Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 21(23)2020 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-33291226

RESUMEN

Various neuroprotective agents have been studied for the treatment of retinal ganglion cell (RGC) diseases, but issues concerning the side effects of systemically administered drugs and the short retention time of intravitreally injected drugs limit their clinical applications. The current study aimed to evaluate the neuroprotective effects of intravitreally injected trichostatin A (TSA)-loaded liposomes in a mouse model of optic nerve crush (ONC) and determine whether TSA-loaded liposomes have therapeutic potential in RGC diseases. The histone deacetylase inhibitor, TSA, was incorporated into polyethylene glycolylated liposomes. C57BL/6J mice were treated with an intravitreal injection of TSA-loaded liposomes and liposomes loaded with a lipophilic fluorescent dye for tracking, immediately after ONC injury. The expression of macroglial and microglial cell markers (glial fibrillary acidic protein and ionized calcium binding adaptor molecule-1), RGC survival, and apoptosis were assessed. We found that the liposomes reached the inner retina. Their fluorescence was detected for up to 10 days after the intravitreal injection, with peak intensity at 3 days postinjection. Intravitreally administered TSA-loaded liposomes significantly decreased reactive gliosis and RGC apoptosis and increased RGC survival in a mouse model of ONC. Our results suggest that TSA-loaded liposomes may help in the treatment of various RGC diseases.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Traumatismos del Nervio Óptico/tratamiento farmacológico , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Apoptosis , Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/uso terapéutico , Inyecciones Intravítreas , Liposomas/química , Ratones , Ratones Endogámicos C57BL , Compresión Nerviosa , Células Ganglionares de la Retina/metabolismo
2.
Brain ; 141(3): 673-687, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29415205

RESUMEN

Peripheral nerve axons require a well-organized axonal microtubule network for efficient transport to ensure the constant crosstalk between soma and synapse. Mutations in more than 80 different genes cause Charcot-Marie-Tooth disease, which is the most common inherited disorder affecting peripheral nerves. This genetic heterogeneity has hampered the development of therapeutics for Charcot-Marie-Tooth disease. The aim of this study was to explore whether histone deacetylase 6 (HDAC6) can serve as a therapeutic target focusing on the mutant glycyl-tRNA synthetase (GlyRS/GARS)-induced peripheral neuropathy. Peripheral nerves and dorsal root ganglia from the C201R mutant Gars mouse model showed reduced acetylated α-tubulin levels. In primary dorsal root ganglion neurons, mutant GlyRS affected neurite length and disrupted normal mitochondrial transport. We demonstrated that GlyRS co-immunoprecipitated with HDAC6 and that this interaction was blocked by tubastatin A, a selective inhibitor of the deacetylating function of HDAC6. Moreover, HDAC6 inhibition restored mitochondrial axonal transport in mutant GlyRS-expressing neurons. Systemic delivery of a specific HDAC6 inhibitor increased α-tubulin acetylation in peripheral nerves and partially restored nerve conduction and motor behaviour in mutant Gars mice. Our study demonstrates that α-tubulin deacetylation and disrupted axonal transport may represent a common pathogenic mechanism underlying Charcot-Marie-Tooth disease and it broadens the therapeutic potential of selective HDAC6 inhibition to other genetic forms of axonal Charcot-Marie-Tooth disease.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/terapia , Glicina-ARNt Ligasa/genética , Histona Desacetilasa 6/metabolismo , Mutación/genética , Animales , Transporte Axonal/genética , Células Cultivadas , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/uso terapéutico , Ganglios Espinales/citología , Histona Desacetilasa 6/genética , Ácidos Hidroxámicos/uso terapéutico , Indoles/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Conducción Nerviosa/genética , Unión Neuromuscular/patología , Unión Neuromuscular/fisiopatología , Desempeño Psicomotor/fisiología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Tubulina (Proteína)/metabolismo
3.
J Neurooncol ; 118(2): 313-319, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24728831

RESUMEN

Perform a phase I study to evaluate the safety, and tolerability of vorinostat, an HDAC inhibitor, when combined with whole brain radiation treatment (WBRT) in patients with brain metastasis. A multi-institutional phase I clinical trial enrolled patients with a histological diagnosis of malignancy and radiographic evidence of brain metastasis. WBRT was 37.5 Gy in 2.5 Gy fractions delivered over 3 weeks. Vorinostat was administrated by mouth, once daily, Monday through Friday, concurrently with radiation treatment. The vorinostat dose was escalated from 200 to 400 mg daily using a 3+3 trial design. Seventeen patients were enrolled, 4 patients were excluded from the analysis due to either incorrect radiation dose (n = 1), or early treatment termination due to disease progression (n = 3). There were no treatment related grade 3 or higher toxicities in the 200 and 300 mg dose levels. In the 400 mg cohort there was a grade 3 pulmonary embolus and one death within 30 days of treatment. Both events were most likely related to disease progression rather than treatment; nonetheless, we conservatively classified the death as a dose limiting toxicity. We found Vorinostat administered with concurrent WBRT to be well tolerated to a dose of 300 mg once daily. This is the recommended dose for phase II study.


Asunto(s)
Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/terapia , Quimioradioterapia , Ácidos Hidroxámicos/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Anciano , Neoplasias Encefálicas/patología , Carcinoma de Pulmón de Células no Pequeñas/patología , Estudios de Cohortes , Progresión de la Enfermedad , Neoplasias de las Trompas Uterinas/patología , Femenino , Inhibidores de Histona Desacetilasas/efectos adversos , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Ácidos Hidroxámicos/efectos adversos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Fármacos Sensibilizantes a Radiaciones/efectos adversos , Resultado del Tratamiento , Vorinostat
4.
Nanomedicine ; 9(8): 1124-34, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23764660

RESUMEN

Nearly 40% of patients with non-invasive bladder cancer will progress to invasive disease despite locally-directed therapy. Overcoming the bladder permeability barrier (BPB) is a challenge for intravesical drug delivery. Using the fluorophore coumarin (C6), we synthesized C6-loaded poly(lactide-co-glycolide) (PLGA) nanoparticles (NPs), which were surface modified with a novel cell penetrating polymer, poly(guanidinium oxanorbornene) (PGON). Addition of PGON to the NP surface improved tissue penetration by 10-fold in intravesically-treated mouse bladder and ex vivo human ureter. In addition, NP-C6-PGON significantly enhanced intracellular uptake of NPs compared to NPs without PGON. To examine biological activity, we synthesized NPs that were loaded with the histone deacetylase (HDAC) inhibitor belinostat (NP-Bel-PGON). NP-Bel-PGON exhibited a significantly lower IC50 in cultured bladder cancer cells, and sustained hyperacetylation, when compared to unencapsulated belinostat. Xenograft tumors treated with NP-Bel-PGON showed a 70% reduction in volume, and a 2.5-fold higher intratumoral acetyl-H4, when compared to tumors treated with unloaded NP-PGON. FROM THE CLINICAL EDITOR: These authors demonstrate that PLGA nanoparticles with PGON surface functionalization result in greatly enhanced cell penetrating capabilities, and present convincing data from a mouse model of bladder cancer for increased chemotherapy efficacy.


Asunto(s)
Portadores de Fármacos/química , Inhibidores de Histona Desacetilasas/administración & dosificación , Ácidos Hidroxámicos/administración & dosificación , Nanopartículas/química , Sulfonamidas/administración & dosificación , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Línea Celular Tumoral , Portadores de Fármacos/metabolismo , Sistemas de Liberación de Medicamentos , Femenino , Inhibidores de Histona Desacetilasas/farmacocinética , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Ácidos Hidroxámicos/farmacocinética , Ácidos Hidroxámicos/uso terapéutico , Ratones , Nanopartículas/metabolismo , Poliglactina 910/química , Poliglactina 910/metabolismo , Sulfonamidas/farmacocinética , Sulfonamidas/uso terapéutico , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Urotelio/efectos de los fármacos , Urotelio/metabolismo , Urotelio/patología
5.
J Periodontal Res ; 46(6): 697-703, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21745207

RESUMEN

BACKGROUND AND OBJECTIVE: Bone loss caused by enhanced osteoclast activity is a significant feature of periodontitis. Histone deacetylase inhibitors (HDACi) can suppress osteoclast-mediated bone loss in vitro and in vivo. This study investigated whether HDACi can suppress bone loss in experimental periodontitis. MATERIAL AND METHODS: Experimental periodontitis was induced in mice by oral inoculation with Porphyromonas gingivalis bacteria. Mice were treated orally with olive oil alone, with olive oil and a novel compound - 1179.4b - which targets both Class I and Class II histone deacetylases (HDACs) or with olive oil and MS-275, which targets Class I HDACs. Micro-computed tomography scans of live mice, stereo imaging and histological analyses were used to detect changes in bone. RESULTS: In the absence of treatment there was a 13.2% increase in bone volume in controls compared with a 7.4% decrease in P. gingivalis-inoculated mice. 1179.4b significantly reduced bone loss, with a 3.4% increase in bone volume (p < 0.01). MS-275 did not have a significant effect on P. gingivalis-induced bone loss. Histological analysis revealed that 1179.4b reduced bone loss despite having no effect on inflammation. CONCLUSION: HDACi were found to effectively suppress bone loss in the mouse model of periodontitis. 1179.4b - the inhibitor of Class I and Class II HDACs - was more effective at suppressing bone loss than MS-275, which targets Class I HDACs only. These compounds may therefore have the potential to be used for the management of periodontitis.


Asunto(s)
Pérdida de Hueso Alveolar/enzimología , Pérdida de Hueso Alveolar/prevención & control , Inhibidores de Histona Desacetilasas/uso terapéutico , Pérdida de Hueso Alveolar/diagnóstico por imagen , Aminoquinolinas/uso terapéutico , Animales , Benzamidas/uso terapéutico , Densidad Ósea , Femenino , Ácidos Hidroxámicos/uso terapéutico , Imagenología Tridimensional , Ratones , Ratones Endogámicos BALB C , Aceite de Oliva , Osteoclastos/patología , Periodontitis/enzimología , Aceites de Plantas/uso terapéutico , Porphyromonas gingivalis , Piridinas/uso terapéutico , Microtomografía por Rayos X
6.
Colloids Surf B Biointerfaces ; 166: 37-44, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29533842

RESUMEN

Histone deacetylases (HDACs) are known to be key enzymes in cancer development and progression through their modulation of chromatin structure and the expression and post-translational modification of numerous proteins. Aggressive dedifferentiated tumors, like glioblastoma, frequently overexpress HDACs, while HDAC inhibition can lead to cell cycle arrest, promote cellular differentiation and induce apoptosis. Although multiple HDAC inhibitors, such as quisinostat, are of interest in oncology due to their potent in vitro efficacy, their failure in the clinic as monotherapies against solid tumors has been attributed to poor delivery. Thus, we were motivated to develop quisinostat loaded poly(D,L-lactide)-b-methoxy poly(ethylene glycol) nanoparticles (NPs) to test their ability to treat orthotopic glioblastoma. In developing our NP formulation, we identified a novel, pH-driven approach for achieving over 9% (w/w) quisinostat loading. We show quisinostat-loaded NPs maintain drug potency in vitro and effectively slow tumor growth in vivo, leading to a prolonged survival compared to control mice.


Asunto(s)
Glioblastoma/tratamiento farmacológico , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/uso terapéutico , Polietilenglicoles/química , Animales , Sistemas de Liberación de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Ratones
7.
Int J Nanomedicine ; 12: 1385-1399, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28260886

RESUMEN

BACKGROUND: The pan-histone deacetylase inhibitor panobinostat is a potential therapy for malignant glioma, but it is water insoluble and does not cross the blood-brain barrier when administered systemically. In this article, we describe the in vitro and in vivo efficacy of a novel water-soluble nano-micellar formulation of panobinostat designed for administration by convection enhanced delivery (CED). MATERIALS AND METHODS: The in vitro efficacy of panobinostat-loaded nano-micelles against rat F98, human U87-MG and M059K glioma cells and against patient-derived glioma stem cells was measured using a cell viability assay. Nano-micelle distribution in rat brain was analyzed following acute CED using rhodamine-labeled nano-micelles, and toxicity was assayed using immunofluorescent microscopy and synaptophysin enzyme-linked immunosorbent assay. We compared the survival of the bioluminescent syngenic F98/Fischer344 rat glioblastoma model treated by acute CED of panobinostat-loaded nano-micelles with that of untreated and vehicle-only-treated controls. RESULTS: Nano-micellar panobinostat is cytotoxic to rat and human glioma cells in vitro in a dose-dependent manner following short-time exposure to drug. Fluorescent rhodamine-labelled nano-micelles distribute with a volume of infusion/volume of distribution (Vi/Vd) ratio of four and five respectively after administration by CED. Administration was not associated with any toxicity when compared to controls. CED of panobinostat-loaded nano-micelles was associated with significantly improved survival when compared to controls (n=8 per group; log-rank test, P<0.001). One hundred percent of treated animals survived the 60-day experimental period and had tumour response on post-mortem histological examination. CONCLUSION: CED of nano-micellar panobinostat represents a potential novel therapeutic option for malignant glioma and warrants translation into the clinic.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Convección , Sistemas de Liberación de Medicamentos , Glioma/tratamiento farmacológico , Ácidos Hidroxámicos/uso terapéutico , Indoles/uso terapéutico , Micelas , Nanopartículas/química , Poloxámero/química , Animales , Muerte Celular , Línea Celular Tumoral , Supervivencia Celular , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Humanos , Ácidos Hidroxámicos/administración & dosificación , Indoles/administración & dosificación , Panobinostat , Ratas Endogámicas F344 , Ratas Wistar , Análisis de Supervivencia
8.
Nanomedicine (Lond) ; 12(1): 43-58, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27879153

RESUMEN

AIM: To improve the bioavailability and anticancer potential of suberoylanilide hydroxamic acid (SAHA) by developing a drug-loaded polymeric nanomicellar system. METHODS: SAHA-loaded Poly(ethylene glycol)-block-poly(caprolactone) (PEG-PCL) micelles were developed, and physico-chemically characterized. In vitro cellular uptake, viability and apoptosis-inducing ability of the SAHA-PEG-PCL micelles were investigated. In vivo anticancer activity was evaluated in C57BL/6 mice-bearing tumor. RESULTS: The SAHA-PEG-PCL micelles had optimum size (∼130 nm) with an entrapment efficiency of approximately 67%. The SAHA-PEG-PCL induced stronger cell cycle arrest in G2/M phase leading to higher rate of apoptosis compared to free SAHA. SAHA-PEG-PCL demonstrated significant tumor suppression compared to free SAHA in vivo. CONCLUSION: The physicochemical properties and the antitumor efficacy of SAHA were improved by encapsulating in polymeric micelles.


Asunto(s)
Antineoplásicos/farmacología , Portadores de Fármacos/química , Ácidos Hidroxámicos/farmacología , Melanoma Experimental/tratamiento farmacológico , Nanopartículas , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Disponibilidad Biológica , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Liberación de Fármacos , Femenino , Humanos , Ácidos Hidroxámicos/farmacocinética , Ácidos Hidroxámicos/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Micelas , Nanopartículas/química , Poliésteres/química , Polietilenglicoles/química , Vorinostat
9.
Clin Lymphoma Myeloma Leuk ; 17(7): 424-432, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28655599

RESUMEN

INTRODUCTION/BACKGROUND: Deacetylase inhibitors have synergistic activity in combination with proteasome inhibitors and anthracyclines in preclinical models of multiple myeloma (MM). We therefore evaluated the safety and efficacy of the deacetylase inhibitor vorinostat in combination with pegylated liposomal doxorubicin (PLD) and bortezomib in relapsed/refractory MM. PATIENTS AND METHODS: Thirty-two patients were treated with PLD and bortezomib in combination with escalating doses of vorinostat on days 4 to 11 or 1 to 14. RESULTS: The maximum tolerated dose of vorinostat was 400 mg on days 4 to 11. Neutropenia and thrombocytopenia attributable to protocol therapy were seen in 59% and 94% of patients, of which 37% and 47% were of grade 3 or higher severity, respectively. Constitutional and gastrointestinal adverse events of all grades were common, the majority of which were less than grade 3 in severity. The overall response rate (partial response rate or better) was 65% and the clinical benefit rate (minimal response rate or better) 74%. The overall response rate was 83%, 71%, and 45% for patients with bortezomib-naive, -sensitive, and -refractory MM, respectively. The median progression-free survival was 13.9 months and the 3-year overall survival 77%. Whole blood proteasome activity assays demonstrated a potential impact of vorinostat on the chymotryptic-like activity of the proteasome. CONCLUSION: Further evaluation of PLD, bortezomib, and deacetylase inhibitor combinations is warranted, with special attention directed toward strategies to improve tolerability.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bortezomib/uso terapéutico , Doxorrubicina/análogos & derivados , Ácidos Hidroxámicos/uso terapéutico , Inhibidores de Proteasoma/uso terapéutico , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bortezomib/administración & dosificación , Bortezomib/farmacología , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos , Femenino , Humanos , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/farmacología , Masculino , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia , Polietilenglicoles/administración & dosificación , Polietilenglicoles/farmacología , Polietilenglicoles/uso terapéutico , Inhibidores de Proteasoma/administración & dosificación , Inhibidores de Proteasoma/farmacología , Vorinostat
10.
Neurotherapeutics ; 14(2): 417-428, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27957719

RESUMEN

Charcot-Marie-Tooth disease (CMT) is the most common inherited peripheral neuropathy, with an estimated prevalence of 1 in 2500. The degeneration of motor and sensory nerve axons leads to motor and sensory symptoms that progress over time and have an important impact on the daily life of these patients. Currently, there is no curative treatment available. Recently, we identified histone deacetylase 6 (HDAC6), which deacetylates α-tubulin, as a potential therapeutic target in axonal CMT (CMT2). Pharmacological inhibition of the deacetylating function of HDAC6 reversed the motor and sensory deficits in a mouse model for mutant "small heat shock protein B1" (HSPB1)-induced CMT2 at the behavioral and electrophysiological level. In order to translate this potential therapeutic strategy into a clinical application, small drug-like molecules that are potent and selective HDAC6 inhibitors are essential. To screen for these, we developed a method that consisted of 3 distinct phases and that was based on the pathological findings in the mutant HSPB1-induced CMT2 mouse model. Three different inhibitors (ACY-738, ACY-775, and ACY-1215) were tested and demonstrated to be both potent and selective HDAC6 inhibitors. Moreover, these inhibitors increased the innervation of the neuromuscular junctions in the gastrocnemius muscle and improved the motor and sensory nerve conduction, confirming that HDAC6 inhibition is a potential therapeutic strategy in CMT2. Furthermore, ACY-1215 is an interesting lead molecule as it is currently tested in clinical trials for cancer. Taken together, these results may speed up the translation of pharmacological inhibition of HDAC6 into a therapy against CMT2.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/tratamiento farmacológico , Enfermedad de Charcot-Marie-Tooth/enzimología , Evaluación Preclínica de Medicamentos , Histona Desacetilasa 6/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Pirimidinas/farmacología , Animales , Transporte Axonal/efectos de los fármacos , Ganglios Espinales/efectos de los fármacos , Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/inervación , Unión Neuromuscular/efectos de los fármacos , Neuronas/efectos de los fármacos , Pirimidinas/uso terapéutico , Células Tumorales Cultivadas
11.
Clin Cancer Res ; 11(9): 3392-401, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15867240

RESUMEN

ErbB2-overexpressing human cancers represent potentially sensitive targets for therapy by candidate histone deacetylase (HDAC) inhibitors as we have shown that HDAC inhibitors can selectively reduce ErbB2 expression by repressing the ErbB2 promoter and accelerating the decay of cytoplasmic ErbB2 transcripts. To extend these in vitro findings and enhance the in vivo pharmacodynamic properties of HDAC inhibitors, we stably encapsulated a potent hydroxamate-based HDAC inhibitor (LAQ824) within long-circulating liposomes (Ls-LAQ824) and immunoliposomes (ILs-LAQ824) bearing >10,000 LAQ824 molecules per nanovesicle. Liposomal LAQ824 exhibits prolonged in vivo stability and, unlike free LAQ824, circulates with a half-life of 10.8 hours following a single i.v. injection. Three weekly i.v. injections of 20 to 25 mg/kg Ls-LAQ824 in nude mice with ErbB2 overexpressing BT-474 breast tumor xenografts significantly impairs tumor growth, and administration of ErbB2-targeted ILs-LAQ824 may further improve this antitumor activity. Studies of tumor-bearing mice 24 hours after single treatment indicate that: (a) >10% of injected liposomal LAQ824 is still circulating (whereas free LAQ824 is undetectable in the blood after 15 minutes); and (b) tumor uptake of Ls-LAQ824 and ILs-LAQ824 is >3% injected drug per gram of tumor, producing levels of acetylated tumor histones that are 5- to 10-fold increased over those following free LAQ824 or saline treatments and resulting in concordantly reduced levels of tumor ErbB2 mRNA. These preclinical results support the clinical evaluation of HDAC inhibitors against ErbB2-overexpressing malignancies, and further indicate that encapsulation into targeted and nontargeted liposomes substantially improves the in vivo pharmacokinetics, tumor uptake, and antitumor properties of hydroxamate-based HDAC inhibitors.


Asunto(s)
Ácidos Hidroxámicos/farmacología , Neoplasias Experimentales/prevención & control , Receptor ErbB-2/inmunología , Animales , Área Bajo la Curva , Northern Blotting , Western Blotting , Cápsulas , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacocinética , Ácidos Hidroxámicos/uso terapéutico , Liposomas/inmunología , Ratones , Ratones Desnudos , Neoplasias Experimentales/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Sci Transl Med ; 8(326): 326ra23, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26888431

RESUMEN

Histone deacetylase inhibitors (HDACi) are approved for treating rare cancers and are of interest as potential therapies for neurodegenerative disorders. We evaluated a triple combination formulation (TCF) comprising the pan-HDACi vorinostat, the caging agent 2-hydroxypropyl-ß-cyclodextrin (HPBCD), and polyethylene glycol (PEG) for treating a mouse model (the Npc1(nmf164) mouse) of Niemann-Pick type C (NPC) disease, a difficult-to-treat cerebellar disorder. Vorinostat alone showed activity in cultured primary cells derived from Npc1(nmf164) mice but did not improve animal survival. However, low-dose, once-weekly intraperitoneal injections of the TCF containing vorinostat increased histone acetylation in the mouse brain, preserved neurites and Purkinje cells, delayed symptoms of neurodegeneration, and extended mouse life span from 4 to almost 9 months. We demonstrate that the TCF boosted the ability of HDACi to cross the blood-brain barrier and was not toxic even when used long term. Further, the TCF enabled dose reduction, which has been a major challenge in HDACi therapy. TCF simultaneously treats neurodegenerative and systemic symptoms of Niemann-Pick type C disease in a mouse model.


Asunto(s)
Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/uso terapéutico , Enfermedad de Niemann-Pick Tipo C/tratamiento farmacológico , 2-Hidroxipropil-beta-Ciclodextrina , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Inhibidores de Histona Desacetilasas/sangre , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/sangre , Ácidos Hidroxámicos/uso terapéutico , Inflamación/sangre , Inflamación/complicaciones , Inflamación/patología , Hígado/efectos de los fármacos , Hígado/patología , Ratones , Mutación/genética , Degeneración Nerviosa/sangre , Degeneración Nerviosa/complicaciones , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/patología , Enfermedad de Niemann-Pick Tipo C/sangre , Enfermedad de Niemann-Pick Tipo C/complicaciones , Enfermedad de Niemann-Pick Tipo C/patología , Polietilenglicoles/química , Células de Purkinje/efectos de los fármacos , Células de Purkinje/metabolismo , Células de Purkinje/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Bazo/efectos de los fármacos , Bazo/patología , Análisis de Supervivencia , Vorinostat , beta-Ciclodextrinas/química
13.
Aliment Pharmacol Ther ; 13(11): 1535-42, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10571613

RESUMEN

BACKGROUND: Recent publications have reported that matrix metalloproteinases (MMPs) are expressed in colonic tissue taken from ulcerative colitis and Crohn's disease patients. AIM: To evaluate the effects of a matrix metalloproteinase inhibitor, marimastat, on colonic inflammation in experimental colitis induced by trinitrobenzenesulphonic acid (TNBS)-ethanol in the rat. METHODS: Rats were dosed (by mouth) for 7 days (b.d.) with either sulphasalazine (50 mg/kg), marimastat (40 mg/kg) or vehicle. TNBS-ethanol was administered rectally on the 4th day of dosing. On the last day of dosing, colons were removed and assessed for inflammation using myeloperoxidase activity, production of soluble TNFalpha (tumour necrosis factor alpha), clinical score and histological assessment. In addition, the bioavailability and effect of marimastat on a range of MMPs were assessed in-vitro. RESULTS: In this study we have confirmed that marimastat is a broad spectrum MMPI with a bioavailability of 5%. TNBS rats dosed with sulphasalazine had a significantly lower (P < 0.05) myeloperoxidase activity, TNFalpha production and a markedly lower clinical score. Similarly, rats dosed with marimastat had a significantly lower (P < 0.05) myeloperoxidase activity and clinical score, but the TNFalpha production was not significantly reduced. CONCLUSIONS: Dosing rats with TNBS-induced colitis using sulphasalazine or marimastat produced a significant reduction in tissue injury and inflammation.


Asunto(s)
Colitis/tratamiento farmacológico , Ácidos Hidroxámicos/uso terapéutico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Inhibidores de la Metaloproteinasa de la Matriz , Inhibidores de Proteasas/uso terapéutico , Ácido Trinitrobencenosulfónico , Animales , Disponibilidad Biológica , Colitis/inducido químicamente , Colitis/patología , Ácidos Hidroxámicos/farmacocinética , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/patología , Masculino , Metaloproteinasas de la Matriz/metabolismo , Peroxidasa/metabolismo , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/metabolismo
14.
J Thorac Cardiovasc Surg ; 125(3): 602-10, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12658202

RESUMEN

BACKGROUND: Global and regional shape changes that occur within the left ventricular wall after myocardial infarction have been termed infarct expansion. A potential mechanism for this postinfarction remodeling is activation of the matrix metalloproteinases. Accordingly, the present study examined the effects of matrix metalloproteinase inhibition on left ventricular global geometry after myocardial infarction in pigs. METHODS: Myocardial infarction was created in pigs by means of occlusion of the first and second obtuse marginal branches of the circumflex coronary artery, resulting in a uniform left ventricular free wall infarct size of 21% +/- 2%. At 5 days after infarction, the pigs were randomized to undergo broad-spectrum matrix metalloproteinase inhibition (n = 9; PD166793, 20 mg. kg(-1). d(-1) by mouth) or myocardial infarction alone (n = 8). Ten pigs served as noninfarction control animals. Left ventricular end-diastolic area, determined by means of echocardiography, was measured 8 weeks after infarction. RESULTS: Left ventricular end-diastolic area increased in both the myocardial infarction plus broad-spectrum matrix metalloproteinase inhibition and myocardial infarction only groups compared to reference control animals (3.7 +/- 0.2 cm(2)), but was reduced with broad-spectrum matrix metalloproteinase inhibition compared to myocardial infarction alone (4.5 +/- 0.2 vs 4.9 +/- 0.2 cm(2), respectively; P <.05). Regional radial stress within the infarct region increased in both infarction groups when compared to values obtained from reference control animals (599 +/- 152 g/cm(2)), but was attenuated in the myocardial infarction plus broad-spectrum matrix metalloproteinase inhibition group compared to the myocardial infarction alone group (663 +/- 108 vs 1242 +/- 251 g/cm(2), respectively; P <.05). Similarly, regional myocardial stiffness increased in both the myocardial infarction plus broad-spectrum matrix metalloproteinase inhibition and the myocardial infarction only groups compared with that observed in reference control animals (14 +/- 1 rkm, P <.05) but was lower with broad-spectrum matrix metalloproteinase inhibition than with myocardial infarction alone (42 +/- 6 vs 68 +/- 10 rkm, respectively; P <.05). CONCLUSIONS: Matrix metalloproteinase inhibition reduced postinfarction left ventricular dilation, reduced regional myocardial wall stress, and modified myocardial material properties. These unique findings suggest that increased myocardial matrix metalloproteinase activation after infarction contributes directly to the left ventricular remodeling process.


Asunto(s)
Modelos Animales de Enfermedad , Ácidos Hidroxámicos/uso terapéutico , Inhibidores de la Metaloproteinasa de la Matriz , Metaloproteinasas de la Matriz/fisiología , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/enzimología , Oligopéptidos/uso terapéutico , Inhibidores Tisulares de Metaloproteinasas/uso terapéutico , Disfunción Ventricular Izquierda/etiología , Remodelación Ventricular/efectos de los fármacos , Análisis de Varianza , Animales , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Ecocardiografía Transesofágica , Hemodinámica/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/diagnóstico , Oligopéptidos/farmacología , Distribución Aleatoria , Volumen Sistólico/efectos de los fármacos , Porcinos , Factores de Tiempo , Inhibidores Tisulares de Metaloproteinasas/farmacología , Disfunción Ventricular Izquierda/prevención & control , Presión Ventricular/efectos de los fármacos
15.
J Endod ; 22(12): 668-73, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9220752

RESUMEN

To investigate the effects of a combination of an antibacterial agent (ofloxacin) and a collagenase inhibitor (FN-439) in the root canal treatment of apical periodontitis, we studied the healing process of experimentally induced periapical lesions in rats by using immunohistochemical methods. With a topical application of a combination of ofloxacin and FN-439 following experimentally induced periapical lesions, both neutrophils and macrophages became significantly decreased in number, while active cementogenesis and extensive bone formation were seen in the periapical region. However, the use of ofloxacin alone also demonstrated a beneficial effect on periapical inflammation and healing. Therefore, it is suggested that ofloxacin is powerful against bacterial infection whether FN-439 is added. The only observed effect of a combination of ofloxacin and FN-439 is that it may more effectively inhibit osteoclastic bone resorption and activate the remodeling of the apical periodontal tissue if this combined medicament is used in a stage of active bone destruction characterized by high production of tissue collagenase.


Asunto(s)
Antiinfecciosos/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Ofloxacino/uso terapéutico , Oligopéptidos/uso terapéutico , Periodontitis Periapical/tratamiento farmacológico , Inhibidores de Proteasas/uso terapéutico , Irrigantes del Conducto Radicular/uso terapéutico , Pérdida de Hueso Alveolar/tratamiento farmacológico , Animales , Antiinfecciosos/metabolismo , Antiinfecciosos/farmacología , Remodelación Ósea/efectos de los fármacos , Cemento Dental/efectos de los fármacos , Combinación de Medicamentos , Formocresoles/administración & dosificación , Formocresoles/metabolismo , Ácidos Hidroxámicos/metabolismo , Ácidos Hidroxámicos/farmacología , Mandíbula , Diente Molar , Ofloxacino/metabolismo , Ofloxacino/farmacología , Oligopéptidos/metabolismo , Oligopéptidos/farmacología , Inhibidores de Proteasas/metabolismo , Ratas , Ratas Wistar , Irrigantes del Conducto Radicular/metabolismo , Irrigantes del Conducto Radicular/farmacología , Estadísticas no Paramétricas
16.
Biomaterials ; 33(17): 4424-30, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22425551

RESUMEN

The chemical conjugate of low molecular weight heparin with taurocholate (LHT7) was previously designed to offer anticancer activity while minimizing the anticoagulant activity. In the present study, we found that the systemic administration of LHT7 in nanolipoplex could substantially enhance tumor vasculature targeting and anticancer effects. Moreover, we found that co-delivery of LHT7 with suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor, in nanolipoplex could provide synergistic antitumor effect. LHT7/SAHA nanolipoplex was formulated by encapsulating SAHA inside cationic liposomes, followed by complexation of negatively charged LHT7 onto the cationic surfaces of SAHA-loaded liposomes (SAHA-L). LHT7/SAHA nanolipoplex was positively charged with a mean diameter of 117.6 nm, and stable in serum. The nanolipoplex form of LHT7 could alter its pharmacokinetics and biodistribution. Compared to the free form of LHT7, LHT7 in the nanolipoplex showed 1.9-fold higher mean residence time, and higher tumor vasculature accumulation after its intravenous administration. LHT7/SAHA nanolipoplex showed highest antitumor efficacy in SCC-bearing mice, compared to LHT7, SAHA-L and sequential co-administration of LHT7 and SAHA-L. Consistent with the enhanced antitumor effect, the reduction of abnormal vessels in the tumor site was also the highest in the LHT7/SAHA nanolipoplex-treated group. These results suggested the potential of LHT7/SAHA nanolipoplex for enhanced tumor vasculature targeting, and the importance of nanolipoplex-mediated co-delivery with a histone deacetylase inhibitor for maximal anticancer effect.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Heparina/análogos & derivados , Ácidos Hidroxámicos/uso terapéutico , Liposomas/química , Nanopartículas/química , Neoplasias/irrigación sanguínea , Neovascularización Patológica/tratamiento farmacológico , Ácido Taurocólico/uso terapéutico , Animales , Antineoplásicos/farmacología , Cationes , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayo de Cambio de Movilidad Electroforética , Heparina/administración & dosificación , Heparina/farmacocinética , Heparina/farmacología , Heparina/uso terapéutico , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/farmacología , Inyecciones Intravenosas , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Ratas , Ratas Sprague-Dawley , Suero/metabolismo , Ácido Taurocólico/administración & dosificación , Ácido Taurocólico/farmacocinética , Ácido Taurocólico/farmacología , Factores de Tiempo , Distribución Tisular/efectos de los fármacos , Vorinostat
17.
J Control Release ; 155(1): 60-6, 2011 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-20971142

RESUMEN

Oligolysine-based cationic lipid derivatives were synthesized for delivery of siRNA, and formulated into cationic liposomes. Among various oligolysine-based lipid derivatives differing in lysine residue number and lipid moiety, trilysinoyl oleylamide (TLO)-based liposomes (TLOL) showed the highest delivery efficiency combined with minimal cytotoxicity. Delivery of siRNA using TLOL silenced target genes both in vitro and in vivo. In green fluorescent protein (GFP)-expressing tumor tissue, a significant reduction of fluorescence was observed after intratumoral administration of siGFP using TLOL compared with control siGL2. Intravenous administration of siMcl1 employing pegylated TLOL (pTLOL) reduced the expression of human Mcl1 protein in KB-xenografted tumor tissue. Despite the reduction in target protein Mcl1 expression following such systemic delivery, tumor growth was only slightly reduced compared to a siGL2-treated control group. To potentiate the anticancer activity of siMcl1, the anticancer drug suberoylanilide hydroxamic acid (SAHA) was additionally encapsulated in pTLOL. After intravenous administration of siMcl1 using SAHA-loaded pTLOL (pSTLOL), a significant reduction in tumor growth was observed compared to that seen in animals treated with free SAHA or siGL2 complexed with pSTLOL. The results indicate that pTLOL could be further developed as a systemic delivery system for synergistic anticancer siRNA and a drug.


Asunto(s)
Antineoplásicos/administración & dosificación , Cationes/química , Ácidos Hidroxámicos/administración & dosificación , Liposomas/química , Neoplasias/tratamiento farmacológico , Ácidos Oléicos/química , ARN Interferente Pequeño/administración & dosificación , Animales , Antineoplásicos/uso terapéutico , Línea Celular , Femenino , Silenciador del Gen , Humanos , Ácidos Hidroxámicos/uso terapéutico , Ratones , Ratones Desnudos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Neoplasias/genética , Neoplasias/patología , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Vorinostat
19.
Int J Pharm ; 397(1-2): 184-93, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20603204

RESUMEN

Histone deacetylase (HDAC) inhibitors (HDACi) of the class I trichostatin A (TSA), CG1521 (CG), and PXD101 (PXD) were incorporated at a high rate (approximately 1mM) in liposomes made of egg phosphatidylcholine/cholesterol/distearoylphosphoethanolamine-polyethylenglycol(2000) (64:30:6). Physicochemical parameters (size, zeta potential, loading, stability, release kinetics) of these HDACi-loaded pegylated liposomes were optimized and their cytotoxicity (MTT test) was measured in MCF-7, T47-D, MDA-MB-231 and SkBr3 breast cancer cell lines. In MCF-7 cells, TSA and PXD were efficient inducers of proteasome-mediated estradiol receptor alpha degradation and they both affected estradiol-induced transcription (TSA>PXD) contrary to CG. Moreover, TSA most efficiently altered breast cancer cell viability as compared to the free drug, CG-liposomes being the weakest, while unloaded liposomes had nearly no cytotoxicity. Pegylated liposomes loaded with TSA or PXD remained stable in size, charge and biological activity for one month when stored at 4 degrees C. All HDACi-loaded liposomes released slowly the encapsulated drug in vitro, CG-loaded liposomes showed the slowest release kinetic. These formulations could improve the efficacy of HDACi not only in breast cancers but also in other solid tumors because most of these drugs are poor water soluble and unstable in vivo, and their administration remains a challenge.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/administración & dosificación , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/uso terapéutico , Liposomas , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Fenómenos Químicos , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Estabilidad de Medicamentos , Estradiol/metabolismo , Receptor alfa de Estrógeno/metabolismo , Femenino , Inhibidores de Histona Desacetilasas/farmacocinética , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Ácidos Hidroxámicos/farmacocinética , Tamaño de la Partícula , Sulfonamidas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA