Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.349
Filtrar
Más filtros











Intervalo de año de publicación
1.
Theranostics ; 14(10): 3810-3826, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38994034

RESUMEN

Rationale: Surgical resection is a primary treatment for solid tumors, but high rates of tumor recurrence and metastasis post-surgery present significant challenges. Manganese (Mn2+), known to enhance dendritic cell-mediated cancer immunotherapy by activating the cGAS-STING pathway, has potential in post-operative cancer management. However, achieving prolonged and localized delivery of Mn2+ to stimulate immune responses without systemic toxicity remains a challenge. Methods: We developed a post-operative microenvironment-responsive dendrobium polysaccharide hydrogel embedded with Mn2+-pectin microspheres (MnP@DOP-Gel). This hydrogel system releases Mn2+-pectin microspheres (MnP) in response to ROS, and MnP shows a dual effect in vitro: promoting immunogenic cell death and activating immune cells (dendritic cells and macrophages). The efficacy of MnP@DOP-Gel as a post-surgical treatment and its potential for immune activation were assessed in both subcutaneous and metastatic melanoma models in mice, exploring its synergistic effect with anti-PD1 antibody. Result: MnP@DOP-Gel exhibited ROS-responsive release of MnP, which could exert dual effects by inducing immunogenic cell death of tumor cells and activating dendritic cells and macrophages to initiate a cascade of anti-tumor immune responses. In vivo experiments showed that the implanted MnP@DOP-Gel significantly inhibited residual tumor growth and metastasis. Moreover, the combination of MnP@DOP-Gel and anti-PD1 antibody displayed superior therapeutic potency in preventing either metastasis or abscopal brain tumor growth. Conclusions: MnP@DOP-Gel represents a promising drug-free strategy for cancer post-operative management. Utilizing this Mn2+-embedding and ROS-responsive delivery system, it regulates surgery-induced immune responses and promotes sustained anti-tumor responses, potentially increasing the effectiveness of surgical cancer treatments.


Asunto(s)
Dendrobium , Hidrogeles , Manganeso , Ratones Endogámicos C57BL , Microesferas , Polisacáridos , Animales , Ratones , Hidrogeles/química , Manganeso/química , Polisacáridos/química , Polisacáridos/farmacología , Dendrobium/química , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Melanoma/inmunología , Melanoma/tratamiento farmacológico , Melanoma/terapia , Inmunoterapia/métodos , Células Dendríticas/inmunología , Células Dendríticas/efectos de los fármacos , Línea Celular Tumoral , Femenino , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Especies Reactivas de Oxígeno/metabolismo , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Melanoma Experimental/tratamiento farmacológico
2.
Egypt J Immunol ; 31(3): 28-40, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38985532

RESUMEN

The study aimed to assess the immunomodulatory effects of Phoenix dactylifera (dates) fruit, a traditional remedy used by Moroccans to enhance immunity against pathogens. This research sought to evaluate the impacts of this fruit on immune cells and their functions. To achieve this, we conducted tests using date extracts on splenocytes, thymocytes, and macrophages, focusing on their functions: antibody production, phagocytosis, and T-lymphocyte toxicity. The results obtained demonstrated that the aqueous extract of P. dactylifera fruit exhibited significant immunostimulatory effects on humoral immunity. It achieved this by enhancing complement activity and increasing splenocyte (including B-lymphocytes) proliferation by 142.5% compared to control cells. Similarly, in the same conditions, there was notable stimulation of cellular immunity through thymocyte activity, resulting in a remarkable increase in cell proliferation (225%) and a boost in thymocyte function (245.9%), which plays a role in safeguarding against cancer. Moreover, the date extract demonstrated anti-inflammatory properties. This was evident in the increased phagocytosis activity mediated by macrophages under the ethyl acetate extract, effectively eliminating pathogens. Assessing the cosmetic potential of date extracts showed that the ethyl acetate extract possesses both anti-inflammatory and strong antioxidant effects, exhibited high photo absorption of ultraviolet-B rays. Based on these findings, we propose to study the utilization of this extract for sun protection as a sunscreen. Furthermore, the Fourier-transform infrared spectroscopy analysis indicated that the most active compounds present were flavonoids. These outcomes substantiate the traditional usage of this fruit for reinforcing immunity.


Asunto(s)
Inmunidad Celular , Inmunidad Humoral , Phoeniceae , Extractos Vegetales , Animales , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Extractos Vegetales/farmacología , Extractos Vegetales/inmunología , Ratones , Phoeniceae/química , Adyuvantes Inmunológicos/farmacología , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Bazo/inmunología , Bazo/efectos de los fármacos , Bazo/citología , Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos , Frutas/química , Frutas/inmunología , Masculino , Proliferación Celular/efectos de los fármacos
3.
Molecules ; 29(13)2024 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-38999177

RESUMEN

A short 19 bp dsRNA with 3'-trinucleotide overhangs acting as immunostimulating RNA (isRNA) demonstrated strong antiproliferative action against cancer cells, immunostimulatory activity through activation of cytokines and Type-I IFN secretion, as well as anti-tumor and anti-metastatic effects in vivo. The aim of this study was to determine the tolerance of chemical modifications (2'-F, 2'-OMe, PS, cholesterol, and amino acids) located at different positions within this isRNA to its ability to activate the innate immune system. The obtained duplexes were tested in vivo for their ability to activate the synthesis of interferon-α in mice, and in tumor cell cultures for their ability to inhibit their proliferation. The obtained data show that chemical modifications in the composition of isRNA have different effects on its individual functions, including interferon-inducing and antiproliferative effects. The effect of modifications depends not only on the type of modification but also on its location and the surrounding context of the modifications. This study made it possible to identify leader patterns of modifications that enhance the properties of isRNA: F2/F2 and F2_S/F2 for interferon-inducing activity, as well as F2_S5/F2_S5, F2-NH2/F2-NH2, and Ch-F2/Ch-F2 for antiproliferative action. These modifications can improve the pharmacokinetic and pharmacodynamic properties, as well as increase the specificity of isRNA action to obtain the desired effect.


Asunto(s)
Proliferación Celular , ARN Bicatenario , ARN Bicatenario/farmacología , ARN Bicatenario/química , Animales , Proliferación Celular/efectos de los fármacos , Ratones , Humanos , Línea Celular Tumoral , Interferón-alfa/metabolismo , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/química , Interferones/metabolismo
4.
Nano Lett ; 24(29): 9017-9026, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39007530

RESUMEN

The development of in situ tumor vaccines offers promising prospects for cancer treatment. Nonetheless, the generation of plenary autologous antigens in vivo and their codelivery to DC cells along with adjuvants remains a significant challenge. Herein, we developed an in situ tumor vaccine using a supramolecular nanoparticle/hydrogel composite (ANPMTO/ALCD) and a deformable nanoadjuvant (PPER848). The ANPMTO/ALCD composite consisted of ß-cyclodextrin-decorated alginate (Alg-g-CD) and MTO-encapsulated adamantane-decorated nanoparticles (ANPMTO) through supramolecular interaction, facilitating the long-term and sustained production of plenary autologous antigens, particularly under a 660 nm laser. Simultaneously, the produced autologous antigens were effectively captured by nanoadjuvant PPER848 and subsequently transported to lymph nodes and DC cells, benefiting from its optimized size and deformability. This in situ tumor vaccine can trigger a robust antitumor immune response and demonstrate significant therapeutic efficacy in inhibiting tumor growth, suppressing tumor metastasis, and preventing postoperative recurrence, offering a straightforward approach to programming in situ tumor vaccines.


Asunto(s)
Adyuvantes Inmunológicos , Vacunas contra el Cáncer , Inmunoterapia , Nanopartículas , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Animales , Ratones , Inmunoterapia/métodos , Nanopartículas/química , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/uso terapéutico , Adyuvantes Inmunológicos/farmacología , Hidrogeles/química , Humanos , Línea Celular Tumoral , Células Dendríticas/inmunología , beta-Ciclodextrinas/química , Neoplasias/terapia , Neoplasias/inmunología , Alginatos/química , Adamantano/química , Adamantano/uso terapéutico
5.
Oncoimmunology ; 13(1): 2373526, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38948931

RESUMEN

Prostate cancer (PCa) is characterized as a "cold tumor" with limited immune responses, rendering the tumor resistant to immune checkpoint inhibitors (ICI). Therapeutic messenger RNA (mRNA) vaccines have emerged as a promising strategy to overcome this challenge by enhancing immune reactivity and significantly boosting anti-tumor efficacy. In our study, we synthesized Tetra, an mRNA vaccine mixed with multiple tumor-associated antigens, and ImmunER, an immune-enhancing adjuvant, aiming to induce potent anti-tumor immunity. ImmunER exhibited the capacity to promote dendritic cells (DCs) maturation, enhance DCs migration, and improve antigen presentation at both cellular and animal levels. Moreover, Tetra, in combination with ImmunER, induced a transformation of bone marrow-derived dendritic cells (BMDCs) to cDC1-CCL22 and up-regulated the JAK-STAT1 pathway, promoting the release of IL-12, TNF-α, and other cytokines. This cascade led to enhanced proliferation and activation of T cells, resulting in effective killing of tumor cells. In vivo experiments further revealed that Tetra + ImmunER increased CD8+T cell infiltration and activation in RM-1-PSMA tumor tissues. In summary, our findings underscore the promising potential of the integrated Tetra and ImmunER mRNA-LNP therapy for robust anti-tumor immunity in PCa.


Asunto(s)
Adyuvantes Inmunológicos , Antígenos de Neoplasias , Vacunas contra el Cáncer , Células Dendríticas , Neoplasias de la Próstata , ARN Mensajero , Animales , Masculino , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/terapia , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/tratamiento farmacológico , Antígenos de Neoplasias/inmunología , Ratones , Células Dendríticas/inmunología , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/administración & dosificación , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Mensajero/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Humanos , Ratones Endogámicos C57BL , Línea Celular Tumoral , Vacunas de ARNm , Linfocitos T CD8-positivos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Inmunoterapia/métodos , Activación de Linfocitos/efectos de los fármacos
6.
Sci Adv ; 10(27): eadg3747, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38959314

RESUMEN

Vaccination can help prevent infection and can also be used to treat cancer, allergy, and potentially even drug overdose. Adjuvants enhance vaccine responses, but currently, the path to their advancement and development is incremental. We used a phenotypic small-molecule screen using THP-1 cells to identify nuclear factor-κB (NF-κB)-activating molecules followed by counterscreening lead target libraries with a quantitative tumor necrosis factor immunoassay using primary human peripheral blood mononuclear cells. Screening on primary cells identified an imidazopyrimidine, dubbed PVP-037. Moreover, while PVP-037 did not overtly activate THP-1 cells, it demonstrated broad innate immune activation, including NF-κB and cytokine induction from primary human leukocytes in vitro as well as enhancement of influenza and SARS-CoV-2 antigen-specific humoral responses in mice. Several de novo synthesis structural enhancements iteratively improved PVP-037's in vitro efficacy, potency, species-specific activity, and in vivo adjuvanticity. Overall, we identified imidazopyrimidine Toll-like receptor-7/8 adjuvants that act in synergy with oil-in-water emulsion to enhance immune responses.


Asunto(s)
Adyuvantes Inmunológicos , Pirimidinas , Receptor Toll-Like 7 , Receptor Toll-Like 8 , Humanos , Receptor Toll-Like 8/agonistas , Receptor Toll-Like 8/metabolismo , Animales , Ratones , Adyuvantes Inmunológicos/farmacología , Receptor Toll-Like 7/agonistas , Pirimidinas/farmacología , Pirimidinas/química , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , Imidazoles/farmacología , Imidazoles/química , Células THP-1 , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/inmunología , COVID-19/virología , COVID-19/inmunología , FN-kappa B/metabolismo , Femenino , Descubrimiento de Drogas/métodos , Inmunidad Innata/efectos de los fármacos
7.
Fish Shellfish Immunol ; 151: 109714, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38906438

RESUMEN

The development of green aquaculture practices has led to the supplementation of fish diets with natural immunostimulants such as organic acids. This study aimed to assess the dietary effects of verjuice (VJ; unfermented unripe grapes; Vitis vinifera) on hematological parameters, skin mucosal immunity, transcriptional immune responses, and antibacterial serum activity against Aeromonas hydrophila in rainbow trout. The fish (51.0 ± 2.4 g) were randomly distributed into 15 tanks and fed ad-libitum thrice daily with diets containing different levels of VJ including 0 (control; VJ-0), 3 (VJ-3), 6 (VJ-6), 9 (VJ-9), and 12 (VJ-12) mL/kg VJ for 56 d. Results showed that immuno-hematological parameters (total white blood cells, neutrophils, and monocytes) were improved in VJ-added groups (P < 0.05). In addition, dietary VJ (9 mL/kg) modulated serum immunological parameters. Skin mucus immunology exhibited a notable increase in alkaline phosphatase, lysozyme activity, alkaline protease, total protein, total immunoglobulin, and esterase levels in VJ-9 group compared with those in the control group (P < 0.05). The mRNA expression of interleukin-1ß, interleukin-6, interleukin-8, and immunoglobulin M were significantly higher in VJ-9 group than in the control (P < 0.05). Furthermore, the results of the antibacterial evaluation showed that A. hydrophila growth was significantly inhibited in the serum samples from VJ-3 to VJ-9 groups after the 56th day and in all VJ-treated groups after the 70th (P < 0.05). In conclusion, dietary VJ is a novel immunostimulant and the optimal dietary supplementation level of 6.65-7.46 mL/kg can effectively improve immune responses in rainbow trout.


Asunto(s)
Aeromonas hydrophila , Alimentación Animal , Dieta , Suplementos Dietéticos , Enfermedades de los Peces , Inmunidad Innata , Oncorhynchus mykiss , Vitis , Animales , Oncorhynchus mykiss/inmunología , Oncorhynchus mykiss/genética , Alimentación Animal/análisis , Dieta/veterinaria , Suplementos Dietéticos/análisis , Aeromonas hydrophila/fisiología , Vitis/química , Enfermedades de los Peces/inmunología , Inmunidad Innata/efectos de los fármacos , Distribución Aleatoria , Infecciones por Bacterias Gramnegativas/veterinaria , Infecciones por Bacterias Gramnegativas/inmunología , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Inmunidad Mucosa/efectos de los fármacos , Relación Dosis-Respuesta a Droga
8.
Acta Biomater ; 183: 330-340, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38838909

RESUMEN

Although vaccination with inactivated vaccines is a popular preventive method against pseudorabies virus (PRV) infection, inactivated vaccines have poor protection efficiency because of their weak immunogenicity. The development of an effective adjuvant is urgently needed to improve the efficacy of inactivated PRV vaccines. In this study, a promising nanocomposite adjuvant named as MIL@A-SW01-C was developed by combining polyacrylic acid-coated metal-organic framework MIL-53(Al) (MIL@A) and squalene (oil)-in-water emulsion (SW01) and then mixing it with a carbomer solution. One part of the MIL@A was loaded onto the oil/water interface of SW01 emulsion via hydrophobic interaction and coordination, while another part was dispersed in the continuous water phase using carbomer. MIL@A-SW01-C showed good biocompatibility, high PRV (antigen)-loading capability, and sustained antigen release. Furthermore, the MIL@A-SW01-C adjuvanted PRV vaccine induced high specific serum antibody titers, increased splenocyte proliferation and cytokine secretion, and a more balanced Th1/Th2 immune response compared with commercial adjuvants, such as alum and biphasic 201. In the mouse challenge experiment, two- and one-shot vaccinations resulted in survival rates of 73.3 % and 86.7 %, respectively. After one-shot vaccination, the host animal pigs were also challenged with wild PRV. A protection rate of 100 % was achieved, which was much higher than that observed with commercial adjuvants. This study not only establishes the superiority of MIL@A-SW01-C composite nanoadjuvant for inactivated PRV vaccine in mice and pigs but also presents an effective method for developing promising nanoadjuvants. STATEMENT OF SIGNIFICANCE: We have developed a nanocomposite of MIL-53(Al) and oil-in-water emulsion (MIL@A-SW01-C) as a promising adjuvant for the inactivated PRV vaccines. MIL@A-SW01-C has good biocompatibility, high PRV (antigen) loading capability, and prolonged antigen release. The developed nanoadjuvant induced much higher specific IgG antibody titers, increased splenocyte proliferation and cytokine secretion, and a more balanced Th1/Th2 immune response than commercial adjuvants alum and biphasic 201. In mouse challenge experiments, survival rates of 73.3 % and 86.7 % were achieved from two-shot and one-shot vaccinations, respectively. At the same time, a protection rate of 100 % was achieved with the host animal pigs challenged with wild PRV.


Asunto(s)
Adyuvantes Inmunológicos , Emulsiones , Animales , Adyuvantes Inmunológicos/farmacología , Emulsiones/química , Ratones , Porcinos , Herpesvirus Suido 1/inmunología , Vacunas contra la Seudorrabia/inmunología , Ratones Endogámicos BALB C , Aceites/química , Femenino , Agua/química , Vacunas de Productos Inactivados/inmunología , Seudorrabia/prevención & control , Seudorrabia/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Citocinas/metabolismo
9.
Int J Biol Macromol ; 273(Pt 1): 133067, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38866287

RESUMEN

Adjuvants, as the essential component of vaccines, are crucial in enhancing the magnitude, breadth and durability of immune responses. Unfortunately, commonly used Alum adjuvants predominantly provoke humoral immune response, but fail to evoke cellular immune response, which is crucial for the prevention of various chronic infectious diseases and cancers. Thus, it is necessary to develop effective adjuvants to simultaneously induce humoral and cellular immune response. In this work, we obtained a water soluble polysaccharide isolated and purified from Poria cocos, named as PCP, and explored the possibility of PCP as a vaccine adjuvant. The PCP, with Mw of 20.112 kDa, primarily consisted of →6)-α-D-Galp-(1→, with a small amount of →3)-ß-D-Glcp-(1 â†’ and →4)-ß-D-Glcp-(1→. Our results demonstrated that the PCP promoted the activation of dendritic cells (DCs) and macrophages in vitro. As the adjuvant to ovalbumin, the PCP facilitated the activation of DCs in lymph nodes, and evoked strong antibody response with a combination of Th1 and Th2 immune responses. Moreover, compared to Alum adjuvant, the PCP markedly induced a potent cellular response, especially the cytotoxic T lymphocytes response. Therefore, we confirmed that the PCP has great potential to be an available adjuvant for simultaneously inducing humoral and cellular immune responses.


Asunto(s)
Adyuvantes Inmunológicos , Células Dendríticas , Polisacáridos , Solubilidad , Agua , Animales , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/química , Polisacáridos/farmacología , Polisacáridos/química , Polisacáridos/aislamiento & purificación , Ratones , Agua/química , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Femenino , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Wolfiporia/química , Ovalbúmina/inmunología , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Poria/química
10.
J Med Chem ; 67(12): 9976-9990, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38886162

RESUMEN

This study describes the design and synthesis of five TF-based cancer vaccine candidates using a lipid A mimetic as the carrier and a built-in adjuvant. All synthesized conjugates elicited robust and consistent TF-specific immune responses in mice without external adjuvants. Immunological studies subsequently conducted in wild-type and TLR4 knockout C57BL/6 mice demonstrated that the activation of TLR4 was the main reason that the synthesized lipid A mimetics increased the TF-specific immune responses. All antisera induced by these conjugates can specifically recognize, bind to, and induce the lysis of TF-positive cancer cells. Moreover, representative conjugates 2 and 3 could effectively reduce the growth of tumors and prolong the survival time of mice in vivo, and the efficacies were better than glycoprotein TF-CRM197 with alum adjuvant. Lipid A mimetics could therefore be a promising platform for the development of new carbohydrate-based vaccine carriers with self-adjuvanting properties for the treatment of cancer.


Asunto(s)
Adyuvantes Inmunológicos , Vacunas contra el Cáncer , Diseño de Fármacos , Lípido A , Ratones Endogámicos C57BL , Animales , Lípido A/análogos & derivados , Lípido A/química , Lípido A/farmacología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacología , Vacunas contra el Cáncer/síntesis química , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/síntesis química , Adyuvantes Inmunológicos/química , Ratones , Ratones Noqueados , Humanos , Femenino , Receptor Toll-Like 4/metabolismo , Línea Celular Tumoral
11.
ACS Nano ; 18(26): 16878-16894, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38899978

RESUMEN

Aluminum salts still remain as the most popular adjuvants in marketed human prophylactic vaccines due to their capability to trigger humoral immune responses with a good safety record. However, insufficient induction of cellular immune responses limits their further applications. In this study, we prepare a library of silicon (Si)- or calcium (Ca)-doped aluminum oxyhydroxide (AlOOH) nanoadjuvants. They exhibit well-controlled physicochemical properties, and the dopants are homogeneously distributed in nanoadjuvants. By using Hepatitis B surface antigen (HBsAg) as the model antigen, doped AlOOH nanoadjuvants mediate higher antigen uptake and promote lysosome escape of HBsAg through lysosomal rupture induced by the dissolution of the dopant in the lysosomes in bone marrow-derived dendritic cells (BMDCs). Additionally, doped nanoadjuvants trigger higher antigen accumulation and immune cell activation in draining lymph nodes. In HBsAg and varicella-zoster virus glycoprotein E (gE) vaccination models, doped nanoadjuvants induce high IgG titer, activations of CD4+ and CD8+ T cells, cytotoxic T lymphocytes, and generations of effector memory T cells. Doping of aluminum salt-based adjuvants with biological safety profiles and immunostimulating capability is a potential strategy to mediate robust humoral and cellular immunity. It potentiates the applications of engineered adjuvants in the development of vaccines with coordinated immune responses.


Asunto(s)
Adyuvantes Inmunológicos , Hidróxido de Aluminio , Calcio , Antígenos de Superficie de la Hepatitis B , Silicio , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacología , Animales , Silicio/química , Ratones , Antígenos de Superficie de la Hepatitis B/inmunología , Antígenos de Superficie de la Hepatitis B/química , Calcio/química , Hidróxido de Aluminio/química , Hidróxido de Aluminio/farmacología , Ratones Endogámicos C57BL , Femenino , Vacunas/inmunología , Vacunas/química , Células Dendríticas/inmunología , Células Dendríticas/efectos de los fármacos , Nanopartículas/química , Humanos , Óxido de Aluminio
12.
ACS Nano ; 18(24): 15557-15575, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38837909

RESUMEN

Tumor vaccines have demonstrated a modest response rate, primarily attributed to their inefficient delivery to dendritic cells (DCs), low cross-presentation, DC-intrinsic immunosuppressive signals, and an immunosuppressive tumor microenvironment (TME). Here, draining lymph node (DLN)-targeted and tumor-targeted nanovaccines were proposed to address these limitations, and heterocyclic lipidoid (A18) and polyester (BR647) were synthesized to achieve dual-targeted cancer immunotherapy. Meanwhile, oligo hyaluronic acid (HA) and DMG-PEG2000-Mannose were incorporated to prepare dual-targeted nanovaccines encapsulated with STAT3 siRNA and model antigens. The nanovaccines were designed to target the DLN and the tumor, facilitating the delivery of cargo into the cytoplasm. These dual-targeted nanovaccines improved antigen presentation and DC maturation, activated the stimulator of interferon genes (STING) pathway, enhanced the pro-apoptotic effect, and stimulated antitumor immune responses. Additionally, these dual-targeted nanovaccines overcame immunosuppressive TME, reduced immunosuppressive cells, and promoted the polarization of tumor-associated neutrophils from N2 to N1. Among the four dual-targeted nanovaccines that induced robust antitumor responses, the heterocyclic lipidoid@polyester hybrid nanovaccines (MALO@HBNS) demonstrated the most promising results. Furthermore, a combination strategy involving MALO@HBNS and an anti-PD-L1 antibody exhibited an immensely powerful anticancer role. This work introduced a dual-targeted nanovaccine platform for antitumor treatment, suggesting its potential combination with an immune checkpoint blockade as a comprehensive anticancer strategy.


Asunto(s)
Vacunas contra el Cáncer , Inmunoterapia , Nanopartículas , Poliésteres , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/química , Animales , Ratones , Poliésteres/química , Nanopartículas/química , Ratones Endogámicos C57BL , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Células Dendríticas/inmunología , Femenino , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/química , Lípidos/química , Humanos , Neoplasias/terapia , Neoplasias/inmunología , Línea Celular Tumoral , ARN Interferente Pequeño/química , Ácido Hialurónico/química , Nanovacunas
13.
Mar Drugs ; 22(6)2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38921545

RESUMEN

Deep seawater (DS), obtained from a depth over 200 m, has health benefits due to its rich nutrients and minerals, and intake of DS has shown diverse immunomodulatory effects in allergies and cancer. Therefore, the immunostimulatory effects of Korean mineral-rich seawaters were examined in a cyclophosphamide (CPA)-induced immunosuppression model. Three samples of Korean seawater, namely DS from the East Sea off the coasts of Pohang (PDS) and Uljin (UDS), and seawater from the West Sea off the coast of Boryeong (BS), were collected. The seawaters were abundant in several minerals (calcium, iron, zinc, selenium, etc.). Mice were orally administered the seawaters for 42 days, followed by CPA-induced immunosuppression. The CPA induction reduced the weight of the spleen and lymph nodes; however, the administration of seawaters increased the weight of the lymphoid organs, accompanied by stimulation of natural killer cells' activity and NF-kB-mediated cytokine production (IFNγ, TNFα, IL1ß, IL6, and IL12). The mouse-derived splenocytes showed lymphoproliferation without cytotoxicity in the seawater groups. Histopathological analysis revealed that the seawaters improved the CPA-induced atrophic changes by promoting lymphoproliferation in the spleen and lymph nodes. These results provide useful information for the use of Korean mineral-rich seawaters, particularly PDS and UDS, as alternative immunostimulants under immunosuppressive conditions.


Asunto(s)
Ciclofosfamida , Agua de Mar , Animales , Ciclofosfamida/farmacología , Ratones , Minerales/farmacología , Citocinas/metabolismo , República de Corea , Terapia de Inmunosupresión , Bazo/efectos de los fármacos , Bazo/inmunología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Masculino , Adyuvantes Inmunológicos/farmacología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Inmunosupresores/farmacología , Ratones Endogámicos BALB C
14.
Pharm Res ; 41(6): 1163-1181, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38839718

RESUMEN

OBJECTIVE: This study aims to utilize PEGylated poly (lactic-co-glycolic acid) (PLGA) nanoparticles as a delivery system for simultaneous administration of the BRAFV600E peptide, a tumor-specific antigen, and imiquimod (IMQ). The objective is to stimulate dendritic cell (DC) maturation, activate macrophages, and facilitate antigen presentation in C57BL6 mice. METHODS: PEG-PLGA-IMQ-BRAFV600E nanoparticles were synthesized using a PLGA-PEG-PLGA tri-block copolymer, BRAFV600E, and IMQ. Characterization included size measurement and drug release profiling. Efficacy was assessed in inhibiting BPD6 melanoma cell growth and activating immature bone marrow DCs, T cells, macrophages, and splenocyte cells through MTT and ELISA assays. In vivo, therapeutic and immunogenic effects potential was evaluated, comparing it to IMQ + BRAFV600E and PLGA-IMQ-BRAFV600E nanoparticles in inhibiting subcutaneous BPD6 tumor growth. RESULTS: The results highlight the successful synthesis of PEG-PLGA-IMQ-BRAFV600E nanoparticles (203 ± 11.1 nm), releasing 73.4% and 63.2% of IMQ and BARFV600E, respectively, within the initial 48 h. In vitro, these nanoparticles demonstrated a 1.3-fold increase in potency against BPD6 cells, achieving ~ 2.8-fold enhanced cytotoxicity compared to PLGA-IMQ-BRAFV600E. Moreover, PEG-PLGA-IMQ-BRAFV600E exhibited a 1.3-fold increase in potency for enhancing IMQ cytotoxic effects and a 1.1- to ~ 2.4-fold increase in activating DCs, T cells, macrophages, and splenocyte cells compared to IMQ-BRAFV600E and PLGA-IMQ-BRAFV600E. In vivo, PEG-PLGA-IMQ-BRAFV600E displayed a 1.3- to 7.5-fold increase in potency for inhibiting subcutaneous BPD6 tumor growth compared to the other formulations. CONCLUSIONS: The findings suggest that PEG-PLGA nanoparticles effectively promote DC maturation, T cell activation, and potentially macrophage activation. The study highlights the promising role of this nanocomposite in vaccine development.


Asunto(s)
Células Dendríticas , Imiquimod , Melanoma , Ratones Endogámicos C57BL , Nanopartículas , Polietilenglicoles , Proteínas Proto-Oncogénicas B-raf , Animales , Células Dendríticas/inmunología , Células Dendríticas/efectos de los fármacos , Polietilenglicoles/química , Proteínas Proto-Oncogénicas B-raf/genética , Melanoma/inmunología , Melanoma/tratamiento farmacológico , Nanopartículas/química , Línea Celular Tumoral , Ratones , Imiquimod/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Femenino , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Liberación de Fármacos , Humanos , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/prevención & control , Neoplasias Cutáneas/tratamiento farmacológico
15.
Int J Biol Macromol ; 269(Pt 2): 132172, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38719009

RESUMEN

Adjuvants including aluminum adjuvant (Alum) and oil-water emulsion have been widely used in inactivated pseudorabies virus (PRV) vaccines to improve their performance, however, they are not sufficient to protect from PRV infection because of the weak immune response and poor Th1-type immune response. Divalent manganese ion (Mn2+) has been reported to increase the cellular immune response significantly. In this work, a xanthan gum and carbomer-dispersed Mn2+-loaded tannic acid-polyethylene glycol (TPMnXC) nanoparticle colloid is developed and used as an adjuvant to improve the performance of the inactivated PRV vaccine. The good in vitro and in vivo biocompatibility of the developed TPMnXC colloid has been confirmed by the cell viability assay, erythrocyte hemolysis, blood routine analysis, and histological analysis of mouse organs and injection site. The TPMnXC-adjuvanted inactivated PRV vaccine (TPMnXC@PRV) significantly promotes higher and more balanced immune responses indicating with an increased specific total IgG antibody and IgG2a/IgG1 ratio, efficient splenocytes proliferation, and elevated Th1- and Th2-type cytokine secretion than those of control groups. Wild PRV challenge experiment is performed using mice as a model animal, achieving a protection rate of up to 86.67 %, which is much higher than those observed from the commercial Alum. This work not only demonstrates the high potentiality of TPMnXC in practical applications but also provides a new way to develop the Mn2+-loaded nanoadjuvant for veterinary vaccines.


Asunto(s)
Adyuvantes Inmunológicos , Herpesvirus Suido 1 , Inmunidad Celular , Inmunidad Humoral , Manganeso , Nanopartículas , Polisacáridos Bacterianos , Taninos , Animales , Ratones , Adyuvantes Inmunológicos/farmacología , Nanopartículas/química , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Taninos/química , Taninos/farmacología , Manganeso/química , Polisacáridos Bacterianos/química , Polisacáridos Bacterianos/farmacología , Polisacáridos Bacterianos/inmunología , Herpesvirus Suido 1/inmunología , Vacunas contra la Seudorrabia/inmunología , Vacunas de Productos Inactivados/inmunología , Seudorrabia/prevención & control , Seudorrabia/inmunología , Femenino , Citocinas/metabolismo , Ratones Endogámicos BALB C , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Polifenoles
16.
J Nanobiotechnology ; 22(1): 267, 2024 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-38764014

RESUMEN

Enhancing immune response activation through the synergy of effective antigen delivery and immune enhancement using natural, biodegradable materials with immune-adjuvant capabilities is challenging. Here, we present NAPSL.p that can activate the Toll-like receptor 4 (TLR4) pathway, an amphiphilic exopolysaccharide, as a potential self-assembly adjuvant delivery platform. Its molecular structure and unique properties exhibited remarkable self-assembly, forming a homogeneous nanovaccine with ovalbumin (OVA) as the model antigen. When used as an adjuvant, NAPSL.p significantly increased OVA uptake by dendritic cells. In vivo imaging revealed prolonged pharmacokinetics of NAPSL. p-delivered OVA compared to OVA alone. Notably, NAPSL.p induced elevated levels of specific serum IgG and isotype titers, enhancing rejection of B16-OVA melanoma xenografts in vaccinated mice. Additionally, NAPSL.p formulation improved therapeutic effects, inhibiting tumor growth, and increasing animal survival rates. The nanovaccine elicited CD4+ and CD8+ T cell-based immune responses, demonstrating the potential for melanoma prevention. Furthermore, NAPSL.p-based vaccination showed stronger protective effects against influenza compared to Al (OH)3 adjuvant. Our findings suggest NAPSL.p as a promising, natural self-adjuvanting delivery platform to enhance vaccine design across applications.


Asunto(s)
Adyuvantes Inmunológicos , Melanoma Experimental , Ratones Endogámicos C57BL , Ovalbúmina , Probióticos , Animales , Ovalbúmina/inmunología , Ovalbúmina/química , Ratones , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/química , Probióticos/farmacología , Melanoma Experimental/inmunología , Femenino , Células Dendríticas/inmunología , Receptor Toll-Like 4/metabolismo , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/química , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Humanos , Nanopartículas/química , Linfocitos T CD4-Positivos/inmunología
17.
Cell Rep Med ; 5(6): 101587, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38781964

RESUMEN

Epstein-Barr virus (EBV) is associated with infectious mononucleosis, cancer, and multiple sclerosis. A vaccine that prevents infection and/or EBV-associated morbidity is an unmet need. The viral gH/gL glycoprotein complex is essential for infectivity, making it an attractive vaccine target. Here, we evaluate the immunogenicity of a gH/gL nanoparticle vaccine adjuvanted with the Sigma Adjuvant System (SAS) or a saponin/monophosphoryl lipid A nanoparticle (SMNP) in rhesus macaques. Formulation with SMNP elicits higher titers of neutralizing antibodies and more vaccine-specific CD4+ T cells. All but one animal in the SMNP group were infected after oral challenge with the EBV ortholog rhesus lymphocryptovirus (rhLCV). Their immune plasma had a 10- to 100-fold lower reactivity against rhLCV gH/gL compared to EBV gH/gL. Anti-EBV neutralizing monoclonal antibodies showed reduced binding to rhLCV gH/gL, demonstrating that EBV gH/gL neutralizing epitopes are poorly conserved on rhLCV gH/gL. Prevention of rhLCV infection despite antigenic disparity supports clinical development of gH/gL nanoparticle vaccines against EBV.


Asunto(s)
Anticuerpos Neutralizantes , Herpesvirus Humano 4 , Lymphocryptovirus , Macaca mulatta , Nanopartículas , Vacunación , Animales , Nanopartículas/química , Herpesvirus Humano 4/inmunología , Lymphocryptovirus/inmunología , Vacunación/métodos , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/prevención & control , Infecciones por Virus de Epstein-Barr/virología , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Humanos , Infecciones por Herpesviridae/prevención & control , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología
18.
Am J Physiol Cell Physiol ; 327(1): C1-C10, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38708521

RESUMEN

The purpose of this study is to investigate the previously unknown connection that succinate has with neutrophils in the setting of adjuvant-mediated immunological enhancement. It has been discovered that succinates stimulate the recruitment of neutrophils in immunization sites, which in turn induces the expression of what is known as neutrophil-derived B cell-activating factor (BAFF). Further amplification of vaccine-induced antibody responses is provided via the succinate receptor 1-interferon regulatory factor 5 (SUCNR1-IRF5)-BAFF signaling pathway, which provides insights into a unique mechanism for immunological enhancement.NEW & NOTEWORTHY This study explores the role of succinate as a vaccine adjuvant, revealing its capacity to enhance neutrophil recruitment at immunization sites, which boosts B cell activation through the succinate receptor 1-interferon regulatory factor 5-B cell-activating factor (SUCNR1-IRF5-BAFF) signaling pathway. Results demonstrate succinate's potential to amplify vaccine-induced antibody responses, highlighting its significance in immunological enhancement and offering new insights into the adjuvant mechanisms of action, particularly in neutrophil-mediated immune responses.


Asunto(s)
Adyuvantes Inmunológicos , Neutrófilos , Transducción de Señal , Ácido Succínico , Neutrófilos/inmunología , Neutrófilos/metabolismo , Animales , Ácido Succínico/metabolismo , Adyuvantes Inmunológicos/farmacología , Humanos , Ratones , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/efectos de los fármacos , Infiltración Neutrófila/efectos de los fármacos , Factor Activador de Células B/metabolismo , Factor Activador de Células B/inmunología , Factor Activador de Células B/genética , Ratones Endogámicos C57BL , Femenino
19.
Bioconjug Chem ; 35(6): 804-815, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38718344

RESUMEN

Adjuvants are essential substances for vaccines and immunotherapies that enhance antigen-specific immune responses. Single-stranded oligodeoxynucleotides containing an unmethylated CpG motif (CpG ODNs) are agonistic ligands for toll-like receptor 9 that initiate an innate immune response. They represent promising adjuvants for antiviral and antitumor immunotherapies; however, CpG ODNs have some limitations, such as poor nuclease resistance and low cell membrane permeability. Therefore, an effective formulation is needed to improve the nuclease resistance and immunostimulatory effects of CpG ODNs. Previously, we demonstrated the selective delivery of a small molecule toll-like receptor 7 ligand to immune cells through sugar-binding receptors using sugar-immobilized gold nanoparticles (SGNPs), which significantly enhanced the potency of the ligand. In this study, we examined SGNPs as carriers for partially phosphorothioated A-type CpG ODN (D35) and an entirely phosphorothioated B-type CpG ODN (K3) and evaluated the functionality of the sugar moiety on SGNPs immobilized with CpG ODN. SGNPs immobilized with D35 (D35-SGNPs) exhibited improved nuclease resistance and the in vitro and in vivo potency was significantly higher compared with that of unconjugated D35. Furthermore, the sugar structure on the GNPs was a significant factor in enhancing the cell internalization ability, and enhanced intracellular delivery of D35 resulted in improving the potencies of the A-type CpG ODN, D35. SGNPs immobilized with K3 (K3-SGNPs) exhibited significantly higher induction activities for both humoral and cellular immunity compared with unconjugated K3 and D35-SGNPs. On the other hand, sugar structure on K3-SGNPs did not affect the immunostimulatory effects. These results indicate that the sugar moiety on K3-SGNPs primarily functions as a hydrophilic dispersant for GNPs and the formulation of K3 to SGNPs contributes to improving the immunostimulatory activity of K3. Because our CpG ODN-SGNPs have superior induction activities for antigen-specific T-cell mediated immune responses, they may be effective adjuvants for vaccines and immunotherapies.


Asunto(s)
Adyuvantes Inmunológicos , Oro , Nanopartículas del Metal , Oligodesoxirribonucleótidos , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/farmacología , Oro/química , Nanopartículas del Metal/química , Animales , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacología , Ratones , Azúcares/química , Humanos , Ratones Endogámicos C57BL
20.
Minerva Urol Nephrol ; 76(4): 474-483, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38757774

RESUMEN

BACKGROUND: The aim of this study is to investigate the impact of the intradermal injection of purified protein derivative (PPD) and PPD skin test reactions on the oncological outcomes of patients with non-muscle invasive bladder cancer (NMIBC) treated by trans-urethral resection of bladder tumor (TURBT) and adjuvant intravesical BCG. METHODS: The study included 100 consecutive patients with NMIBC prospectively given intradermal PPD 1-2 weeks before starting BCG therapy. Another 100 patients with NMIBC not given intradermal PPD before starting BCG were chosen as a historical control. The control group was chosen to be matching with the study group regarding baseline characteristics. The study group was divided into 2 subgroups with positive and negative reaction to PPD skin test. Oncological outcomes, immunological markers (TNF-α and IL-6) changes and BCG side effects were evaluated. RESULTS: There were no significant differences between patients who received PPD or not regarding the 2-year recurrence free survival (RFS) rates and progression-free survival (PFS) rates and immunological markers changes. The 2-year RFS and PFS rates were significantly higher in patients with positive reactions. Post-induction values of immunological markers increased in all patients with a significant increase in patients with positive reactions. BCG side effects were significantly higher in patients with positive reactions. CONCLUSIONS: The intradermal injection of PPD before intravesical BCG has no impact on oncological outcomes of patients with NMIBC treated with TURBT and intravesical BCG. However, the PPD skin test reactions before BCG therapy can predict the oncological outcomes, BCG side effects and the immunological outcomes of patients.


Asunto(s)
Vacuna BCG , Neoplasias de la Vejiga Urinaria , Humanos , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/patología , Vacuna BCG/uso terapéutico , Vacuna BCG/administración & dosificación , Vacuna BCG/inmunología , Masculino , Femenino , Administración Intravesical , Anciano , Persona de Mediana Edad , Estudios Prospectivos , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/uso terapéutico , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/efectos adversos , Invasividad Neoplásica , Tuberculina/inmunología , Tuberculina/administración & dosificación , Inyecciones Intradérmicas , Resultado del Tratamiento , Neoplasias Vesicales sin Invasión Muscular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA