Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Clin Cancer Res ; 12(9): 2869-78, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16675583

RESUMEN

PURPOSE: The purpose of this study was to evaluate a fiber knob replacement strategy to improve infectivity and efficacy of Ad5 fiber chimeric oncolytic viruses for treatment of melanoma and head and neck cancers (HNC). EXPERIMENTAL DESIGN: Adenoviral receptors and transduction levels were used to determine the level of infectivity of fiber-modified, green fluorescent protein-expressing, replication-deficient viruses in a panel of melanoma and HNC cell lines in vitro. Virus yield and cytotoxicity assays were used to determine the tumor specificity and virus replication-mediated cytotoxicity of the fiber-modified oncolytic viruses in the same panel of melanoma and HNC in vitro. Xenograft tumor models were used to assess the antitumor activity of those fiber-modified chimeric viruses compared with the parental virus. RESULTS: Marker gene expression following gene transfer of the fiber chimeric vectors in melanoma and HNC cell lines was approximately 10-fold higher than that obtained with parental Ad5 vector. The fiber chimeric oncolytic variants mediated killing of melanoma and HNC cells that was 2- to 576-fold better than with the parental virus. In addition, fiber chimeric variants produced 2- to 7-fold more progeny virus in tumor cells than the parental virus. Moreover, a high multiplicity of infection was needed for the fiber chimeric viruses to produce cytotoxicity in normal cells. A significantly stronger antitumor response and survival advantage were shown in the tested melanoma and HNC xenograft models following i.t. injections. CONCLUSIONS: In vitro and in vivo studies showed the improved transduction, replication, cytotoxicity, antitumor efficacy, and survival advantage in melanoma and HNC tumor models, suggesting a potential use of these oncolytic agents for the treatment of melanoma and HNCs.


Asunto(s)
Adenovirus Humanos/genética , Técnicas de Transferencia de Gen , Neoplasias de Cabeza y Cuello/terapia , Melanoma/terapia , Línea Celular Tumoral , Quimera , Cartilla de ADN , Marcadores Genéticos , Humanos , Reacción en Cadena de la Polimerasa , Transducción Genética
12.
Clin Cancer Res ; 12(1): 305-13, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16397056

RESUMEN

PURPOSE: The purpose of this study was to examine the tumor specificity, cytotoxicity, and granulocyte macrophage colony-stimulating factor expression of CG0070, a conditionally replicating oncolytic adenovirus, in human bladder transitional cell carcinoma (TCC) cell lines and determine its antitumor efficacy in bladder TCC tumor models. EXPERIMENTAL DESIGN: Virus yield and cytotoxicity assays were used to determine tumor specificity and virus replication-mediated cytotoxicity of CG0070 in a panel of human bladder TCC cell lines and primary cells in vitro. Two s.c. and one orthotopic bladder TCC xenograft tumor models were used to assess antitumor activity of CG0070. RESULTS: In a matched isogenic pair of cell lines with differing retinoblastoma (Rb) pathway status, CG0070 showed selective E1a and granulocyte macrophage colony-stimulating factor (GM-CSF) expression in Rb pathway-defective cells. CG0070 replicated in Rb-defective bladder TCC cell lines as efficiently as wild-type adenovirus but produced 100-fold less virus in normal human cells. CG0070 was up to 1,000-fold more cytotoxic in Rb pathway-defective bladder TCC cells in comparison with normal human cells. Antitumor activity of CG0070 was shown in two bladder TCC s.c. xenograft tumor models following intratumoral injections and intravesical treatment in an orthotopic xenograft tumor model when compared with PBS treatment. CONCLUSIONS: In vitro and in vivo studies showed the selective replication, cytotoxicity, GM-CSF production, and antitumor efficacy of CG0070 in several bladder TCC models, suggesting a potential utility of this oncolytic agent for the treatment of bladder cancer. Further studies are warranted to show the role of human GM-CSF in the antitumor efficacy of CG0070.


Asunto(s)
Carcinoma de Células Transicionales/virología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Viroterapia Oncolítica , Neoplasias de la Vejiga Urinaria/virología , Adenoviridae/genética , Proteínas E1A de Adenovirus/biosíntesis , Animales , Antineoplásicos Fitogénicos/uso terapéutico , Carcinoma de Células Transicionales/terapia , Línea Celular Tumoral , Docetaxel , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Virus Oncolíticos , Proteína de Retinoblastoma/biosíntesis , Proteína de Retinoblastoma/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Taxoides/uso terapéutico , Neoplasias de la Vejiga Urinaria/terapia , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Methods Mol Med ; 130: 61-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17401164

RESUMEN

One of the most time-consuming steps in the generation of adenoviral vectors is the construction of recombinant plasmids. This chapter describes a detailed method for the rapid construction of adenoviral vectors. The method described here uses homologous recombination machinery of Escherichia coli BJ5183 to construct plasmids used in generation of adenoviral vectors. With this method, no ligation steps are involved in generating the plasmids, and any region of the adenoviral genome can be easily modified. Briefly, the full-length adenoviral genome flanked by unique restriction enzyme sites is first cloned into a bacterial plasmid. Next, the region of the viral genome to be modified is subcloned into a bacterial shuttle plasmid, and the desired changes are introduced by molecular biology techniques. The modified viral DNA fragment is gel-purified and cotransformed with the full-length plasmid, linearized in the targeted region, into BJ5183 cells. Homologous recombination in E. coli generates plasmids containing the modified adenoviral genome. Recombinant virus is generated following release of the viral DNA sequences from the plasmid backbone and transfection into a producer cell line. With this method, homogeneous recombinant adenoviruses can be obtained without plaque purification.


Asunto(s)
Adenoviridae/genética , Escherichia coli/genética , Recombinación Genética , ADN Viral/genética , Vectores Genéticos , Genoma Viral , Indicadores y Reactivos , Plásmidos , Transfección/métodos
14.
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
15.
Virus Res ; 116(1-2): 119-28, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16242804

RESUMEN

Out of 51 human adenoviral serotypes recognized to date, 32 of them belong to species D. Members of species D adenoviruses are commonly isolated from immune suppressed patients (organ transplant) and patients suffering from AIDS. The role of species D adenoviruses in pathogenesis is currently unclear. To derive new insights into the genetic content and evolution of species D adenoviruses and as a first step towards development of human adenovirus serotype 46 (Ad46) as vector, the complete nucleotide sequence of the virus was determined. The size of the genome is 35,178 bp in length with a G+C content of 56.9%. All the early and late region genes are present in the expected locations of the genome. The deduced amino acid sequences of all late region genes, with the exception of fiber, exhibited high degree of homology with the corresponding proteins of other adenoviruses. The deduced amino acid sequences of early regions E1, E3 and E4 showed a high degree of homology with the corresponding proteins of adenoviruses belonging to species D and less homology with the corresponding proteins of adenoviruses of other species. The homologues of Ad5 E3 region genes encoding 12.5K, gp19K, 10.4K, 14.5K and 14.7K are conserved in the genome of Ad46. However, the E3 region of Ad46 lacks genes encoding 6.7K and adenovirus death protein (ADP) but contains two additional open reading frames with a coding capacity of 433 and 281 amino acids. The fiber protein of Ad46 is 200 amino acids smaller than the fiber protein of Ad5 and contains only 10 pseudo-repeats in the shaft region. To facilitate the manipulation of the genome, the complete genome of Ad46 was cloned into a single bacterial plasmid. Following transfection into E1 complementing cell lines, the virus was recovered demonstrating the feasibility of viral genome manipulation for generation of recombinant viruses.


Asunto(s)
Adenovirus Humanos/genética , Genoma Viral , Proteínas Precoces de Adenovirus/genética , Composición de Base , Secuencia de Bases , Línea Celular , Secuencia Conservada , Orden Génico , Humanos , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Filogenia , ARN Viral , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Serotipificación , Proteínas Virales/genética
16.
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
17.
Hum Gene Ther ; 14(8): 749-61, 2003 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-12804138

RESUMEN

Transcriptional regulation that is rapid, reversible, and repeatedly inducible would greatly enhance the safety and efficacy of many gene therapy strategies. We developed a chimeric ligand-inducible regulation system based on the human estrogen receptor. This system has two components, the responsive promoter driving expression of the transgene of interest, and the ligand-inducible chimeric transcription factor. The transcription factor is composed of a novel DNA binding domain and a modified estrogen receptor ligand-binding domain. A point mutation in the ligand-binding domain significantly reduces estrogen binding while allowing binding of the estrogen antagonist, tamoxifen. We used a gutless adenoviral vector system and incorporated both components into two separate vectors. A single gutless vector encoding both system components was also generated. The tamoxifen-mediated induciblity of transgene expression of the gutless vector system was compared in vitro and in vivo with the analogous components incorporated into early generation, E1/E2a/E3-deficient adenoviral vectors. In normal mice, both the gutless vector and early generation systems displayed inducibility in the presence of tamoxifen. Importantly, the gutless vector system was inducible to extremely high levels, at least four times over a 2-month period. In contrast, the early generation vector system was inducible only once. Furthermore, the early generation system displayed significant toxicity, as evidenced by extremely high liver enzyme levels, abnormal liver pathology, and rapid loss of vector DNA from the liver, while the gutless vector system displayed minimal toxicity. These data directly demonstrate the improved in vivo function of the tamoxifen-inducible transcriptional regulation system in the context of the gutless adenoviral vectors.


Asunto(s)
Adenoviridae/genética , Regulación de la Expresión Génica , Vectores Genéticos , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas , Virus Defectuosos/genética , Endostatinas/biosíntesis , Endostatinas/genética , Vectores Genéticos/toxicidad , Células HeLa , Humanos , Ligandos , Hígado/metabolismo , Hígado/patología , Hepatopatías/metabolismo , Hepatopatías/patología , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/farmacología , Transcripción Genética
18.
Hum Gene Ther ; 14(3): 243-54, 2003 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-12639304

RESUMEN

Adenoviral vectors devoid of all viral coding regions are referred to by many names, including gutless vectors. Gutless vectors display reduced toxicity and immunogenicity, increased duration of transgene expression, and increased coding capacity compared to early generation vectors, which contain the majority of the viral backbone genes. However, the production of gutless vectors at a scale and purity suitable for clinical use has limited the utility of this technology. In this work we describe the optimization of the production of gutless vectors. We constructed an improved helper virus and generated an alternative gutless vector producer cell line, PERC6-Cre. We demonstrated increased gutless vector yields, minimal helper virus contamination, and no replication-competent adenovirus contamination using the optimized system. Furthermore, the PERC6-Cre cells were adapted to serum-free suspension culture and high-titer gutless vector preparations were produced using bioreactor technology, suggesting the feasibility of gutless vector scale-up for clinical use. Finally, we observed that helper virus lacking a packaging signal could be packaged at a low frequency, revealing an inherent limitation to the differential packaging strategy for gutless vector propagation.


Asunto(s)
Adenoviridae , Vectores Genéticos , Virus Helper , Genes Reporteros , Reacción en Cadena de la Polimerasa
19.
Cancer Gene Ther ; 11(8): 555-69, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15232601

RESUMEN

A potentially promising treatment of metastatic cancer is the systemic delivery of oncolytic adenoviruses. This requires engineering viruses which selectively replicate in tumors. We have constructed such an oncolytic adenovirus, OAS403, in which two early region genes are under the control of tumor-selective promoters that play a role in two key pathways involved in tumorigenesis. The early region E1A is controlled by the promoter for the E2F-1 gene, a transcription factor that primarily upregulates genes for cell growth. The E4 region is under control of the promoter for human telomerase reverse transcriptase, a gene upregulated in most cancer cells. OAS403 was evaluated in vitro on a panel of human cells and found to elicit tumor-selective cell killing. Also, OAS403 was less toxic in human hepatocyte cultures, as well as in vivo when compared to an oncolytic virus that lacked selective E4 control. A single intravenous injection of 3 x 10(12) vp/kg in a Hep3B xenograft mouse tumor model led to significant antitumor efficacy. Additionally, systemic administration in a pre-established LNCaP prostate tumor model resulted in over 80% complete tumor regressions at a tolerable dose. Vector genome copy number was measured in tumors and livers at various times following tail vein injection and showed a selective time-dependent increase in tumors but not livers over 29 days. Furthermore, efficacy was significantly improved when OAS403 treatment was combined with doxorubicin. This virus holds promise for the treatment of a broad range of human cancers including metastatic disease.


Asunto(s)
Adenoviridae/genética , Neoplasias/terapia , Adenoviridae/metabolismo , Animales , Proteínas de Unión al ADN , Doxorrubicina/uso terapéutico , Vectores Genéticos/administración & dosificación , Hepatocitos/metabolismo , Humanos , Concentración 50 Inhibidora , Inyecciones , Ratones , Ratones SCID , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/metabolismo , Regiones Promotoras Genéticas , Telomerasa/genética , Telomerasa/metabolismo , Replicación Viral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Oncoscience ; 6(1-2): 294-295, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30800717
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA