Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cancer Immunol Immunother ; 71(7): 1611-1623, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34731283

RESUMEN

We tested the concept that host preexisting influenza A virus immunity can be redirected to inhibit tumor growth and metastasis through systemic administration of influenza A virus-related peptides to targeted tumors. Mice infected with influenza A virus strain A/Puerto Rico/8/34 (PR8) were used as a model of a host with preexisting viral immunity. The extent to which preexisting influenza A immunity in PR8-immunized mice can be redirected to inhibit tumor growth and metastasis was first examined by ectopic expression of influenza A nucleoprotein (NP) and hemagglutinin (HA) in syngeneic mammary tumor cells via lentiviral transduction. Then, the feasibility of implementing this strategy using a systemic therapy approach was assessed by systemic delivery of major histocompatibility complex class I (MHC-I)-compatible peptides to targeted mammary tumors overexpressing human epidermal growth factor receptor-2 (HER2) in mice using a novel HER2-targeting single-lipid nanoparticle (SLNP). Our results show that preexisting influenza A immunity in PR8-immunized mice could be quickly redirected to syngeneic tumors expressing influenza A NP and HA, leading to strong inhibition of tumor growth and metastasis and improvement of survival compared to the findings in antigen-naïve control mice. MHC-I-compatible peptides could be delivered to targeted mammary tumors in mice using the HER2-targeting SLNP for antigen presentation, which subsequently redirected preexisting influenza A immunity to the tumors to exert antitumor activities. In conclusion, preexisting influenza A immunity can be repurposed for cancer immunotherapy through systemic delivery of influenza A-related peptides to targeted tumors. Further development of the strategy for clinical translation is warranted.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Neoplasias , Infecciones por Orthomyxoviridae , Animales , Anticuerpos Antivirales , Humanos , Inmunoterapia , Gripe Humana/prevención & control , Liposomas , Ratones , Ratones Endogámicos BALB C , Nanopartículas , Neoplasias/terapia , Infecciones por Orthomyxoviridae/prevención & control , Péptidos
2.
J Obstet Gynaecol ; 42(6): 2139-2143, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35166183

RESUMEN

Uterine leiomyomas are complex tumours with limited medical treatment options. Simvastatin is used to treat hypercholesterolaemia and has shown promising effects as a treatment option for leiomyomas. Previously, our group demonstrated a promising effect of simvastatin treatment in a patient-derived xenograft mouse model. Here, we tested the efficacy of simvastatin liposomal nanoparticles (NPs). After bilateral leiomyoma xenograft implantation, mice (N = 12) were divided into three treatment arms: control, simvastatin and simvastatin-loaded liposome NPs (simvastatin-NPs). Treatment with simvastatin significantly reduced tumour volume and inhibited the Ki67 expression when compared to the control group. There was a trend of reduced tumour volume and Ki67 expression after treatment with simvastatin-NP; however, the results were not significant. Due to low bioavailability and short half-life of simvastatin, liposomal NPs have the potential to enhance drug delivery, however, in this study NP did not provide improvement over simvastatin, but did demonstrate their potential for the delivery of simvastatin.Impact statementWhat is already known on this subject? Simvastatin treatment in a patient-derived xenograft mouse model reduced tumour growth and decreased proliferation.What do the results of this study add? Treatment with simvastatin significantly reduced tumour volume and inhibited the Ki67 expression when compared to the control group. There was a trend of reduced tumour volume and Ki67 expression after treatment with simvastatin-NP, however, it did not improve the efficacy of simvastatin at reducing tumour growth and proliferation.What are the implications of these findings for clinical practice and/or further research? More studies are needed to optimise the formulation of NPs to further enhance the sustainable delivery of simvastatin.


Asunto(s)
Leiomioma , Nanopartículas , Animales , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Antígeno Ki-67 , Leiomioma/tratamiento farmacológico , Leiomioma/patología , Liposomas , Ratones , Proyectos Piloto , Simvastatina/farmacología
3.
Prostate ; 80(1): 65-73, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31614005

RESUMEN

BACKGROUND: The TMPRSS2/ERG (TE) fusion gene is present in half of the prostate cancers (PCas). The TMPRSS2 and ERG junction of the fusion messenger RNA (mRNA) constitutes a cancer-specific target. Although docetaxel-based chemotherapy is the second line of therapy following development resistance to androgen ablation therapies, it is not curative. Therefore, the development of nontoxic novel monotherapies for targeting TE mRNA in PCa patients and for increasing the clinical efficacy of docetaxel treatment are needed. METHODS: We evaluated multiple approaches to enhance the delivery of TE small interfering RNA (siRNA) containing liposomes including PEGylation, topical treatment with nitroglycerin (NG) to increase permeability and retention, and three different PEG modifications: folate, RGD cyclic peptide, and a bFGF fibroblast growth factor receptor-targeting peptide. The efficacy of the optimized TE siRNA liposome in combination with docetaxel was then evaluated in vivo with or without topical NG in vivo using a VCaP xenograft model. TE fusion protein knockdown in residual tumors was assessed using Western blotting and immunohistochemistry. RESULTS: In vivo therapeutic targeting of TE fusion gene by systemic delivery of RGD-peptide-coated liposomal siRNA nanovectors led to sustained target silencing, suppressed tumor growth in xenograft models and enhanced the efficacy of docetaxel chemotherapy. Simultaneous application of the vasodilator NG to the skin further increased tissue the delivery of siRNA and enhanced target knockdown. CONCLUSION: TE-targeted gene silencing therapy using liposomal nanovectors is a potential therapeutic strategy as a monotherapy and to enhance the efficacy of chemotherapy in patients with advanced PCa.


Asunto(s)
Docetaxel/farmacología , Proteínas de Fusión Oncogénica/genética , Neoplasias de la Próstata Resistentes a la Castración/terapia , ARN Mensajero/genética , ARN Interferente Pequeño/administración & dosificación , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Terapia Combinada , Técnicas de Silenciamiento del Gen/métodos , Humanos , Liposomas/administración & dosificación , Masculino , Ratones , Terapia Molecular Dirigida , Nanopartículas/administración & dosificación , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , ARN Interferente Pequeño/genética , Distribución Aleatoria , Regulador Transcripcional ERG/deficiencia , Regulador Transcripcional ERG/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Pharm Res ; 37(3): 63, 2020 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-32133571

RESUMEN

PURPOSE: To evalauted natural polymeric biomaterials including hyaluronic acid (HA) and its copolymeric form HA:Suc nanoparticles (NPs) as drug carrier systems for delivery of hydrophobic small molecule kinase EF2-kinase inhibitor in breast and pancreatic cancer cells. METHODS: In vitro cellular uptake studies of Rhodamine 6G labaled HA:Suc nanoparticles were evaluated by using flow cytometry analysis and fluorescent microscopy in breast (MDA-MB-231 and MDA-MB-436) and pancreatic cancer cells (PANC-1 and MiaPaca-2). Besides, in vitro release study of compound A (an EF2-kinase inhibitor) as a model hydrophobic drug was performed in the cancer cells. RESULTS: These biological evaluation studies indicated that HA and HA:Suc NPs provided a highly effective delivery of compound A were into breast and pancreatic cancer cells, leading to significant inhibition of cell proliferation and colony formation of breast and pancreatic cancer cells. CONCLUSION: HA-sucrose NPs incorporating an EF2-Kinase inhibitor demonstrate significant biologic activity in breast and pancreatic cancer cells. This is the first study that shows natural polymeric drug carriers succesfully deliver a hydrofobic cancer drug into cancer cells. Graphical Abstract Nanoparticles based on HA:Suc are effective in delivering hydrofobic cancer drugs in breast and pancreatic cancers.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Quinasa del Factor 2 de Elongación/antagonistas & inhibidores , Ácido Hialurónico/química , Nanogeles/química , Neoplasias Pancreáticas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Quinasa del Factor 2 de Elongación/metabolismo , Femenino , Humanos , Neoplasias Pancreáticas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología
5.
Reprod Sci ; 28(1): 271-277, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32632769

RESUMEN

Uterine leiomyomas represent a challenging problem with limited medical treatment options. The anti-tumor agent 2-methoxyestradiol (2-ME) shows promising results but its efficacy is limited by inadequate pharmacokinetics. We previously demonstrated that 2-ME nanoparticles can be successfully formulated and that they show improved in vitro anti-leiomyoma cell activity. Here, we examined the effects of the in vivo delivery of 2-ME nanoparticles in a patient-derived xenograft (PDX) leiomyoma mouse model. Patient-derived leiomyoma tumor tissues were xenografted subcutaneously in estrogen/progesterone pretreated immunodeficient NOG mice. Animals (n = 12) were treated with liposomal 2-ME nanoparticles by intra-peritoneal (IP) injection (50 mg/kg/dose, three times weekly) or control for 28 days. Tumor volume was measured weekly by calipers and prior to sacrifice by ultrasound. In addition, the expression of the cell proliferation marker Ki67 and the apoptosis marker cleaved caspase-3 in tumor tissues after treatment were measured by immunohistochemistry. Liposomal 2-ME treatment was associated with a significant tumor growth inhibition (30.5% less than controls as early as 2 weeks, p = 0.025). In addition, injections of liposomal 2-ME inhibited the expression of the proliferation marker Ki67 (55.8% reduction, p < 0.001). Furthermore, liposomal 2-ME treatment was associated with a 67.5% increase of cleaved caspase-3 expression of increase (p = 0.048). Our findings suggest that liposomal nanoparticle formulation can successfully deliver 2-ME and can be a promising therapeutic strategy for uterine leiomyoma. Further characterization of the liposomal-2ME, including pharmacokinetics, maximal tolerated dose, and safety, is needed in preclinical models prior to clinical trials.


Asunto(s)
2-Metoxiestradiol/farmacología , Antineoplásicos/farmacología , Leiomioma/tratamiento farmacológico , Neoplasias Uterinas/tratamiento farmacológico , 2-Metoxiestradiol/química , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Proliferación Celular/efectos de los fármacos , Composición de Medicamentos , Femenino , Humanos , Antígeno Ki-67/metabolismo , Leiomioma/metabolismo , Leiomioma/patología , Liposomas , Ratones Endogámicos NOD , Ratones SCID , Nanopartículas , Carga Tumoral/efectos de los fármacos , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Int J Biol Macromol ; 126: 1150-1157, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30625351

RESUMEN

Porous and biodegradable hyaluronic acid (HA) nanogel and their copolymeric forms with sucrose (Suc), HA:Sucrose (HA:Suc) nanogels, were synthesized by employing glycerol diglycidyl ether (GDE) as crosslinker with a single step reaction in surfactant-free medium. The size of the nanogels was determined as 150 ±â€¯50 nm in dried state from SEM images and found to increase to about 540 ±â€¯47 nm in DI water measured with DLS measurements. The surface areas of HA and HA:Suc nanogels were measured as 18.07 ±â€¯2.4 and 32.30 ±â€¯6.1 m2/g with porosities of 3.58 ±â€¯1.8, and 9.44 ±â€¯3.1 nm via BET analysis, respectively. The zeta potentials for HA and HA:Suc nanogels were measured as -33 ±â€¯1.4 and - 30 ±â€¯1.2 mV, respectively. The thermal degradation of both types of nanogels revealed similar trends, while hydrolytic degradation of the nanogels was about 22.7 ±â€¯0.2 wt% in 15 days. Both HA and HA:Suc nanogels were stable in blood up to 250 µg/mL concentration with approximately 0.5 ±â€¯0.1% hemolysis ratio and 76 ±â€¯12% blood clotting indices, respectively. Finally, these nanogels were used as a sustained slow-release or long-term delivery system over 2 days for a hydrophobic cancer drug, 3­((E)­3­(4­hydroxyphenyl)acryloyl)­2H­chromen­2­on (A#) established by our group. The nanogels successfully delivered the model drug A at 10.43 ±â€¯2.12 mg/g for 2 days.


Asunto(s)
Antineoplásicos/farmacología , Sistemas de Liberación de Medicamentos , Ácido Hialurónico/química , Interacciones Hidrofóbicas e Hidrofílicas , Polietilenglicoles/química , Polietileneimina/química , Sacarosa/química , Espectroscopía de Resonancia Magnética con Carbono-13 , Hemólisis , Humanos , Nanogeles , Porosidad , Espectroscopía Infrarroja por Transformada de Fourier , Electricidad Estática , Temperatura
7.
Nanomedicine (Lond) ; 12(16): 1961-1973, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28745127

RESUMEN

AIM: In this study, we aimed to therapeutically target eukaryotic elongation factor 2 kinase (eEF-2K) in an in vivo triple-negative breast cancer (TNBC) tumor model. MATERIALS & METHODS: We synthesized a highly monodisperse nanoformulation using polyethylenimine-modified gold nanoparticles (AuNP-PEI) as siRNA delivery vehicle and evaluated gene downregulation. RESULTS: We found that AuNP-PEI/eEF-2K nanoformulation was highly effective for in vitro and in vivo gene downregulation and showed remarkable antitumor efficacy that was associated with eEF-2K knockdown, inhibition of Src and MAPK-ERK signaling pathways in a TNBC orthotopic tumor model. CONCLUSION: Our study suggests that eEF-2K plays an important role in TNBC tumorigenesis and its inhibition by AuNP-PEI/eEF-2K siRNA-based nanotherapeutics may be a potential therapeutic strategy for TNBC.


Asunto(s)
Oro/química , Nanopartículas del Metal/química , ARN Interferente Pequeño/administración & dosificación , Neoplasias de la Mama Triple Negativas/terapia , Animales , Línea Celular Tumoral , Regulación hacia Abajo , Liberación de Fármacos , Quinasa del Factor 2 de Elongación/genética , Quinasa del Factor 2 de Elongación/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Expresión Génica , Técnicas de Silenciamiento del Gen , Técnicas de Transferencia de Gen , Humanos , Nanopartículas del Metal/toxicidad , Ratones , Terapia Molecular Dirigida , Nanomedicina , Tamaño de la Partícula , Polietileneimina/química , Transducción de Señal , Propiedades de Superficie
8.
Oncotarget ; 8(7): 11641-11658, 2017 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-28036267

RESUMEN

Triple negative breast cancer (TNBC) is an aggressive type of breast cancer characterized by the absence of defined molecular targets, including estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) and is associated with high rates of relapse and distant metastasis despite surgery and adjuvant chemotherapy. The lack of effective targeted therapies for TNBC represents an unmet therapeutic challenge. Eukaryotic elongation factor 2 kinase (eEF2K) is an atypical calcium/calmodulin-dependent serine/threonine kinase that promotes TNBC tumorigenesis, progression, and drug resistance, representing a potential novel molecular target. However, the mechanisms regulating eEF2K expression are unknown. Here, we report that eEF2K protein expression is highly up-regulated in TNBC cells and patient tumors and it is associated with poor patient survival and clinical outcome. We found that loss/reduced expression of miR-603 leads to eEF2K overexpression in TNBC cell lines. Its expression results in inhibition of eEF2K by directly targeting the 3-UTR and the inhibition of tumor cell growth, migration and invasion in TNBC. In vivo therapeutic gene delivery of miR-603 into TNBC xenograft mouse models by systemic administration of miR-603-nanoparticles led to a significant inhibition of eEF2K expression and tumor growth, which was associated with decreased activity of the downstream targets of eEF2K, including Src, Akt, cyclin D1 and c-myc. Our findings suggest that miR-603 functions as a tumor suppressor and loss of miR-603 expression leads to increase in eEF2K expression and contributes to the growth, invasion, and progression of TNBC. Taken together, our data suggest that miR-603-based gene therapy is a potential strategy against TNBC.


Asunto(s)
Carcinogénesis/genética , Quinasa del Factor 2 de Elongación/antagonistas & inhibidores , MicroARNs/administración & dosificación , MicroARNs/genética , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/terapia , Animales , Línea Celular Tumoral , Quinasa del Factor 2 de Elongación/genética , Femenino , Humanos , Liposomas/administración & dosificación , Ratones , Ratones Desnudos , MicroARNs/biosíntesis , Nanopartículas/administración & dosificación , Transfección , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Pharm Pharm Sci ; 6(2): 292-301, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12935441

RESUMEN

PURPOSE: To determine single-and multiple-dose pharmacokinetics of liposomal-all- trans -retinoic acid (Atragen) following intravenous and oral ATRA (Vesanoid) administration in healthy volunteers. METHODS: This was a randomized, prospective, open-label, parallel pharmacokinetic study in which 29 subjects were given 90 mg/m (2) i.v. liposomal (L)-ATRA (16 subjects) every other day or 45 mg/m (2) oral ATRA (13 subjects) daily over 15 days. Pharmacokinetic parameters were assessed on days 1, 9, and 15. RESULTS: Twenty-two subjects (11 in each group) completed the study and were evaluated. Area under the plasma concentration-time curve [AUC(0, infinity)] and maximum plasma concentration (C(max) ) of ATRA did not decrease during the 15 days of L-ATRA treatment. However, the oral ATRA regimen resulted in a significant decrease in the AUC(0, infinity) and C(max) of 45.3% and 31.8%, respectively, on day 9 as compared with that to day 1 (p< 0.05). In addition, the mean AUC(0, infinity) was 13- and 22-fold greater for L-ATRA than for oral ATRA on days 1 and 9, respectively. Despite the significantly higher plasma concentrations after L-ATRA treatment, the side effects of each formulation were similar, except for dermal exfoliation, which was seen in 31% of the subjects after L-ATRA dosing, and abnormal liver function tests that were seen in 23% of the subjects after oral ATRA administration. CONCLUSIONS: These findings suggest that i.v. administration of L-ATRA maintains higher and stable plasma ATRA concentrations than oral ATRA in healthy subjects after repetitive administration. L-ATRA with a favorable pharmacokinetic profile may have potential advantages over oral ATRA and may be more efficacious in the treatment of acute promyelocytic leukemia or other retinoid-responsive cancers.


Asunto(s)
Antineoplásicos/farmacocinética , Liposomas , Tretinoina/farmacocinética , Administración Oral , Adulto , Antineoplásicos/administración & dosificación , Portadores de Fármacos , Femenino , Humanos , Masculino , Estudios Prospectivos , Tretinoina/administración & dosificación
10.
Adv Drug Deliv Rev ; 66: 110-6, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24384374

RESUMEN

Small interfering RNAs (siRNA) have recently emerged as a new class of therapeutics with a great potential to revolutionize the treatment of cancer and other diseases. A specifically designed siRNA binds and induces post-transcriptional silencing of target genes (mRNA). Clinical applications of siRNA-based therapeutics have been limited by their rapid degradation, poor cellular uptake, and rapid renal clearance following systemic administration. A variety of synthetic and natural nanoparticles composed of lipids, polymers, and metals have been developed for siRNA delivery, with different efficacy and safety profiles. Liposomal nanoparticles have proven effective in delivering siRNA into tumor tissues by improving stability and bioavailability. While providing high transfection efficiency and a capacity to form complexes with negatively charged siRNA, cationic lipids/liposomes are highly toxic. Negatively charged liposomes, on the other hand, are rapidly cleared from circulation. To overcome these problems we developed highly safe and effective neutral lipid-based nanoliposomes that provide robust gene silencing in tumors following systemic (intravenous) administration. This delivery system demonstrated remarkable antitumor efficacy in various orthotopic human cancer models in animals. Here, we briefly overview this and other lipid-based approaches with preclinical applications in different tumor models for cancer therapy and potential applications as siRNA-nanotherapeutics in human cancers.


Asunto(s)
Neoplasias/terapia , ARN Interferente Pequeño/administración & dosificación , Animales , Humanos , Liposomas , Nanopartículas/administración & dosificación
11.
Clin Cancer Res ; 18(24): 6648-57, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23052253

RESUMEN

PURPOSE: The TMPRSS2/ERG (T/E) fusion gene is present in half of all prostate cancer tumors. Fusion of the oncogenic ERG gene with the androgen-regulated TMPRSS2 gene promoter results in expression of fusion mRNAs in prostate cancer cells. The junction of theTMPRSS2- and ERG-derived portions of the fusion mRNA constitutes a cancer-specific target in cells containing the T/E fusion gene. Targeting the most common alternatively spliced fusion gene mRNA junctional isoforms in vivo using siRNAs in liposomal nanovectors may potentially be a novel, low-toxicity treatment for prostate cancer. EXPERIMENTAL DESIGN: We designed and optimized siRNAs targeting the two most common T/E fusion gene mRNA junctional isoforms (type III or type VI). Specificity of siRNAs was assessed by transient co-transfection in vitro. To test their ability to inhibit growth of prostate cancer cells expressing these fusion gene isoforms in vivo, specific siRNAs in liposomal nanovectors were used to treat mice bearing orthotopic or subcutaneous xenograft tumors expressing the targeted fusion isoforms. RESULTS: The targeting siRNAs were both potent and highly specific in vitro. In vivo they significantly inhibited tumor growth. The degree of growth inhibition was variable and was correlated with the extent of fusion gene knockdown. The growth inhibition was associated with marked inhibition of angiogenesis and, to a lesser degree, proliferation and a marked increase in apoptosis of tumor cells. No toxicity was observed. CONCLUSIONS: Targeting the T/E fusion junction in vivo with specific siRNAs delivered via liposomal nanovectors is a promising therapy for men with prostate cancer.


Asunto(s)
Proteínas de Fusión Oncogénica/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Transfección/métodos , Animales , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Liposomas , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Proteínas de Fusión Oncogénica/metabolismo , Neoplasias de la Próstata/terapia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Regulador Transcripcional ERG
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA