Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.336
Filtrer
1.
Annu Rev Biomed Eng ; 26(1): 273-306, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38959389

RÉSUMÉ

Nanomaterials are becoming important tools for vaccine development owing to their tunable and adaptable nature. Unique properties of nanomaterials afford opportunities to modulate trafficking through various tissues, complement or augment adjuvant activities, and specify antigen valency and display. This versatility has enabled recent work designing nanomaterial vaccines for a broad range of diseases, including cancer, inflammatory diseases, and various infectious diseases. Recent successes of nanoparticle vaccines during the coronavirus disease 2019 (COVID-19) pandemic have fueled enthusiasm further. In this review, the most recent developments in nanovaccines for infectious disease, cancer, inflammatory diseases, allergic diseases, and nanoadjuvants are summarized. Additionally, challenges and opportunities for clinical translation of this unique class of materials are discussed.


Sujet(s)
COVID-19 , Nanostructures , SARS-CoV-2 , Développement de vaccin , Humains , Nanostructures/composition chimique , COVID-19/prévention et contrôle , SARS-CoV-2/immunologie , Vaccins contre la COVID-19/composition chimique , Animaux , Adjuvants immunologiques/composition chimique , Tumeurs/immunologie , Tumeurs/prévention et contrôle , Nanoparticules/composition chimique , Vaccins , Pandémies/prévention et contrôle
2.
Molecules ; 29(13)2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-38999177

RÉSUMÉ

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.


Sujet(s)
Prolifération cellulaire , ARN double brin , ARN double brin/pharmacologie , ARN double brin/composition chimique , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris , Humains , Lignée cellulaire tumorale , Interféron alpha/métabolisme , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/composition chimique , Interférons/métabolisme
3.
Methods Mol Biol ; 2821: 65-70, 2024.
Article de Anglais | MEDLINE | ID: mdl-38997480

RÉSUMÉ

Conjugation to carrier proteins is necessary for peptides to be able to induce antibody formation when injected into animals together with a suitable adjuvant. This is usually performed by conjugation in solution followed by mixing with the adjuvant. Alternatively, the carrier may be adsorbed onto a solid support followed by activation and conjugation with the peptide by solid-phase chemistry. Different reagents can be used for conjugation through peptide functional groups (-SH, -NH2, -COOH), and various carrier proteins may be used depending on the peptides and the intended use of the antibodies. The solid phase may be an ion exchange matrix, from which the conjugate can subsequently be eluted and mixed with adjuvant. Alternatively, the adjuvant aluminum hydroxide may be used as the solid-phase matrix, whereupon the carrier is immobilized and conjugated with peptide. The resulting adjuvant-carrier-peptide complexes may then be used directly for immunization.


Sujet(s)
Peptides , Peptides/composition chimique , Animaux , Adjuvants immunologiques/composition chimique , Hydroxyde d'aluminium/composition chimique , Protéines de transport/composition chimique , Protéines de transport/métabolisme , Techniques de synthèse en phase solide/méthodes
4.
Methods Mol Biol ; 2821: 111-127, 2024.
Article de Anglais | MEDLINE | ID: mdl-38997484

RÉSUMÉ

Immune stimulants (adjuvants) enhance immune system recognition to provide an effective and individualized immune response when delivered with an antigen. Synthetic cyclic deca-peptides, co-administered with a toll-like receptor targeting lipopeptide, have shown self-adjuvant properties, dramatically boosting the immune response in a murine model as a subunit peptide-based vaccine containing group A Streptococcus peptide antigens.Here, we designed a novel peptide and lipid adjuvant system for the delivery of group A Streptococcus peptide antigen and a T helper peptide epitope. Following linear peptide synthesis on 2-chlorotrityl chloride resin, the linear peptide was cleaved and head-to-tail cyclized in solution. The selective arrangement of amino acids in the deca-peptide allowed for selective conjugation of lipids and/or peptide antigens following cyclisation. Using both solution-phase peptide chemistry and copper-catalyzed azide-alkyne cycloaddition reaction were covalently (and selectively) ligated lipid and/or peptide antigens onto the cyclic deca-peptide core. Subcutaneous administration of the vaccine design to mice resulted in the generation of a large number of serum immunoglobulin (Ig) G antibodies.


Sujet(s)
Adjuvants immunologiques , Immunisation , Peptides cycliques , Vaccins conjugués , Animaux , Souris , Peptides cycliques/immunologie , Peptides cycliques/composition chimique , Vaccins conjugués/immunologie , Vaccins conjugués/composition chimique , Vaccins conjugués/administration et posologie , Immunisation/méthodes , Adjuvants immunologiques/composition chimique , Adjuvants immunologiques/administration et posologie , Injections sous-cutanées , Vaccins sous-unitaires/immunologie , Vaccins sous-unitaires/administration et posologie , Vaccins sous-unitaires/composition chimique , Streptococcus pyogenes/immunologie , Immunoglobuline G/immunologie , Immunoglobuline G/sang , Antigènes bactériens/immunologie , Antigènes bactériens/composition chimique ,
5.
Int J Nanomedicine ; 19: 6589-6602, 2024.
Article de Anglais | MEDLINE | ID: mdl-38979535

RÉSUMÉ

Orthopedic infection is one of the most intractable orthopedic problems. Bacteria resistant to antibiotics also develop gradually. Chitosan is widely used in the Biomedical field because of its high biocompatibility, biodegradability, and antibacterial activity. Chitosan-based drug delivery systems are frequently utilized to produce controlled medication release. When combined with antibiotics, synergistic antibacterial effects can be achieved. Chitosan-based nanoparticles are one of the most widely used applications in drug delivery systems. The focus of this review is to provide information on new methods being developed for chitosan-based nanoparticles in the field of bone infection treatment, including chitosan nanoparticles for antibacterial purposes, Ch-loaded with antibiotics, Ch-loaded with metal, and used as immune adjuvants. It may Provide ideas for the fundamental research and the prospects of future clinical applications of orthopedic infections.


Sujet(s)
Antibactériens , Chitosane , Nanoparticules , Chitosane/composition chimique , Chitosane/pharmacologie , Humains , Nanoparticules/composition chimique , Antibactériens/composition chimique , Antibactériens/pharmacologie , Antibactériens/administration et posologie , Antibactériens/pharmacocinétique , Animaux , Systèmes de délivrance de médicaments/méthodes , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/composition chimique , Adjuvants immunologiques/administration et posologie , Vecteurs de médicaments/composition chimique
6.
Nano Lett ; 24(29): 9017-9026, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39007530

RÉSUMÉ

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.


Sujet(s)
Adjuvants immunologiques , Vaccins anticancéreux , Immunothérapie , Nanoparticules , Vaccins anticancéreux/composition chimique , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Animaux , Souris , Immunothérapie/méthodes , Nanoparticules/composition chimique , Adjuvants immunologiques/composition chimique , Adjuvants immunologiques/administration et posologie , Adjuvants immunologiques/usage thérapeutique , Adjuvants immunologiques/pharmacologie , Hydrogels/composition chimique , Humains , Lignée cellulaire tumorale , Cellules dendritiques/immunologie , Cyclodextrines bêta/composition chimique , Tumeurs/thérapie , Tumeurs/immunologie , Alginates/composition chimique , Adamantane/composition chimique , Adamantane/usage thérapeutique
7.
ACS Appl Bio Mater ; 7(6): 3877-3889, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38832760

RÉSUMÉ

Adjuvants and immunomodulators that effectively drive a Th17-skewed immune response are not part of the standard vaccine toolkit. Vaccine adjuvants and delivery technologies that can induce Th17 or Th1/17 immunity and protection against bacterial pathogens, such as tuberculosis (TB), are urgently needed. Th17-polarized immune response can be induced using agonists that bind and activate C-type lectin receptors (CLRs) such as macrophage inducible C-type lectin (Mincle). A simple but effective strategy was developed for codelivering Mincle agonists with the recombinant Mycobacterium tuberculosis fusion antigen, M72, using tunable silica nanoparticles (SNP). Anionic bare SNP, hydrophobic phenyl-functionalized SNP (P-SNP), and cationic amine-functionalized SNP (A-SNP) of different sizes were coated with three synthetic Mincle agonists, UM-1024, UM-1052, and UM-1098, and evaluated for adjuvant activity in vitro and in vivo. The antigen and adjuvant were coadsorbed onto SNP via electrostatic and hydrophobic interactions, facilitating multivalent display and delivery to antigen presenting cells. The cationic A-SNP showed the highest coloading efficiency for the antigen and adjuvant. In addition, the UM-1098-adsorbed A-SNP formulation demonstrated slow-release kinetics in vitro, excellent stability over 12 months of storage, and strong IL-6 induction from human peripheral blood mononuclear cells. Co-adsorption of UM-1098 and M72 on A-SNP significantly improved antigen-specific humoral and Th17-polarized immune responses in vivo in BALB/c mice relative to the controls. Taken together, A-SNP is a promising platform for codelivery and proper presentation of adjuvants and antigens and provides the basis for their further development as a vaccine delivery platform for immunization against TB or other diseases for which Th17 immunity contributes to protection.


Sujet(s)
Antigènes bactériens , Lectines de type C , Nanoparticules , Silice , Cellules Th17 , Lectines de type C/métabolisme , Lectines de type C/immunologie , Lectines de type C/agonistes , Nanoparticules/composition chimique , Cellules Th17/immunologie , Animaux , Silice/composition chimique , Souris , Antigènes bactériens/immunologie , Antigènes bactériens/administration et posologie , Antigènes bactériens/composition chimique , Mycobacterium tuberculosis/immunologie , Adjuvants immunologiques/composition chimique , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/administration et posologie , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Taille de particule , Test de matériaux , Humains , Femelle , Protéines membranaires/immunologie , Protéines membranaires/agonistes
8.
Int J Biol Macromol ; 272(Pt 2): 132913, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38851606

RÉSUMÉ

Nasal vaccine is a non-invasive vaccine that activates systemic and mucosal immunity in the presence of an adjuvant, thereby enhancing immune function. In this work, chitosan/oligochitosan/tween 80 (CS-COS-T80) co-stabilized emulsion was designed and further used as the nasal adjuvant. CS-COS-T80 emulsion exhibited outstanding stability under pH 6-8 with uniformly dispersed droplets and nano-scale particle size (<0.25 µm), and maintained stable at 4 °C for 150-day storage. Addition of model antigen ovalbumin (OVA) had no effect on the stability of CS-COS-T80 emulsion. In vivo nasal immunity indicated that CS-COS-T80 emulsion prolonged the retention time of OVA in the nasal cavity (from 4 to 8 h to >12 h), as compared to T80-emulsion. CS-COS-T80 emulsion produced a stronger mucosal immune response to OVA, with secretory IgA levels 5-fold and 2-fold higher than those of bare OVA and commercial adjuvant MF59, respectively. Compared to MF59, CS-COS-T80 induced a stronger humoral immune response and a mixed Th1/Th2 immune response of OVA after immunization. Furthermore, in the presence of CS-COS-T80 emulsion, the secretion of IL-4 and IFN-γ and the activation of splenocyte memory T-cell differentiation increased from 173.98 to 210.21 pg/mL and from 75.46 to 104.01 pg/mL, respectively. Therefore, CS-COS-T80 emulsion may serve as a promising adjuvant platform.


Sujet(s)
Adjuvants immunologiques , Chitosane , Émulsions , Immunité muqueuse , Muqueuse nasale , Ovalbumine , Chitosane/composition chimique , Animaux , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/composition chimique , Immunité muqueuse/effets des médicaments et des substances chimiques , Souris , Ovalbumine/immunologie , Ovalbumine/composition chimique , Muqueuse nasale/immunologie , Femelle , Administration par voie nasale , Souris de lignée BALB C , Cytokines/métabolisme , Taille de particule , Oligosaccharides
9.
ACS Nano ; 18(24): 15557-15575, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38837909

RÉSUMÉ

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.


Sujet(s)
Vaccins anticancéreux , Immunothérapie , Nanoparticules , Polyesters , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/composition chimique , Animaux , Souris , Polyesters/composition chimique , Nanoparticules/composition chimique , Souris de lignée C57BL , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Cellules dendritiques/immunologie , Femelle , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/composition chimique , Lipides/composition chimique , Humains , Tumeurs/thérapie , Tumeurs/immunologie , Lignée cellulaire tumorale , Petit ARN interférent/composition chimique , Acide hyaluronique/composition chimique ,
10.
J Med Chem ; 67(12): 9976-9990, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38886162

RÉSUMÉ

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.


Sujet(s)
Adjuvants immunologiques , Vaccins anticancéreux , Conception de médicament , Lipide A , Souris de lignée C57BL , Animaux , Lipide A/analogues et dérivés , Lipide A/composition chimique , Lipide A/pharmacologie , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/pharmacologie , Vaccins anticancéreux/synthèse chimique , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/synthèse chimique , Adjuvants immunologiques/composition chimique , Souris , Souris knockout , Humains , Femelle , Récepteur de type Toll-4/métabolisme , Lignée cellulaire tumorale
11.
ACS Nano ; 18(26): 16878-16894, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38899978

RÉSUMÉ

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.


Sujet(s)
Adjuvants immunologiques , Hydroxyde d'aluminium , Calcium , Antigènes de surface du virus de l'hépatite B , Silicium , Adjuvants immunologiques/composition chimique , Adjuvants immunologiques/pharmacologie , Animaux , Silicium/composition chimique , Souris , Antigènes de surface du virus de l'hépatite B/immunologie , Antigènes de surface du virus de l'hépatite B/composition chimique , Calcium/composition chimique , Hydroxyde d'aluminium/composition chimique , Hydroxyde d'aluminium/pharmacologie , Souris de lignée C57BL , Femelle , Vaccins/immunologie , Vaccins/composition chimique , Cellules dendritiques/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Humains , Oxyde d'aluminium
12.
ACS Nano ; 18(26): 16589-16609, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38885198

RÉSUMÉ

Adjuvants are effective tools to enhance vaccine efficacy and control the type of immune responses such as antibody and T helper 1 (Th1)- or Th2-type responses. Several studies suggest that interferon (IFN)-γ-producing Th1 cells play a significant role against infections caused by intracellular bacteria and viruses; however, only a few adjuvants can induce a strong Th1-type immune response. Recently, several studies have shown that lipid nanoparticles (LNPs) can be used as vaccine adjuvants and that each LNP has a different adjuvant activity. In this study, we screened LNPs to develop an adjuvant that can induce Th1 cells and antibodies using a conventional influenza split vaccine (SV) as an antigen in mice. We observed that LNP with 1,2-di-O-octadecenyl-3-trimethylammonium-propane (DOTMA) as a component lipid (DOTMA-LNP) elicited robust SV-specific IgG1 and IgG2 responses compared with SV alone in mice and was as efficient as SV adjuvanted with other adjuvants in mice. Furthermore, DOTMA-LNPs induced robust IFN-γ-producing Th1 cells without inflammatory responses compared to those of other adjuvants, which conferred strong cross-protection in mice. We also demonstrated the high versatility of DOTMA-LNP as a Th1 cell-inducing vaccine adjuvant using vaccine antigens derived from severe acute respiratory syndrome coronavirus 2 and Streptococcus pneumoniae. Our findings suggest the potential of DOTMA-LNP as a safe and effective Th1 cell-inducing adjuvant and show that LNP formulations are potentially potent adjuvants to enhance the effectiveness of other subunit vaccines.


Sujet(s)
Nanoparticules , Composés d'ammonium quaternaire , Lymphocytes auxiliaires Th1 , Animaux , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Souris , Composés d'ammonium quaternaire/composition chimique , Composés d'ammonium quaternaire/pharmacologie , Femelle , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/composition chimique , Lipides/composition chimique , Souris de lignée BALB C , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/composition chimique , Adjuvants vaccinaux/composition chimique , Adjuvants vaccinaux/pharmacologie , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/composition chimique , COVID-19/prévention et contrôle , COVID-19/immunologie , Liposomes
13.
Int J Biol Macromol ; 273(Pt 1): 133067, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38866287

RÉSUMÉ

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.


Sujet(s)
Adjuvants immunologiques , Cellules dendritiques , Polyosides , Solubilité , Eau , Animaux , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/composition chimique , Polyosides/pharmacologie , Polyosides/composition chimique , Polyosides/isolement et purification , Souris , Eau/composition chimique , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Femelle , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Wolfiporia/composition chimique , Ovalbumine/immunologie , Immunité cellulaire/effets des médicaments et des substances chimiques , Immunité humorale/effets des médicaments et des substances chimiques , Poria/composition chimique
14.
Int J Biol Macromol ; 270(Pt 1): 132258, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38735613

RÉSUMÉ

Covalently linking an adjuvant to an antigenic protein enhances its immunogenicity by ensuring a synergistic delivery to the immune system, fostering a more robust and targeted immune response. Most adjuvant-protein conjugate vaccines incorporate only one adjuvant due to the difficulties in its synthesis. However, there is a growing interest in developing vaccines with multiple adjuvants designed to elicit a more robust and targeted immune response by engaging different aspects of the immune system for complex diseases where traditional vaccines fall short. Here, we pioneer the synthesis of a dual-adjuvants protein conjugate Vaccine 1 by assembling a toll-like receptor 7/8 (TLR7/8) agonist, an invariant natural killer T cell (iNKT) agonist with a clickable bicyclononyne (BCN). The BCN group can bio-orthogonally react with azide-modified severe acute respiratory syndrome coronavirus-2 receptor-binding domain (SARS-CoV-2 RBD) trimer antigen to give the three-component Vaccine 1. Notably, with a mere 3 µg antigen, it elicited a balanced subclass of IgG titers and 20-fold more IgG2a than control vaccines, highlighting its potential for enhancing antibody-dependent cellular cytotoxicity. This strategy provides a practicable way to synthesize covalently linked dual immunostimulants. It expands the fully synthetic self-adjuvant protein vaccine that uses a single adjuvant to include two different types of adjuvants.


Sujet(s)
Adjuvants immunologiques , Vaccins contre la COVID-19 , COVID-19 , Cellules T tueuses naturelles , SARS-CoV-2 , Récepteur de type Toll-7 , Récepteur de type Toll-8 , Récepteur de type Toll-7/agonistes , Récepteur de type Toll-7/immunologie , SARS-CoV-2/immunologie , Animaux , Cellules T tueuses naturelles/immunologie , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/pharmacologie , Récepteur de type Toll-8/agonistes , Récepteur de type Toll-8/immunologie , Humains , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/composition chimique , Souris , COVID-19/prévention et contrôle , COVID-19/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Anticorps antiviraux/immunologie , Femelle , Adjuvants vaccinaux/composition chimique , Adjuvants vaccinaux/pharmacologie , Immunoglobuline G/immunologie
15.
J Med Chem ; 67(11): 9389-9405, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38787938

RÉSUMÉ

TLR7/8 agonists are versatile immune stimulators capable of treating various diseases such as viral infections, autoimmune, and cancer. Despite the structural similarity of TLR7/8, their immune stimulation mechanisms and time-course responses significantly differ. In this study, a new series of TLR7-selective agonists was synthesized utilizing the economical building block 2,6-dichloropurine. Compound 27b showed the most potent activity on hTLR7 with an EC50 of 17.53 nM and demonstrated high hTLR7 selectivity (224 folds against TLR8). 27b effectively stimulated the secretion of proinflammatory cytokines in mouse macrophages and enhanced intranasal vaccine efficacy against influenza A virus in vivo. Assessment of humoral and mucosal antibody titers confirmed that 27b elevates IgG and IgA levels, protecting against both homologous and heterologous influenza viral infections. These findings suggest that 27b is a promising candidate as a vaccine adjuvant to prevent viral infections or as a robust immunomodulator with prolonged activity for treating immune-suppressed diseases.


Sujet(s)
Administration par voie nasale , Conception de médicament , Vaccins antigrippaux , Purines , Récepteur de type Toll-7 , Récepteur de type Toll-7/agonistes , Animaux , Souris , Humains , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Purines/pharmacologie , Purines/composition chimique , Adjuvants vaccinaux/pharmacologie , Adjuvants vaccinaux/composition chimique , Relation structure-activité , Souris de lignée BALB C , Femelle , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Cytokines/métabolisme , Cellules RAW 264.7 , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/synthèse chimique , Adjuvants immunologiques/composition chimique
16.
Carbohydr Res ; 541: 109166, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38815341

RÉSUMÉ

Triacedimannose (TADM) is a synthetic trivalent acetylated glycocluster comprising ß-1,2-linked mannobioses that in humans induces TNF in vitro and in vivo. The purpose of this study was to analyze whether uptake of acetylated glycoclusters of such ß-1,2-linked mannobioses by human macrophages is dependent on the mannose receptor (CD206) or if it is mediated by transmembrane activation. In mannose receptor blocking assays, monocyte-derived polarized macrophages were incubated with carbohydrate test-compounds and their binding to the mannose receptor was demonstrated as inhibition of FITC-Dextran binding. For 1H NMR spectroscopy, macrophages were incubated with TADM. The cells were collected at 6 and 24 h of incubation, centrifuged and washed twice with PBS. We found dose-dependent blocking of the mannose receptor in macrophage carbohydrate constructs containing free hydroxyl groups, but not by the trivalent acetylated glycocluster molecules. NMR spectroscopic analyses demonstrated that TADM was found in washed cellular pellets after 6-h co-culture, while after 24-h co-culture TADM was no more detectable, suggesting cleavage of the acetyl groups in vitro. The Type 1 immune response enhancing effects of TADM and other, stereochemically and structurally similar, trivalent acetylated glycoclusters may be due to transmembrane uptake of macrophages independent of the mannose receptor.


Sujet(s)
Lectines de type C , Macrophages , Récepteur du mannose , Lectines liant le mannose , Récepteurs de surface cellulaire , Lectines de type C/métabolisme , Lectines de type C/composition chimique , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Récepteurs de surface cellulaire/métabolisme , Lectines liant le mannose/métabolisme , Lectines liant le mannose/composition chimique , Humains , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/composition chimique , Acétylation
17.
Nature ; 629(8013): 937-944, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38720067

RÉSUMÉ

QS-21 is a potent vaccine adjuvant and remains the only saponin-based adjuvant that has been clinically approved for use in humans1,2. However, owing to the complex structure of QS-21, its availability is limited. Today, the supply depends on laborious extraction from the Chilean soapbark tree or on low-yielding total chemical synthesis3,4. Here we demonstrate the complete biosynthesis of QS-21 and its precursors, as well as structural derivatives, in engineered yeast strains. The successful biosynthesis in yeast requires fine-tuning of the host's native pathway fluxes, as well as the functional and balanced expression of 38 heterologous enzymes. The required biosynthetic pathway spans seven enzyme families-a terpene synthase, P450s, nucleotide sugar synthases, glycosyltransferases, a coenzyme A ligase, acyl transferases and polyketide synthases-from six organisms, and mimics in yeast the subcellular compartmentalization of plants from the endoplasmic reticulum membrane to the cytosol. Finally, by taking advantage of the promiscuity of certain pathway enzymes, we produced structural analogues of QS-21 using this biosynthetic platform. This microbial production scheme will allow for the future establishment of a structure-activity relationship, and will thus enable the rational design of potent vaccine adjuvants.


Sujet(s)
Adjuvants immunologiques , Génie métabolique , Saccharomyces cerevisiae , Saponines , Adjuvants immunologiques/biosynthèse , Adjuvants immunologiques/composition chimique , Adjuvants immunologiques/génétique , Adjuvants immunologiques/métabolisme , Voies de biosynthèse/génétique , Conception de médicament , Enzymes/génétique , Enzymes/métabolisme , Génie métabolique/méthodes , Plantes/enzymologie , Plantes/génétique , Plantes/métabolisme , Saccharomyces cerevisiae/cytologie , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Saponines/biosynthèse , Saponines/composition chimique , Saponines/génétique , Saponines/métabolisme , Relation structure-activité
18.
J Control Release ; 370: 691-706, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38723671

RÉSUMÉ

Vaccination is essential for preventing and controlling infectious diseases, along with reducing mortality. Developing safe and versatile adjuvants to enhance humoral and cellular immune responses to vaccines remains a key challenge in vaccine development. Here, we designed hierarchical mesoporous MOF-801 (HM801) using a Cocamidopropyl betaine (CAPB) and a Pluronics F127 in an aqueous phase system. Meanwhile, we synthesized a novel SARS-CoV-2 nanovaccine (R@M@HM801) with a high loading capacity for both the STING agonist (MSA-2) and the Delta receptor binding domain (Delta-RBD) antigen. R@M@HM801 enhanced MSA-2 and RBD utilization and effectively co-delivered MSA-2 and RBD antigens to antigen-presenting cells in the draining lymph nodes, thereby promoting the activation of both T and B cells. Lymphocyte single-cell analysis showed that R@M@HM801 stimulated robust CD11b+CD4+ T cells, CXCR5+CD4+ T follicular helper (Tfh), and durable CD4+CD44+CD62L-, CD8+CD44+CD62L- effector memory T cell (TEM) immune responses, and promoted the proliferative activation of CD26+ B cells in vivo. Meanwhile, R@M@HM801 induced stronger specific antibodies and neutralization of pseudovirus against Delta compared to the RBD + MAS-2 and RBD + MAS-2 + Alum vaccines. Our study demonstrated the efficacy of a hierarchical mesoporous HM801 and its potential immune activation mechanism in enhancing adaptive immune responses against viruses and other diseases.


Sujet(s)
Adjuvants immunologiques , Immunité cellulaire , Immunité humorale , Protéines membranaires , Réseaux organométalliques , Animaux , Immunité humorale/effets des médicaments et des substances chimiques , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/administration et posologie , Adjuvants immunologiques/composition chimique , Immunité cellulaire/effets des médicaments et des substances chimiques , Protéines membranaires/immunologie , Souris , Réseaux organométalliques/composition chimique , Femelle , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , SARS-CoV-2/immunologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Porosité , Souris de lignée C57BL , Lymphocytes B/immunologie , Lymphocytes B/effets des médicaments et des substances chimiques
19.
Bioconjug Chem ; 35(6): 804-815, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38718344

RÉSUMÉ

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.


Sujet(s)
Adjuvants immunologiques , Or , Nanoparticules métalliques , Oligodésoxyribonucléotides , Oligodésoxyribonucléotides/composition chimique , Oligodésoxyribonucléotides/pharmacologie , Or/composition chimique , Nanoparticules métalliques/composition chimique , Animaux , Adjuvants immunologiques/composition chimique , Adjuvants immunologiques/pharmacologie , Souris , Sucres/composition chimique , Humains , Souris de lignée C57BL
20.
J Nanobiotechnology ; 22(1): 267, 2024 May 19.
Article de Anglais | MEDLINE | ID: mdl-38764014

RÉSUMÉ

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.


Sujet(s)
Adjuvants immunologiques , Mélanome expérimental , Souris de lignée C57BL , Ovalbumine , Probiotiques , Animaux , Ovalbumine/immunologie , Ovalbumine/composition chimique , Souris , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/composition chimique , Probiotiques/pharmacologie , Mélanome expérimental/immunologie , Femelle , Cellules dendritiques/immunologie , Récepteur de type Toll-4/métabolisme , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/composition chimique , Lymphocytes T CD8+/immunologie , Lignée cellulaire tumorale , Humains , Nanoparticules/composition chimique , Lymphocytes T CD4+/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE