Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 32(6): 1895-1916, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38549376

RESUMEN

Malignant tumors are often associated with an immunosuppressive tumor microenvironment (TME), rendering most of them resistant to standard-of-care immune checkpoint inhibitors (CPIs). Signal transducer and activator of transcription 3 (STAT3), a ubiquitously expressed transcription factor, has well-defined immunosuppressive functions in several leukocyte populations within the TME. Since the STAT3 protein has been challenging to target using conventional pharmaceutical modalities, we investigated the feasibility of applying systemically delivered RNA interference (RNAi) agents to silence its mRNA directly in tumor-associated immune cells. In preclinical rodent tumor models, chemically stabilized acylated small interfering RNAs (siRNAs) selectively silenced Stat3 mRNA in multiple relevant cell types, reduced STAT3 protein levels, and increased cytotoxic T cell infiltration. In a murine model of CPI-resistant pancreatic cancer, RNAi-mediated Stat3 silencing resulted in tumor growth inhibition, which was further enhanced in combination with CPIs. To further exemplify the utility of RNAi for cancer immunotherapy, this technology was used to silence Cd274, the gene encoding the immune checkpoint protein programmed death-ligand 1 (PD-L1). Interestingly, silencing of Cd274 was effective in tumor models that are resistant to PD-L1 antibody therapy. These data represent the first demonstration of systemic delivery of RNAi agents to the TME and suggest applying this technology for immuno-oncology applications.


Asunto(s)
Antígeno B7-H1 , Interferencia de ARN , ARN Interferente Pequeño , Factor de Transcripción STAT3 , Microambiente Tumoral , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Animales , Ratones , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Línea Celular Tumoral , Humanos , Microambiente Tumoral/inmunología , ARN Interferente Pequeño/genética , Inmunoterapia/métodos , Resistencia a Antineoplásicos/genética , Inhibidores de Puntos de Control Inmunológico/farmacología , Modelos Animales de Enfermedad , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/genética
2.
Nanomedicine (Lond) ; 16(7): 535-551, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33683145

RESUMEN

Aim: To evaluate the role of vitronectin-enriched protein corona on systemic delivery of siRNA-encapsulated cationic lipid nanoparticles (LNPs) to αvß3 integrin expressing solid tumors. Materials & methods: 1,2-Dioleoyl-3-trimethylammonium-propane LNPs were formulated, protein corona formed in nude mice serum and its impact on drug delivery were analyzed. Results: 1,2-Dioleoyl-3-trimethylammonium-propane-containing LNP led to enhanced recruitment of vitronectin and showed preferential transfection to αvß3-expressed cells relative to controls. Upon systemic administration in mice, the LNPs accumulated in the αvß3-expressing endothelial lining of the tumor blood vessels before reaching tumor cells. Conclusion: These results present an optimized LNP that selectively recruits endogenous proteins in situ to its corona which may lead to enhanced delivery and transfection in tissues of interest.


Asunto(s)
Nanopartículas , Animales , Lípidos , Ratones , Ratones Desnudos , Corona de Proteínas , ARN Interferente Pequeño/genética , Transfección , Vitronectina
3.
AAPS J ; 22(4): 83, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32495039

RESUMEN

Upon systemic administration, nanoparticles encounter serum proteins in the biological system resulting in the formation of "protein corona" on the surface. Increased understanding of the relationship between nanoparticles' "chemical identity" and "biological identity" can contribute to improved clinical translation. Recent studies of protein corona composition on nanoparticles, including from our group, suggest that a strategic choice of materials can influence the types of protein adsorbed from plasma and lead to improved delivery efficiency. This mini-review reflects on the fundamental knowledge of nanoparticle protein corona and highlights the recent applications of protein corona on nanoparticles' systemic circulation, cell, and tissue-specific delivery. Important considerations on the safety and efficacy aspects pertaining to the exploration of nanoparticle protein corona's targeting effect are also summarized. Finally, the future perspectives of protein corona research are discussed.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/metabolismo , Corona de Proteínas/metabolismo , Corona de Proteínas/métodos , Animales , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Nanopartículas/administración & dosificación , Tamaño de la Partícula , Unión Proteica/fisiología , Propiedades de Superficie/efectos de los fármacos
4.
Nanoscale ; 11(40): 18806-18824, 2019 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-31595922

RESUMEN

The application of lipid-based nanoparticle (LNP) delivery systems remains a popular strategy for the systemic delivery of gene therapies to specific disease targets, including solid tumors. It is now well acknowledged that upon systemic administration, biomolecules from blood will adsorb onto nanoparticles' surfaces, forming a "protein corona", affording nanoparticles a "biological identity" on top of their "synthetic identity". Detailed analysis of nanoparticle protein corona is gradually revealing the "missing link" between nanoparticle chemical properties and the biological identity. Nevertheless, the discovery of nanoparticle protein corona's impact on tumor delivery is limited. In this study, we demonstrate that protein corona can be manipulated by formulation composition and particle surface charge changes, and a single lipid switch could switch the nanoparticle protein corona profile. The protein corona composition differences had a profound impact on cell transfection, in vivo biodistribution as well as tumor-specific delivery efficiency. Nanoparticles with apolipoprotein-rich corona showed better delivery to hepatocellular carcinoma (HepG2) as compared to those with vitronectin-rich corona. In addition, we found that, the PEG conjugated lipid chain length and PEG amount in LNPs were key factors to consider in successful RNA interference therapy for solid tumors.


Asunto(s)
Apolipoproteínas , Carcinoma Hepatocelular/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Lípidos , Neoplasias Hepáticas/tratamiento farmacológico , Nanopartículas , Oligonucleótidos , Transfección , Vitronectina , Animales , Apolipoproteínas/química , Apolipoproteínas/farmacología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Femenino , Células Hep G2 , Humanos , Lípidos/química , Lípidos/farmacología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Desnudos , Nanopartículas/química , Nanopartículas/uso terapéutico , Oligonucleótidos/química , Oligonucleótidos/farmacología , Vitronectina/química , Vitronectina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Oncoscience ; 6(1-2): 294-295, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30800717
6.
Nanoscale ; 11(18): 8760-8775, 2019 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-30793730

RESUMEN

Delivery of genetic medicines, such as small interfering RNA (siRNA), by lipid nanoparticles (LNPs) is a promising approach towards the treatment of diseases, such as solid tumors. However, in vitro and in vivo nanoparticle delivery efficiency is influenced by the formation of a protein corona in biological media. In this study, we have formulated four types of EnCore nanoparticles (F1 to F4) with a similar composition, but different polyethylene glycol (PEG) conjugated lipid chain lengths (carbon 14 vs. carbon 18) and molar ratios (6% vs. 3%). These LNPs showed dramatic differences in cellular delivery and transfection in hepatocellular carcinoma (HepG2) cells in the absence and presence of fetal bovine serum (FBS). The presence of proteins inhibited the cellular uptake of C18 (3%) nanoparticles, while it facilitated the cellular uptake of C14 nanoparticles. Among the adsorbed proteins from FBS, apolipoprotein E, but not apolipoprotein A1, affected the cellular uptake of the carbon 14 LNPs. Additionally, surface PEG was one of the determinants for the protein corona amount and composition. Finally, different serum to LNP volume ratios resulted in different protein enrichment patterns. Overall, the results showed a correlation between surface chemistry of LNPs and the protein corona composition suggesting a potential use for targeted delivery.


Asunto(s)
Proteínas Sanguíneas/química , Lípidos/química , Nanopartículas/química , Corona de Proteínas/química , Interferencia de ARN , Apolipoproteína A-I/química , Células Hep G2 , Humanos , ARN Interferente Pequeño/química , ARN Interferente Pequeño/metabolismo , Propiedades de Superficie , Transfección/métodos , beta Catenina/antagonistas & inhibidores , beta Catenina/genética , beta Catenina/metabolismo
7.
Mol Ther ; 26(11): 2567-2579, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30274786

RESUMEN

Wnt/ß-catenin signaling mediates cancer immune evasion and resistance to immune checkpoint therapy, in part by blocking cytokines that trigger immune cell recruitment. Inhibition of ß-catenin may be an effective strategy for increasing the low response rate to these effective medicines in numerous cancer populations. DCR-BCAT is a nanoparticle drug product containing a chemically optimized RNAi trigger targeting CTNNB1, the gene that encodes ß-catenin. In syngeneic mouse tumor models, ß-catenin inhibition with DCR-BCAT significantly increased T cell infiltration and potentiated the sensitivity of the tumors to checkpoint inhibition. The combination of DCR-BCAT and immunotherapy yielded significantly greater tumor growth inhibition (TGI) compared to monotherapy in B16F10 melanoma, 4T1 mammary carcinoma, Neuro2A neuroblastoma, and Renca renal adenocarcinoma. Response to the RNAi-containing combination therapy was not dependent on Wnt activation status of the tumor. Importantly, this drug combination was associated with elevated levels of biomarkers of T cell-mediated cytotoxicity. Finally, when CTLA-4 and PD-1 antibodies were combined with DCR-BCAT in MMTV-Wnt1 transgenic mice, a genetic model of spontaneous Wnt-driven tumors, complete regressions were achieved in the majority of treated subjects. These data support RNAi-mediated ß-catenin inhibition as an effective strategy to increase response rates to cancer immunotherapy.


Asunto(s)
Antígeno CTLA-4/antagonistas & inhibidores , Carcinoma de Células Renales/tratamiento farmacológico , Melanoma Experimental/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , beta Catenina/genética , Animales , Antígeno CTLA-4/genética , Antígeno CTLA-4/inmunología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Terapia Combinada , Femenino , Humanos , Inmunoterapia/métodos , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Transgénicos , Receptor de Muerte Celular Programada 1/inmunología , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Linfocitos T/inmunología , Vía de Señalización Wnt/genética , Proteína Wnt1/genética , beta Catenina/antagonistas & inhibidores
8.
Mol Cancer Ther ; 17(2): 544-553, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29282298

RESUMEN

Colorectal carcinomas harbor well-defined genetic abnormalities, including aberrant activation of Wnt/ß-catenin and MAPK pathways, often simultaneously. Although the MAPK pathway can be targeted using potent small-molecule drugs, including BRAF and MEK inhibitors, ß-catenin inhibition has been historically challenging. RNAi approaches have advanced to the stage of clinical viability and are especially well suited for transcriptional modulators, such as ß-catenin. In this study, we report therapeutic effects of combined targeting of these pathways with pharmacologic agents. Using a recently described tumor-selective nanoparticle containing a ß-catenin-targeting RNAi trigger, in combination with the FDA-approved MEK inhibitor (MEKi) trametinib, we demonstrate synergistic tumor growth inhibition in in vivo models of colorectal cancer, melanoma, and hepatocellular carcinoma. At dose levels that were insufficient to significantly impact tumor growth as monotherapies, combination regimens resulted in synergistic efficacy and complete tumor growth inhibition. Importantly, dual MEKi/RNAi therapy dramatically improved survival of mice bearing colorectal cancer liver metastases. In addition, pharmacologic silencing of ß-catenin mRNA was effective against tumors that are inherently resistant or that acquire drug-induced resistance to trametinib. These results provide a strong rationale for clinical evaluation of this dual-targeting approach for cancers harboring Wnt/ß-catenin and MAPK pathway mutations. Mol Cancer Ther; 17(2); 544-53. ©2017 AACR.


Asunto(s)
Neoplasias Colorrectales/terapia , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Piridonas/farmacología , Pirimidinonas/farmacología , ARN Mensajero/genética , ARN Interferente Pequeño/administración & dosificación , beta Catenina/genética , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Sinergismo Farmacológico , Silenciador del Gen , Xenoinjertos , Humanos , Neoplasias Hepáticas Experimentales/secundario , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Desnudos , Nanopartículas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo
9.
Nanomedicine (Lond) ; 12(17): 2113-2135, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28805542

RESUMEN

Although a variety of nanoparticles (NPs) have been used for drug delivery applications, their surfaces are immediately covered by plasma protein corona upon systemic administration. As a result, the adsorbed proteins create a unique biological identity of the NPs that lead to unpredictable performance. The protein corona on NPs could also impede active targeting, induce off-target effects, trigger particle clearance and even provoke toxicity. This article reviews the fundamentals of NP-plasma protein interaction, the consequences of the interactions, and provides insights into the correlations of protein corona with biodistribution and cellular delivery. We hope that this review will trigger additional questions and possible solutions that lead to more favorable developments in NP-based targeted delivery systems.


Asunto(s)
Proteínas Sanguíneas/química , Proteínas Sanguíneas/metabolismo , Nanopartículas/química , Nanopartículas/metabolismo , Administración Oral , Adsorción , Animales , Portadores de Fármacos , Liberación de Fármacos , Humanos , Terapia Molecular Dirigida , Tamaño de la Partícula , Corona de Proteínas/metabolismo , Propiedades de Superficie , Distribución Tisular
10.
Hepatology ; 65(5): 1581-1599, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27981621

RESUMEN

Recently, we have shown that coexpression of hMet and mutant-ß-catenin using sleeping beauty transposon/transposase leads to hepatocellular carcinoma (HCC) in mice that corresponds to around 10% of human HCC. In the current study, we investigate whether Ras activation, which can occur downstream of Met signaling, is sufficient to cause HCC in association with mutant-ß-catenin. We also tested therapeutic efficacy of targeting ß-catenin in an HCC model. We show that mutant-K-Ras (G12D), which leads to Ras activation, cooperates with ß-catenin mutants (S33Y, S45Y) to yield HCC in mice. Affymetrix microarray showed > 90% similarity in gene expression in mutant-K-Ras-ß-catenin and Met-ß-catenin HCC. K-Ras-ß-catenin tumors showed up-regulation of ß-catenin targets like glutamine synthetase (GS), leukocyte cell-derived chemotaxin 2, Regucalcin, and Cyclin-D1 and of K-Ras effectors, including phosphorylated extracellular signal-regulated kinase, phosphorylated protein kinase B, phosphorylated mammalian target of rapamycin, phosphorylated eukaryotic translation initiation factor 4E, phosphorylated 4E-binding protein 1, and p-S6 ribosomal protein. Inclusion of dominant-negative transcription factor 4 at the time of K-Ras-ß-catenin injection prevented HCC and downstream ß-catenin and Ras signaling. To address whether targeting ß-catenin has any benefit postestablishment of HCC, we administered K-Ras-ß-catenin mice with EnCore lipid nanoparticles (LNP) loaded with a Dicer substrate small interfering RNA targeting catenin beta 1 (CTNNB1; CTNNB1-LNP), scrambled sequence (Scr-LNP), or phosphate-buffered saline for multiple cycles. A significant decrease in tumor burden was evident in the CTNNB1-LNP group versus all controls, which was associated with dramatic decreases in ß-catenin targets and some K-Ras effectors, leading to reduced tumor cell proliferation and viability. Intriguingly, in relatively few mice, non-GS-positive tumors, which were evident as a small subset of overall tumor burden, were not affected by ß-catenin suppression. CONCLUSION: Ras activation downstream of c-Met is sufficient to induce clinically relevant HCC in cooperation with mutant ß-catenin. ß-catenin suppression by a clinically relevant modality is effective in treatment of ß-catenin-positive, GS-positive HCCs. (Hepatology 2017;65:1581-1599).


Asunto(s)
Carcinoma Hepatocelular/etiología , Genes ras , Neoplasias Hepáticas Experimentales/etiología , beta Catenina/metabolismo , Animales , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Serina-Treonina Quinasas TOR/metabolismo , beta Catenina/antagonistas & inhibidores , beta Catenina/genética
11.
Mol Cancer Ther ; 15(9): 2143-54, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27390343

RESUMEN

The Wnt/ß-catenin pathway is among the most frequently altered signaling networks in human cancers. Despite decades of preclinical and clinical research, efficient therapeutic targeting of Wnt/ß-catenin has been elusive. RNA interference (RNAi) technology silences genes at the mRNA level and therefore can be applied to previously undruggable targets. Lipid nanoparticles (LNP) represent an elegant solution for the delivery of RNAi-triggering oligonucleotides to disease-relevant tissues, but have been mostly restricted to applications in the liver. In this study, we systematically tuned the composition of a prototype LNP to enable tumor-selective delivery of a Dicer-substrate siRNA (DsiRNA) targeting CTNNB1, the gene encoding ß-catenin. This formulation, termed EnCore-R, demonstrated pharmacodynamic activity in subcutaneous human tumor xenografts, orthotopic patient-derived xenograft (PDX) tumors, disseminated hematopoietic tumors, genetically induced primary liver tumors, metastatic colorectal tumors, and murine metastatic melanoma. DsiRNA delivery was homogeneous in tumor sections, selective over normal liver and independent of apolipoprotein-E binding. Significant tumor growth inhibition was achieved in Wnt-dependent colorectal and hepatocellular carcinoma models, but not in Wnt-independent tumors. Finally, no evidence of accelerated blood clearance or sustained liver transaminase elevation was observed after repeated dosing in nonhuman primates. These data support further investigation to gain mechanistic insight, optimize dose regimens, and identify efficacious combinations with standard-of-care therapeutics. Mol Cancer Ther; 15(9); 2143-54. ©2016 AACR.


Asunto(s)
Neoplasias/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , beta Catenina/genética , Animales , Apolipoproteínas E/química , Apolipoproteínas E/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Lípidos/química , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Melanoma Experimental , Ratones , Nanopartículas/química , Metástasis de la Neoplasia , Neoplasias/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/química , Relación Estructura-Actividad , Vía de Señalización Wnt , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
12.
J Control Release ; 172(3): 699-706, 2013 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-24161254

RESUMEN

Multidrug resistance (MDR) is a significant problem in the clinical management of several cancers. Overcoming MDR generally involves multi-modal therapeutic approaches that integrate enhancement of delivery efficiency using targeted nano-platforms as well as strategies that can sensitize cancer cells to drug treatments. We recently demonstrated that tandem delivery of siRNAs that downregulate anti-apoptotic genes overexpressed in cisplatin resistant tumors followed by therapeutic challenge using cisplatin loaded CD44 targeted hyaluronic acid (HA) nanoparticle (NP) induced synergistic antitumor response CD44 expressing tumors that are resistant to cisplatin. In the current study, a near infrared (NIR) dye-loaded HA NP was employed to image the whole body localization of NPs after intravenous (i.v.) injection into live mice bearing human lung tumors that were sensitive and resistant to cisplatin. In addition, we quantified the siRNA duplexes and cisplatin dose distribution in various tissues and organs using an ultra-sensitive quantitative PCR method and inductively coupled plasma-mass spectrometry (ICP-MS), respectively, after i.v. injection of the payload loaded HA NPs in tumor bearing mice. Our findings demonstrate that the distribution pattern of the siRNA and cisplatin using specifically engineered CD44 targeting HA NPs correlated well with the tumor targeting capability as well as the activity and efficacy obtained with combination treatments.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Sistemas de Liberación de Medicamentos , Ácido Hialurónico/química , Nanopartículas/química , ARN Interferente Pequeño/administración & dosificación , Animales , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Cisplatino/farmacocinética , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Ratones , Neoplasias/tratamiento farmacológico , ARN Interferente Pequeño/farmacocinética , Distribución Tisular
13.
Mol Ther Nucleic Acids ; 2: e110, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23900224

RESUMEN

One of the most challenging aspects of lung cancer therapy is the rapid acquisition of multidrug-resistant (MDR) phenotype. One effective approach would be to identify and downregulate resistance-causing genes in tumors using small interfering RNAs (siRNAs) to increase the sensitivity of tumor cells to chemotherapeutic challenge. After identifying the overexpressed resistance-related antiapoptotic genes (survivin and bcl-2) in cisplatin-resistant cells, the siRNA sequences were designed and screened to select the most efficacious candidates. Modifications were introduced in them to minimize off-target effects. Subsequently, the combination of siRNA and cisplatin that gave the maximum synergy was identified in resistant cells. We then demonstrated that the combination treatment of the selected siRNAs and cisplatin encapsulated in CD44-targeting hyaluronic acid (HA)-based self-assembling nanosystems reversed the resistance to cisplatin and delayed the tumor growth significantly (growth inhibition increased from 30 to 60%) in cisplatin-resistant tumors. In addition, no abnormalities in body weights, liver enzyme levels or histopathology of liver/spleen tissues were observed in any of the treatment groups during the study period. Overall, we demonstrate that the combination of siRNA-mediated gene-silencing strategy with chemotherapeutic agents constitutes a valuable and safe approach for the treatment of MDR tumors.Molecular Therapy-Nucleic Acids (2013) 2, e110; doi:10.1038/mtna.2013.29; published online 30 July 2013.

14.
Biomaterials ; 34(13): 3489-502, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23410679

RESUMEN

Anticancer therapeutics employing RNA interference mechanism holds promising potentials for sequence-specific silencing of target genes. However targeted delivery of siRNAs to tumor tissues and cells and more importantly, their intracellular release at sites of interest still remains a major challenge that needs to be addressed before this technique could become a clinically viable option. In the current study, we have engineered and screened a series of CD44 targeting hyaluronic acid (HA) based self-assembling nanosystems for targeted siRNA delivery. The HA polymer was functionalized with lipids of varying carbon chain lengths/nitrogen content, as well as polyamines for assessing siRNA encapsulation. From the screens, several HA-derivatives were identified that could stably encapsulate/complex siRNAs and form self-assembled nanosystems, as determined by gel retardation assays and dynamic light scattering. Many HA derivatives could transfect siRNAs into cancer cells overexpressing CD44 receptors. Interestingly, blocking the CD44 receptors on the cells using free excess soluble HA prior to incubation of cy3-labeled-siRNA loaded HA nano-assemblies resulted in >90% inhibition of the receptor mediated uptake, confirming target specificity. In addition, SSB/PLK1 siRNA encapsulated in HA-PEI/PEG nanosystems demonstrated dose dependent and target specific gene knockdown in both sensitive and resistant A549 lung cancer cells overexpressing CD44 receptors. More importantly, these siRNA encapsulated nanosystems demonstrated tumor selective uptake and target specific gene knock down in vivo in solid tumors as well as in metastatic tumors. The HA based nanosystems thus portend to be promising siRNA delivery vectors for systemic targeting of CD44 overexpressing cancers including tumor initiating (stem-) cells and metastatic lesions.


Asunto(s)
Técnicas de Transferencia de Gen , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/química , Nanopartículas/química , Neoplasias/metabolismo , ARN Interferente Pequeño/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Endocitosis , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Ácido Hialurónico/síntesis química , Ratones , Nanopartículas/ultraestructura , Metástasis de la Neoplasia , Neoplasias/irrigación sanguínea , Neoplasias/patología , Polietileneimina/química , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa Tipo Polo 1
15.
Int J Pharm ; 427(1): 21-34, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21621597

RESUMEN

Gene therapy has shown a tremendous potential to benefit patients in a variety of disease conditions. However, finding a safe and effective systemic delivery system is the major obstacle in this area. Although viral vectors showed promise for high transfection rate, the immunogenicity associated with these systems has hindered further development. As an alternative to viral gene delivery, this review focuses on application of novel safe and effective non-condensing polymeric systems that have shown high transgene expression when administered systemically or by the oral route. Type B gelatin-based engineered nanocarriers were evaluated for passive and active tumor-targeted delivery and transfection using both reporter and therapeutic plasmid DNA. Additionally, we have shown that nanoparticles-in-microsphere oral system (NiMOS) can efficiently deliver reporter and therapeutic gene constructs in the gastrointestinal tract. Additionally, there has been a significant recent interest in the use small interfering RNA (siRNA) as a therapeutic system for gene silencing. Both gelatin nanoparticles and NiMOS have shown activity in systemic and oral delivery of siRNA, respectively.


Asunto(s)
Portadores de Fármacos/administración & dosificación , Marcación de Gen/métodos , Terapia Genética/métodos , Nanopartículas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Administración Oral , Animales , Tracto Gastrointestinal/metabolismo , Gelatina/administración & dosificación , Técnicas de Transferencia de Gen , Humanos , Ratones , Microesferas , Neoplasias/terapia , Plásmidos/administración & dosificación , Plásmidos/química , Polímeros/administración & dosificación
16.
J Control Release ; 155(2): 237-47, 2011 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-21497176

RESUMEN

Inefficiencies in systemic drug delivery and tumor residence as well as micro-environmental selection pressures contribute to the development of multidrug resistance (MDR) in cancer. Characteristics of MDR include abnormal vasculature, regions of hypoxia, up-regulation of ABC-transporters, aerobic glycolysis, and an elevated apoptotic threshold. Nano-sized delivery vehicles are ideal for treating MDR cancer as they can improve the therapeutic index of drugs and they can be engineered to achieve multifunctional parameters. The multifunctional ability of nanocarriers makes them more adept at treating heterogeneous tumor mass than traditional chemotherapy. Nanocarriers also have preferential tumor accumulation via the EPR effect; this accumulation can be further enhanced by actively targeting the biological profile of MDR cells. Perhaps the most significant benefit of using nanocarrier drug delivery to treat MDR cancer is that nanocarrier delivery diverts the effects of ABC-transporter mediated drug efflux; which is the primary mechanism of MDR. This review discusses the capabilities, applications, and examples of multifunctional nanocarriers for the treatment of MDR. This review emphasizes multifunctional nanocarriers that enhance drug delivery efficiency, the application of RNAi, modulation of the tumor apoptotic threshold, and physical approaches to overcome MDR.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Glucólisis/efectos de los fármacos , Nanotecnología , Neoplasias/terapia , Células Madre Neoplásicas/efectos de los fármacos , Adenosina Trifosfato/biosíntesis , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Terapia Combinada , Portadores de Fármacos/química , Resistencia a Múltiples Medicamentos/fisiología , Resistencia a Antineoplásicos/fisiología , Glucólisis/fisiología , Humanos , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neoplasias/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología
17.
Hum Gene Ther ; 20(10): 1201-13, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19572803

RESUMEN

The limited efficacy of adenovirus type 5 (Ad5)-based oncolytic viruses seen in the clinic thus far may be attributable in part to variable expression of its receptor on tumor cells. Replacement of the Ad5 fiber knob with the Ad35 fiber knob generated the Ad5/35 chimeric virus, which has previously been demonstrated to have significant antitumor activity in murine tumor models, presumably by virtue of its recognition of the CD46 receptor, which is abundant on many types of tumor cells. In the current study, a CD46 receptor transgenic mouse strain (hCD46Ge) that expresses the CD46 receptor in a pattern closely mirroring that in humans was used to study the in vivo properties of Ad5/Ad35 chimeric viruses. Vector distribution was evaluated after intravenous administration to hCD46Ge mice of an Ad5-based oncolytic adenovirus or an Ad5/35 chimeric oncolytic adenovirus (designated OV-5 and OV-5T35H, respectively), a wild-type Ad5 virus (Ad5wt), or an Ad5-based, E1-deleted adenovirus (Addl312) at 1.25 x 10(12) viral particles/kg. The amount of OV-5T35H vector genomes in the liver was at least two orders of magnitude lower than that of Ad5-based viruses. Moreover, animals injected with OV-5T35H virus had significantly lower elevations of serum proinflammatory cytokines and liver enzyme levels. Mice injected with Ad5wt lost more than 20% of their body weight and died or required euthanasia because of poor clinical condition within 4 days of virus administration. Mice treated with OV-5 lost as much as 15% of their body weight over 8-9 days, but recovered within 14 days. Mice that were treated with Addl312 or OV-5T35H exhibited no body weight loss during the study period. These studies suggest that the Ad5/35-based chimeric viruses may have a better safety profile after intravenous injection compared with Ad5-based viruses.


Asunto(s)
Adenoviridae/genética , Adenoviridae/fisiología , Proteína Cofactora de Membrana/genética , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Replicación Viral/fisiología , Animales , Biomarcadores/metabolismo , Recuento de Células Sanguíneas , Células Sanguíneas/metabolismo , Células Endoteliales/virología , Proteínas Fluorescentes Verdes/metabolismo , Heterocigoto , Homocigoto , Humanos , Inmunidad Innata/inmunología , Hígado/enzimología , Hígado/inmunología , Hígado/virología , Ratones , Viroterapia Oncolítica/métodos , Distribución Tisular , Transducción Genética
18.
Clin Cancer Res ; 14(12): 3933-41, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18559615

RESUMEN

PURPOSE: Evaluate the codelivery of hyaluronidase enzyme with oncolytic adenoviruses to determine whether it improves the spread of the virus throughout tumors, thereby leading to a greater overall antitumor efficacy in tumor models. EXPERIMENTAL DESIGN: The optimal dose of hyaluronidase that provided best transduction efficiency and spread of a green fluorescent protein (GFP)-expressing adenovirus within tumors was combined with oncolytic viruses in tumor models to determine whether the combination treatment results in an improvement of antitumor efficacy. RESULTS: In mice injected with the adenovirus Ad5/35GFP and an optimal dose of hyaluronidase (50 U), a significant increase in the number of GFP-expressing cells was observed when compared with animals injected with virus only (P < 0.0001). When the oncolytic adenoviruses Ad5OV or Ad5/35 OV (OV-5 or OV5T35H) were codelivered with 50 U of hyaluronidase, a significant delay in tumor progression was observed, which translated into a significant increase in the mean survival time of tumor-bearing mice compared with either of the monotherapy-treated groups (P < 0.0001). Furthermore, the mice that received the combination of Ad5/35 OV and hyaluronidase showed the best antitumor efficacy. Importantly, the combination treatment did not increase the metastatic potential of the tumors. Lastly, the increase in virus potency observed in animals injected with both enzyme and virus correlated with enhanced virus spread throughout tumors. CONCLUSION: Antitumor activity and overall survival of mice bearing highly aggressive tumors are significantly improved by codelivery of oncolytic adenoviruses and hyaluronidase when compared with either of the monotherapy-treated groups, and it may prove to be a potent and novel approach to treating patients with cancer.


Asunto(s)
Adenoviridae/efectos de los fármacos , Hialuronoglucosaminidasa/administración & dosificación , Hialuronoglucosaminidasa/farmacología , Virus Oncolíticos/efectos de los fármacos , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia , Adenoviridae/fisiología , Animales , Línea Celular Tumoral , Terapia Combinada , Femenino , Humanos , Inyecciones Intralesiones , Masculino , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Resultado del Tratamiento , Replicación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Natl Cancer Inst ; 99(21): 1623-33, 2007 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-17971529

RESUMEN

BACKGROUND: Numerous clinical trials have demonstrated that oncolytic viruses can elicit antitumor responses when they are administered directly into localized cancers. However, the treatment of metastatic disease with oncolytic viruses has been challenging due to the inactivation of viruses by components of human blood and/or to inadequate tumor selectivity. METHODS: We determined the cytolytic potential and selectivity of Seneca Valley Virus-001 (SVV-001), a newly discovered native picornavirus, in neuroendocrine and pediatric tumor cell lines and normal cells. Suitability of the virus for intravenous delivery in humans was assessed by blood inactivation assays. Safety was evaluated in vivo using an immune-competent mouse model, and efficacy was evaluated in vivo in athymic mice bearing tumors derived from human small-cell lung cancer and retinoblastoma cell lines. RESULTS: Cell lines derived from small-cell lung cancers and solid pediatric cancers were at least 10,000-fold more sensitive to the cytolytic activity of SVV-001 than were any of the adult normal human cells tested. Viral infectivity was not inhibited by human blood components. Intravenous doses up to 1 x 10(14) virus particles (vp) per kg were well tolerated, and no dose-limiting toxicity was observed in immune-competent mice. A single intravenous dose of 1 x 10(8) vp per kg into athymic mice bearing preestablished small-cell lung or retinoblastoma tumors resulted in complete, durable responses in ten of ten and five of eight mice, respectively. CONCLUSIONS: SVV-001 has potent cytolytic activity and high selectivity for tumor cell lines having neuroendocrine properties versus adult normal cells. Systemically administered SVV-001 has potential for the treatment of metastatic neuroendocrine cancers.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Neuroendocrino/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos , Picornaviridae , Animales , Carcinoma Neuroendocrino/tratamiento farmacológico , Carcinoma de Células Pequeñas/terapia , Línea Celular Tumoral , Modelos Animales de Enfermedad , Pruebas de Hemaglutinación , Humanos , Inmunohistoquímica , Inyecciones Intravenosas , Neoplasias Pulmonares/terapia , Ratones , Ratones Desnudos , Virus Oncolíticos/clasificación , Virus Oncolíticos/patogenicidad , Picornaviridae/clasificación , Picornaviridae/patogenicidad , Proyectos de Investigación , Retinoblastoma/terapia , Trasplante Heterólogo
20.
Cancer Res ; 67(9): 4399-407, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17483354

RESUMEN

Selective replication of oncolytic viruses in tumor cells provides a promising approach for the treatment of human cancers. One of the limitations observed with oncolytic viruses currently used in the treatment of solid tumors is the inefficient spread of virus throughout the tumor mass following intratumoral injection. Data are presented showing that oncolytic adenoviruses expressing the relaxin gene and containing an Ad5/Ad35 chimeric fiber showed significantly enhanced transduction and increased virus spread throughout the tumor when compared with non-relaxin-expressing, Ad5-based viruses. The increased spread of such viruses throughout tumors correlated well with improved antitumor efficacy and overall survival in two highly metastatic tumor models. Furthermore, nonreplicating viruses expressing relaxin did not increase metastases, suggesting that high level expression of relaxin will not enhance metastatic spread of tumors. In summary, the data show that relaxin may play a role in rearranging matrix components within tumors, which helps recombinant oncolytic adenoviruses to spread effectively throughout the tumor mass and thereby increase the extent of viral replication within the tumor. Expressing relaxin from Ad5/Ad35 fiber chimeric adenoviruses may prove a potent and novel approach to treating patients with cancer.


Asunto(s)
Adenoviridae/fisiología , Viroterapia Oncolítica/métodos , Neoplasias de la Próstata/terapia , Relaxina/biosíntesis , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Línea Celular Tumoral , Colágeno/metabolismo , Femenino , Vectores Genéticos/genética , Humanos , Masculino , Ratones , Metástasis de la Neoplasia , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/virología , Relaxina/genética , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA