Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Mater Chem B ; 9(36): 7435-7446, 2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-34551058

RESUMEN

Cancer vaccines based on DNA encoding oncogenes have shown great potential in preclinical studies. However, the efficacy of DNA vaccines is limited by their weak immunogenicity because of low cellular internalisation and insufficient activation of dendritic cells (DCs). Calcium phosphate (CP) nanoparticles (NPs) are biodegradable vehicles with low toxicity and high loading capacity of DNA but suffer from stability issues. Here we employed adenosine triphosphate (ATP) as a dual functional agent, i.e. stabiliser for CP and immunological adjuvant, and applied the ATP-modified CP (ACP) NPs to the DNA vaccine. ACP NP-enhanced cellular uptake and improved transfection efficiency of DNA vaccine, and further showed the ability to activate DCs that are critical for them to prime T cells in cancer immunotherapy. As a result, a higher level of antigen-specific antibody with stronger tumour growth inhibition was achieved in mice immunised with the ACP-DNA vaccine. Overall, this one-step synthesised ACP NPs are an efficient nano-delivery system and nano-adjuvant for cancer DNA vaccines.


Asunto(s)
Adenosina Trifosfato/química , Adyuvantes Inmunológicos/química , Fosfatos de Calcio/química , Nanopartículas/química , Vacunas de ADN/química , Animales , Reacciones Antígeno-Anticuerpo , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Inmunoterapia , Ratones , Ratones Endogámicos C57BL , Neoplasias/terapia , Trasplante Homólogo , Vacunación , Vacunas de ADN/inmunología , Vacunas de ADN/farmacología
2.
Acc Chem Res ; 53(10): 2094-2105, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-33017150

RESUMEN

The immune system has evolved over time to protect the host from foreign microorganisms. Activation of the immune system is predicated on a distinction between self and nonself. Unfortunately, cancer is characterized by genetic alterations in the host's cells, leading to uncontrolled cellular proliferation and evasion of immune surveillance. Cancer immunotherapy aims to educate the host's immune system to not only recognize but also attack and kill mutated cancer cells. While immune checkpoint blockers have been proven to be effective against multiple types of advanced cancer, the overall patient response rate still remains below 30%. Therefore, there is an urgent need to improve current cancer immunotherapies. In this Account, we present an overview of our recent progress on nanoparticle-based strategies for improving cancer vaccines and immunotherapies. We also present other complementary strategies to give a well-rounded snapshot of the field of combination cancer immunotherapy. The versatility and tunability of nanoparticles make them promising platforms for addressing individual challenges posed by various cancers. For example, nanoparticles can deliver cargo materials to specific cells, such as vaccines delivered to antigen-presenting cells for strong immune activation. Nanoparticles also allow for stimuli-responsive delivery of various therapeutics to cancer cells, thus forming the basis for combination cancer immunotherapy. Here, we focus on nanoparticle platforms engineered to deliver tumor antigens, whole tumor cells, and chemotherapeutic or phototherapeutic agents in a manner to effectively and safely trigger the host's immune system against tumor cells. For each work, we discuss the nanoparticle platform developed, synthesis chemistry, and in vivo applications. Nanovaccines offer a unique platform for codelivery of personalized tumor neoantigens and adjuvants and elicitation of robust immune responses against aggressive tumors. Nanovaccines either delivering whole tumor cell lysate or formed from tumor cell lysate may increase the repertoire of tumor antigens as immune targets while exploiting immunogenic cell death to prime antitumor immune responses. We also discuss how antigen- and whole tumor cell-based approaches may open the door for personalized cancer vaccination and immunotherapy. On the other hand, chemotherapy, phototherapy, and radiotherapy are more standardized cancer therapies, and nanoparticle-based approaches may promote their ability to initiate T cell activation against tumor cells and improve antitumor efficacy with minimal toxicity. Finally, building on the recent progress in nanoparticle-based cancer immunotherapy, the field should set the ultimate goal to be clinical translation and clinical efficacy. We will discuss regulatory, analytical, and manufacturing hurdles that should be addressed to expedite the clinical translation of nanomedicine-based cancer immunotherapy.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Inmunoterapia , Nanopartículas/química , Neoplasias/terapia , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos de Neoplasias/química , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Grafito/química , Humanos , Neoplasias/inmunología , Neoplasias/prevención & control , Polímeros/química
3.
J Med Chem ; 63(20): 11691-11706, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-32960056

RESUMEN

Self-adjuvanting vaccines, wherein an antigenic peptide is covalently bound to an immunostimulating agent, have been shown to be promising tools for immunotherapy. Synthetic Toll-like receptor (TLR) ligands are ideal adjuvants for covalent linking to peptides or proteins. We here introduce a conjugation-ready TLR4 ligand, CRX-527, a potent powerful lipid A analogue, in the generation of novel conjugate-vaccine modalities. Effective chemistry has been developed for the synthesis of the conjugation-ready ligand as well as the connection of it to the peptide antigen. Different linker systems and connection modes to a model peptide were explored, and in vitro evaluation of the conjugates showed them to be powerful immune-activating agents, significantly more effective than the separate components. Mounting the CRX-527 ligand at the N-terminus of the model peptide antigen delivered a vaccine modality that proved to be potent in activation of dendritic cells, in facilitating antigen presentation, and in initiating specific CD8+ T-cell-mediated killing of antigen-loaded target cells in vivo. Synthetic TLR4 ligands thus show great promise in potentiating the conjugate vaccine platform for application in cancer vaccination.


Asunto(s)
Vacunas contra el Cáncer/síntesis química , Glucosamina/análogos & derivados , Lípido A/análogos & derivados , Compuestos Organofosforados/química , Ovalbúmina/química , Receptor Toll-Like 4/inmunología , Adyuvantes Inmunológicos , Animales , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Citocinas/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Glucosamina/química , Glucosamina/inmunología , Inmunoglobulina G/sangre , Ligandos , Activación de Linfocitos/efectos de los fármacos , Ratones , Compuestos Organofosforados/inmunología , Ovalbúmina/inmunología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Vacunas Conjugadas
4.
Bioconjug Chem ; 31(9): 2147-2157, 2020 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-32786363

RESUMEN

Triple-negative breast cancer (TNBC) is an immune-enriched subset of breast cancer that has recently demonstrated clinical responsiveness to combinatorial immunotherapy. However, the lack of targeted interventions against hormone receptors or HER2 continues to limit treatment options for these patients. To begin expanding available interventions for patients with metastatic TNBC, we previously reported a therapeutic vaccine regimen that significantly reduced spontaneous lung metastases in a preclinical TNBC model. This heterologous vaccine approach "primed" mice with tumor lysate antigens encapsulated within poly(lactic-co-glycolic) acid microparticles (PLGA MPs), and then "boosted" mice with tumor lysates plus adjuvant. The use of the PLGA MP prime as monotherapy demonstrated no efficacy, suggesting that improving this component of our therapy would achieve greater vaccine efficacy. Here, we functionally improved the PLGA MP prime by coating microparticles with biotinylated streptavidin-conjugated using 1-ethyl-3-(3-dimethylaminoproplyl) carbodiimide/N-hydroxysuccinimide (EDC/Sulfo-NHS) linkers. This modification enhanced the immunostimulatory potential of our PLGA MPs, as evidenced by increased phagocytosis, maturation, and stimulatory ligand expression by antigen-presenting cells (APCs). Therapeutic prime/boost vaccination of TNBC-bearing mice with surfaced-coated PLGA MPs significantly reduced spontaneous lung metastases by an average of 56% relative to mice primed with unmodified PLGA MPs, and a significant 88% average reduction in spontaneous lung metastases relative to untreated control mice. These findings illustrate that relatively common biotin-streptavidin conjugation formulations can positively affect microparticle-based vaccine immunogenicity resulting in enhanced therapeutic efficacy against established preclinical mammary tumors.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Estreptavidina/uso terapéutico , Neoplasias de la Mama Triple Negativas/prevención & control , Adyuvantes Inmunológicos/química , Animales , Biotinilación , Vacunas contra el Cáncer/química , Portadores de Fármacos/química , Portadores de Fármacos/uso terapéutico , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/uso terapéutico , Estreptavidina/química , Neoplasias de la Mama Triple Negativas/inmunología
5.
ACS Appl Mater Interfaces ; 12(37): 41127-41137, 2020 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-32808767

RESUMEN

Weak T cell responses and immune checkpoints within tumors could be two key factors for limiting antitumor efficacy in the field of cancer immunotherapy. Thus, the combined strategy of tumor vaccines and immune checkpoint blockade has been widely studied and expected to boost antitumor immune responses. Herein, we first developed a two-barreled strategy to combine the nanovaccine with a gene-mediated PD-L1 blockade. On the one hand, polyethyleneimine (PEI) worked as a vaccine carrier to codeliver the antigen ovalbumin (OVA) and the adjuvant unmethylated cytosine-phosphate-guanine (CpG) to formulate the PEI/OVA/CpG nanovaccine through electrostatic binding, which realized both dendritic cell activation and antigen cross-presentation enhancement. On the other hand, the PD-L1 silence gene was loaded by PEI to form PEI/pshPD-L1 complexes, which were further in situ shielded by aldehyde-modified polyethylene glycol (OHC-PEG-CHO) via pH-responsive Schiff base bonds. The formed pshPD-L1@NPs could decrease PD-L1 expression on the tumor cells. However, such a combined two-barreled strategy improved feebly for tumor inhibition in comparison with monotherapy, exhibiting the antagonistic effect, which might be due to the limited T cell response enhancement in the tumor microenvironment. To solve this problem, we have further developed a three-barreled strategy to combine oral administration of l-arginine, which worked as an amplifier to induce robust T cell response enhancement, without causing the upregulation of other negative immune regulators. Superior antitumor behavior and tumor rechallenge protection were realized by the three-barreled strategy in B16F10-OVA (B16-OVA)-bearing mice. The unique three-barreled strategy we developed might offer a novel clinical therapeutic treatment.


Asunto(s)
Arginina/inmunología , Antígeno B7-H1/antagonistas & inhibidores , Vacunas contra el Cáncer/inmunología , Inmunoterapia , Nanopartículas/química , Animales , Arginina/química , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Vacunas contra el Cáncer/química , Citosina/química , Citosina/inmunología , Guanina/química , Guanina/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Ovalbúmina/química , Ovalbúmina/inmunología , Tamaño de la Partícula , Fosfatos/química , Fosfatos/inmunología , Polietileneimina/química , Propiedades de Superficie
6.
ACS Appl Mater Interfaces ; 12(31): 34658-34666, 2020 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-32662625

RESUMEN

Owing to the limitations of conventional cancer therapies, cancer immunotherapy has emerged for the prevention of cancer recurrence. To provoke adaptive immune responses that are antigen-specific, it is important to develop an efficient antigen delivery system that can enhance the activation and maturation of the dendritic cells (DCs) in the human body. In this study, we synthesize hollow mesoporous silica nanoparticles with extra-large mesopores (H-XL-MSNs) based on a single-step synthesis from core-shell mesoporous silica nanoparticles with a core composed of an assembly of iron oxide nanoparticles. The hollow void inside the mesoporous silica nanoparticles with large mesopores allows a high loading efficiency of various model proteins of different sizes. The H-XL-MSNs are coated with a poly(ethyleneimine) (PEI) solution to provide an immune adjuvant and change the surface charge of the particles for loading and slow release of a model antigen. An in vitro study using a cancer vaccine based on the PEI-coated H-XL-MSNs with the loading of the model antigen showed an enhanced activation of the DCs. An in vivo study demonstrated that the resulting cancer vaccine increased the antigen-specific cytotoxic T cells, enhanced the suppression of tumor growth, and improved the survival rate after challenging cancer to mice. These findings suggest that these hollow MSNs with extra-large pores can be used as excellent antigen carriers for immunotherapy.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Melanoma Experimental/inmunología , Nanopartículas/química , Dióxido de Silicio/química , Adyuvantes Inmunológicos , Animales , Vacunas contra el Cáncer/química , Células Dendríticas/inmunología , Melanoma Experimental/patología , Ratones , Tamaño de la Partícula , Porosidad , Propiedades de Superficie , Linfocitos T Citotóxicos/inmunología
7.
Int J Nanomedicine ; 15: 2685-2697, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32368049

RESUMEN

BACKGROUND: Nanocarriers could deliver significantly higher amounts of antigen to antigen-presenting cells (APCs), which have great potential to stimulate humoral and cellular response in cancer immunotherapy. Thereafter, silica solid nanosphere (SiO2) was prepared, and a model antigen (ovalbumin, OVA) was covalently conjugated on the surface of SiO2 to form nanovaccine (OVA@SiO2). And the application of OVA@SiO2 for cancer immunotherapy was evaluated. MATERIALS AND METHODS: SiO2 solid nanosphere was prepared by the Stöber method, then successively aminated by aminopropyltriethoxysilane and activated with glutaraldehyde. OVA was covalently conjugated on the surface of activated SiO2 to obtain nanovaccine (OVA@SiO2). Dynamic light scattering, scanning electron microscope, and transmission electron microscope were conducted to identify the size distribution, zeta potential and morphology of OVA@SiO2. The OVA loading capacity was investigated by varying glutaraldehyde concentration. The biocompatibility of OVA@SiO2 to DC2.4 and RAW246.7 cells was evaluated by a Cell Counting Kit-8 assay. The uptake of OVA@SiO2 by DC2.4 and its internalization pathway were evaluated in the absence or presence of different inhibitors. The activation and maturation of bone marrow-derived DC cells by OVA@SiO2 were also investigated. Finally, the in vivo transport of OVA@SiO2 and its toxicity to organs were appraised. RESULTS: All results indicated the successful covalent conjugation of OVA on the surface of SiO2. The as-prepared OVA@SiO2 possessed high antigen loading capacity, which had good biocompatibility to APCs and major organs. Besides, OVA@SiO2 facilitated antigen uptake by DC2.4 cells and its cytosolic release. Noteworthily, OVA@SiO2 significantly promoted the maturation of dendritic cells and up-regulation of cytokine secretion by co-administration of adjuvant CpG-ODN. CONCLUSION: The as-prepared SiO2 shows promising potential for use as an antigen delivery carrier.


Asunto(s)
Antígenos/metabolismo , Vacunas contra el Cáncer/farmacología , Inmunoterapia/métodos , Nanosferas/química , Ovalbúmina/química , Adyuvantes Inmunológicos/administración & dosificación , Animales , Presentación de Antígeno , Antígenos/administración & dosificación , Antígenos/química , Antígenos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/química , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Femenino , Ratones , Ratones Endogámicos C57BL , Nanosferas/administración & dosificación , Oligodesoxirribonucleótidos/administración & dosificación , Ovalbúmina/inmunología , Ovalbúmina/farmacocinética , Células RAW 264.7 , Dióxido de Silicio/química
8.
Int J Pharm ; 582: 119310, 2020 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-32276088

RESUMEN

Antigen-adjuvant combination could induce a protective and long-lasting anti-tumor immune response. However, exploiting system which could co-deliver melanoma antigen peptide Trp2 (Tyrosinase-related protein 2) and Toll-like-receptor-7 (TLR7) agonists imiquimod (R837) both are poor aqueous solubility is still challenging. Our new nanocomplex was explored for specific delivery of Trp2 and R837 into antigen-presenting cells (APCs). R837 was loaded into mannosylated-ß-cyclodextrin (Man-CD) to target dendritic cells (DCs) by binding mannose receptors (MR) on DCs. A fusion peptide (WT) was constructed by incorporating the amino acid region of TAT (cell-penetrating peptide) into Trp2 to improve the TAT-mediated intracellular efficiency. Negatively charged sodium alginate (SA), a biocompatible polymer, which can induce adjuvant responses by affecting the functions of DCs, was complexed with Man-CD/R837 and WT via physical adsorption. The optimized nanocomplex promoted the cellular uptake and showed remarkable efficacy to enhance the secreting of Th1-cytokines. This multi-functional nanocomplex system may allow effective targeting-codelivery of multi-hydrophobic drugs and be a promising subunit vaccine candidate as a potential prevention action of tumor.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacunas contra el Cáncer/farmacología , Portadores de Fármacos , Imiquimod/farmacología , Manosa/química , Melanoma Experimental/tratamiento farmacológico , Proteínas de la Membrana/farmacología , Nanopartículas , Fragmentos de Péptidos/farmacología , beta-Ciclodextrinas/química , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/química , Alginatos/química , Alginatos/farmacología , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/química , Línea Celular Tumoral , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Composición de Medicamentos , Femenino , Interacciones Hidrofóbicas e Hidrofílicas , Imiquimod/administración & dosificación , Imiquimod/química , Lectinas Tipo C/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Proteínas de la Membrana/administración & dosificación , Proteínas de la Membrana/química , Ratones Endogámicos C57BL , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/química , Receptores de Superficie Celular/metabolismo , Solubilidad , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/metabolismo
9.
Adv Mater ; 32(16): e1908185, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32108390

RESUMEN

Cytomembrane-derived nanoplatforms are an effective biomimetic strategy in cancer therapy. To improve their functionality and expandability for enhanced vaccination, a eukaryotic-prokaryotic vesicle (EPV) nanoplatform is designed and constructed by fusing melanoma cytomembrane vesicles (CMVs) and attenuated Salmonella outer membrane vesicles (OMVs). Inheriting the virtues of the parent components, the EPV integrates melanoma antigens with natural adjuvants for robust immunotherapy and can be readily functionalized with complementary therapeutics. In vivo prophylactic testing reveals that the EPV nanoformulation can be utilized as a prevention vaccine to stimulate the immune system and trigger the antitumor immune response, combating tumorigenesis. In the melanoma model, the poly(lactic-co-glycolic acid)-indocyanine green (ICG) moiety (PI)-implanted EPV (PI@EPV) in conjunction with localized photothermal therapy with durable immune inhibition shows synergetic antitumor effects as a therapeutic vaccine. The eukaryotic-prokaryotic fusion strategy provides new perspectives for the design of tumor-immunogenic, self-adjuvanting, and expandable vaccine platforms.


Asunto(s)
Melanoma/prevención & control , Nanomedicina/métodos , Fototerapia , Salmonella/química , Vacunación/métodos , Animales , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Verde de Indocianina/química , Melanoma/patología , Ratones , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química
10.
Theranostics ; 10(4): 1814-1832, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32042338

RESUMEN

Melanoma is one of the deadliest malignancies with a high risk of relapse and metastasis. Long-term, tumor-specific, and systemic immunity induced by local intervention is ideal for personalized cancer therapy. Laser immunotherapy (LIT), a combination of local irradiation of laser and local administration of an immunostimulant, was developed to achieve such an immunity. Although LIT showed promising efficacy on tumors, its immunological mechanism is still not understood, especially its spatio-temporal dynamics. Methods: In this study, we investigated LIT-induced immunological responses using a 980-nm laser and a novel immunostimulant, N-dihydrogalactochitosan (GC). Then we followed the functions of key immune cells spatially and temporally using intravital imaging and immunological assays. Results: Immediately after LIT, GC induced a rapid infiltration of neutrophils which ingested most GC in tumors. The cytokines released to the serum peaked at 12 h after LIT. Laser irradiations produced photothermal effects to ablate the tumor, release damage-associated molecular patterns, and generate whole-cell tumor vaccines. LIT-treated tumor-bearing mice efficiently resisted the rechallenged tumor and prevented lung metastasis. Intravital imaging of tumor at rechallenging sites in LIT-treated mice revealed that the infiltration of tumor-infiltrating lymphocytes (TILs) increased with highly active motility. Half of TILs with arrest and confined movements indicated that they had long-time interactions with tumor cells. Furthermore, LIT has synergistic effect with checkpoint blockade to improve antitumor efficacy. Conclusion: Our research revealed the important role of LIT-induced neutrophil infiltration on the in situ whole-cell vaccine-elicited antitumor immune response and long-term T cell immune memory.


Asunto(s)
Memoria Inmunológica/efectos de la radiación , Inmunoterapia/métodos , Melanoma/patología , Infiltración Neutrófila/efectos de la radiación , Linfocitos T/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Femenino , Neoplasias Pulmonares/secundario , Melanoma/mortalidad , Melanoma/terapia , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/prevención & control , Fototerapia/métodos
11.
ACS Appl Mater Interfaces ; 11(51): 47798-47809, 2019 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-31773941

RESUMEN

Conventional adjuvants (e.g., aluminum) are insufficient to trigger cell-mediated immunity, which plays a crucial role in triggering specific immunity against cancer. Therefore, developing appropriate adjuvants for cancer vaccines is a central way to stimulate the antitumor immune response. Hollow mesoporous silica nanoparticles (HMSNs) have been proven to stimulate Th1 antitumor immunity in vivo and promote immunological memory in the formulation of novel cancer vaccines. Yet, immune response rates of existing HMSNs for anticancer immunity still remain low. Here, we demonstrate the generation of polyethylenimine (PEI)-incorporated thin-shell HMSNs (THMSNs) through a facile PEI etching strategy for cancer immunotherapy. Interestingly, incorporation of PEI and thin-shell hollow structures of THMSNs not only improved the antigen-loading efficacy and sustained drug release profiles but also enhanced the phagocytosis efficiency by dendritic cells (DCs), enabled DC maturation and Th1 immunity, and sustained immunological memory, resulting in the enhancement of the adjuvant effect of THMSNs. Moreover, THMSNs vaccines without significant side effects can significantly reduce the potentiality of tumor growth and metastasis in tumor challenge and rechallenge models, respectively. THMSNs are considered to be promising vehicles and excellent adjuvants for the formulation of cancer vaccines for immunotherapy.


Asunto(s)
Adyuvantes Inmunológicos/química , Vacunas contra el Cáncer/química , Inmunoterapia/métodos , Nanopartículas/química , Polietileneimina/química , Dióxido de Silicio/química , Animales , Células Dendríticas/metabolismo , Humanos
12.
ACS Nano ; 13(11): 12553-12566, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31689085

RESUMEN

Efficient cancer vaccines not only require the co-delivery of potent antigens and highly immunostimulatory adjuvants to initiate robust tumor-specific host immune response but also solve the spatiotemporal consistency of host immunity and tumor microenvironment (TME) immunomodulation. Here, we designed a biomaterials-based strategy for converting tumor-derived antigenic microparticles (T-MPs) into a cancer vaccine to meet this conundrum and demonstrated its therapeutic potential in multiple murine tumor models. The internal cavity of T-MPs was employed to store nano-Fe3O4 (Fe3O4/T-MPs), and then dense adjuvant CpG-loaded liposome arrays (CpG/Lipo) were tethered on the surface of Fe3O4/T-MP through mild surface engineering to get a vaccine (Fe3O4/T-MPs-CpG/Lipo), demonstrating that co-delivery of Fe3O4/T-MPs and CpG/Lipo to antigen presenting cells (APCs) could elicit strong tumor antigen-specific host immune response. Meanwhile, vaccines distributed in the TME could reverse infiltrated tumor-associated macrophages into a tumor-suppressive M1 phenotype by nano-Fe3O4, amazingly induce abundant infiltration of cytotoxic T lymphocytes, and transform a "cold" tumor into a "hot" tumor. Furthermore, amplified antitumor immunity was realized by the combination of an Fe3O4/T-MPs-CpG/Lipo vaccine and immune checkpoint PD-L1 blockade, specifically inhibiting ∼83% of the progression of B16F10-bearing mice and extending the median survival time to 3 months. Overall, this study synergistically modulates the tumor immunosuppressive network and host antitumor immunity in a spatiotemporal manner, which suggests a general cell-engineering strategy tailored to a personalized vaccine from autologous cancer cell materials of each individual patient.


Asunto(s)
Antígenos de Neoplasias , Vacunas contra el Cáncer , Inmunomodulación/inmunología , Inmunoterapia/métodos , Microambiente Tumoral/inmunología , Animales , Antígenos de Neoplasias/química , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Ingeniería Celular , Sistemas de Liberación de Medicamentos , Femenino , Óxido Ferrosoférrico/química , Ratones , Ratones Endogámicos C57BL
13.
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
14.
ACS Nano ; 13(3): 2956-2968, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30789699

RESUMEN

Personalized cancer vaccines show great potential in cancer immunotherapy by inducing an effective and durable antitumor response. However, the limitation of neoantigen identification, low immunogenicity, and weak immune response hamper the development of personalized cancer vaccines. The surgically removed tumor contains tumor antigens specific to the patient, which provides a promising source for personalized cancer vaccines. Here, we utilized the surgically removed tumor to prepare a personalized photothermal vaccine combined with the PD-1 checkpoint blockade antibody to prevent tumor relapse and metastasis. Black phosphorus quantum dot nanovesicles (BPQD-CCNVs) coated with surgically removed tumor cell membrane were prepared and loaded into a thermosensitive hydrogel containing GM-CSF and LPS. The sustained release of GM-CSF from the hypodermic injection of Gel-BPQD-CCNVs effectively recruited dendritic cells to capture tumor antigen. NIR irradiation and LPS stimulated the expansion and activation of DCs, which then traveled to the lymph nodes to present antigen to CD8+ T cells. Moreover, the combination with PD-1 antibody significantly enhanced tumor-specific CD8+ T cell elimination of the surgical residual and lung metastatic tumor. Hence, our work may provide a promising strategy for the clinical development of a personalized cancer vaccine.


Asunto(s)
Neoplasias de la Mama/terapia , Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia , Melanoma Experimental/terapia , Fototerapia , Medicina de Precisión , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Membrana Celular/efectos de los fármacos , Membrana Celular/inmunología , Modelos Animales de Enfermedad , Composición de Medicamentos , Femenino , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nanopartículas/química , Fósforo/química , Puntos Cuánticos/química , Células Tumorales Cultivadas
15.
Hum Vaccin Immunother ; 15(3): 611-619, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30427754

RESUMEN

We previously found that Pleurotus ferulae polysaccharides (PFPS) improved the maturation and function of dendritic cells (DCs). In this study, we investigated the effects of PFPS on the antitumor efficacy of therapeutic human papillomavirus (HPV) DC-based vaccine. PFPS stimulated DCs pulsed with HPV E6/E7 peptides were used to treat tumor mice on day 5 & 12 (HPV + PFPS-DCs early) and day 12 & 19 (HPV + PFPS-DCs late) after TC-1 cell injection. Compared to control group, both HPV + PFPS-DCs early and HPV + PFPS-DCs late strategies significantly inhibited tumor growth, which was significantly correlated with the increased activation status of both CD4+ and CD8+ T cells, the decreased frequencies of myeloid-derived suppressor cells, and the induction of HPV-specific CD8+ T cell responses. The survival of tumor mice was also greatly improved by HPV + PFPS-DCs early. Moreover, HPV + PFPS-DCs early completely inhibited the growth of second challenged TC-1 cells in tumor free mice. The results showed that PFPS improved the antitumor efficacy of therapeutic HPV DC-based vaccine, suggesting that PFPS might be a potential adjuvant for DC-based vaccines. This study provides a potential strategy for developing the therapeutic DC-based vaccine against cervical cancer.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Polisacáridos Fúngicos/inmunología , Neoplasias/terapia , Vacunas contra Papillomavirus/inmunología , Pleurotus/química , Adyuvantes Inmunológicos/administración & dosificación , Animales , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/química , Línea Celular Tumoral , Células Dendríticas/efectos de los fármacos , Femenino , Polisacáridos Fúngicos/farmacología , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Papillomaviridae , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/química
16.
Adv Mater ; 31(34): e1803953, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30417454

RESUMEN

Immunotherapy has been recognized for decades as a promising therapeutic method for cancer treatment. To enhance host immune responses against cancer, antigen-presenting cells (APCs; e.g., dendritic cells) or T cells are educated using immunomodulatory agents including tumor-associated antigens and adjuvants, and manipulated to induce a cascading adaptive immune response targeting tumor cells. Mesoporous silica materials are promising candidates to improve cancer immunotherapy based on their attractive properties that include high porosity, high biocompatibility, facile surface modification, and self-adjuvanticity. Here, the recent progress on mesoporous-silica-based immunotherapies based on two material forms is summarized: 1) mesoporous silica nanoparticles (MSNs), which can be internalized into APCs, and 2) micrometer-sized mesoporous silica rods (MSRs) that can form a 3D space to recruit APCs. Subcutaneously injected MSN-based cancer vaccines can be taken up by peripheral APCs or by APCs in lymphoid organs to educate the immune system against cancer cells. MSR cancer vaccines can recruit immune cells into the MSR scaffold to induce cancer-specific immunity. Both vaccine systems successfully stimulate the adaptive immune response to eradicate cancer in vivo. Thus, mesoporous silica has potential value as a material platform for the treatment of cancer or infectious diseases.


Asunto(s)
Materiales Biocompatibles/química , Vacunas contra el Cáncer/química , Inmunoterapia/métodos , Nanopartículas/química , Neoplasias/terapia , Dióxido de Silicio/química , Adyuvantes Inmunológicos , Animales , Células Presentadoras de Antígenos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Terapia Combinada , Humanos , Factores Inmunológicos/administración & dosificación , Neoplasias/inmunología , Porosidad , Linfocitos T/inmunología
17.
Theranostics ; 8(3): 860-873, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29344312

RESUMEN

Recently, near-infrared (NIR) light-based photothermal therapy (PTT) has been widely applied in cancer treatment. However, in most cases, the tissue penetration depth of NIR light is not sufficient and thus photothermal therapy is unable to completely eradicate deep, seated tumors inevitably leading to recurrence of the tumor. Due to this significant limitation of NIR, improved therapeutic strategies are urgently needed. Methods: We developed an endogenous vaccine based on a novel nanoparticle platform for combinatorial photothermal ablation and immunotherapy. The design was based on fluorophore-loaded liposomes (IR-7-lipo) coated with a multivalent immunoadjuvant (HA-CpG). In vitro PTT potency was assessed in cells by LIVE/DEAD and Annexin V-FITC/PI assays. The effect on bone marrow-derived dendritic cells (BMDC) maturation and antigen presentation was evaluated by flow cytometry (FCM) with specific antibodies. After treatment, the immune cell populations in tumor micro-environment and the cytokines in the serum were detected by FCM and Elisa assay, respectively. Finally, the therapeutic outcome was investigated in an animal model. Results: Upon irradiation with 808 nm laser, IR-7-lipo induced tumor cell necrosis and released tumor-associated antigens, while the multivalent immunoadjuvant improved the expression of co-stimulatory molecules on BMDC and promoted antigen presentation. The combination therapy of PTT and immunotherapy regulated the tumor micro-environment, decreased immunosuppression, and potentiated host antitumor immunity. Most significantly, due to an enhanced antitumor immune response, combined photothermal immunotherapy was effective in eradicating tumors in mice and inhibiting tumor metastasis. Conclusion: This endogenous vaccination strategy based on synergistic photothermal and immunotherapy may provide a potentially effective approach for treatment of cancers, especially those difficult to be surgically removed.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Hipertermia Inducida/métodos , Inmunoterapia/métodos , Neoplasias Experimentales/terapia , Fototerapia/métodos , Microambiente Tumoral , Adyuvantes Inmunológicos/química , Animales , Presentación de Antígeno , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/uso terapéutico , Línea Celular Tumoral , Células Cultivadas , Células Dendríticas/inmunología , Femenino , Liposomas/química , Ratones , Ratones Endogámicos BALB C , Fármacos Fotosensibilizantes/química
18.
J Control Release ; 248: 1-9, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28057523

RESUMEN

Cell surface engineering is an expanding field and whilst extensive research has been performed decorating cell surfaces with biomolecules, the engineering of cell surfaces with particles has been a largely unexploited area. This study reports on the assembly of cell-particle hybrids where irradiated tumor cells were surface engineered with adjuvant-loaded, biodegradable, biocompatible, polymeric particles, with the aim of generating a construct capable of functioning as a therapeutic cancer vaccine. Successfully assembled cell-particle hybrids presented here comprised either melanoma cells or prostate cancer cells stably adorned with Toll-like receptor-9 ligand-loaded particles using streptavidin-biotin cross-linking. Both cell-particle assemblies were tested in vivo for their potential as therapeutic cancer vaccines yielding promising therapeutic results for the prostate cancer model. The ramifications of results obtained for both tumor models are openly discussed.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacunas contra el Cáncer/farmacología , Portadores de Fármacos/química , Neoplasias/prevención & control , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/química , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Reactivos de Enlaces Cruzados/química , Femenino , Ácido Láctico/química , Masculino , Melanoma/inmunología , Melanoma/prevención & control , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/prevención & control , Estreptavidina/química , Propiedades de Superficie
19.
Carbohydr Polym ; 155: 1-10, 2017 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-27702491

RESUMEN

A novel powder-form combination adjuvant system containing two immunostimulatory compounds was firstly developed and evaluated as a therapeutic intervention for cancer immunotherapy. With the help of hyaluronic acid (HA), water insoluble monophosphoryl lipid A (MPL), QS21 and imiquimod (R837), could be easily dispersed in aqueous solution and lyophilized as powder-form, which have an advantage in room-temperature storage stability compared with those conventional liquid formulation that requires cold storage. Two kinds of HA-based combination vaccine adjuvants (HA/MPL/QS21, HMQ and HA/MPL/R837, HMR) contributed to the increase of both humoral and cellular immunity, which is very important for efficient cancer immunotherapy. Through the challenge experiments in EG7-OVA (mouse lymphoma-expressing OVA) tumor-bearing mice model, we found out that the immunostimulatory effects of HMQ and HMR were successful in the inhibition of tumor proliferation. Taken together, both HA-based powder-form combination adjuvant systems are expected to be used as potent prophylactic and therapeutic cancer vaccine.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Ácido Hialurónico/farmacología , Linfoma/terapia , Adyuvantes Inmunológicos/química , Aminoquinolinas/química , Aminoquinolinas/inmunología , Aminoquinolinas/uso terapéutico , Animales , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Portadores de Fármacos , Femenino , Ácido Hialurónico/química , Enlace de Hidrógeno , Imiquimod , Inmunoterapia , Lípido A/análogos & derivados , Lípido A/química , Lípido A/inmunología , Lípido A/uso terapéutico , Linfoma/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Saponinas/química , Saponinas/inmunología , Saponinas/uso terapéutico , Solubilidad
20.
Small ; 12(26): 3510-5, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27191183

RESUMEN

Hollow and non-hollow mesoporous silica nanospheres are synthesized and used for cancer vaccine adjuvants. The hollow structure of mesoporous silica nanospheres significantly promote cellular uptake of a model cancer antigen by macrophage-like cells in vitro, improve anti-cancer immunity, CD4(+) and CD8(+) T cell populations in splenocytes of mice in vivo.


Asunto(s)
Adyuvantes Inmunológicos/química , Nanosferas/química , Dióxido de Silicio/química , Adyuvantes Inmunológicos/efectos adversos , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/química , Supervivencia Celular/efectos de los fármacos , Ratones , Microscopía Electroquímica de Rastreo , Microscopía Electrónica de Transmisión , Células 3T3 NIH , Nanosferas/efectos adversos , Nanosferas/ultraestructura , Porosidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA