaminases/antagonistas & inibidores , Acrilamidas/química , Acrilamidas/farmacologia , Animais , Células CACO-2 , Permeabilidade da Membrana Celular , Células HEK293 , Humanos , Técnicas In Vitro , Masculino , Camundongos , Microssomos Hepáticos/metabolismo , Modelos Moleculares , Piperazinas/síntese química , Piperazinas/química , Piperazinas/farmacologia , Proteína 2 Glutamina gama-Glutamiltransferase , Piridinas/síntese química , Piridinas/química , Piridinas/farmacologia , Pirimidinas/síntese química , Pirimidinas/química , Pirimidinas/farmacologia , Ratos , Relação Estrutura-Atividade , Sulfonamidas/química , Sulfonamidas/farmacologia
14.
J Med Chem ; 54(1): 312-9, 2011 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-21128645

RESUMO

The inhibition of Aurora kinases in order to arrest mitosis and subsequently inhibit tumor growth via apoptosis of proliferating cells has generated significant discussion within the literature. We report a novel class of Aurora kinase inhibitors based upon a phthalazinone pyrazole scaffold. The development of the phthalazinone template resulted in a potent Aurora-A selective series of compounds (typically >1000-fold selectivity over Aurora-B) that display good pharmacological profiles with significantly improved oral bioavailability compared to the well studied Aurora inhibitor VX-680.


Assuntos
Antineoplásicos/síntese química , Ftalazinas/síntese química , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Pirazóis/síntese química , Administração Oral , Antineoplásicos/química , Antineoplásicos/farmacologia , Aurora Quinase B , Aurora Quinases , Disponibilidade Biológica , Linhagem Celular Tumoral , Cristalografia por Raios X , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Modelos Moleculares , Estrutura Molecular , Ftalazinas/química , Ftalazinas/farmacologia , Pirazóis/química , Pirazóis/farmacologia , Relação Estrutura-Atividade
15.
J Biomol Screen ; 15(5): 478-87, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20395409

RESUMO

Huntington's disease (HD) is associated with increased expression levels and activity of tissue transglutaminase (TG2), an enzyme primarily known for its cross-linking of proteins. To validate TG2 as a therapeutic target for HD in transgenic models and for eventual clinical development, a selective and brain-permeable inhibitor is required. Here, a comprehensive profiling platform of biochemical and cellular assays is presented which has been established to evaluate the potency, cellular efficacy, subtype selectivity and the mechanism-of-action of known and novel TG2 inhibitors. Several classes of inhibitors have been characterized including: the commonly used pseudo-substrate inhibitors, cystamine and putrescine (which are generally nonspecific for TG2 and therefore not practical for drug development), the various peptidic inhibitors that target the active site cysteine residue (which display excellent selectivity but in general have poor cellular activity), and the allosteric reversible small-molecule hydrazides (which show poor selectivity and a lack of cellular activity and could not be improved despite considerable medicinal chemistry efforts). In addition, a set of inhibitors identified from a collection of pharmacologically active compounds was found to be unselective for TG2. Moreover, inhibition at the guanosine triphosphate binding site has been examined, but apart from guanine nucleotides, no such inhibitors have been identified. In addition, the promising pharmacological profile of a TG2 inhibitor is presented which is currently in lead optimization to be developed as a tool compound.


Assuntos
Bioensaio/métodos , Inibidores Enzimáticos/metabolismo , Proteínas de Ligação ao GTP/antagonistas & inibidores , Transglutaminases/antagonistas & inibidores , Animais , Linhagem Celular , Inibidores Enzimáticos/química , Humanos , Doença de Huntington/enzimologia , Doença de Huntington/patologia , Camundongos , Estrutura Molecular , Proteína 2 Glutamina gama-Glutamiltransferase
16.
Org Biomol Chem ; 2(17): 2483-95, 2004 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-15326529

RESUMO

The first total syntheses of the title fungal metabolites preussomerins F, K and L are described and their structures confirmed thereby. The syntheses were achieved following a versatile, unified, non-biomimetic approach, which is easily extendable to prepare other known and novel members of this family. Key steps include the functionalisation of a 2-arylacetal anion, tandem one-pot Friedel-Crafts cyclisation-deprotection, regioselective substrate-directable hydrogenation and reductive-opening of epoxides.


Assuntos
Compostos de Epóxi/síntese química , Compostos Heterocíclicos de 4 ou mais Anéis/síntese química , Modelos Moleculares , Conformação Molecular , Estereoisomerismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA