Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 66(4): 2744-2760, 2023 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-36762932

RESUMEN

Enveloped viruses depend on the host endoplasmic reticulum (ER) quality control (QC) machinery for proper glycoprotein folding. The endoplasmic reticulum quality control (ERQC) enzyme α-glucosidase I (α-GluI) is an attractive target for developing broad-spectrum antivirals. We synthesized 28 inhibitors designed to interact with all four subsites of the α-GluI active site. These inhibitors are derivatives of the iminosugars 1-deoxynojirimycin (1-DNJ) and valiolamine. Crystal structures of ER α-GluI bound to 25 1-DNJ and three valiolamine derivatives revealed the basis for inhibitory potency. We established the structure-activity relationship (SAR) and used the Site Identification by Ligand Competitive Saturation (SILCS) method to develop a model for predicting α-GluI inhibition. We screened the compounds against SARS-CoV-2 in vitro to identify those with greater antiviral activity than the benchmark α-glucosidase inhibitor UV-4. These host-targeting compounds are candidates for investigation in animal models of SARS-CoV-2 and for testing against other viruses that rely on ERQC for correct glycoprotein folding.


Asunto(s)
1-Desoxinojirimicina , Antivirales , COVID-19 , Inhibidores de Glicósido Hidrolasas , alfa-Glucosidasas , Animales , 1-Desoxinojirimicina/química , 1-Desoxinojirimicina/farmacología , alfa-Glucosidasas/efectos de los fármacos , Antivirales/química , Antivirales/farmacología , Retículo Endoplásmico/enzimología , Glicoproteínas , Inhibidores de Glicósido Hidrolasas/química , Inhibidores de Glicósido Hidrolasas/farmacología , SARS-CoV-2/metabolismo , Relación Estructura-Actividad Cuantitativa
2.
PLoS Negl Trop Dis ; 16(8): e0010636, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35939501

RESUMEN

BACKGROUND: UV-4 (N-(9'-methoxynonyl)-1-deoxynojirimycin, also called MON-DNJ) is an iminosugar small-molecule oral drug candidate with in vitro antiviral activity against diverse viruses including dengue, influenza, and filoviruses and demonstrated in vivo efficacy against both dengue and influenza viruses. The antiviral mechanism of action of UV-4 is through inhibition of the host endoplasmic reticulum-resident α-glucosidase 1 and α-glucosidase 2 enzymes. This inhibition prevents proper glycan processing and folding of virus glycoproteins, thereby impacting virus assembly, secretion, and the fitness of nascent virions. METHODOLOGY/PRINCIPAL FINDINGS: Here we report a first-in-human, single ascending dose Phase 1a study to evaluate the safety, tolerability, and pharmacokinetics of UV-4 hydrochloride (UV-4B) in healthy subjects (ClinicalTrials.gov Identifier NCT02061358). Sixty-four subjects received single oral doses of UV-4 as the hydrochloride salt equivalent to 3, 10, 30, 90, 180, 360, 720, or 1000 mg of UV-4 (6 subjects per cohort), or placebo (2 subjects per cohort). Single doses of UV-4 hydrochloride were well tolerated with no serious adverse events or dose-dependent increases in adverse events observed. Clinical laboratory results, vital signs, and physical examination data did not reveal any safety signals. Dose-limiting toxicity was not observed; the maximum tolerated dose of UV-4 hydrochloride in humans has not yet been determined (>1000 mg). UV-4 was rapidly absorbed and distributed after dosing with the oral solution formulation used in this study. Median time to reach maximum plasma concentration ranged from 0.5-1 hour and appeared to be independent of dose. Exposure increased approximately in proportion with dose over the 333-fold dose range. UV-4 was quantifiable in pooled urine over the entire collection interval for all doses. CONCLUSIONS/SIGNIFICANCE: UV-4 is a host-targeted broad-spectrum antiviral drug candidate. At doses in humans up to 1000 mg there were no serious adverse events reported and no subjects were withdrawn from the study due to treatment-emergent adverse events. These data suggest that therapeutically relevant drug levels of UV-4 can be safely administered to humans and support further clinical development of UV-4 hydrochloride or other candidate antivirals in the iminosugar class. TRIAL REGISTRATION: ClinicalTrials.gov NCT02061358 https://clinicaltrials.gov/ct2/show/NCT02061358.


Asunto(s)
Dengue , alfa-Glucosidasas , 1-Desoxinojirimicina/efectos adversos , Antivirales/farmacología , Área Bajo la Curva , Dengue/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Voluntarios Sanos , Humanos , alfa-Glucosidasas/metabolismo , alfa-Glucosidasas/uso terapéutico
3.
Biochemistry ; 61(10): 822-832, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35476408

RESUMEN

All viruses depend on host cell proteins for replication. Denying viruses' access to the function of critical host proteins can result in antiviral activity against multiple virus families. In particular, small-molecule drug candidates which inhibit the α-glucosidase enzymes of the endoplasmic reticulum (ER) translation quality control (QC) pathway have demonstrated broad-spectrum antiviral activities and low risk for development of viral resistance. However, antiviral drug discovery focused on the ERQC enzyme α-glucosidase I (α-GluI) has been hampered by difficulties in obtaining crystal structures of complexes with inhibitors. We report here the identification of an orthologous enzyme from a thermophilic fungus, Chaetomium thermophilum (Ct), as a robust surrogate for mammalian ER α-GluI and a platform for inhibitor design. Previously annotated only as a hypothetical protein, the Ct protein was validated as a bona fide α-glucosidase by comparing its crystal structure to that of mammalian α-GluI, by demonstrating enzymatic activity on the unusual α-d-Glcp-(1 → 2)-α-d-Glcp-(1 → 3) substrate glycan, and by showing that well-known inhibitors of mammalian α-GluI (1-DNJ, UV-4, UV-5) also inhibit Ct α-GluI. Crystal structures of Ct α-GluI in complex with three such inhibitors (UV-4, UV-5, EB-0159) revealed extensive interactions with all four enzyme subsites and provided insights into the catalytic mechanism. Identification of ER Ct α-GluI as a surrogate for mammalian α-GluI will accelerate the structure-guided discovery of broad-spectrum antivirals. This study also highlights Ct as a source of thermostable eukaryotic proteins, such as ER α-Glu I, that lack orthologs in bacterial or archaeal thermophiles.


Asunto(s)
Virus , alfa-Glucosidasas , Animales , Antivirales/metabolismo , Antivirales/farmacología , Retículo Endoplásmico/metabolismo , Hongos/metabolismo , Inhibidores de Glicósido Hidrolasas/farmacología , Mamíferos/metabolismo , Virus/metabolismo , alfa-Glucosidasas/metabolismo
4.
J Med Chem ; 64(24): 18010-18024, 2021 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-34870992

RESUMEN

Most enveloped viruses rely on the host cell endoplasmic reticulum (ER) quality control (QC) machinery for proper folding of glycoproteins. The key ER α-glucosidases (α-Glu) I and II of the ERQC machinery are attractive targets for developing broad-spectrum antivirals. Iminosugars based on deoxynojirimycin have been extensively studied as ER α-glucosidase inhibitors; however, other glycomimetic compounds are less established. Accordingly, we synthesized a series of N-substituted derivatives of valiolamine, the iminosugar scaffold of type 2 diabetes drug voglibose. To understand the basis for up to 100,000-fold improved inhibitory potency, we determined high-resolution crystal structures of mouse ER α-GluII in complex with valiolamine and 10 derivatives. The structures revealed extensive interactions with all four α-GluII subsites. We further showed that N-substituted valiolamines were active against dengue virus and SARS-CoV-2 in vitro. This study introduces valiolamine-based inhibitors of the ERQC machinery as candidates for developing potential broad-spectrum therapeutics against the existing and emerging viruses.


Asunto(s)
Antivirales/farmacología , Inhibidores de Glicósido Hidrolasas/farmacología , Iminoazúcares/farmacología , Inositol/análogos & derivados , alfa-Glucosidasas/metabolismo , Animales , Antivirales/síntesis química , Antivirales/metabolismo , Sitios de Unión , Chlorocebus aethiops , Cristalografía por Rayos X , Virus del Dengue/efectos de los fármacos , Retículo Endoplásmico/enzimología , Inhibidores de Glicósido Hidrolasas/síntesis química , Inhibidores de Glicósido Hidrolasas/metabolismo , Humanos , Iminoazúcares/síntesis química , Iminoazúcares/metabolismo , Inositol/síntesis química , Inositol/metabolismo , Inositol/farmacología , Ratones , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Unión Proteica , SARS-CoV-2/efectos de los fármacos , Células Vero , alfa-Glucosidasas/química
5.
J Med Chem ; 63(8): 4205-4214, 2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32227946

RESUMEN

Influenza and dengue viruses present a growing global threat to public health. Both viruses depend on the host endoplasmic reticulum (ER) glycoprotein folding pathway. In 2014, Sadat et al. reported two siblings with a rare genetic defect in ER α-glucosidase I (ER Glu I) who showed resistance to viral infections, identifying ER Glu I as a key antiviral target. Here, we show that a single dose of UV-4B (the hydrochloride salt form of N-(9'-methoxynonyl)-1-deoxynojirimycin; MON-DNJ) capable of inhibiting Glu I in vivo is sufficient to prevent death in mice infected with lethal viral doses, even when treatment is started as late as 48 h post infection. The first crystal structure of mammalian ER Glu I will constitute the basis for the development of potent and selective inhibitors. Targeting ER Glu I with UV-4B-derived compounds may alter treatment paradigms for acute viral disease through development of a single-dose therapeutic regime.


Asunto(s)
Dengue/prevención & control , Retículo Endoplásmico/efectos de los fármacos , Inhibidores de Glicósido Hidrolasas/administración & dosificación , Gripe Humana/prevención & control , alfa-Glucosidasas , Animales , Dengue/tratamiento farmacológico , Dengue/enzimología , Virus del Dengue/efectos de los fármacos , Virus del Dengue/enzimología , Relación Dosis-Respuesta a Droga , Retículo Endoplásmico/enzimología , Humanos , Gripe Humana/tratamiento farmacológico , Gripe Humana/enzimología , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Estructura Secundaria de Proteína , alfa-Glucosidasas/metabolismo
6.
Antimicrob Agents Chemother ; 63(12)2019 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-31548183

RESUMEN

Burkholderia pseudomallei (B. pseudomallei), the etiological agent of melioidosis, is a Gram-negative bacterium with additional concern as a biothreat pathogen. The mortality rate from B. pseudomallei varies depending on the type of infection and extent of available health care, but in the case of septicemia left untreated it can range from 50 - 90%. Current therapy for melioidosis is biphasic, consisting of parenteral acute-phase treatment for two weeks or longer, followed by oral eradication-phase treatment lasting several months. An effective oral therapeutic for outpatient treatment of acute-phase melioidosis is needed. GC-072 is a potent, 4-oxoquinolizine antibiotic with selective inhibitory activity against bacterial topoisomerases. GC-072 has demonstrated in vitro potency against susceptible and drug-resistant strains of B. pseudomallei and is also active against Burkholderia mallei, Bacillus anthracis, Yersinia pestis, and Francisella tularensis GC-072 is bactericidal both extra- and intracellularly, with rapid killing noted within a few hours and reduced development of resistance compared to ceftazidime. GC-072, delivered intragastrically to mimic oral administration, promoted dose-dependent survival in mice using lethal inhalational models of B. pseudomallei infection following exposure to a 24 or 339 LD50 challenge with B. pseudomallei strain 1026b. Overall, GC-072 appears to be a strong candidate for first-line, oral treatment of melioidosis.

7.
Antiviral Res ; 138: 22-31, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27908828

RESUMEN

Iminosugars are host-directed antivirals with broad-spectrum activity. The iminosugar, N-butyl-deoxynojirimycin (NB-DNJ or Miglustat®), is used in humans for treatment of Gaucher's disease and has mild antiviral properties. More potent analogs of NB-DNJ have been generated and have demonstrated activity against a variety of viruses including flaviviruses, influenza, herpesviruses and filoviruses. In the current study, a panel of analogs based on NB-DNJ was analyzed for activity against Ebola (EBOV) and Marburg viruses (MARV). The antiviral activity of NB-DNJ (UV-1), UV-2, UV-3, UV-4 and UV-5 against both EBOV and MARV was demonstrated in Vero cells. Subsequent studies to examine the activity of UV-4 and UV-5 using rodent models of EBOV and MARV were performed. In vivo efficacy studies provided inconsistent data following treatment with iminosugars using filovirus mouse models. A tolerability study in nonhuman primates demonstrated that UV-4 could be administered at much higher dose levels than rodents. Since UV-4 was active in vitro, had been demonstrated to be active against influenza and dengue in vivo, and was being tested in a Phase 1 clinical trial, a small proof-of-concept nonhuman primate trial was performed to determine whether this antiviral candidate could provide clinical benefit to EBOV-infected individuals. Administration of UV-4B did not provide a clinical or survival benefit to macaques infected with EBOV-Makona; however, dosing of animals was not optimal in this study. Efficacy may be improved by thrice daily dosing (e.g. by nasogastric tube feeding) to match the efficacious dosing regimens demonstrated against dengue and influenza viruses.


Asunto(s)
Antivirales/farmacología , Antivirales/uso terapéutico , Ebolavirus/efectos de los fármacos , Iminoazúcares/farmacología , Iminoazúcares/uso terapéutico , Marburgvirus/efectos de los fármacos , 1-Desoxinojirimicina/administración & dosificación , 1-Desoxinojirimicina/agonistas , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacología , 1-Desoxinojirimicina/uso terapéutico , Animales , Antivirales/administración & dosificación , Antivirales/química , Chlorocebus aethiops , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Iminoazúcares/administración & dosificación , Iminoazúcares/química , Macaca , Ratones , Modelos Animales , Células Vero
8.
Antiviral Res ; 129: 93-98, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26946111

RESUMEN

The antiviral activity of UV-4 was previously demonstrated against dengue virus serotype 2 (DENV2) in multiple mouse models. Herein, step-wise minimal effective dose and therapeutic window of efficacy studies of UV-4B (UV-4 hydrochloride salt) were conducted in an antibody-dependent enhancement (ADE) mouse model of severe DENV2 infection in AG129 mice lacking types I and II interferon receptors. Significant survival benefit was demonstrated with 10-20 mg/kg of UV-4B administered thrice daily (TID) for seven days with initiation of treatment up to 48 h after infection. UV-4B also reduced infectious virus production in in vitro antiviral activity assays against all four DENV serotypes, including clinical isolates. A set of purified enzyme, in vitro, and in vivo studies demonstrated that inhibition of endoplasmic reticulum (ER) α-glucosidases and not the glycosphingolipid pathway appears to be responsible for the antiviral activity of UV-4B against DENV. Along with a comprehensive safety package, these and previously published data provided support for an Investigational New Drug (IND) filing and Phases 1 and 2 clinical trials for UV-4B with an indication of acute dengue disease.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Inhibidores de Glicósido Hidrolasas/farmacología , Dengue Grave/tratamiento farmacológico , alfa-Glucosidasas/metabolismo , 1-Desoxinojirimicina/administración & dosificación , 1-Desoxinojirimicina/farmacología , 1-Desoxinojirimicina/uso terapéutico , Animales , Anticuerpos Antivirales/sangre , Acrecentamiento Dependiente de Anticuerpo/efectos de los fármacos , Antivirales/administración & dosificación , Antivirales/uso terapéutico , Células Cultivadas , Chlorocebus aethiops , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Drogas en Investigación , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/enzimología , Inhibidores de Glicósido Hidrolasas/administración & dosificación , Inhibidores de Glicósido Hidrolasas/química , Inhibidores de Glicósido Hidrolasas/uso terapéutico , Humanos , Concentración 50 Inhibidora , Ratones , Monocitos/virología , Receptores de Interferón/deficiencia , Serogrupo , Dengue Grave/virología , Células Vero
9.
Invest New Drugs ; 29(2): 340-6, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20084425

RESUMEN

BACKGROUND: 2-methoxyestradiol (2ME2) is an estradiol-17ß metabolite with antiproliferative and antiangiogenic activities. ENMD-1198 is an analog of 2ME2 which was developed to decrease the metabolism and increase both the bioavailability and antitumor activities of the parent molecule. This first-in-human phase I study evaluated the tolerability, pharmacokinetics and preliminary evidence of activity of ENMD-1198 in advanced cancer patients. METHODS: Eligible patients received ENMD-1198 orally once daily in Part A (standard 3 + 3 dose escalation design), or in Part B (accelerated dose escalation design). Cycle 1 consisted of 28 days daily dosing followed by a 14-(Part A) or 7-(Part B) day observation period, then continuously in 28 day cycles thereafter. RESULTS: A total of 29 patients were enrolled in 12 dose cohorts (5 to 550 mg/m²)/d). The most common drug-related toxicities were Grade 1/2 fatigue (55%), nausea and vomiting (37%), and constipation (34%). Two DLTs (Grade 4 neutropenia) occurred at 550 mg/m²/day, and 425 mg/m²/d was declared the maximum tolerated dose. ENMD-1198 was absorbed rapidly with a T(max) of 1-2 h. Exposure to ENMD-1198 (C(max) and AUC0₋24 hr increased linearly with dose. The mean terminal half-life was 15 h. A 3-fold accumulation was found after multiple doses. Five patients achieved stabilization of disease for at least 2 cycles, three of whom (with neuroendocrine carcinoma of pancreas, prostate cancer and ovarian cancer) demonstrated prolonged stabilization ranging from 8-24.5 cycles. CONCLUSION: ENMD-1198 is well-tolerated with a pharmacokinetic exposure profile compatible with once daily dosing. The recommended phase II dose of ENMD-1198 is 425 mg/m²/d. Early evidence of prolonged disease stabilization in pre-treated patients suggests ENMD-1198 is worthy of additional investigation.


Asunto(s)
Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Estradiol/análogos & derivados , Estrenos/farmacocinética , Estrenos/uso terapéutico , Neoplasias/tratamiento farmacológico , 2-Metoxiestradiol , Administración Oral , Adulto , Anciano , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Demografía , Perros , Relación Dosis-Respuesta a Droga , Estradiol/administración & dosificación , Estradiol/efectos adversos , Estradiol/farmacocinética , Estradiol/uso terapéutico , Estrenos/administración & dosificación , Estrenos/efectos adversos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/sangre , Ratas , Especificidad de la Especie
10.
Bioorg Med Chem ; 17(20): 7344-52, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19762246

RESUMEN

A novel series of 17-modified and 2,17-modified analogs of 2-methoxyestradiol (2ME2) were synthesized and characterized. These analogs were designed to retain or potentiate the biological activities of 2ME2 and have diminished metabolic liability. The analogs were evaluated for antiproliferative activity against MDA-MB-231 breast tumor cells, antiangiogenic activity in HUVEC, and estrogenic activity on MCF-7 cell proliferation. Several analogs were evaluated for metabolic stability in human liver microsomes and in vivo in a rat cassette dosing model. This study lead to several 17-modified analogs of 2ME2 that have similar or improved antiproliferative and antiangiogenic activity, lack estrogenic properties and have improved metabolic stability compared to 2ME2.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Estradiol/análogos & derivados , Estrona/síntesis química , Estrona/farmacología , 2-Metoxiestradiol , Animales , Línea Celular , Estradiol/farmacología , Estrona/química , Humanos , Ratas , Relación Estructura-Actividad
11.
Bioorg Med Chem Lett ; 19(21): 6241-4, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19782564

RESUMEN

The syntheses of 21 analogs of 2-methoxyestradiol are presented, including ENMD-1198 which was selected for advancement into Phase 1 clinical trials in oncology. These analogs were evaluated for antiproliferative activity using breast tumor MDA-MB-231 cells, for antiangiogenic activity in HUVEC proliferation assays, and for estrogenic activity in MCF-7 cell proliferation. The most active analogs were evaluated for iv and oral pharmacokinetic properties via cassette dosing in rat and in mice pharmacokinetic models.


Asunto(s)
Antineoplásicos Hormonales/síntesis química , Estradiol/análogos & derivados , 2-Metoxiestradiol , Animales , Antineoplásicos Hormonales/química , Antineoplásicos Hormonales/farmacocinética , Línea Celular Tumoral , Estradiol/síntesis química , Estradiol/química , Estradiol/farmacocinética , Estrenos/química , Estrenos/farmacocinética , Humanos , Ratones , Ratas , Relación Estructura-Actividad
12.
Bioorg Med Chem Lett ; 19(22): 6459-62, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19782568

RESUMEN

The syntheses of 2-methoxyestradiol analogs with modifications at the 3-position are described. The analogs were assessed for their antiproliferative, antiangiogenic, and estrogenic activities. Several lead substituents were identified with similar or improved antitumor activities and reduced metabolic liability compared to 2-methoxyestradiol.


Asunto(s)
Estradiol/análogos & derivados , Huso Acromático/efectos de los fármacos , 2-Metoxiestradiol , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ensayos de Selección de Medicamentos Antitumorales , Estradiol/farmacología , Estradiol/uso terapéutico , Humanos , Masculino , Ratones , Ratones Desnudos , Modelos Moleculares , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Ensayos Antitumor por Modelo de Xenoinjerto/estadística & datos numéricos
13.
Mol Cancer Ther ; 7(6): 1472-82, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18566218

RESUMEN

Clinical studies using the microtubule-targeting agent 2-methoxyestradiol (2ME2; Panzem) in cancer patients show that treatment is associated with clinical benefit, including prolonged stable disease, complete and partial responses, and an excellent safety profile. Studies have shown that 2ME2 is metabolized by conjugation at positions 3 and 17 and oxidation at position 17. To define structure-activity relationships for these positions of 2ME2 and to generate metabolically stable analogues with improved anti-tubulin properties, a series of analogues was generated and three lead analogues were selected, ENMD-1198, ENMD-1200, and ENMD-1237. These molecules showed improved metabolic stability with >65% remaining after 2-h incubation with hepatocytes. Pharmacokinetic studies showed that oral administration of the compounds resulted in increased plasma levels compared with 2ME2. All three analogues bind the colchicine binding site of tubulin, induce G(2)-M cell cycle arrest and apoptosis, and reduce hypoxia-inducible factor-1alpha levels. ENMD-1198 and ENMD-1200 showed improved in vitro antiproliferative activities. Significant reductions in tumor volumes compared with vehicle-treated mice were observed in an orthotopic breast carcinoma (MDA-MB-231) xenograft model following daily oral treatment with all compounds (ANOVA, P < 0.05). Significantly improved median survival time was observed with ENMD-1198 and ENMD-1237 (200 mg/kg/d) in a Lewis lung carcinoma metastatic model (P < 0.05). In both tumor models, the high-dose group of ENMD-1198 showed antitumor activity equivalent to that of cyclophosphamide. ENMD-1198 was selected as the lead molecule in this analogue series and is currently in a phase I clinical trial in patients with refractory solid tumors.


Asunto(s)
Antineoplásicos/farmacología , Estrenos/farmacología , Microtúbulos/efectos de los fármacos , Moduladores de Tubulina/farmacología , 2-Metoxiestradiol , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Unión Competitiva/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colchicina/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Estradiol/análogos & derivados , Estradiol/química , Estrenos/administración & dosificación , Estrenos/química , Estrenos/farmacocinética , Fase G2/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Concentración 50 Inhibidora , Masculino , Ratones , Ratones Endogámicos C57BL , Mitosis/efectos de los fármacos , Ratas , Análisis de Supervivencia , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/administración & dosificación , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacocinética
14.
J Med Chem ; 50(26): 6700-5, 2007 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-18052315

RESUMEN

A prodrug strategy was investigated to address the problem of limited aqueous solubility and the resulting limited bioavailability of the antitumor agent 2-methoxyestradiol. The 3-phosphate, 17-phosphate, and 3,17-diphosphate of 2-methoxyestradiol were synthesized. 2-methoxyestradiol 3-phosphate was metabolized more efficiently to the parent compound in vivo than 2-methoxyestradiol 17-phosphate, and it was also more cytotoxic in cancer cell cultures than either the 17-phosphate or the 3,17-diphosphate. These results agree with the in vivo anticancer activity of 2-methoxyestradiol 3-phosphate in a mouse Lewis lung carcinoma experimental metastasis model as opposed to the 17-phosphate and 3,17-diphosphate, both of which were inactive. The in vivo antitumor activity of 2-methoxyestradiol 3-phosphate at a dose of 200 mg/kg per day was comparable to that of a maximally tolerated dose of cyclophosphamide.


Asunto(s)
Antineoplásicos/síntesis química , Estradiol/análogos & derivados , Organofosfatos/síntesis química , Profármacos/síntesis química , 2-Metoxiestradiol , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/secundario , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Estradiol/síntesis química , Estradiol/farmacocinética , Estradiol/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Organofosfatos/farmacocinética , Organofosfatos/farmacología , Profármacos/farmacocinética , Profármacos/farmacología , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
15.
Bioorg Med Chem ; 15(24): 7524-37, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17910916

RESUMEN

A series of 16-modified 2-methoxyestradiol analogs were synthesized and evaluated for antiproliferative activity toward HUVEC and MDA-MB-231 cells, and for susceptibility to conjugation. In addition, the estrogenicity of these analogs was accessed by measuring cell proliferation of the estrogen-dependent cell line MCF7 in response to compound treatment. It was observed that antiproliferative activity dropped as the size of the 16 substituent increased. Selected analogs tested in glucuronidation assays had similar rates of clearance to 2-methoxyestradiol, but had enhanced clearance in sulfonate conjugation assays.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma/tratamiento farmacológico , Estradiol/análogos & derivados , 2-Metoxiestradiol , Antineoplásicos/química , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Estradiol/química , Estradiol/uso terapéutico , Femenino , Humanos , Estructura Molecular , Relación Estructura-Actividad
16.
Invest New Drugs ; 25(1): 41-8, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16969706

RESUMEN

PURPOSE: We report the first phase I trials of 2-methoxyestradiol (2ME2, Panzem Capsules, EntreMed, Rockville, MD), alone and in combination with docetaxel, in patients with metastatic breast cancer (MBC). PATIENTS AND METHODS: In Trial 001, 2ME2 monotherapy was administered orally once (200-1000 mg/d, cohorts 1-5) or twice daily (200-800 mg/q12h, cohorts 6-9) for 28 days followed by a 14-day observation period, continuously thereafter. In Trial 002, docetaxel 35 mg/m(2) was administered weekly for four of six weeks for a maximum six cycles; 2ME2 (200-1000 mg/d) was given orally once daily for 28 days followed by a 13-day observation period in cycle one, continuously thereafter. In both trials, responding or stable patients continued 2ME2 until progression. RESULTS: Trial 001 enrolled 31 patients; there were no objective responses. Trial 002 enrolled 15 patients; ORR was 20% including one CR. There were no Grade IV toxicities; MTD was not reached in either study. When combined with docetaxel, three patients had significant transaminase elevations that returned to normal with continued treatment (in two of three patients). There was significant inter-patient variability and extensive metabolism to 2-methoxyestrone (2ME1). Steady-state AUC and trough concentrations of 2ME2 increased linearly up to 400-600 mg/d; doses above 400-600 mg/d did not increase 2ME2 levels. The target trough concentration (3-25 ng/mL) was not attained. Combined administration did not alter docetaxel or 2ME2 pharmacokinetics. CONCLUSION: 2ME2, alone or in combination with docetaxel, was well tolerated in patients with MBC but systemic exposure remained below the expected therapeutic range.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Estradiol/análogos & derivados , Taxoides/uso terapéutico , 2-Metoxiestradiol , Administración Oral , Adulto , Anciano , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacocinética , Antineoplásicos Fitogénicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Área Bajo la Curva , Neoplasias de la Mama/patología , Docetaxel , Esquema de Medicación , Estradiol/efectos adversos , Estradiol/farmacocinética , Estradiol/uso terapéutico , Fatiga/inducido químicamente , Femenino , Semivida , Humanos , Metaloproteinasa 2 de la Matriz/sangre , Metaloproteinasa 9 de la Matriz/sangre , Persona de Mediana Edad , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia , Taxoides/efectos adversos , Taxoides/farmacocinética , Resultado del Tratamiento , Moduladores de Tubulina/administración & dosificación , Moduladores de Tubulina/farmacocinética , Moduladores de Tubulina/uso terapéutico , Urinálisis , Molécula 1 de Adhesión Celular Vascular/sangre , Factor A de Crecimiento Endotelial Vascular/orina
17.
Cancer Res ; 62(8): 2300-5, 2002 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11956087

RESUMEN

Thalidomide has recently been shown to be useful in the treatment of multiple myeloma and may also be useful in the treatment of other hematological malignancies. We have identified a new derivative of thalidomide, S-3-[3-amino-phthalimido]-glutarimide (S-3APG) with dual activity against B-cell neoplasias. S-3APG was able to directly inhibit the proliferation of myeloma and Burkitt's lymphoma cell lines in vitro without showing toxicity to normal bone marrow stromal cells or hematopoietic progenitor cells. In vivo, S-3APG treatment of drug resistant myeloma cell tumors in mice was able to produce complete and sustained regressions without any observed toxicity. Additionally, S-3APG induced complete regressions of Burkitt's lymphoma cell tumors. Furthermore, S-3APG inhibited angiogenesis more potently than thalidomide in the murine corneal micropocket model. We conclude that S-3APG is a powerful anti-myeloma and anti-B-cell-lymphoma agent that has both antiproliferative and antiangiogenic effects.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Linfoma de Burkitt/tratamiento farmacológico , Mieloma Múltiple/tratamiento farmacológico , Talidomida/farmacología , Células 3T3 , Animales , Linfoma de Burkitt/patología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/patología , División Celular/efectos de los fármacos , Inhibidores de Crecimiento/farmacología , Humanos , Interleucina-6/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Mieloma Múltiple/patología , Neovascularización Fisiológica/efectos de los fármacos , Talidomida/análogos & derivados , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA