Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Res ; 80(22): 4972-4985, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32978168

RESUMEN

Lung squamous carcinoma (LUSC) is a highly metastatic disease with a poor prognosis. Using an integrated screening approach, we found that miR-671-5p reduces LUSC metastasis by inhibiting a circular RNA (circRNA), CDR1as. Although the putative function of circRNA is through miRNA sponging, we found that miR-671-5p more potently silenced an axis of CDR1as and its antisense transcript, cerebellar degeneration related protein 1 (CDR1). Silencing of CDR1as or CDR1 significantly inhibited LUSC metastases and CDR1 was sufficient to promote migration and metastases. CDR1, which directly interacted with adaptor protein 1 (AP1) complex subunits and coatomer protein I (COPI) proteins, no longer promoted migration upon blockade of Golgi trafficking. Therapeutic inhibition of the CDR1as/CDR1 axis with miR-671-5p mimics reduced metastasis in vivo. This report demonstrates a novel role for CDR1 in promoting metastasis and Golgi trafficking. These findings reveal an miRNA/circRNA axis that regulates LUSC metastases through a previously unstudied protein, CDR1. SIGNIFICANCE: This study shows that circRNA, CDR1as, promotes lung squamous migration, metastasis, and Golgi trafficking through its complimentary transcript, CDR1.


Asunto(s)
Autoantígenos/metabolismo , Carcinoma de Células Escamosas/secundario , Aparato de Golgi/metabolismo , Neoplasias Pulmonares/patología , Proteínas del Tejido Nervioso/metabolismo , ARN Circular/antagonistas & inhibidores , ARN Largo no Codificante/metabolismo , Complejo 1 de Proteína Adaptadora/metabolismo , Animales , Autoantígenos/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/mortalidad , Línea Celular Tumoral , Movimiento Celular/fisiología , Proteína Coat de Complejo I/metabolismo , Retículo Endoplásmico/metabolismo , Femenino , Humanos , Ácido Hialurónico/uso terapéutico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidad , Ratones , Ratones Desnudos , MicroARNs/metabolismo , Nanopartículas/uso terapéutico , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/genética
2.
Mol Ther ; 27(3): 507-517, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30545600

RESUMEN

Local immunomodulation can be a promising strategy to augment the efficacy and decrease off-target toxicities associated with cancer treatment. Pancreatic cancer is resistant to immunotherapies due to the immunosuppressive tumor microenvironment. Herein, we investigated a therapeutic approach involving delivery of a short interfering double-stranded RNA (dsRNA), specific to Bcl2, with 5' triphosphate ends, by lipid calcium phosphate nanoparticles, in an orthotopic allograft KPC model of pancreatic cancer. Retinoic acid-inducible gene I (RIG-I)-like receptors can bind to 5' triphosphate dsRNA (ppp dsRNA), a pathogen-associated molecular pattern, producing type I interferon, while Bcl2 silencing can drive apoptosis of cancer cells. Our approach demonstrated a robust enrichment of tumor tissue with therapeutic nanoparticles and enabled a significant tumor growth inhibition, prolonging median overall survival. Nanoparticles encapsulating dual-therapeutic ppp dsRNA allowed strong induction in levels of pro-inflammatory Th1 cytokines, further increasing proportions of CD8+ T cells over regulatory T cells, M1 over M2 macrophages, and decreased levels of immunosuppressive B regulatory and plasma cells in the tumor microenvironment. Thus, these results provide a new immunotherapy approach for pancreatic cancer.


Asunto(s)
Nanopartículas/química , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Apoptosis/fisiología , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Fosfatos de Calcio/química , Proteína 58 DEAD Box/metabolismo , Femenino , Inmunidad Innata/fisiología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo
3.
Nat Commun ; 9(1): 2237, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29884866

RESUMEN

Although great success has been obtained in the clinic, the current immune checkpoint inhibitors still face two challenging problems: low response rate and immune-related adverse effects (irAEs). Here we report the combination of immunogenic chemotherapy and locally expressed PD-L1 trap fusion protein for efficacious and safe cancer immunotherapy. We demonstrate that oxaliplatin (OxP) boosts anti-PD-L1 mAb therapy against murine colorectal cancer. By design of a PD-L1 trap and loading its coding plasmid DNA into a lipid-protamine-DNA nanoparticle, PD-L1 trap is produced transiently and locally in the tumor microenvironment, and synergizes with OxP for tumor inhibition. Significantly, unlike the combination of OxP and anti-PD-L1 mAb, the combination of OxP and PD-L1 trap does not induce obvious Th17 cells accumulation in the spleen, indicating better tolerance and lower tendency to irAEs. The reports here may highlight the potential of applying PD-L1 inhibitor, especially locally expressed PD-L1 trap, in cancer therapy following OxP-based chemotherapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Colorrectales/terapia , Inmunoterapia/métodos , Nanopartículas/química , Animales , Antígeno B7-H1/química , Antígeno B7-H1/genética , Secuencia de Bases , Línea Celular Tumoral , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , ADN/química , ADN/genética , Sinergismo Farmacológico , Femenino , Humanos , Lípidos/química , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nanopartículas/administración & dosificación , Oxaliplatino/administración & dosificación , Protaminas/química , Análisis de Supervivencia
4.
ACS Nano ; 11(9): 8690-8706, 2017 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-28809532

RESUMEN

Pancreatic tumors are known to be resistant to immunotherapy due to the extensive immune suppressive tumor microenvironment (TME). We hypothesized that CXCL12 and PD-L1 are two key molecules controlling the immunosuppressive TME. Fusion proteins, called traps, designed to bind with these two molecules with high affinity (Kd = 4.1 and 0.22 nM, respectively) were manufactured and tested for specific binding with the targets. Plasmid DNA encoding for each trap was formulated in nanoparticles and intravenously injected to mice bearing orthotopic pancreatic cancer. Expression of traps was mainly seen in the tumor, and secondarily, accumulations were primarily in the liver. Combination trap therapy shrunk the tumor and significantly prolonged the host survival. Either trap alone only brought in a partial therapeutic effect. We also found that CXCL12 trap allowed T-cell penetration into the tumor, and PD-L1 trap allowed the infiltrated T-cells to kill the tumor cells. Combo trap therapy also significantly reduced metastasis of the tumor cells to other organs. We conclude that the trap therapy significantly modified the immunosuppressive TME to allow the host immune system to kill the tumor cells. This can be an effective therapy in clinical settings.


Asunto(s)
Antígeno B7-H1/inmunología , Carcinoma Ductal Pancreático/terapia , Quimiocina CXCL12/inmunología , ADN/uso terapéutico , Inmunoterapia/métodos , Animales , Antígeno B7-H1/antagonistas & inhibidores , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Quimiocina CXCL12/antagonistas & inhibidores , Terapia Genética/métodos , Ratones , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Linfocitos T/patología , Escape del Tumor , Microambiente Tumoral
5.
Biomaterials ; 141: 260-271, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28700955

RESUMEN

The ability to generate potent immunotherapies locally and transiently for the treatment of cancers is a promising strategy to improve efficacy and decrease off-target toxicities. Here, we explored an alternative approach for the delivery of immunotherapeutic agents, in which we deliver the pDNA of an engineered PD-L1 trap and/or CXCL12 trap to the nucleus of liver hepatocytes via a lipid calcium phosphate nanoparticle. This strategy greatly increased the concentrations of immunotherapeutic agents in the local tissue, allowing the therapy to inhibit the accumulation of immune suppressive cells and liver metastasis. Furthermore, we find that the lipid calcium phosphate nanoparticles containing the pCXCL12 trap resolved the formation of immune suppressive ectopic lymphoid structures, while the pPD-L1 trap promoted T-cell survival and migration into the liver following vaccination against tumor antigens (>180% increase in survival). This approach showed superior efficacy in the treatment of the liver metastasis compared to free protein immunotherapies. This strategy should be considered as an approach to support liver metastasis therapies as well as for future research interested in manipulating the chemokine/cytokine immune factors within the liver. SIGNIFICANCE: Our approach results in transient liver specific expression of gene immunotherapies with improved efficacy and reduced off-target toxicities over traditional systemically administered immunotherapies. This approach would allow clinicians to manipulate the liver and immune microenvironment to resist cancer invasion, improve organ health, and prolong patient survival.


Asunto(s)
Antígeno B7-H1/genética , Vacunas contra el Cáncer/uso terapéutico , Quimiocina CXCL12/genética , Neoplasias Colorrectales/patología , ADN/uso terapéutico , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/terapia , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/patología , Fosfatos de Calcio/química , ADN/administración & dosificación , ADN/genética , Femenino , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Humanos , Inmunoterapia/métodos , Lípidos/química , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Plásmidos/administración & dosificación , Plásmidos/genética , Plásmidos/uso terapéutico
6.
Vaccine ; 35(19): 2550-2557, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28385609

RESUMEN

The lipid calcium phosphate nanoparticle is a versatile platform capable of encapsulating a wide range of phosphorylated molecules from single nucleotides to pDNA. The use of this platform has shown great success as an immunotherapeutic vaccine carrier, capable of delivering co-encapsulated phosphorylated adjuvants and peptides. Three potent vaccine formulations were investigated for anti-cancer efficacy. The phosphorylated adjuvants, CpG, 2'3'cGAMP, and 5'pppdsRNA were co-encapsulated with a model phosphorylated tumor specific peptide antigen (p-AH1-A5). The anti-cancer efficacy of these adjuvants was assessed using an orthotopic colorectal liver metastasis model based on highly aggressive and metastatic CT-26 FL3 cells implanted into the cecum wall. The results clearly indicate that the RIG-1 ligand, 5'pppdsRNA, co-encapsulated with the p-AH1-A5 peptide antigen greatly reduced the growth rate of the primary colon cancer as well as arrested the establishment of liver metastasis in comparison to the other adjuvant formulations and unvaccinated controls. Further evaluation of the immune cell populations within the primary tumor confirms the ability of the 5'pppdsRNA adjuvant to boost the adaptive CD8+ T-cell population, while not inciting increased populations of immune suppressive cell types such as T-regulatory cells or myeloid derived suppressor cells. Furthermore, to our knowledge this is the first study to investigate the anti-cancer efficacy of a specific RIG-1 receptor ligand, 5'pppdsRNA, alongside more established TLR 9 (CpG) and STING (2'3'cGAMP) adjuvants in a cancer vaccine. The 5'pppdsRNA vaccine formulation can be a potent immunotherapy, especially when combined with agents that remodel the immune suppressive microenvironment of the tumor.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antígenos de Neoplasias/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/terapia , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/terapia , Inmunidad Adaptativa , Adyuvantes Inmunológicos/efectos adversos , Animales , Antígenos de Neoplasias/efectos adversos , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/efectos adversos , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Femenino , Neoplasias Hepáticas/patología , Ratones Endogámicos BALB C , Metástasis de la Neoplasia/terapia , Péptidos/administración & dosificación , Péptidos/efectos adversos , Resultado del Tratamiento
7.
ACS Nano ; 11(5): 4916-4925, 2017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28414916

RESUMEN

Our previous work demonstrated that Wnt16 expression in cisplatin-damaged tumor-associated fibroblasts is a key factor contributing to cisplatin resistance in malignancies. Natural antifibrotic compounds with low toxicities are promising candidates to downregulate Wnt16 expression, improving the antitumor effect of cisplatin nanoparticles. Upon screening several natural chemicals, we found that a dietary flavonoid, quercetin, significantly suppresses Wnt16 expression in activated fibroblasts. To facilitate drug delivery, we have prepared a targeted lipid/calcium/phosphate nanoparticle formulation consisting of a prodrug of quercetin, i.e., quercetin phosphate, with a high loading efficiency (26.6% w/w). This quercetin nanoparticle with a particle size of around 35 nm significantly improved the bioavailability and metabolic stability of the parent quercetin. Quercetin phosphate is released from the nanoparticles and converted back to the parent quercetin under physiological conditions. Following systemic administration of quercetin phosphate nanoparticles, a significant downregulation in Wnt16 expression was observed and further yielded a synergistic antitumor effect with cisplatin nanoparticles in a stroma-rich bladder carcinoma model. The α-SMA-positive fibroblast and collagen within the tumor decreased significantly after combination treatment. This suggests that the remodeling of the tumor microenvironment induced by quercetin plays a critical role in promoting the synergy. Indeed, our data further confirmed that quercetin phosphate alone significantly remodeled the tumor microenvironment and increased the penetration of second-wave nanoparticles into the tumor nests. Collectively, quercetin phosphate nanoparticles may be a safe and effective way to improve therapeutic treatment for desmoplastic tumors.


Asunto(s)
Nanopartículas/química , Quercetina/farmacología , Microambiente Tumoral/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Disponibilidad Biológica , Línea Celular Tumoral , Cisplatino/farmacología , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Lípidos , Ratones , Ratones Desnudos , Células 3T3 NIH , Tamaño de la Partícula , Quercetina/metabolismo , Proteínas Wnt/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
Biochemistry ; 56(2): 421-440, 2017 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-28000448

RESUMEN

DNA-alkylating drugs continue to remain an important weapon in the arsenal against cancers. However, they typically suffer from several shortcomings because of the indiscriminate DNA damage that they cause and their inability to specifically target cancer cells. We have developed a strategy for overcoming the deficiencies in current DNA-alkylating chemotherapy drugs by designing a site-specific DNA-methylating agent that can target cancer cells because of its selective uptake via glucose transporters, which are overexpressed in most cancers. The design features of the molecule, its synthesis, its reactivity with DNA, and its toxicity in human glioblastoma cells are reported here. In this molecule, a glucosamine unit, which can facilitate uptake via glucose transporters, is conjugated to one end of a bispyrrole triamide unit, which is known to bind to the minor groove of DNA at A/T-rich regions. A methyl sulfonate moiety is tethered to the other end of the bispyrrole unit to serve as a DNA-methylating agent. This molecule produces exclusively N3-methyladenine adducts upon reaction with DNA and is an order of magnitude more toxic to treatment resistant human glioblastoma cells than streptozotocin is, a Food and Drug Administration-approved, glycoconjugated DNA-methylating drug. Cellular uptake studies using a fluorescent analogue of our molecule provide evidence of uptake via glucose transporters and localization within the nucleus of cells. These results demonstrate the feasibility of our strategy for developing more potent anticancer chemotherapeutics, while minimizing common side effects resulting from off-target damage.


Asunto(s)
Antineoplásicos Alquilantes/síntesis química , Aductos de ADN/biosíntesis , ADN de Neoplasias/antagonistas & inhibidores , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Glicoconjugados/síntesis química , Neuroglía/efectos de los fármacos , Adenina/análogos & derivados , Adenina/química , Adenina/metabolismo , Alcanosulfonatos/química , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacología , Transporte Biológico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Aductos de ADN/química , Daño del ADN , Metilación de ADN , ADN de Neoplasias/química , ADN de Neoplasias/metabolismo , Expresión Génica , Glucosamina/química , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Glicoconjugados/metabolismo , Glicoconjugados/farmacología , Humanos , Simulación de Dinámica Molecular , Terapia Molecular Dirigida , Neuroglía/metabolismo , Neuroglía/patología , Conformación de Ácido Nucleico , Pirroles/química , Estreptozocina/farmacología
9.
Sci Transl Med ; 8(364): 364ra153, 2016 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-27831902

RESUMEN

The liver is the primary site of metastasis for gastrointestinal cancers and is a location highly susceptible to the establishment of metastasis in numerous other primary cancers, including breast, lung, and pancreatic cancers. The current standard of care typically consists of primary tumor resection and systemic administration of potent but toxic chemotherapeutics, yielding a minimal improvement in the median survival rate. CXCL12, a chemokine, is a key factor for activating the migration/survival pathways of CXCR4+ cancer cells and for recruiting immunosuppressive cells to areas of inflammation. Therefore, reducing CXCL12 concentrations within the liver has the potential to decrease tumor and immunosuppressive cell activation/migration within the liver. However, because of off-target toxicities associated with systemic administration of anti-CXCL12 therapies, transient and liver-specific expression of a CXCL12 trap is necessary. To address this challenge, we developed a lipid calcium phosphate nanoparticle optimized for delivering plasmid DNA, encoding an engineered CXCL12 protein trap, to the nucleus of liver hepatocytes. This pCXCL12-trap formulation yielded transient (4 days) liver-specific expression, which greatly decreased the occurrence of liver metastasis in two aggressive liver metastasis models, including colorectal [CT-26(FL3)] and breast (4T1) cancers. Subsequent studies in an aggressive human colorectal liver metastasis model (HT-29) decreased the establishment of liver metastasis more effectively than did systemic administration of the CXCL12 protein trap and to a level comparable to a high-dose regimen of a potent CXCR4 antagonist (AMD3100).


Asunto(s)
Quimiocina CXCL12/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/secundario , Terapia Molecular Dirigida , Metástasis de la Neoplasia/prevención & control , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , ADN/química , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Inmunosupresores/uso terapéutico , Inflamación , Interferometría , Masculino , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Invasividad Neoplásica , Proteínas Recombinantes/química , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA