Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Neurosurg ; 134(3): 701-710, 2020 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-32168477

RESUMEN

OBJECTIVE: Glutamatergic signaling significantly promotes proliferation, migration, and invasion in glioblastoma (GBM). Riluzole, a metabotropic glutamate receptor 1 inhibitor, reportedly suppresses GBM growth. However, the effects of combining riluzole with the primary GBM chemotherapeutic agent, temozolomide (TMZ), are unknown. This study aimed to investigate the efficacy of combinatorial therapy with TMZ/riluzole for GBM in vitro and in vivo. METHODS: Three GBM cell lines, T98G (human; O6-methylguanine DNA methyltransferase [MGMT] positive), U87MG (human; MGMT negative), and GL261 (murine; MGMT positive), were treated with TMZ, riluzole, or a combination of both. The authors performed cell viability assays, followed by isobologram analysis, to evaluate the effects of combinatorial treatment for each GBM cell line. They tested the effect of riluzole on MGMT, a DNA repair enzyme causing chemoresistance to TMZ, through quantitative real-time reverse transcription polymerase chain reaction in T98G cells. Furthermore, they evaluated the efficacy of combinatorial TMZ/riluzole treatment in an orthotopic mouse allograft model of MGMT-positive GBM using C57BL/6 J mice and GL261 cells. RESULTS: Riluzole displayed significant time- and dose-dependent growth-inhibitory effects on all GBM cell lines assessed independently. Riluzole enhanced the antitumor effect of TMZ synergistically in MGMT-positive but not in MGMT-negative GBM cell lines. Riluzole singularly suppressed MGMT expression, and it significantly suppressed TMZ-induced MGMT upregulation (p < 0.01). Furthermore, combinatorial TMZ/riluzole treatment significantly suppressed tumor growth in the intracranial MGMT-positive GBM model (p < 0.05). CONCLUSIONS: Riluzole attenuates TMZ-induced MGMT upregulation and enhances the antitumor effect of TMZ in MGMT-positive GBMs. Therefore, combinatorial TMZ/riluzole treatment is a potentially promising novel therapeutic regimen for MGMT-positive GBMs.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Metilasas de Modificación del ADN/antagonistas & inhibidores , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Glioblastoma/tratamiento farmacológico , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Animales , Antineoplásicos Alquilantes/administración & dosificación , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Metilasas de Modificación del ADN/biosíntesis , Metilasas de Modificación del ADN/genética , Reparación del ADN/efectos de los fármacos , Enzimas Reparadoras del ADN/biosíntesis , Enzimas Reparadoras del ADN/genética , Sinergismo Farmacológico , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Riluzol/administración & dosificación , Temozolomida/administración & dosificación , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Oncol Rep ; 43(1): 169-176, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31789402

RESUMEN

Osteosarcomas are the most commonly occurring malignant bone cancer in young individuals. The survival rate of patients with metastatic osteosarcoma is low and has been stagnant for over two decades. We previously demonstrated that the glutamate release inhibitor, riluzole inhibits osteosarcoma cell growth. Towards the development of more effective therapy, we investigated the delivery of riluzole in human metastatic osteosarcoma xenografts in mice. We compared the efficacy of riluzole delivery by intraperitoneally injecting either free riluzole or riluzole released via two different shapes of iron oxide nanoparticles (nanocage or nanosphere) of size 15±2.5 nm. We monitored tumor size using Vernier calipers and bioluminescence assay and found a significant reduction in tumor size in the riluzole­treated groups when injected, either in free form or via nanoparticles, compared to the control groups (PBS, nanosphere or nanocage). Importantly, nanocage­delivered riluzole was most effective in reducing tumor size in the xenograft nude mice. While riluzole delivery induced apoptosis in tumor tissues in all three groups of riluzole­treated animals, it was highest in tumors from the nanocage­delivered riluzole group. Therefore, we conclude that riluzole is an effective drug to reduce tumor size in osteosarcoma and the efficacy of riluzole as a apoptotic and tumor­reducing drug is enhanced when delivered via nanocage.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Compuestos Férricos/química , Osteosarcoma/tratamiento farmacológico , Riluzol/administración & dosificación , Animales , Neoplasias Óseas/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Composición de Medicamentos , Humanos , Inyecciones Intraperitoneales , Masculino , Nanopartículas del Metal , Ratones , Ratones Desnudos , Osteosarcoma/patología , Riluzol/química , Riluzol/farmacología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Clin Ther ; 41(12): 2490-2499, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31635890

RESUMEN

PURPOSE: During amyotrophic lateral sclerosis progression, up to 85% of patients develop dysphagia. Riluzole oral suspension 50 mg/10 mL is bioequivalent to riluzole 50-mg film-coated tablets administered orally under fasting conditions. Here, we compare the bioavailability of a single 50-mg dose of riluzole oral suspension via intragastric tube, a proxy for percutaneous endoscopic gastrostomy administration, with that of oral administration in healthy volunteers under fasting conditions. Secondary objectives included the plasma pharmacokinetic and safety profiles of each administration route. METHODS: This was a single-center, single-dose, open-label, randomized, 2-period, 2-sequence, crossover bioequivalence/bioavailability study. Healthy volunteers were randomized to riluzole oral suspension 50 mg/10 mL either via nasogastric tube or orally, with a 5-day washout before crossover. FINDINGS: A total of 32 subjects were randomized (safety population); 30 were eligible for pharmacokinetic analysis. The ratios (nasogastric tube/oral) of the geometric least squares means and the geometric 90% CIs of AUC0-t, AUC0-inf, and Cmax were calculated to be 90.60% (85.66%-95.82%), 90.43% (85.47%-95.67%), and 96.99% (89.40%-105.23%), respectively, indicating bioequivalence. No significant differences in Cmax, Tmax, Kel, and t1/2el between treatments were found. Overall, riluzole oral suspension was well tolerated. No deaths or other serious adverse events were reported. IMPLICATIONS: In this study, riluzole oral suspension was bioequivalent when administered intragastrically and orally in healthy subjects under fasting conditions. Both administration methods were well tolerated. These results show that intragastric administration of riluzole oral suspension may provide an important formulation option in people with amyotrophic lateral sclerosis who have a percutaneous endoscopic gastrostomy tube.


Asunto(s)
Gastrostomía/instrumentación , Riluzol , Administración Oral , Disponibilidad Biológica , Nutrición Enteral , Ayuno , Humanos , Intubación Gastrointestinal , Riluzol/administración & dosificación , Riluzol/sangre , Riluzol/farmacocinética , Suspensiones
4.
J Psychopharmacol ; 33(3): 305-315, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30526230

RESUMEN

BACKGROUND: Riluzole is a glutamate regulator and effective in treatment of neuropsychiatric conditions. AIMS: We assessed riluzole for treatment of methamphetamine dependence. METHODS: In this randomized, double-blind, placebo-controlled clinical trial, male outpatients with methamphetamine dependence who were 18-65 years old received either 50 mg riluzole ( n=34) or placebo ( n=54) twice daily for 12 weeks. Patients were excluded in case of comorbid serious medical conditions or neurologic disorders, comorbid psychiatric disorders other than methamphetamine dependence requiring specific treatment interventions, simultaneous positive urine test result for substances of abuse other than methamphetamine, smoking >3 days per week, simultaneous consumption of medications which are contraindicated or have interaction with riluzole. RESULTS: Concerning primary outcomes, the cumulative mean number of attended weekly visits was higher in the riluzole arm compared with the placebo arm approaching a statistically significant difference (riluzole, median (range)=13.00 (2.00-13.00); placebo=4.00 (2.00-13.00); Mann-Whitney U=505.00, p-value=0.073), and the weekly measured rate of positive methamphetamine urine test results was significantly lower in the riluzole arm by the end of the study (riluzole=1 (5.00%), placebo=9 (45.00%), p-value=0.004). Patients in the riluzole arm experienced significantly greater improvement on all the craving, withdrawal, and depression measures regarding mean score changes from baseline to endpoint. No significant difference was detected between the two arms in terms of incidence of adverse events. CONCLUSION: Future randomized clinical trials are needed to investigate proper dosing strategy in a more inclusive sample.


Asunto(s)
Trastornos Relacionados con Anfetaminas/tratamiento farmacológico , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Metanfetamina/efectos adversos , Riluzol/administración & dosificación , Adolescente , Adulto , Anciano , Ansia/efectos de los fármacos , Depresión/epidemiología , Método Doble Ciego , Antagonistas de Aminoácidos Excitadores/efectos adversos , Antagonistas de Aminoácidos Excitadores/farmacología , Humanos , Masculino , Persona de Mediana Edad , Riluzol/efectos adversos , Riluzol/farmacología , Resultado del Tratamiento , Adulto Joven
5.
Anticancer Res ; 39(1): 207-214, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30591460

RESUMEN

BACKGROUND/AIM: Glioblastoma multiforme (GBM) is a malignant primary brain tumor with high rates of recurrence. This study aimed to investigate the effect of repurposed drug combinations on GBM. MATERIALS AND METHODS: Viability of U87 MG and 11ST patient-derived GMB cell lines, after valproic acid, tranylcypromine or riluzole alone, in different combinations, as well as combined with standard temozolomide chemotherapy was examined using the MTT assay. Proliferation, mRNA level of tissue factor pathway inhibitor 2 (TFPI2), and cell invasion were evaluated using anti-Ki-67 antibody staining, reverse transcriptase-polymerase chain reaction and xCELLigence system. RESULTS: The strongest effect on cell viability was achieved by the combination of riluzole with valproic acid (U87MG: 27.2%, 11ST: 25.99%). Tranylcypromine significantly enhanced the effect of temozolomide when used in combination, as did valproic acid. The normally high proliferation of GBM significantly declined under treatment with valproic acid with tranylcypromine (p=0.01). Finally, we observed reduction of invasion comparing single tranylcypromine to its combination with valproic acid or riluzole. CONCLUSION: These results support the idea that combinations of drugs could increase the treatment efficiency of GBM.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Glioblastoma/tratamiento farmacológico , Glicoproteínas/genética , Recurrencia Local de Neoplasia/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Glioblastoma/patología , Humanos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Riluzol/administración & dosificación , Temozolomida/administración & dosificación , Tranilcipromina/administración & dosificación , Ácido Valproico/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Eur J Pharm Sci ; 120: 30-39, 2018 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-29704642

RESUMEN

Riluzole is currently one of two approved medications for the treatment of amyotrophic lateral sclerosis (ALS). However, brain disposition of riluzole, as a substrate of P-glycoprotein (P-gp), is limited by the efflux transporters at the blood-brain barrier (BBB). We propose to develop a liposomal co-delivery system that could effectively transport riluzole to brain cells by reducing efflux pumps with a P-gp inhibitor, verapamil. Riluzole and verapamil cocktail liposomes were prepared by lipid film hydration. The average particle size of cocktail liposomes was 194.3 ±â€¯6.0 nm and their polydispersity index (PDI) was 0.272 ±â€¯0.017. The encapsulation efficiencies of verapamil and riluzole in the cocktail liposomes were 86.0 ±â€¯1.4% and 85.6 ±â€¯1.1%, respectively. The drug release from cocktail liposomes after 8 h in PBS at 37 °C was 78.4 ±â€¯6.2% of riluzole and 76.7 ±â€¯3.8% of verapamil. The average particle size of liposomes did not show significant changes at 4 °C after three months. Verapamil cocktail liposomes inhibited P-gp levels measured by western blotting in dose and time-dependent manners in brain endothelial bEND.3 cells. Increased drug efflux transporters were detected in bEND.3 and astrocytes C8D1A cells, promoted by tumor necrosis factor (TNF-α) or hydrogen peroxide (H2O2). Restored accumulations of riluzole and fluorescent dye rhodamine 123 were observed in bEND.3 cells after treatments with cocktail liposomes. It indicated that inhibitory potential of co-delivery liposome system towards P-gp could mediate the transport of both P-gp substrates. Verapamil and riluzole co-loaded liposomes may be used to overcome pharmacoresistance of riluzole for improving ALS therapy.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Astrocitos/efectos de los fármacos , Encéfalo/efectos de los fármacos , Resistencia a Medicamentos/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Riluzol/farmacología , Verapamilo/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Liberación de Fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Cinética , Liposomas , Ratones , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/metabolismo , Tamaño de la Partícula , Riluzol/administración & dosificación , Riluzol/metabolismo , Solubilidad , Verapamilo/administración & dosificación
7.
Pigment Cell Melanoma Res ; 31(4): 534-540, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29453787

RESUMEN

Studies demonstrate that GRM, expressed by >60% of human melanomas, may be a therapeutic target. We performed a phase II trial of 100 mg PO bid of riluzole, an inhibitor of GRM1 signaling, in patients with advanced melanoma with the primary endpoint of response rate. Thirteen patients with GRM1-positive tumors were enrolled. No objective responses were observed, and accrual was stopped. Stable disease was noted in six (46%) patients, with one patient on study for 42 weeks. Riluzole was well tolerated, with fatigue (62%) as the most common adverse event. Downregulation of MAPK and PI3K/AKT was noted in 33% of paired tumor biopsies. Hypothesis-generating correlative studies suggested that downregulation of angiogenic markers and increased leukocytes at the active edge of tumor correlate with clinical benefit. Pharmacokinetic analysis showed interpatient variability consistent with prior riluzole studies. Future investigations should interrogate mechanisms of biologic activity and advance the development of agents with improved bioavailability.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Riluzol/administración & dosificación , Adulto , Femenino , Humanos , Masculino , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Melanoma/patología , Persona de Mediana Edad , Receptores de Glutamato Metabotrópico/biosíntesis , Riluzol/efectos adversos
8.
Ulus Travma Acil Cerrahi Derg ; 23(6): 452-458, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29115658

RESUMEN

BACKGROUND: We investigated the effects of an N-methyl-D-aspartate receptor antagonist, riluzole, and a pancaspase inhibitor and basic apoptosis mediator, Q-VD-OPh, in combination or alone in posttraumatic spinal cord injury. METHODS: In our study, 45 healthy male Sprague Dawley rats were used. Spinal trauma was induced by the clip compression technique via thoracal 7, 8, 9 laminectomies. After inducing the trauma, the drug was continuously administered intraperitoneally for 5 days. After inducing the trauma, the subjects were assessed using Tarlov's motor grading scale and inclined plane test. Five days after the trauma, the spinal cord specimens were harvested, and a histopathological examination was performed. RESULTS: Compared with the other groups, a statistically significant difference with regard to better results for necrosis, inflammation, and apoptosis was observed in the riluzole only and combination groups. Statistically better motor function scores were observed in the Q-VD-OPh only group than in the other groups. CONCLUSION: With regard to limiting secondary damage after trauma, statistically significant results were observed in the Q-VDOPh only and Q-VD-OPh-riluzole combination groups. More extensive laboratory studies are required to limit and control the effects of secondary damage after spinal cord trauma.


Asunto(s)
Clorometilcetonas de Aminoácidos , Fármacos Neuroprotectores , Quinolinas , Riluzol , Traumatismos de la Médula Espinal , Clorometilcetonas de Aminoácidos/administración & dosificación , Clorometilcetonas de Aminoácidos/farmacología , Clorometilcetonas de Aminoácidos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Necrosis/tratamiento farmacológico , Necrosis/prevención & control , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Quinolinas/administración & dosificación , Quinolinas/farmacología , Quinolinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Riluzol/administración & dosificación , Riluzol/farmacología , Riluzol/uso terapéutico , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/fisiopatología
9.
Breast Cancer Res Treat ; 166(2): 407-419, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28780701

RESUMEN

PURPOSE: One in eight women will develop breast cancer, 15-20% of whom will have triple-negative breast cancer (TNBC), an aggressive breast cancer with no current targeted therapy. We have demonstrated that riluzole, an FDA-approved drug for treating amyotrophic lateral sclerosis, inhibits growth of TNBC. In this study, we explore potential synergism between riluzole and paclitaxel, a chemotherapeutic agent commonly used to treat TNBC, in regulating TNBC proliferation, cell cycle arrest, and apoptosis. METHODS: TNBC cells were treated with paclitaxel and/or riluzole and synergistic effects on cell proliferation were quantified via MTT assay and CompuSyn analysis. Apoptosis was observed morphologically and by measuring cleaved PARP/caspase three products. Microarray analysis was performed using MDA-MB-231 cells to examine cell cycle genes regulated by riluzole and any enhanced effects on paclitaxel-mediated cell cycle arrest, determined by FACS analysis. These results were confirmed in vivo using a MDA-MB-231 xenograft model. RESULTS: Strong enhanced or synergistic effects of riluzole on paclitaxel regulation of cell cycle progression and apoptosis was demonstrated in all TNBC cells tested as well as in the xenograft model. The MDA-MB-231, SUM149, and SUM229 cells, which are resistant to paclitaxel treatment, demonstrated the strongest synergistic or enhanced effect. Key protein kinases were shown to be upregulated in this study by riluzole as well as downstream cell cycle genes regulated by these kinases. CONCLUSIONS: All TNBC cells tested responded synergistically to riluzole and paclitaxel strongly suggesting the usefulness of this combinatorial treatment strategy in TNBC, especially for patients whose tumors are relatively resistant to paclitaxel.


Asunto(s)
Antineoplásicos/administración & dosificación , Proteínas de Ciclo Celular/genética , Paclitaxel/administración & dosificación , Riluzol/administración & dosificación , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Paclitaxel/farmacología , Riluzol/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Sci Rep ; 7(1): 9661, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28851920

RESUMEN

Oxaliplatin, a chemotherapy medication, causes severe peripheral neuropathy. Although oxaliplatin-induced peripheral neuropathy is a dose-limiting toxicity, a therapeutic strategy against its effects has not been established. We previously reported the involvement of N-methyl-D-aspartate receptors and their intracellular signalling pathway in oxaliplatin-induced mechanical allodynia in rats. The aim of this study was to clarify the involvement of spinal glutamate transmission in oxaliplatin-induced mechanical allodynia. In vivo spinal microdialysis revealed that the baseline glutamate concentration was elevated in oxaliplatin-treated rats, and that mechanical stimulation of the hind paw markedly increased extracellular glutamate concentration in the same rats. In these rats, the expression of glutamate transporter 1 (GLT-1), which plays a major role in glutamate uptake, was decreased in the spinal cord. Moreover, we explored the potential of pharmacological therapy targeting maintenance of extracellular glutamate homeostasis. The administration of riluzole, an approved drug for amyotrophic lateral sclerosis, suppressed the increase of glutamate concentration, the decrease of GLT-1 expression and the development of mechanical allodynia. These results suggest that oxaliplatin disrupts the extracellular glutamate homeostasis in the spinal cord, which may result in neuropathic symptoms, and support the use of riluzole for prophylaxis of oxaliplatin-induced mechanical allodynia.


Asunto(s)
Antineoplásicos/efectos adversos , Quimioprevención/métodos , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Glutamatos/metabolismo , Hiperalgesia/inducido químicamente , Oxaliplatino/efectos adversos , Riluzol/administración & dosificación , Animales , Transporte Biológico , Modelos Animales de Enfermedad , Hiperalgesia/fisiopatología , Hiperalgesia/prevención & control , Ratas , Resultado del Tratamiento
11.
Eur J Trauma Emerg Surg ; 43(5): 691-699, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27277212

RESUMEN

PURPOSE: Peripheral nerve injury (PNI) is common disorder that represents more than 3 % of all traumatic injury cases. One type of PNI, sciatic nerve injury, leads to considerable motoneuron dysfunction. Because Riluzole is clinically approved for the treatment of motoneuron disease, we evaluated whether Riluzole treatment could enhance the nerve regeneration process and improve functional outcome after sciatic nerve crush in rats. METHODS: In acute treatment groups, a single dose of Riluzole (6 and 8 mg/kg) was administered intra-peritoneally 15 min after the crush nerve injury. In the chronic treatment groups, animals were treated with Riluzole (4 and 6 mg/kg/d) for 8 days. Sciatic functional index (SFI) was evaluated for 9 weeks after injury. Furthermore, electrophysiological and morphometric evaluations were performed at the 9th week following injury. RESULTS: Acute and chronic administrations of Riluzole immediately after sciatic nerve crush result in significantly delayed regeneration and reduced motor function outcome. CONCLUSIONS: These findings suggest that early administration of even a single dose of Riluzole after sciatic nerve crush injury can delay motor function recovery. This effect may not depend on its anti-nociceptive activity.


Asunto(s)
Lesiones por Aplastamiento/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Riluzol/uso terapéutico , Nervio Ciático/efectos de los fármacos , Neuropatía Ciática/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Inyecciones Intraperitoneales , Locomoción , Masculino , Regeneración Nerviosa/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Wistar , Recuperación de la Función , Riluzol/administración & dosificación , Riluzol/farmacología , Resultado del Tratamiento
12.
Nano Lett ; 16(12): 7357-7363, 2016 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-27960523

RESUMEN

Although a range of nanoparticles have been developed as drug delivery systems in cancer therapeutics, this approach faces several important challenges concerning nanocarrier circulation, clearance, and penetration. The impact of reducing nanoparticle size on penetration through leaky blood vessels around tumor microenvironments via enhanced permeability and retention (EPR) effect has been extensively examined. Recent research has also investigated the effect of nanoparticle shape on circulation and target binding affinity. However, how nanoparticle shape affects drug release and therapeutic efficacy has not been previously explored. Here, we compared the drug release and efficacy of iron oxide nanoparticles possessing either a cage shape (IO-NCage) or a solid spherical shape (IO-NSP). Riluzole cytotoxicity against metastatic cancer cells was enhanced 3-fold with IO-NCage. The shape of nanoparticles (or nanocages) affected the drug release point and cellular internalization, which in turn influenced drug efficacy. Our study provides evidence that the shape of iron oxide nanoparticles has a significant impact on drug release and efficacy.


Asunto(s)
Dextranos , Portadores de Fármacos , Compuestos Férricos , Nanopartículas , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Humanos , Riluzol/administración & dosificación
13.
Breast Cancer Res Treat ; 151(1): 57-73, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25859923

RESUMEN

Metabotropic glutamate receptor 1 (mGluR1/Grm1) is a member of the G-protein-coupled receptor superfamily, which was once thought to only participate in synaptic transmission and neuronal excitability, but has more recently been implicated in non-neuronal tissue functions. We previously described the oncogenic properties of Grm1 in cultured melanocytes in vitro and in spontaneous melanoma development with 100 % penetrance in vivo. Aberrant mGluR1 expression was detected in 60-80 % of human melanoma cell lines and biopsy samples. As most human cancers are of epithelial origin, we utilized immortalized mouse mammary epithelial cells (iMMECs) as a model system to study the transformative properties of Grm1. We introduced Grm1 into iMMECs and isolated several stable mGluR1-expressing clones. Phenotypic alterations in mammary acinar architecture were assessed using three-dimensional morphogenesis assays. We found that mGluR1-expressing iMMECs exhibited delayed lumen formation in association with decreased central acinar cell death, disrupted cell polarity, and a dramatic increase in the activation of the mitogen-activated protein kinase pathway. Orthotopic implantation of mGluR1-expressing iMMEC clones into mammary fat pads of immunodeficient nude mice resulted in mammary tumor formation in vivo. Persistent mGluR1 expression was required for the maintenance of the tumorigenic phenotypes in vitro and in vivo, as demonstrated by an inducible Grm1-silencing RNA system. Furthermore, mGluR1 was found be expressed in human breast cancer cell lines and breast tumor biopsies. Elevated levels of extracellular glutamate were observed in mGluR1-expressing breast cancer cell lines and concurrent treatment of MCF7 xenografts with glutamate release inhibitor, riluzole, and an AKT inhibitor led to suppression of tumor progression. Our results are likely relevant to human breast cancer, highlighting a putative role of mGluR1 in the pathophysiology of breast cancer and the potential of mGluR1 as a novel therapeutic target.


Asunto(s)
Neoplasias de la Mama/genética , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Receptores de Glutamato Metabotrópico/genética , Animales , Apoptosis/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Humanos , Células MCF-7 , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/patología , Ratones , Receptores de Glutamato Metabotrópico/biosíntesis , Riluzol/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell Physiol Biochem ; 35(2): 419-32, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25613036

RESUMEN

BACKGROUND: Metabotropic glutamate receptors (mGluRs) are G-protein-coupled receptors that mediate neuronal excitability and synaptic plasticity in the central nervous system, and emerging evidence suggests a role of mGluRs in the biology of cancer. Previous studies showed that mGluR1 was a potential therapeutic target for the treatment of breast cancer and melanoma, but its role in human glioma has not been determined. METHODS: In the present study, we investigated the effects of mGluR1 inhibition in human glioma U87 cells using specific targeted small interfering RNA (siRNA) or selective antagonists Riluzole and BAY36-7620. The anti-cancer effects of mGluR1 inhibition were measured by cell viability, lactate dehydrogenase (LDH) release, TUNEL staining, cell cycle assay, cell invasion and migration assays in vitro, and also examined in a U87 xenograft model in vivo. RESULTS: Inhibition of mGluR1 significantly decreased the cell viability but increased the LDH release in a dose-dependent fashion in U87 cells. These effects were accompanied with the induction of caspase-dependent apoptosis and G0/G1 cell cycle arrest. In addition, the results of Matrigel invasion and cell tracking assays showed that inhibition of mGluR1 apparently attenuated cell invasion and migration in U87 cells. All these anti-cancer effects were ablated by the mGluR1 agonist L-quisqualic acid. The results of western blot analysis showed that mGluR1 inhibition overtly decreased the phosphorylation of PI3K, Akt, mTOR and P70S6K, indicating the mitigated activation of PI3K/Akt/mTOR pathway. Moreover, the anti-tumor activity of mGluR1 inhibition in vivo was also demonstrated in a U87 xenograft glioma model in athymic nude mice. CONCLUSION: The remarkable efficiency of mGluR1 inhibition to induce cell death in U87 cells may find therapeutic application for the treatment of glioma patients.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Glioma/metabolismo , Humanos , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Naftalenos/administración & dosificación , Naftalenos/farmacología , Ácido Quiscuálico/farmacología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/farmacología , Receptores de Glutamato Metabotrópico/metabolismo , Riluzol/administración & dosificación , Riluzol/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Neurosurg ; 120(4): 854-63, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24484234

RESUMEN

OBJECT: The poor outcome of malignant gliomas is largely due to local invasiveness. Previous studies suggest that gliomas secrete excess glutamate and destroy surrounding normal peritumoral brain by means of excitotoxic mechanisms. In this study the authors assessed the effect on survival of 2 glutamate modulators (riluzole and memantine) in rodent glioma models. METHODS: In an in vitro growth inhibition assay, F98 and 9L cells were exposed to riluzole and memantine. Mouse cerebellar organotypic cultures were implanted with F98 glioma cells and treated with radiation, radiation + riluzole, or vehicle and assessed for tumor growth. Safety and tolerability of intracranially implanted riluzole and memantine CPP:SA polymers were tested in F344 rats. The efficacy of these drugs was tested against the 9L model and riluzole was further tested with and without radiation therapy (RT). RESULTS: In vitro assays showed effective growth inhibition of both drugs on F98 and 9L cell lines. F98 organotypic cultures showed reduced growth of tumors treated with radiation and riluzole in comparison with untreated cultures or cultures treated with radiation or riluzole alone. Three separate efficacy experiments all showed that localized delivery of riluzole or memantine is efficacious against the 9L gliosarcoma tumor in vivo. Systemic riluzole monotherapy was ineffective; however, riluzole given with RT resulted in improved survival. CONCLUSIONS: Riluzole and memantine can be safely and effectively delivered intracranially via polymer in rat glioma models. Both drugs demonstrate efficacy against the 9L gliosarcoma and F98 glioma in vitro and in vivo. Although systemic riluzole proved ineffective in increasing survival, riluzole acted synergistically with radiation and increased survival compared with RT or riluzole alone.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Glioma/tratamiento farmacológico , Memantina/uso terapéutico , Riluzol/uso terapéutico , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Memantina/administración & dosificación , Ratones , Polímeros/administración & dosificación , Polímeros/uso terapéutico , Ratas , Riluzol/administración & dosificación
17.
Clin Exp Pharmacol Physiol ; 40(12): 856-63, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24256163

RESUMEN

The cardiac persistent sodium current (IN aP ) presents a novel target for cardiac ischaemic protection. Herein we investigated the effects of the IN aP blocker riluzole in a pig model of regional myocardial ischaemia. Landrace or Large White pigs were subjected to 3 h ligation of the left anterior descending coronary artery (LAD). Pigs received either saline (500 mL/h, i.v.) throughout the experiment (control; n = 7) or riluzole (2 mg/kg in 2 mL propylene glycol in 100 mL saline, i.v.; RIL; n = 7) between 15 and 5 min prior to ligation. The arrhythmia score was calculated in 5 min epochs. Myocardial damage was assessed using epicardial image analysis and histological sectioning. In the control group, all seven pigs developed premature ventricular contractions (PVC), seven developed non-sustained arrhythmias and six of seven developed sustained arrhythmias. Of the sustained arrhythmias, 23 of 28 instances were initiated by R-on-T extrasytoles (extrasystoles within the vulnerable period that can trigger re-entrant arrhythmias). In the RIL group, all seven pigs developed PVC, six of seven developed non-sustained arrhythmias and only three developed sustained arrhythmias, of which two of five instances were R-on-T initiated. The riluzole-treated pigs exhibited less myocardial damage than pigs in the control group (65% smaller surface area (P = 0.008) on gross epicardial inspection, 51% less oedema (P = 0.01), 53% less fibre waviness (P = 0.029) assessed by haematoxylin and eosin staining and 79% fewer fragmented nuclei (P = 0.009) assessed by terminal deoxyribonucleotidyl transferase-mediated dUTP-digoxigenin nick end-labelling). In conclusion, riluzole significantly reduced Phase 2 (the period associated with irreversible damage) ischaemic R-on-T triggered and non-R-on-T arrhythmias and myocardial damage occurring during the 3 h period of regional ischaemia.


Asunto(s)
Antiarrítmicos/uso terapéutico , Arritmias Cardíacas/prevención & control , Oclusión Coronaria/tratamiento farmacológico , Isquemia Miocárdica/tratamiento farmacológico , Miocardio/patología , Riluzol/uso terapéutico , Bloqueadores de los Canales de Sodio/uso terapéutico , Animales , Antiarrítmicos/administración & dosificación , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patología , Oclusión Coronaria/complicaciones , Oclusión Coronaria/metabolismo , Oclusión Coronaria/patología , Modelos Animales de Enfermedad , Electrocardiografía , Femenino , Masculino , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Miocardio/metabolismo , Riluzol/administración & dosificación , Bloqueadores de los Canales de Sodio/administración & dosificación , Porcinos
18.
Adv Healthc Mater ; 2(9): 1236-45, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23495216

RESUMEN

Current cancer therapies are challenged by weakly soluble drugs and by drug combinations that exhibit non-uniform biodistribution and poor bioavailability. In this study, we have presented a new platform of advanced healthcare materials based on albumin nanoparticles (ANPs) engineered as tumor penetrating, delivery vehicles of combinatorially applied factors to solid tumors. These materials were designed to overcome three sequential key barriers: tissue level transport across solid tumor matrix; uptake kinetics into individual cancer cells; therapeutic resistance to single chemotherapeutic drugs. The ANPs were designed to penetrate deeper into solid tumor matrices using collagenase decoration and evaluated using a three-dimensional multicellular melanoma tumor spheroid model. Collagenase modified ANPs exhibited 1-2 orders of magnitude greater tumor penetration than unmodified ANPs into the spheroid mass after 96 hours, and showed preferential uptake into individual cancer cells for smaller sized ANPs (<100 nm). For enhanced efficacy, collagenase coated ANPs were modified with two therapeutic agents, curcumin and riluzole, with complementary mechanisms of action for combined cell cycle arrest and apoptosis in melanoma. The collagenase coated, drug loaded nanoparticles induced significantly more cell death within 3-D tumor models than the unmodified, dual drug loaded ANP particles and the kinetics of cytotoxicity was further influenced by the ANP size. Thus, multifunctional nanoparticles can be imbued with complementary size and protease activity features that allow them to penetrate solid tumors and deliver combinatorial therapeutic payload with enhanced cancer cytotoxicity but minimal collateral damage to healthy primary cells.


Asunto(s)
Portadores de Fármacos/química , Nanopartículas/química , Albúmina Sérica/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Colagenasas/metabolismo , Curcumina/administración & dosificación , Curcumina/química , Curcumina/toxicidad , Humanos , Nanopartículas/ultraestructura , Neoplasias/tratamiento farmacológico , Tamaño de la Partícula , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Riluzol/administración & dosificación , Riluzol/química , Riluzol/toxicidad , Albúmina Sérica/genética , Albúmina Sérica/metabolismo
19.
Praxis (Bern 1994) ; 101(14): 907-14, 2012 Jul 04.
Artículo en Alemán | MEDLINE | ID: mdl-22763933

RESUMEN

Amyotrophic lateral sclerosis (ALS) represents the most common motoneuron disorder in adulthood. It is characterized by selective degeneration of the motoneurons. About 10% of patients have a genetically determined ALS. Clinically, ALS is characterized by coexistence of signs of the first motoneuron, such as spasticity and hyperreflexia, as well as the second motoneuron, such as muscular atrophy and fasciculations. If such signs are present in at least three regions and if other possible causes have been excluded, a definite diagnosis of ALS can be made based on the revised El-Escorial criteria. Initial manifestations are often focalized and generalization develops during the course. The glutamate antagonist riluzole is worldwide the only approved ALS treatment. However, symptomatic treatments to ameliorate spasticity, drooling, speech and swallowing problems, and assisted ventilation to treat respiratory failure are essential.


Asunto(s)
Esclerosis Amiotrófica Lateral/diagnóstico , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Actividades Cotidianas/clasificación , Anciano , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/mortalidad , Animales , Causas de Muerte , Estudios Transversales , Análisis Mutacional de ADN , Diagnóstico Diferencial , Modelos Animales de Enfermedad , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Antagonistas de Aminoácidos Excitadores/efectos adversos , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Persona de Mediana Edad , Neuronas Motoras/patología , Examen Neurológico , Cuidados Paliativos , Riluzol/administración & dosificación , Riluzol/efectos adversos , Médula Espinal/patología , Análisis de Supervivencia , Proteinopatías TDP-43/patología
20.
Breast Cancer Res Treat ; 132(2): 565-73, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21681448

RESUMEN

Metabotropic glutamate receptors are G-protein-coupled receptors normally expressed in the central nervous system where they mediate neuronal excitability, synaptic plasticity, and feedback inhibition of neurotransmitter release. However, recent data suggest that these receptors are also expressed and functional in some cancers, most notably melanoma. We detected the expression of metabotropic glutamate receptor-1 (gene: GRM1; protein: mGluR1) in triple negative breast cancer cells and evaluated its role in regulating the pro-proliferative phenotype of these cells. mGluR1 inhibitors (Riluzole or BAY36-7620) inhibited the proliferation of triple negative breast cancer cells in a time- and dose-dependent manner and this inhibition correlated with increased apoptosis as demonstrated by increase in PARP cleavage products and Annexin V staining. mGluR1 knockdown using Lentiviral constructs expressing shRNA targeting GRM1 also inhibited proliferation compared to non-silencing controls. In addition, treatment of mice bearing MDA-MB-231 xenografts with Riluzole or BAY36-7620, by intraperitoneal injection, resulted in a significant reduction in tumor volume of up to 80%. Moreover, Riluzole was effective against triple negative breast cancer xenografts in mice at doses equivalent to those currently being used in humans for the treatment of amyotrophic lateral sclerosis. Our observations implicate mGluR1 and glutamate signaling as a promising new molecular target for the treatment of breast cancer. Even more promising, Riluzole, because it is an oral drug that can be administered with low toxicity, represents a promising approach in the treatment of triple negative breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Antagonistas de Aminoácidos Excitadores/farmacología , Naftalenos/farmacología , Receptores de Glutamato Metabotrópico/efectos de los fármacos , Riluzol/farmacología , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Femenino , Humanos , Inyecciones Intraperitoneales , Ratones , Ratones Desnudos , Naftalenos/administración & dosificación , Fenotipo , Ácido Quiscuálico/farmacología , Interferencia de ARN , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Riluzol/administración & dosificación , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA