Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cancer Ther ; 22(3): 333-342, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36808277

RESUMEN

PARP inhibitors have been approved by the FDA for use in the treatment of patients with ovarian, breast, pancreatic, and prostate cancers. PARP inhibitors show diverse suppressive effects on PARP family members and PARP-DNA trapping potency. These properties are associated with distinct safety/efficacy profiles. Here, we report the nonclinical characteristics of venadaparib (also known as IDX-1197 or NOV140101), a novel potent PARP inhibitor. The physiochemical properties of venadaparib were analyzed. Furthermore, the efficacy of venadaparib against PARP enzymes, PAR formation, and PARP trapping activities, and growth inhibition of cell lines with BRCA mutations were evaluated. Ex vivo and in vivo models were also established to study pharmacokinetics/pharmacodynamics, efficacy, and toxicity. Venadaparib specifically inhibits PARP-1 and -2 enzymes. Oral administration of venadaparib HCl at doses above 12.5 mg/kg significantly reduced tumor growth in the OV_065 patient-derived xenograft model. Intratumoral PARP inhibition remained at over 90% until 24 hours after dosing. Venadaparib had wider safety margins than olaparib. Notably, venadaparib showed favorable physicochemical properties and superior anticancer effects in homologous recombination-deficient in vitro and in vivo models with improved safety profiles. Our results suggest the possibility of venadaparib as a next-generation PARP inhibitor. On the basis of these findings, phase Ib/IIa studies on the efficacy and safety of venadaparib have been initiated.


Asunto(s)
Antineoplásicos , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Masculino , Humanos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Recombinación Homóloga
2.
Ther Adv Med Oncol ; 12: 1758835920926796, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32536979

RESUMEN

BACKGROUND: YYB101, a humanized monoclonal antibody against hepatocyte growth factor (HGF), has shown safety and efficacy in vitro and in vivo. This is a first-in-human trial of this antibody. MATERIALS AND METHODS: YYB101 was administered intravenously to refractory cancer patients once every 4 weeks for 1 month, and then once every 2 weeks until disease progression or intolerable toxicity, at doses of 0.3, 1, 3, 5, 10, 20, 30 mg/kg, according to a 3+3 dose escalation design. Maximum tolerated dose, safety, pharmacokinetics, and pharmacodynamics were studied. HGF, MET, PD-L1, and ERK expression was evaluated for 9 of 17 patients of the expansion cohort (20 mg/kg). RESULTS: In 39 patients enrolled, no dose-limiting toxicity was observed at 0.3 mg/kg, and the most commonly detected toxicity was generalized edema (n = 7, 18.9%) followed by pruritis and nausea (n = 5, 13.5%, each), fatigue, anemia, and decreased appetite (n = 4, 10.8%, each). No patient discontinued treatment because of adverse events. YYB101 showed dose-proportional pharmacokinetics up to 30 mg/kg. Partial response in 1 (2.5%) and stable disease in 17 (43.5%) were observed. HGF, MET, PD-L1, and ERK proteins were not significant predictors for treatment response. However, serum HGF level was significantly lowered in responders upon drug administration. RNA sequencing revealed a mesenchymal signature in two long-term responders. CONCLUSION: YYB101 showed favorable safety and efficacy in patients with refractory solid tumors. Based on this phase I trial, a phase II study on the YYB101 + irinotecan combination in refractory metastatic colorectal cancer patients is planned. CONCLUSION: ClinicalTrials.gov Identifier: NCT02499224.

3.
Pharmacol Res Perspect ; 8(2): e00568, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32162844

RESUMEN

We report a phase I pharmacological study of an oral formulation of CKD-516, a vascular-disrupting agent, in patients with refractory solid tumors. Twenty-seven patients (16 in the dose-escalation cohort and 11 in the expansion cohort) received a single daily dose (5-25 mg) of CKD-516 five days per week. Nausea (67%) and diarrhea (63%) were the most common treatment-related adverse events. The recommended phase II dose of oral CKD-516 was 20 mg/d (15 mg/d with a body surface area (BSA) <1.65 m2 ). Notably, S-516 half-lives in patients receiving 15-20 mg CKD-516/d significantly differed between patients with and without splenomegaly that is suggestive of portal hypertension associated with liver cirrhosis (6.1 vs 4.6 hours, respectively). Of 11 patients without splenomegaly who completed at least one cycle of a daily CKD-516 dose of either 15 or 20 mg, only one patient (9.1%) suffered from any dose-limiting toxicity. We conclude that a daily oral dose of 15 or 20 mg CKD-516 five days per week could be tolerable in patients without liver cirrhosis.


Asunto(s)
Antineoplásicos/farmacocinética , Benzofenonas/farmacocinética , Neoplasias/metabolismo , Valina/análogos & derivados , Administración Oral , Adulto , Anciano , Antineoplásicos/efectos adversos , Antineoplásicos/sangre , Benzofenonas/efectos adversos , Benzofenonas/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/sangre , Neoplasias/tratamiento farmacológico , Resultado del Tratamiento , Valina/efectos adversos , Valina/sangre , Valina/farmacocinética , Adulto Joven
4.
Front Oncol ; 9: 571, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31355133

RESUMEN

Current chemotherapy regimens have certain limitations in improving the survival rates of patients with advanced ovarian cancer. Hepatocyte growth factor (HGF) is important in ovarian cancer cell migration and invasion. This study assessed the effects of YYB-101, a humanized monoclonal anti-HGF antibody, on the growth and metastasis of ovarian cancer cells. YYB-101 suppressed the phosphorylation of the HGF receptor c-MET and inhibited the migration and invasion of SKOV3 and A2780 ovarian cancer cells. Moreover, the combination of YYB-101 and paclitaxel synergistically inhibited tumor growth in an in vivo ovarian cancer mouse xenograft model and significantly increased the overall survival (OS) rate compared with either paclitaxel or YYB-101 alone. Taken together, these findings suggest that YYB-101 has therapeutic potential in ovarian cancer when combined with conventional chemotherapy agents.

5.
Neuro Oncol ; 21(2): 222-233, 2019 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29939324

RESUMEN

BACKGROUND: Cancer is a complex disease with profound genomic alterations and extensive heterogeneity. Recent studies on large-scale genomics have shed light on the impact of core oncogenic pathways, which are frequently dysregulated in a wide spectrum of cancer types. Aberrant activation of the hepatocyte growth factor (HGF) signaling axis has been associated with promoting various oncogenic programs during tumor initiation, progression, and treatment resistance. As a result, HGF-targeted therapy has emerged as an attractive therapeutic approach. However, recent clinical trials involving HGF-targeted therapies have demonstrated rather disappointing results. Thus, an alternative, in-depth assessment of new patient stratification is necessary to shift the current clinical course. METHODS: To address such challenges, we have evaluated the therapeutic efficacy of YYB-101, an HGF-neutralizing antibody, in a series of primary glioblastoma stem cells (GSCs) both in vitro and in vivo. Furthermore, we performed genome and transcriptome analysis to determine genetic and molecular traits that exhibit therapeutic susceptibility to HGF-mediated therapy. RESULTS: We have identified several differentially expressed genes, including MET, KDR, and SOX3, which are associated with tumor invasiveness, malignancy, and unfavorable prognosis in glioblastoma patients. We also demonstrated the HGF-MET signaling axis as a key molecular determinant in GSC invasion, and we discovered that a significant association in HGF expression existed between mesenchymal phenotype and immune cell recruitment. CONCLUSIONS: Upregulation of MET and mesenchymal cellular state are essential in generating HGF-mediated therapeutic responses. Our results provide an important framework for evaluating HGF-targeted therapy in future clinical settings.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genómica/métodos , Glioblastoma/tratamiento farmacológico , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Transcriptoma , Animales , Apoptosis , Movimiento Celular , Proliferación Celular , Femenino , Glioblastoma/genética , Glioblastoma/patología , Factor de Crecimiento de Hepatocito/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fenotipo , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Exp Mol Med ; 49(3): e309, 2017 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-28336956

RESUMEN

Hepatocyte growth factor (HGF) and its receptor, cMET, play critical roles in cell proliferation, angiogenesis and invasion in a wide variety of cancers. We therefore examined the anti-tumor activity of the humanized monoclonal anti-HGF antibody, YYB-101, in nude mice bearing human glioblastoma xenografts as a single agent or in combination with temozolomide. HGF neutralization, The extracellular signal-related kinases 1 and 2 (ERK1/2) phosphorylation, and HGF-induced scattering were assessed in HGF-expressing cell lines treated with YYB-101. To support clinical development, we also evaluated the preclinical pharmacokinetics and toxicokinetics in cynomolgus monkeys, and human and cynomolgus monkey tissue was stained with YYB-101 to test tissue cross-reactivity. We found that YYB-101 inhibited cMET activation in vitro and suppressed tumor growth in the orthotopic mouse model of human glioblastoma. Combination treatment with YYB-101 and temozolomide decreased tumor growth and increased overall survival compared with the effects of either agent alone. Five cancer-related genes (TMEM119, FST, RSPO3, ROS1 and NBL1) were overexpressed in YYB-101-treated mice that showed tumor regrowth. In the tissue cross-reactivity assay, critical cross-reactivity was not observed. The terminal elimination half-life was 21.7 days. Taken together, the in vitro and in vivo data demonstrated the anti-tumor efficacy of YYB-101, which appeared to be mediated by blocking the HGF/cMET interaction. The preclinical pharmacokinetics, toxicokinetics and tissue cross-reactivity data support the clinical development of YYB-101 for advanced cancer.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Antineoplásicos Inmunológicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Neutralizantes/efectos adversos , Anticuerpos Neutralizantes/inmunología , Antineoplásicos Inmunológicos/efectos adversos , Antineoplásicos Inmunológicos/uso terapéutico , Perros , Femenino , Células Hep G2 , Factor de Crecimiento de Hepatocito/inmunología , Humanos , Macaca fascicularis , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/inmunología
7.
Bioorg Med Chem Lett ; 24(3): 780-5, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24433862

RESUMEN

ß-Lactamase inhibitors with a bicyclic urea core and a variety of heterocyclic side chains were prepared and evaluated as potential partners for combination with imipenem to overcome class A and C ß-lactamase mediated antibiotic resistance. The piperidine analog 3 (MK-7655) inhibited both class A and C ß-lactamases in vitro. It effectively restored imipenem's activity against imipenem-resistant Pseudomonas and Klebsiella strains at clinically achievable concentrations. A combination of MK-7655 and Primaxin® is currently in phase II clinical trials for the treatment of Gram-negative bacterial infections.


Asunto(s)
Compuestos de Azabiciclo/química , Compuestos de Azabiciclo/farmacología , Cilastatina/química , Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Imipenem/química , Inhibidores de beta-Lactamasas , Cilastatina/farmacología , Combinación Cilastatina e Imipenem , Cristalografía por Rayos X , Combinación de Medicamentos , Farmacorresistencia Bacteriana/efectos de los fármacos , Imipenem/farmacología , Concentración 50 Inhibidora , Klebsiella/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Modelos Biológicos , Pseudomonas/efectos de los fármacos , Relación Estructura-Actividad
8.
Antimicrob Agents Chemother ; 56(9): 4662-70, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22710113

RESUMEN

The resistance of methicillin-resistant Staphylococcus aureus (MRSA) to all ß-lactam classes limits treatment options for serious infections involving this organism. Our goal is to discover new agents that restore the activity of ß-lactams against MRSA, an approach that has led to the discovery of two classes of natural product antibiotics, a cyclic depsipeptide (krisynomycin) and a lipoglycopeptide (actinocarbasin), which potentiate the activity of imipenem against MRSA strain COL. We report here that these imipenem synergists are inhibitors of the bacterial type I signal peptidase SpsB, a serine protease that is required for the secretion of proteins that are exported through the Sec and Tat systems. A synthetic derivative of actinocarbasin, M131, synergized with imipenem both in vitro and in vivo with potent efficacy. The in vitro activity of M131 extends to clinical isolates of MRSA but not to a methicillin-sensitive strain. Synergy is restricted to ß-lactam antibiotics and is not observed with other antibiotic classes. We propose that the SpsB inhibitors synergize with ß-lactams by preventing the signal peptidase-mediated secretion of proteins required for ß-lactam resistance. Combinations of SpsB inhibitors and ß-lactams may expand the utility of these widely prescribed antibiotics to treat MRSA infections, analogous to ß-lactamase inhibitors which restored the utility of this antibiotic class for the treatment of resistant Gram-negative infections.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Compuestos de Bifenilo/farmacología , Depsipéptidos/farmacología , Glicopéptidos/farmacología , Glicósidos/farmacología , Lipopéptidos/farmacología , Proteínas de la Membrana/antagonistas & inhibidores , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Oligopéptidos/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , beta-Lactamas/farmacología , Animales , Antibacterianos/aislamiento & purificación , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Transporte Biológico , Compuestos de Bifenilo/síntesis química , Depsipéptidos/aislamiento & purificación , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Glicopéptidos/síntesis química , Glicopéptidos/aislamiento & purificación , Glicósidos/aislamiento & purificación , Humanos , Lipopéptidos/aislamiento & purificación , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/crecimiento & desarrollo , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Familia de Multigenes , Oligopéptidos/síntesis química , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Infecciones Estafilocócicas/microbiología , Resistencia betalactámica/efectos de los fármacos , Resistencia betalactámica/genética , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
10.
Bioorg Med Chem Lett ; 21(14): 4363-5, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21664132

RESUMEN

The preparation and characterization of a series of thiophenyl oxime phosphonate beta-lactamase inhibitors is described. A number of these analogs were potent and selective inhibitors of class C beta-lactamases from Pseudomonas aeruginosa and Enterobacter cloacae. Compounds 3b and 7 reduced the MIC of imipenem against an AmpC expressing strain of imipenem-resistant P. aeruginosa. A number of the title compounds retained micromolar potency against the class D OXA-40 beta-lactamase from Acinetobacter baumannii and at high concentrations compound 3b was shown to reduce the MIC of imipenem against a highly imipenem-resistant strain of A. baumanii expressing the OXA-40 beta-lactamase. In mice compound 3b exhibited phamacokinetics similar to imipenem.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/química , Inhibidores Enzimáticos/química , Organofosfonatos/química , Oximas/química , Pseudomonas aeruginosa/efectos de los fármacos , Inhibidores de beta-Lactamasas , Antibacterianos/síntesis química , Antibacterianos/farmacología , Farmacorresistencia Bacteriana , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Imipenem/farmacología , Pruebas de Sensibilidad Microbiana , Organofosfonatos/síntesis química , Organofosfonatos/farmacología , Oximas/síntesis química , Oximas/farmacología , Tiofenos/química , beta-Lactamasas/metabolismo
11.
Bioorg Med Chem Lett ; 20(8): 2622-4, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20299220

RESUMEN

4,7-Dichloro-1-benzothien-2-yl sulfonylaminomethyl boronic acid (DSABA, Compound I) was discovered as the first boronic acid-based class D beta-lactamase inhibitor. It exhibited an IC(50) of 5.6 microM against OXA-40. The compound also inhibited class A and C beta-lactamases with sub to low microM IC(50), and synergized with imipenem against Acinetobacter baumannii.


Asunto(s)
Antibacterianos/farmacología , Ácidos Borónicos/farmacología , Inhibidores Enzimáticos/farmacología , Inhibidores de beta-Lactamasas , Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/química , Ácidos Borónicos/química , Inhibidores Enzimáticos/química , Concentración 50 Inhibidora , Pruebas de Sensibilidad Microbiana
12.
Bioorg Med Chem Lett ; 20(3): 918-21, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20044254
13.
Chem Biol ; 16(8): 837-48, 2009 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-19716474

RESUMEN

Methicillin-resistant Staphylococcus aureus (MRSA) is a major nosocomial and community-acquired pathogen for which few existing antibiotics are efficacious. Here we describe two structurally related synthetic compounds that potentiate beta-lactam activity against MRSA. Genetic studies indicate that these agents target SAV1754 based on the following observations: (i) it has a unique chemical hypersensitivity profile, (ii) overexpression or point mutations are sufficient to confer resistance, and (iii) genetic inactivation phenocopies the potentiating effect of these agents in combination with beta-lactams. Further, we demonstrate these agents inhibit peptidoglycan synthesis. Because SAV1754 is essential for growth and structurally related to the recently reported peptidoglycan flippase of Escherichia coli, we speculate it performs an analogous function in S. aureus. These results suggest that SAV1754 inhibitors might possess therapeutic potential alone, or in combination with beta-lactams to restore MRSA efficacy.


Asunto(s)
Antibacterianos/farmacología , Carbapenémicos/farmacología , Indoles/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Peptidoglicano/química , Piperidinas/farmacología , Antibacterianos/química , Carbapenémicos/química , Sinergismo Farmacológico , Indoles/química , Staphylococcus aureus Resistente a Meticilina/genética , Peptidoglicano/metabolismo , Piperidinas/química , Interferencia de ARN , ARN sin Sentido/metabolismo
15.
Anal Biochem ; 349(1): 129-35, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16325756

RESUMEN

Current immunoassays for the measurement of leukotriene B(4) (LTB(4)) typically utilize an enzyme-linked immunosorbent assay (ELISA) format that requires multiple incubations and washing steps and often expensive immunoassay kits. We have developed a bead-based, mix and read, indirect fluorescence-linked immunosorbent assay utilizing fluorometric microvolume assay technology (FMAT). The assay employs a monoclonal anti-LTB(4) antibody-coated onto goat antimouse antibody coupled polystyrene beads and an AlexaFluor-647-coupled LTB(4) ligand. Because the FMAT measurement is made only in the portion of the well volume containing the settled beads coated with AF647-LTB(4), the free label in the solution is not measured. Similarly, substances present in plasma that interfere with other immunoassays are largely ignored. The assay is robust (Z=0.8; S/N=250) and can be measured in the presence of relatively high concentrations of dimethyl sulfoxide or serum. It is inexpensive (<0.10 dollars/assay) and amenable to robotics and has a sensitivity comparable to that of the most sensitive ELISA assays; the concentration of LTB(4) giving 50% inhibition (IC(50)) was ca. 55pg/ml. Cross-reactivity in the FMAT assay was comparable to that of the ELISA assay with significant cross-reactivity found only with 20-hydroxy LTB(4) and 12-epi LTB(4). Measurements of LTB(4) determined by FMAT were equivalent to those measured by standard ELISA in samples of ionophore-stimulated human neutrophils or whole blood.


Asunto(s)
Fluorometría , Inmunoensayo , Leucotrieno B4/análisis , Humanos , Inmunoensayo/métodos , Leucotrieno B4/sangre , Neutrófilos/metabolismo
16.
J Am Chem Soc ; 127(9): 3031-7, 2005 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-15740141

RESUMEN

Acyclic o-phenylene-bridged bis(anilido-aldimine) compounds, o-C(6)H(4){C(6)H(2)R(2)N=CH-C(6)H(4)-(H)N(C(6)H(3)R'(2))}(2) and related 30-membered macrocyclic compounds, o-C(6)H(4){C(6)H(2)R'(2)N=CH-C(6)H(4)-(H)N-C(6)H(2)R(2)}(2) (o-C(6)H(4)) are prepared. Successive additions of Me(2)Zn and SO(2) gas to the bis(anilido-aldimine) compounds afford quantitatively dinuclear mu-methylsulfinato zinc complexes, o-C(6)H(4){(C(6)H(2)R(2)N=CH-C(6)H(4)-N(C(6)H(3)R'(2))-kappa(2)-N,N)Zn(mu-OS(O)Me)}(2) (R = iPr and R' = iPr, 29; R = Et and R' = Et, 30; R = Me and R'= Me, 31; R = Me and R' = iPr, 32; R = Et and R' = Me, 33; R = Et and R' = iPr, 34; R = iPr and R' = Et, 35) and o-C(6)H(4){C(6)H(2)R'(2)N=CH-C(6)H(4)-N-C(6)H(2)R(2)-kappa(2)-N,N)Zn(mu-OS(O)Me)}(2) (o-C(6)H(4)) (R = Et and R'= Et, 36; R = Me and R' = Me, 37; R = iPr and R' = Me, 38; R = Et and R' = Me, 39; R = Me and R'= iPr, 40). Molecular structures of 34 and 40 are confirmed by X-ray crystallography. Complexes 30-35 show high activity for cyclohexene oxide/CO(2) copolymerization at low [Zn]/[monomer] ratio (1:5600), whereas the complex of mononucleating beta-diketiminate {[(C(6)H(3)Et(2))N=C(Me)CH=C(Me)N(C(6)H(3)Et(2))]Zn(mu-OS(O)Et)}(2) shows negligible activity in the same condition. Activity is sensitive to the N-aryl ortho substituents and the highest activity is observed with 32. Turnover number up to 2980 and molecular weight (M(n)) up to 284 000 are attained with 32 at such a highly diluted condition as [Zn]/[monomer] = 1:17 400. Macrocyclic complexes 36-40 show negligible activity for copolymerization.

17.
Assay Drug Dev Technol ; 1(4): 521-5, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15090248

RESUMEN

We describe a novel, "mix and read" immunoassay for insulin in biological samples using FMAT. Current commercial assays for insulin require multiple washing steps and can be expensive. The insulin assay described is a simple two-step, time-saving assay and amenable to robotics. The linear response for the fluorometric signal is comparable to that observed using classical ELISA and RIA. A series of mouse plasma samples were tested for insulin levels and yielded results comparable to that measured using a commercial ELISA for insulin.


Asunto(s)
Fluorometría/métodos , Insulina/análisis , Microquímica/métodos , Animales , Ensayo de Inmunoadsorción Enzimática , Fluorometría/economía , Humanos , Ratones , Ratas
18.
Proc Natl Acad Sci U S A ; 99(10): 6603-6, 2002 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-11997440

RESUMEN

A fluorescence resonance energy transfer assay has been developed for monitoring Bacillus anthracis lethal factor (LF) protease activity. A fluorogenic 16-mer peptide based on the known LF protease substrate MEK1 was synthesized and found to be cleaved by the enzyme at the anticipated site. Extension of this work to a fluorogenic 19-mer peptide, derived, in part, from a consensus sequence of known LF protease targets, produced a much better substrate, cleaving approximately 100 times more efficiently. This peptide sequence was modified further on resin to incorporate donor/quencher pairs to generate substrates for use in fluorescence resonance energy transfer-based appearance assays. All peptides cleaved at similar rates with signal/background ranging from 9-16 at 100% turnover. One of these substrates, denoted (Cou)Consensus(K(QSY-35)GG)-NH(2), was selected for additional assay optimization. A plate-based assay requiring only low nanomolar levels of enzyme was developed for screening and inhibitor characterization.


Asunto(s)
Antígenos Bacterianos , Bacillus anthracis/enzimología , Toxinas Bacterianas/metabolismo , Metaloendopeptidasas/metabolismo , MAP Quinasa Quinasa 1 , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Péptidos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Espectrometría de Fluorescencia/métodos , Espectrofotometría Ultravioleta/métodos , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA