Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cancer Immunol Immunother ; 71(2): 433-444, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34218294

RESUMEN

BACKGROUND: Standard care for patients with high-risk myelodysplastic syndrome (MDS) is hypomethylating agents such as azacitidine (AZA), which can induce expression of methylated tumor-associated antigens and therefore potentiate immunotherapeutic targeting. METHOD: In this phase 1 trial, we combined AZA with a therapeutic peptide vaccine targeting antigens encoded from NY-ESO-1, MAGE-A3, PRAME, and WT-1, which have previously been demonstrated to be upregulated by AZA treatment. RESULT: Five patients who had responded to AZA monotherapy were included in the study and treated with the vaccine. The combination therapy showed only few adverse events during the study period, whereof none classified as serious. However, no specific immune responses could be detected using intracellular cytokine staining or ELISpot assays. Minor changes in the phenotypic composition of immune cells and their expression of stimulatory and inhibitory markers were detected. All patients progressed to AML with a mean time to progression from inclusion (TTP) of 5.2 months (range 2.8 to 7.6). Mean survival was 18.1 months (range 10.9 to 30.6) from MDS diagnosis and 11.3 months (range 4.3 to 22.2) from inclusion. Sequencing of bone marrow showed clonal expansion of malignant cells, as well as appearance of novel mutations. CONCLUSION: The patients progressed to AML with an average time of only five months after initiating the combination therapy. This may be unrelated to the experimental treatment, but the trial was terminated early as there was no sign of clinical benefit or immunological response. Why the manuscript is especially interesting This study is the first to exploit the potential synergistic effects of combining a multi-peptide cancer vaccine with epigenetic therapy in MDS. Although our results are negative, they emphasize challenges to induce immune reactivity in patients with high-risk MDS.


Asunto(s)
Antígenos de Neoplasias/inmunología , Azacitidina/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Epigénesis Genética , Síndromes Mielodisplásicos/tratamiento farmacológico , Anciano , Antimetabolitos Antineoplásicos/uso terapéutico , Azacitidina/farmacocinética , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacocinética , Quimioterapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Masculino , Dosis Máxima Tolerada , Síndromes Mielodisplásicos/inmunología , Síndromes Mielodisplásicos/patología , Pronóstico , Distribución Tisular
2.
Nat Commun ; 11(1): 5828, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33203928

RESUMEN

Stimuli-sensitive nanomaterials with cooperative response are capable of converting subtle and gradual biological variations into robust outputs to improve the precision of diagnostic or therapeutic outcomes. In this study, we report the design, synthesis and characterization of a series of degradable ultra-pH sensitive (dUPS) polymers that amplify small acidic pH changes to efficacious therapeutic outputs. A hydrolytically active polycarbonate backbone is used to construct the polymer with pH-dependent degradation kinetics. One dUPS polymer, PSC7A, can achieve activation of the stimulator of interferon genes and antigen delivery upon endosomal pH activation, leading to T cell-mediated antitumor immunity. While a non-degradable UPS polymer induces granulomatous inflammation that persists over months at the injection site, degradable PSC7A primes a transient acute inflammatory response followed by polymer degradation and complete tissue healing. The improved therapeutic window of the dUPS polymers opens up opportunities in pH-targeted drug and protein therapy.


Asunto(s)
Vacunas contra el Cáncer/farmacocinética , Nanopartículas/química , Cemento de Policarboxilato/química , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacocinética , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/efectos adversos , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Concentración de Iones de Hidrógeno , Inmunoterapia/métodos , Espectroscopía de Resonancia Magnética , Melanoma/terapia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Nanopartículas/administración & dosificación , Nanopartículas/efectos adversos , Nanopartículas/uso terapéutico , Cemento de Policarboxilato/metabolismo , Polietilenglicoles/química , Polímeros/síntesis química , Polímeros/química , Polímeros/metabolismo , Linfocitos T/inmunología
3.
Mol Pharm ; 17(12): 4603-4615, 2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33175556

RESUMEN

Cancer nanovaccines have been widely explored to enhance immunotherapy efficiency, in which the significant irritation of antigen-specific cytotoxic T cells (CTLs) is the critical point. In this study, we developed a pH and reduction dual-sensitive nanovaccine (PMSN@OVA-MPN) composed of two parts. The inner part was made up of polyethyleneimine (PEI)-modified mesoporous silica nanoparticles (MSNs) loaded with model antigen ovalbumin (OVA) and the outer part was made up of disulfide bond-involved metal-phenolic networks (MPNs) as a protective corona. In vitro release experiments proved that PMSN@OVA-MPN could intelligently release OVA in the presence of reductive glutathione, but not in neutral phosphate-buffered saline (PBS). Moreover, in vitro cell assays indicated that the nanovaccine promoted not only the OVA uptake efficiency by DC2.4 cells but also antigen lysosome escape due to the proton sponge effect of PEI. Furthermore, in vivo animal experiments indicated that PMSN@OVA-MPN induced a large tumor-specific cellular immune response so as to effectively inhibit the growth of an existing tumor. Finally, the immune memory effect caused by the nanovaccine afforded conspicuous prophylaxis efficacy in neonatal tumors. Hence, the multifunctional vaccine delivery system prepared in this work exhibits a great application potential in cancer immunotherapy and offers a platform for the development of nanovaccines.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Portadores de Fármacos/química , Inmunoterapia Activa/métodos , Nanosferas/química , Neoplasias/terapia , Animales , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacocinética , Línea Celular Tumoral , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Composición de Medicamentos/métodos , Liberación de Fármacos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Inmunogenicidad Vacunal , Memoria Inmunológica , Estructuras Metalorgánicas/química , Ratones , Neoplasias/inmunología , Polietileneimina/química , Dióxido de Silicio/química
4.
Mol Pharm ; 17(10): 3845-3856, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32902989

RESUMEN

Ionic liquids (ILs) attract significant attention as novel solvents for drug delivery systems because of their ability to solubilize poorly soluble drugs and tune the physiological properties of active pharmaceutical ingredients. For the next generation of IL-based drug delivery systems, biocompatibility is a high priority. In the current study, choline-fatty acids ([Cho][FA]) were used as a biocompatible IL to mediate the dissolution of a water-soluble antigen peptide in an oil-based skin penetration enhancer. Among the candidate fatty acids (C8, C10, C12, C14, C16, C18:0, and C18:1), C18:1 was selected because of its low cytotoxicity and mediation of skin permeability for an antigen peptide. Using IL[Cho][C18:1] and an oil-based penetration enhancer, the flux of transdermal delivery of the peptide increased 28-fold compared with delivery using an aqueous vehicle. Furthermore, the IL-mediated transcutaneous vaccination succeeded in suppressing tumor growth in vivo compared to injection. The skin irritation produced by this formulation was tested using an in vitro 3D constructed skin tissue model and an in vivo histological study, which concluded that the formulation did not cause skin irritation. The results suggest that biocompatible IL-mediated dissolution in an oil-based skin penetration enhancer is a promising strategy for transdermal drug delivery.


Asunto(s)
Antígenos de Neoplasias/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Portadores de Fármacos/química , Líquidos Iónicos/química , Neoplasias/prevención & control , Administración Cutánea , Animales , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/farmacocinética , Línea Celular Tumoral/trasplante , Colina/química , Modelos Animales de Enfermedad , Ácidos Grasos/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Neoplasias/inmunología , Permeabilidad , Piel , Absorción Cutánea , Solventes/química , Vacunación/métodos , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/química , Vacunas de Subunidad/farmacocinética
5.
Adv Drug Deliv Rev ; 161-162: 145-160, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32827558

RESUMEN

Vaccines hold enormous potential in cancer immunotherapy by stimulating the body's immune response; unfortunately, the clinical response rates of cancer vaccines are less than 30%. Nanovaccines show the potential to enhance the treatment efficacy of conventional vaccines due to their unique properties, such as efficient co-delivery of cocktail to the secondary lymphatic system, high tumor accumulation and penetration, and customizable delivery of antigens and adjuvants. Meanwhile, the non-invasive visualization of vaccines after their delivery can yield information about in vivo distribution and performance, and aid in their subsequent optimization and translational studies. In this review, we summarize the strategies for the spatiotemporal visualization of nanovaccines in lymph nodes, including whole-body in vivo imaging, intravital organ/cell imaging, and ex vivo tissue/cell imaging. The application of imaging modalities in nanovaccine development is discussed. Moreover, strategies to achieve different combinations of imaging modalities are proposed.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/farmacología , Diagnóstico por Imagen/métodos , Inmunoterapia/métodos , Ganglios Linfáticos/inmunología , Adyuvantes Inmunológicos , Animales , Células Presentadoras de Antígenos/metabolismo , Antígenos de Neoplasias/biosíntesis , Vacunas contra el Cáncer/farmacocinética , Células Dendríticas/metabolismo , Humanos , Ganglios Linfáticos/efectos de los fármacos , Imagen Multimodal , Nanopartículas , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/metabolismo
6.
J Microencapsul ; 37(1): 14-28, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31718364

RESUMEN

Aim: MUC-1 lipopeptide vaccine exhibited immense potential in the treatment of non-small cell lung cancer (NSCLC) in both preclinical and clinical trials. However, it lacks triggering of mucosal immunity at the site of action. Therefore, in present investigation, MUC-1 peptide-loaded poly(lactide-co-glycolide) nanoparticles (MUC-1 peptide-PLGA-NPs) and MUC-1 peptide-loaded poly(lactide-co-glycolide) non-aggregated nanoparticles (MUC-1 peptide-PLGA-NA-NPs) using Central Composite Design (CCD) were customised.Methods and Results: The mean particle size of MUC-1 peptide PLGA-NPs was estimated to be 176.7 ± 32.7 nm, significantly (p < 0.05) higher than 100.3 ± 24.3 nm of MUC-1 peptide-PLGA-NA-NPs. Furthermore, integrity and stability of MUC-1 were maintained in MUC-1 peptide PLGA-NA-NPs. MUC-1 peptide-PLGA-NA-NPs exhibited augmented cellular uptake in mouse RAW264.7 macrophages preferably by clathrin-mediated endocytosis pathway as compared to phagocytosis followed by MUC-1-peptide PLGA-NPs owing to size ≤100 nm, and spherical shape.Conclusion: MUC-1 peptide-PLGA-NA-NPs may be a potential candidate to study antitumor potential in xenograft model of NSCLC through inhalation route of administration.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Portadores de Fármacos/química , Mucina-1/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Animales , Vacunas contra el Cáncer/farmacocinética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Endocitosis , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Macrófagos/inmunología , Ratones , Nanopartículas/química , Fagocitosis , Células RAW 264.7
7.
Nanoscale ; 11(45): 21782-21789, 2019 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-31709434

RESUMEN

For efficient cancer vaccines, the antitumor function largely relies on cytotoxic T cells, whose activation can be effectively induced via antigen-encoding mRNA, making mRNA-based cancer vaccines an attractive approach for personalized cancer therapy. While the liposome-based delivery system enables the systemic delivery and transfection of mRNA, incorporating an adjuvant that is non-lipid like remains challenging, although the co-delivery of mRNA (antigen) and effective adjuvant is key to the activation of the cytotoxic T cells. This is because the presence of an adjuvant is important for dendritic cell maturation-another necessity for cytotoxic T cell activation. In the present work, we designed a poly (lactic-co-glycolic acid) (PLGA)-core/lipid-shell hybrid nanoparticle carrier for the co-delivery of mRNA and gardiquimod (adjuvant that cannot be incorporated into the lipid shell). We demonstrated in the present work that the co-delivery of mRNA and gardiquimod led to the effective antigen expression and DC maturation in vitro. The intravenous administration of the hybrid nanovaccine resulted in the enrichment of mRNA expression in the spleen and a strong immune response in vivo. The simultaneous delivery of the antigen and adjuvant both spatially and temporally via the core/shell nanoparticle carrier is found to be beneficial for tumor growth inhibition.


Asunto(s)
Adyuvantes Inmunológicos , Aminoquinolinas , Vacunas contra el Cáncer , Imidazoles , Nanopartículas , Neoplasias Experimentales , ARN Neoplásico , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacocinética , Adyuvantes Inmunológicos/farmacología , Aminoquinolinas/química , Aminoquinolinas/farmacocinética , Aminoquinolinas/farmacología , Animales , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/inmunología , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/farmacocinética , Vacunas contra el Cáncer/farmacología , Línea Celular Tumoral , Células Dendríticas/inmunología , Células Dendríticas/patología , Imidazoles/química , Imidazoles/farmacocinética , Imidazoles/farmacología , Liposomas , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Nanopartículas/uso terapéutico , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Ácido Poliglicólico/química , Ácido Poliglicólico/farmacocinética , Ácido Poliglicólico/farmacología , ARN Neoplásico/química , ARN Neoplásico/farmacocinética , ARN Neoplásico/farmacología , Linfocitos T Citotóxicos/inmunología
8.
Nat Biotechnol ; 37(10): 1174-1185, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31570898

RESUMEN

Therapeutic messenger RNA vaccines enable delivery of whole antigens, which can be advantageous over peptide vaccines. However, optimal efficacy requires both intracellular delivery, to allow antigen translation, and appropriate immune activation. Here, we developed a combinatorial library of ionizable lipid-like materials to identify mRNA delivery vehicles that facilitate mRNA delivery in vivo and provide potent and specific immune activation. Using a three-dimensional multi-component reaction system, we synthesized and evaluated the vaccine potential of over 1,000 lipid formulations. The top candidate formulations induced a robust immune response, and were able to inhibit tumor growth and prolong survival in melanoma and human papillomavirus E7 in vivo tumor models. The top-performing lipids share a common structure: an unsaturated lipid tail, a dihydroimidazole linker and cyclic amine head groups. These formulations induce antigen-presenting cell maturation via the intracellular stimulator of interferon genes (STING) pathway, rather than through Toll-like receptors, and result in limited systemic cytokine expression and enhanced anti-tumor efficacy.


Asunto(s)
Antineoplásicos , Vacunas contra el Cáncer , Lípidos/farmacocinética , Vehículos Farmacéuticos , ARN Mensajero , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/farmacocinética , Vacunas contra el Cáncer/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Femenino , Células HeLa , Humanos , Interferones/genética , Interferones/metabolismo , Ratones , Ratones Endogámicos C57BL , Vehículos Farmacéuticos/química , Vehículos Farmacéuticos/farmacocinética , ARN Mensajero/genética , ARN Mensajero/farmacocinética , ARN Mensajero/farmacología , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas
9.
Drug Deliv Transl Res ; 9(6): 1095-1105, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31228097

RESUMEN

Poly(lactic-co-glycolic) acid (PLGA) has been successfully used in drug delivery and biomaterial applications, but very little attention has been directed towards the potential in vivo effects of peptide-loaded PLGA nanoparticles (NPs), specifically the potency of intravenous (IV) STEAP peptide-loaded PLGA-NP (nanovaccine) dosing and whether STEAP-specific CD8+ T cells directly play a key role in tumor inhibition. To address these concerns, syngeneic prostate cancer mouse models were established and treated with either mSTEAP peptide emulsified in incomplete Freund's adjuvant (IFA) via subcutaneous (SC) injection or mSTEAP peptide nanovaccine containing the same amount of peptide via IV or SC injection. Meanwhile, mice were treated with either CD8b mAb followed by nanovaccine treatment, free mSTEAP peptide, or empty PLGA-NPs. Immune responses in these mice were examined using cytotoxicity assays at 14 days after treatment. Tumor size and survival in various treatment groups were measured and monitored. The results demonstrated that mSTEAP peptide nanovaccine resulted in tumor inhibition by eliciting a significantly stronger CD8+ T cell immune response when compared with the controls. Moreover, the survival periods of mice treated with mSTEAP nanovaccine were significantly longer than those of mice treated with mSTEAP peptide emulsified in IFA or the treatment controls. Additionally, it was observed that the peptide nanovaccine was mainly distributed in the mouse liver and lungs after IV injection. These findings suggest that the peptide nanovaccine is a promising immunotherapeutic approach and offers a new opportunity for prostate cancer therapies.


Asunto(s)
Antígenos de Neoplasias/administración & dosificación , Linfocitos T CD8-positivos/efectos de los fármacos , Vacunas contra el Cáncer/administración & dosificación , Nanopartículas/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Antígenos de Neoplasias/farmacología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/farmacocinética , Línea Celular Tumoral , Hígado/metabolismo , Pulmón/metabolismo , Masculino , Ratones Endogámicos C57BL , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacocinética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo
10.
Biomed Microdevices ; 21(2): 39, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30949852

RESUMEN

Effective migration of dendritic cells into the lymphatic system organs is the prerequisite for a functional dendritic cell vaccine. We have previously developed a porous silicon microparticle (PSM)-based therapeutic dendritic cell vaccine (Nano-DC vaccine) where PSM serves both as the vehicle for antigen peptides and an adjuvant. Here, we analyzed parameters that determined dendritic cell uptake of PSM particles and Nano-DC vaccine accumulation in lymphatic tissues in a murine model of HER2-positive breast cancer. Our study revealed a positive correlation between sphericity of the PSM particles and their cellular uptake by circulating dendritic cells. In addition, the intravenously administered vaccines accumulated more in the spleens and inguinal lymph nodes, while the intradermally inoculated vaccines got enriched in the popliteal lymph nodes. Furthermore, mice with large tumors received more vaccines in the lymph nodes than those with small to medium size tumors. Information from this study will provide guidance on design and optimization of future therapeutic cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/metabolismo , Células Dendríticas/metabolismo , Nanomedicina , Animales , Transporte Biológico , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacocinética , Línea Celular Tumoral , Células Dendríticas/inmunología , Ratones , Microesferas , Fagocitos/inmunología , Silicio/química , Distribución Tisular , Carga Tumoral/inmunología
11.
Nat Commun ; 10(1): 1899, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31015397

RESUMEN

Nanoparticles can potentially stimulate tumour microenvironments to elicit antitumour immunity. Herein, we demonstrate effective immunotherapy of colorectal cancer via systemic delivery of an immunostimulatory chemotherapeutic combination in nanoscale coordination polymer (NCP) core-shell particles. Oxaliplatin and dihydroartemesinin have contrasting physicochemical properties but strong synergy in reactive oxygen species (ROS) generation and anticancer activity. The combined ROS generation is harnessed for immune activation to synergize with an anti-PD-L1 antibody for the treatment of murine colorectal cancer tumours. The favourable biodistribution and tumour uptake of NCPs and the absence of peripheral neuropathy allow for repeated dosing to afford 100% tumour eradication. The involvement of innate and adaptive immune systems elicit strong and long lasting antitumour immunity which prevents tumour formation when cured mice are challenged with cancer cells. The intrinsically biodegradable, well tolerated, and systemically available immunostimulatory NCP promises to enter clinical testing as an immunotherapy against colorectal cancer.


Asunto(s)
Adenocarcinoma/terapia , Vacunas contra el Cáncer/farmacología , Neoplasias Colorrectales/terapia , Factores Inmunológicos/farmacología , Nanopartículas/administración & dosificación , Polímeros/administración & dosificación , Inmunidad Adaptativa/efectos de los fármacos , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Adenocarcinoma/mortalidad , Animales , Anticuerpos Neutralizantes/farmacología , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Artemisininas/farmacocinética , Artemisininas/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Vacunas contra el Cáncer/síntesis química , Vacunas contra el Cáncer/farmacocinética , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/mortalidad , Composición de Medicamentos/métodos , Humanos , Inmunidad Innata/efectos de los fármacos , Factores Inmunológicos/síntesis química , Factores Inmunológicos/farmacocinética , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Trasplante de Neoplasias , Oxaliplatino/farmacocinética , Oxaliplatino/farmacología , Polímeros/síntesis química , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/agonistas , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Análisis de Supervivencia , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
12.
Nano Lett ; 19(7): 4237-4249, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-30868883

RESUMEN

Among approaches of current cancer immunotherapy, a dendritic cell (DC)-targeted vaccine based on nanotechnology could be a promising way to efficiently induce potent immune responses. To enhance DC targeting and vaccine efficiency, we included imiquimod (IMQ), a toll-like receptor 7/8 (TLR 7/8) agonist, and monophosphoryl lipid A (MPLA), a TLR4 agonist, to synthesize lipid-polymer hybrid nanoparticles using PCL-PEG-PCL and DOTAP (IMNPs) as well as DSPE-PEG-mannose (MAN-IMNPS). The spatiotemporal delivery of MPLA (within the outer lipid layer) to extracellular TLR4 and IMQ (in the hydrophobic core of NPs) to intracellular TLR7/8 can activate DCs synergistically to improve vaccine efficacy. Ovalbumin (OVA) as a model antigen was readily absorbed by positively charged DOTAP and showed a quick release in vitro. Our results demonstrated that this novel nanovaccine enhanced cellular uptake, cytokine production, and maturation of DCs. Compared with the quick metabolism of free OVA-agonists, the depot effect of OVA-IMNPs was observed, whereas MAN-OVA-IMNPs promoted trafficking to secondary lymphoid organs. After immunization with a subcutaneous injection, the nanovaccine, especially MAN-OVA-IMNPs, induced more antigen-specific CD8+ T cells, greater lymphocyte activation, stronger cross-presentation, and more generation of memory T cells, antibody, IFN-γ, and granzyme B. Prophylactic vaccination of MAN-OVA-IMNPs significantly delayed tumor development and prolonged the survival in mice. The therapeutic tumor challenge indicated that MAN-OVA-IMNPs prohibited tumor progression more efficiently than other formulations, and the combination with an immune checkpoint blockade further enhanced antitumor effects. Hence, the DC-targeted vaccine codelivery with IMQ and MPLA adjuvants by hybrid cationic nanoparticles in a spatiotemporal manner is a promising multifunctional antigen delivery system in cancer immunotherapy.


Asunto(s)
Antígenos de Neoplasias , Vacunas contra el Cáncer , Células Dendríticas/inmunología , Sistemas de Liberación de Medicamentos , Imiquimod , Inmunoterapia , Lípido A/análogos & derivados , Nanopartículas , Neoplasias Experimentales , Receptores Toll-Like/agonistas , Animales , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/farmacología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacocinética , Vacunas contra el Cáncer/farmacología , Células Dendríticas/patología , Imiquimod/inmunología , Imiquimod/farmacocinética , Imiquimod/farmacología , Lípido A/inmunología , Lípido A/farmacocinética , Lípido A/farmacología , Ratones , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Receptores Toll-Like/inmunología
13.
ACS Appl Mater Interfaces ; 11(5): 4876-4888, 2019 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-30628437

RESUMEN

Codelivery nanovaccines of antigens and adjuvants have achieved positive therapy for cancer immunotherapy. The insufficient immunogenicity of these vaccines leads to the difficulty of eliciting robust immune effects for immune clearance due to the inadequate loading efficiency, complex preparation processes, low safety concerns, and weak immune responses. Herein, a visible codelivery nanovaccine of an antigen and adjuvant based on self-cross-linked antigen nanoparticles (ovalbumin nanoparticles (ONPs)) combined with the adjuvant (CpG) for cancer immunotherapy was prepared using antigens themselves as carriers. ONPs not only provide sufficient antigens for continuous simulation of the immune response with high antigen loading efficiency but also serve as natural carriers of CpG. In vitro and in vivo experiments proved that ONPs-CpG can elicit a robust immune response including DC maturity, T cell activation, and IFN-γ production. ONPs-CpG induced strong tumor-specific immunity and exhibited remarkable antitumor immunotherapy effects in vivo using mouse models of lymphoma. Furthermore, to perform the precise vaccine delivery, the dual fluorescent codelivery nanovaccine was monitored in real time in vivo by the visible imaging method. With regard to migration tracking, fluorescence imaging allowed for both high resolution and sensitivity of visible detection based on the fluorescence of ONPs and CpG. The multifunctional nanovaccine could function as a robust platform for cancer immunotherapy and a visible system for antigen-adjuvant tracking.


Asunto(s)
Adyuvantes Inmunológicos/química , Vacunas contra el Cáncer , Sistemas de Liberación de Medicamentos/métodos , Inmunoterapia/métodos , Nanopartículas/química , Animales , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacocinética , Células Cultivadas , Fosfatos de Dinucleósidos/química , Fosfatos de Dinucleósidos/inmunología , Modelos Animales de Enfermedad , Colorantes Fluorescentes/química , Linfoma/diagnóstico por imagen , Linfoma/inmunología , Linfoma/terapia , Ratones , Nanopartículas/metabolismo , Imagen Óptica/métodos , Ovalbúmina/química , Ovalbúmina/inmunología , Tamaño de la Partícula
14.
Acta Biomater ; 76: 193-207, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29940370

RESUMEN

α-Galactosylceramide (GalCer) is a glycolipid widely known as an activator of Natural killer T (NKT) cells, constituting a promising adjuvant against cancer, including melanoma. However, limited clinical outcomes have been obtained so far. This study evaluated the synergy between GalCer and major histocompatibility complex (MHC) class I and MHC class II melanoma-associated peptide antigens and the Toll-Like Receptor (TLR) ligands CpG and monophosphoryl lipid A (MPLA), which we intended to maximize following their co-delivery by a nanoparticle (NP). This is expected to improve GalCer capture by dendritic cells (DCs) and subsequent presentation to NKT cells, simultaneously inducing an anti-tumor specific T-cell mediated immunity. The combination of GalCer with melanoma peptides and TLR ligands successfully restrained tumor growth. The tumor volume in these animals was 5-fold lower than the ones presented by mice immunized with NPs not containing GalCer. However, tumor growth was controlled at similar levels by GalCer entrapped or in its soluble form, when mixed with antigens and TLR ligands. Those two groups showed an improved infiltration of T lymphocytes into the tumor, but only GalCer-loaded nano-vaccine induced a prominent and enhanced infiltration of NKT and NK cells. In addition, splenocytes of these animals secreted levels of IFN-γ and IL-4 at least 1.5-fold and 2-fold higher, respectively, than those treated with the mixture of antigens and adjuvants in solution. Overall, the combined delivery of the NKT agonist with TLR ligands and melanoma antigens via this multivalent nano-vaccine displayed a synergistic anti-tumor immune-mediated efficacy in B16F10 melanoma mouse model. STATEMENT OF SIGNIFICANCE: Combination of α-galactosylceramide (GalCer), a Natural Killer T (NKT) cell agonist, with melanoma-associated antigens presented by MHC class I (Melan-A:26) and MHC class II (gp100:44) molecules, and Toll-like Receptor (TLR) ligands (MPLA and CpG), within nanoparticle matrix induced a prominent anti-tumor immune response able to restrict melanoma growth. An enhanced infiltration of NKT and NK cells into tumor site was only achieved when the combination GalCer, antigens and TLR ligands were co-delivered by the nanovaccine.


Asunto(s)
Vacunas contra el Cáncer , Galactosilceramidas , Inmunidad Celular/efectos de los fármacos , Melanoma Experimental/terapia , Nanopartículas , Péptidos , Animales , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/farmacocinética , Vacunas contra el Cáncer/farmacología , Línea Celular Tumoral , Células Dendríticas/inmunología , Células Dendríticas/patología , Galactosilceramidas/química , Galactosilceramidas/farmacocinética , Galactosilceramidas/farmacología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Masculino , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , Nanopartículas/uso terapéutico , Péptidos/química , Péptidos/farmacocinética , Péptidos/farmacología , Linfocitos T/inmunología , Linfocitos T/patología , Receptores Toll-Like/inmunología
15.
Expert Opin Biol Ther ; 18(7): 829-835, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29936901

RESUMEN

INTRODUCTION: Lung cancer represents the most common cause of cancer death worldwide. While the prognosis remains poor, immunotherapy is giving a positive impact on survival. Cancer vaccines represent a form of active immunotherapy that historically has given modest results in terms of efficacy. The overexpression of the EGFR by tumor cells was reported in more than half of cases of lung cancer, representing a mechanism of cancerogenesis. CIMAvax-EGF, a therapeutic vaccine for non-small cell lung cancer (NSCLC) developed in Cuba, consists of a human recombinant EGF able to induce antibodies against the autologous EGF, resulting in serum EGF withdrawal and lower EGF-EGFR interaction. Area covered: We critically reviewed the existing literature about CIMAvax-EGF, from the Pilot studies to the efficacy controlled studies. We also overviewed the ongoing trials. Expert opinion: CIMAvax-EGF demonstrated to be safe and immunogenic. In a phase III randomized study CIMAvax-EGF, used as a switch maintenance treatment after platinum-based chemotherapy, did not significantly improve survival. Current data are not sufficient to recommend CIMAvax-EGF as a treatment option for advanced stage NSCLC. Further studies, conducted in a context of worldwide standardized clinical practice, are needed to better define if a subpopulation of patients can benefit from the vaccination.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/terapia , Animales , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacocinética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Evaluación de Medicamentos , Humanos , Inmunoterapia Activa/métodos , Neoplasias Pulmonares/inmunología , Vacunación/métodos
16.
Bioconjug Chem ; 29(3): 776-785, 2018 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-29436221

RESUMEN

Peptide subunit vaccines are desirable because they increase control over the immune response and safety of the vaccine by reducing the risk of off-target responses to molecules other than the target antigen. The immunogenicity of most peptides, however, is low. Peptide nanoclusters (PNC) are proposed as a subunit peptide vaccine delivery system made completely of cross-linked peptide antigen that could improve the immunogenicity of a peptide vaccine. Proof of concept is demonstrated with oncofetal antigen (OFA), an immature laminin receptor protein expressed by many hematologic cancer cells but not by healthy cells. Peptide epitopes from this protein, called OFA 1, 2, and 3, were synthesized into PNC as a potential cancer peptide vaccine delivery system. PNC were formed by desolvation and stabilized with disulfide bonds using a trithiol cross-linker. Cysteines were added to the C-terminus of each peptide to assist in this cross-linking step, denoted OFA 1C, 2C, and 3C PNC. OFA 2C was found to form the smallest PNC, 148 ± 15 nm in diameter and stable in solution. This size is in the range where particles are readily internalized by dendritic cells (DCs) and may also passively diffuse to regional lymph nodes. OFA 2C PNC and soluble OFA 2C were internalized similarly by DCs in vitro, but only PNC resulted in significant peptide presentation by DCs. This indicates the potential for PNC to improve immune activation against this antigen. Additionally, PNC displayed higher retention at the intradermal injection site in vivo than soluble peptide, allowing more time to interact with DCs in an area of increased DC activity. While offering traditional nanoparticle benefits such as increased DC recognition, slower diffusion, and potential for multivalent cellular interactions, PNC also maximize antigen delivered per particle while minimizing off-target material delivery because the antigens are the main building blocks of the particle. With these properties, PNC are a delivery system with potential to increase peptide subunit vaccine immunogenicity for OFA and other peptide antigens.


Asunto(s)
Antígenos de Neoplasias/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Péptidos/administración & dosificación , Animales , Antígenos de Neoplasias/química , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacocinética , Línea Celular , Reactivos de Enlaces Cruzados/química , Células Dendríticas/inmunología , Humanos , Ratones , Neoplasias/inmunología , Neoplasias/prevención & control , Péptidos/química , Péptidos/inmunología , Péptidos/farmacocinética , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/química , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/farmacocinética
17.
Bioconjug Chem ; 28(2): 565-573, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27951636

RESUMEN

Cancer vaccine has the ability to directly eradicate tumor cells by creating and activating cytotoxic T lymphocytes (CTLs). To achieve efficient CTL activity and to induce Th1 responses, it is essential to administer an appropriate adjuvant as well as an antigen. CpG-ODN is known as a ligand of Toll-like receptor 9 (TLR9) and strongly induces Th1 responses. In our previous study, we developed a CpG-ODN delivery system by use of the formation of complexes between ODN and a ß-glucan SPG, denoted as CpG/SPG, and demonstrated that CpG/SPG induces high Th1 responses. In this study, we created a nanogel made from CpG/SPG complexes through DNA-DNA hybridization (cross-linked (CL)-CpG). Immunization with CL-CpG induced much stronger antigen-specific Th1 responses in combination with the antigenic protein ovalbumin (OVA) than that with CpG/SPG. Mice preimmunized with CL-CpG and OVA exhibited a long delay in tumor growth and an improved survival rate after tumor inoculation. These immune inductions can be attributed to the improvement of cellular uptake by the combination of increased size and the cluster effect of the ß-glucan recognition site in the nanogel structure. In other words, the particle nature of CL-CpG, instead of the semiflexible rod conformation of CpG/SPG, enhanced the efficacy of a cancer vaccine. The present results indicate that CL-CpG can be used as a potent vaccine adjuvant for the treatment of cancers and infectious diseases.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Geles/química , Neoplasias/prevención & control , Oligodesoxirribonucleótidos/administración & dosificación , beta-Glucanos/química , Adyuvantes Inmunológicos/farmacocinética , Adyuvantes Inmunológicos/uso terapéutico , Animales , Vacunas contra el Cáncer/farmacocinética , Vacunas contra el Cáncer/uso terapéutico , Reactivos de Enlaces Cruzados/química , Citocinas/inmunología , Inmunización/métodos , Ratones , Neoplasias/inmunología , Oligodesoxirribonucleótidos/farmacocinética , Oligodesoxirribonucleótidos/uso terapéutico , Ovalbúmina/inmunología , Células RAW 264.7 , Receptor Toll-Like 9/inmunología
18.
Semin Oncol ; 43(4): 501-13, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27663482

RESUMEN

Immunotherapy has become a major modality of cancer treatment, with multiple new classes of immunotherapeutics recently entering the clinic and obtaining market approval from regulatory agencies. While the promise of these therapies is great, so is the number of possible combinations not only with each other but also with small molecule therapeutics. Furthermore, the observation of unusual dose-response relationships suggests a critical dependency of drug effectiveness on the dosage regimen (dose and schedule). Clinical pharmacodynamic (PD) biomarkers will be useful endpoints for confirming drug mechanism of action, evaluating combination therapies for synergy or antagonism, and identifying optimal dosage regimens. In contrast to conventional PD in which drug action occurs entirely within a single target cell (ie, is self-contained within the malignant cell), immunotherapy involves a complex mechanism of action with sequential steps that propagate through multiple cell types, both normal and malignant. Its intercellular pharmacology begins with molecular target engagement either on an immune effector cell or a malignant cell, followed by stimulatory biochemical and biological signals in immune effector cells, and then finally ends with activation of cell death mechanisms in malignant cells lying within a certain distance from the activated effector cells (immune cell-tumor cell proximity). Evaluating such "trans-cellular pharmacology," in which different steps of drug action are distributed across multiple cell types, requires novel microscopy and image analysis tools capable of quantifying PD-biomarker responses, mapping the responses onto the cellular geography of the tumor using phenotypic biomarkers to identify specific cell types, and finally analyzing the spatial relationships between biomarkers in the context of each cell's biological role. We have termed this form of nearest neighbor image analysis of drug action "proximity PD microscopy," to indicate the importance of the location of the PD-biomarker response within the cellular landscape of a tumor specimen. We discuss herein the major modes of immunotherapy, and lay out a blueprint for using PD assessment to optimize dosage regimens of single agents and guide development of combination immunotherapy regimens, using PD1/PD-L1 immune checkpoint inhibition as a case study.


Asunto(s)
Biomarcadores de Tumor/análisis , Vacunas contra el Cáncer/farmacocinética , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Anticuerpos Monoclonales/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Biomarcadores de Tumor/inmunología , Vacunas contra el Cáncer/uso terapéutico , Terapia Combinada , Citocinas/inmunología , Humanos , Inmunidad Humoral , Terapia Molecular Dirigida/métodos , Neoplasias/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Resultado del Tratamiento
19.
Int J Nanomedicine ; 11: 2417-29, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27313455

RESUMEN

BACKGROUND: Therapeutic human papillomavirus (HPV) vaccines are currently being developed. However, no therapeutic efficacy has been achieved in clinical trials for the treatment of cervical intraepithelial neoplasia or cancer. One of the important issues in increasing vaccine efficacy is determining the best way to enhance tumor antigen-specific cellular immune responses. This study aimed to explore the virus-like particles (VLPs) of hepatitis B core antigen (HBcAg) as potential therapeutic vaccine carriers and to assess its immunological characteristics. METHODS: Chimeric VLPs presenting a HPV 16 cytotoxic T lymphocytes epitope E749-57 (amino acid 49-57 of the E7 protein) were prepared using recombinant genes. C57BL/6 mice were immunized with VLPs and grafted with tumor cells TC-1 which is an E7-expressing tumorigenic cell line. The dynamic tumor growth was monitored and anti-tumor immune responses were investigated. RESULTS: Using a preventive strategy, immunization with VLPs resulted in nearly complete suppression of tumor growth. In treatment studies, VLP immunization significantly suppressed the tumor progression in mice carrying 2-3 mm tumors and in those bearing even larger tumors with diameters up to 8-9 mm. The VLP structure was shown to be important to induce vigorous antitumor immunity and effects. In immunized mice, enhanced E749-57-specific cellular immune responses were evidenced by increased interferon (IFN)-γ expression and decreased interleukin (IL)-4 expression in splenic lymphocytes, as well as an elevated number of effector cells expressing IFN-γ in response to the in vitro stimulation of the specific peptide E749-57. In addition, effective immune memory after VLP immunization was maintained for at least 16 weeks, preventing significant tumor growth after subsequent TC-1 challenge. CONCLUSION: While VLPs were highly immunogenic in stimulating humoral immunity, our results strongly indicated that VLPs, such as HBcAg particles, might also be potent therapeutic vaccine carriers to elicit robust cellular immune responses, even in the immunosuppressive microenvironment of a tumor.


Asunto(s)
Vacunas contra el Cáncer/farmacocinética , Antígenos del Núcleo de la Hepatitis B/inmunología , Papillomavirus Humano 16/inmunología , Vacunas contra Papillomavirus/inmunología , Animales , Antígenos de Neoplasias , Vacunas contra el Cáncer/inmunología , Epítopos/genética , Epítopos/inmunología , Femenino , Antígenos del Núcleo de la Hepatitis B/genética , Humanos , Inmunidad Celular , Ratones Endogámicos C57BL , Vacunas contra Papillomavirus/genética , Linfocitos T Citotóxicos/inmunología , Vacunación , Ensayos Antitumor por Modelo de Xenoinjerto
20.
AAPS J ; 18(1): 261-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26589309

RESUMEN

The need for multiple vaccinations to enhance the immunogenicity of subunit vaccines may be reduced by delivering the vaccine over an extended period of time. Here, we report two novel injectable pentablock copolymer based thermoresponsive hydrogels made of polyethyleneglycol-polycaprolactone-polylactide-polycaprolactone-polyethyleneglycol (PEG-PCL-PLA-PCL-PEG) with varying ratios of polycaprolactone (PCL) and polylactide (PLA), as single shot sustained release vaccines. Pentablock copolymer hydrogels were loaded with vaccine-encapsulated poly lactic-co-glycolic acid nanoparticles (PLGA-NP) or with the soluble vaccine components. Incorporation of PLGA-NP into the thermoresponsive hydrogels increased the complex viscosity of the gels, lowered the gelation temperature, and minimized the burst release of antigen and adjuvants. The two pentablock hydrogels stimulated both cellular and humoral responses. The addition of PLGA-NP to the hydrogels sustained immune responses for up to 49 days. The polymer with a higher ratio of PCL to PLA formed a more rigid gel, induced stronger immune responses, and stimulated effective anti-tumor responses in a prophylactic melanoma tumor model.


Asunto(s)
Hidrogeles/química , Vacunas/administración & dosificación , Vacunas/farmacocinética , Adyuvantes Inmunológicos , Animales , Antígenos/administración & dosificación , Antígenos/química , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacocinética , Química Farmacéutica , Preparaciones de Acción Retardada , Sistemas de Liberación de Medicamentos , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Masculino , Melanoma/inmunología , Melanoma/prevención & control , Ratones , Ratones Endogámicos C57BL , Nanopartículas , Poliésteres/química , Polietilenglicoles , Polímeros , Temperatura , Viscosidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA