Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42.588
Filter
1.
Biomaterials ; 312: 122751, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39121726

ABSTRACT

Tumor immunotherapies have emerged as a promising frontier in the realm of cancer treatment. However, challenges persist in achieving localized, durable immunostimulation while counteracting the tumor's immunosuppressive environment. Here, we develop a natural mussel foot protein-based nanomedicine with spatiotemporal control for tumor immunotherapy. In this nanomedicine, an immunoadjuvant prodrug and a photosensitizer are integrated, which is driven by their dynamic bonding and non-covalent assembling with the protein carrier. Harnessing the protein carrier's bioadhesion, this nanomedicine achieves a drug co-delivery with spatiotemporal precision, by which it not only promotes tumor photothermal ablation but also broadens tumor antigen repertoire, facilitating in situ immunotherapy with durability and maintenance. This nanomedicine also modulates the tumor microenvironment to overcome immunosuppression, thereby amplifying antitumor responses against tumor progression. Our strategy underscores a mussel foot protein-derived design philosophy of drug delivery aimed at refining combinatorial immunotherapy, offering insights into leveraging natural proteins for cancer treatment.


Subject(s)
Immunotherapy , Nanomedicine , Animals , Immunotherapy/methods , Nanomedicine/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/pharmacology , Photothermal Therapy/methods , Mice , Humans , Tumor Microenvironment/drug effects , Cell Line, Tumor , Proteins/chemistry , Female , Neoplasms/therapy , Neoplasms/immunology , Adhesives/chemistry , Mice, Inbred C57BL , Adjuvants, Immunologic/pharmacology
2.
Vaccine ; 42(24): 126269, 2024 Oct 24.
Article in English | MEDLINE | ID: mdl-39241354

ABSTRACT

Recombinant influenza virus neuraminidase (NA) is a promising broadly protective influenza vaccine candidate. However, the recombinant protein alone is not sufficient to induce durable and protective immune responses and requires the coadministration of immunostimulatory molecules. Here, we evaluated the immunogenicity and cross-protective potential of a recombinant influenza virus N2 neuraminidase vaccine construct, adjuvanted with a toll-like receptor 9 (TLR9) agonist (CpG 1018® adjuvant), and alum. The combination of CpG 1018 adjuvant and alum induced a balanced and robust humoral and T-cellular immune response against the NA, which provided protection and reduced morbidity against homologous and heterologous viral challenges in mouse and hamster models. This study supports Syrian hamsters as a useful complementary animal model to mice for pre-clinical evaluation of influenza virus vaccines.


Subject(s)
Adjuvants, Immunologic , Antibodies, Viral , Influenza Vaccines , Neuraminidase , Orthomyxoviridae Infections , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Neuraminidase/immunology , Neuraminidase/genetics , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Mice , Adjuvants, Immunologic/administration & dosage , Female , Cricetinae , Antibodies, Viral/immunology , Antibodies, Viral/blood , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Adjuvants, Vaccine , Mice, Inbred BALB C , Cross Protection/immunology , Mesocricetus , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Alum Compounds/administration & dosage , Disease Models, Animal , Immunity, Cellular
3.
Microb Pathog ; 195: 106913, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39236968

ABSTRACT

Bacterial septicemia represents a significant disease affecting cultured grass carp culture, with the primary etiological agent identified as the Gram-negative bacterium Aeromonas veronii. In response to an outbreak of septicemia in Guangzhou, we developed a formaldehyde-inactivated vaccine against an A. veronii strain designated AV-GZ21-2. This strain exhibited high pathogenicity in experimental infections across at all developmental stages of grass carp. Mortality rates for grass carp weighing 15 ± 5 g ranged from 16 % to 92 % at exposure temperatures of 19 °C-34 °C, respectively. The median lethal dose (LD50) for grass carp groups weighing 15 ± 5 g, 60 ± 10 g, 150 ± 30 g and 500 ± 50 g were determined to be 1.43, 2.52, 4.65 and 7.12 × 107(CFU/mL), respectively. We investigated the inactivated vaccine in conbination with aluminum hydroxide gel (AV-AHG), Montanide ISA201VG (AV-201VG), and white oil (AV-WO) adjuvants. This study aimed to optimize inactivation conditions and identify the adjuvant that elicits the most robust immune response. The AV-GZ21-2 inactivated bacterial solution (AV),when combined with various adjuvants, was capable of inducing a strong specific immune response in grass carp. The relative percent survival (RPS) following a lethal challenge with AV-GZ21-2 were 94 % for AV-AHG, 88 % for AV-201VG, 84 % for AV-WO and 78 % for AV alone. The minimum immunization dose of the AV-AHG vaccine was determined to be 6.0 × 107 CFU per fish, providing immunity for a duration of six months with an immune protection level exceeding 75 %. Furthermore, the AV-AHG vaccine demonstrated significant protective efficacy against various epidemic isolates of A. veronii. Consequently, we developed an inactivated vaccine targeting a highly pathogenic strain of A. veronii, incorporating an aluminum hydroxide gel adjuvant, which resulted in high immune protection and a duration of immunity exceeding six months. These findings suggest that the AV-AHG vaccine holds substantial potential for industrial application.


Subject(s)
Adjuvants, Immunologic , Aeromonas veronii , Bacterial Vaccines , Carps , Fish Diseases , Gram-Negative Bacterial Infections , Vaccines, Inactivated , Animals , Carps/microbiology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Aeromonas veronii/immunology , Fish Diseases/prevention & control , Fish Diseases/microbiology , Fish Diseases/immunology , Gram-Negative Bacterial Infections/prevention & control , Gram-Negative Bacterial Infections/veterinary , Gram-Negative Bacterial Infections/immunology , Virulence , Adjuvants, Immunologic/administration & dosage , Lethal Dose 50 , Temperature , China/epidemiology , Aluminum Hydroxide/administration & dosage
4.
Microb Cell Fact ; 23(1): 250, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39272136

ABSTRACT

BACKGROUND: Bordetella pertussis is the causative agent of whooping cough or pertussis. Although both acellular (aP) and whole-cell pertussis (wP) vaccines protect against disease, the wP vaccine, which is highly reactogenic, is better at preventing colonization and transmission. Reactogenicity is mainly attributed to the lipid A moiety of B. pertussis lipooligosaccharide (LOS). Within LOS, lipid A acts as a hydrophobic anchor, engaging with TLR4-MD2 on host immune cells to initiate both MyD88-dependent and TRIF-dependent pathways, thereby influencing adaptive immune responses. Lipid A variants, such as monophosphoryl lipid A (MPLA) can also act as adjuvants. Adjuvants may overcome the shortcomings of aP vaccines. RESULTS: This work used lipid A modifying enzymes from other bacteria to produce an MPLA-like adjuvant strain in B. pertussis. We created B. pertussis strains with distinct lipid A modifications, which were validated using MALDI-TOF. We engineered a hexa-acylated monophosphorylated lipid A that markedly decreased human TLR4 activation and activated the TRIF pathway. The modified lipooligosaccharide (LOS) promoted IRF3 phosphorylation and type I interferon production, similar to MPLA responses. We generated three other variants with increased adjuvanticity properties and reduced endotoxicity. Pyrogenicity studies using the Monocyte Activation Test (MAT) revealed that these four lipid A variants significantly decreased the IL-6, a marker for fever, response in peripheral blood mononuclear cells (PBMCs). CONCLUSION: These findings pave the way for developing wP vaccines that are possibly less reactogenic and designing adaptable adjuvants for current vaccine formulations, advancing more effective immunization strategies against pertussis.


Subject(s)
Adjuvants, Immunologic , Bordetella pertussis , Lipid A , Toll-Like Receptor 4 , Lipid A/analogs & derivatives , Lipid A/immunology , Bordetella pertussis/immunology , Humans , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/immunology , Adjuvants, Immunologic/pharmacology , Adaptor Proteins, Vesicular Transport/metabolism , Adaptor Proteins, Vesicular Transport/immunology , Pertussis Vaccine/immunology , Lipopolysaccharides , Interferon Regulatory Factor-3/metabolism , Whooping Cough/prevention & control , Whooping Cough/immunology , Interleukin-6/metabolism , Interleukin-6/immunology
5.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273405

ABSTRACT

SpikoGen® vaccine is a subunit COVID-19 vaccine composed of an insect cell expressed recombinant spike protein extracellular domain formulated with Advax-CpG55.2™ adjuvant. A randomized double-blind, placebo-controlled Phase II clinical trial was conducted in 400 adult subjects who were randomized 3:1 to receive two intramuscular doses three weeks apart of either SpikoGen® vaccine 25 µg or saline placebo, as previously reported. This study reports a post hoc analysis of the trial data to explore potential immune correlates of SpikoGen® vaccine protection. A range of humoral markers collected pre- and post-vaccination, including spike- and RBD-binding IgG and IgA, surrogate (sVNT), and conventional (cVNT) virus neutralization tests were compared between participants who remained infection-free or got infected over three months of follow-up. From 2 weeks after the second vaccine dose, 21 participants were diagnosed with SARS-CoV-2 infection, 13 (4.2%) in the SpikoGen® group and 8 (9%) in the placebo group. Those in the vaccinated group who experienced breakthrough infections had significantly lower sVNT titers (GMT 5.75 µg/mL, 95% CI; 3.72-8.91) two weeks after the second dose (day 35) than those who did not get infected (GMT 21.06 µg/mL, 95% CI; 16.57-26.76). Conversely, those who did not develop SARS-CoV-2 infection during follow-up had significantly higher baseline sVNT, cVNT, spike-binding IgG and IgA, and RBD-binding IgG, consistent with a past SARS-CoV-2 infection. SpikoGen® further reduced the risk of re-infection (OR 0.29) in baseline seropositive (previously infected) as well as baseline seronegative participants. This indicates that while SpikoGen vaccine is protective in seronegative individuals, those with hybrid immunity have the most robust protection.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , Spike Glycoprotein, Coronavirus/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , SARS-CoV-2/immunology , Female , Male , Adult , Antibodies, Viral/immunology , Antibodies, Viral/blood , Middle Aged , Double-Blind Method , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin A/immunology , Immunoglobulin A/blood , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Adjuvants, Vaccine , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Adjuvants, Immunologic/administration & dosage , Aged
6.
Eur J Med Chem ; 278: 116792, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39217861

ABSTRACT

Enhancing the efficacy of subunit vaccines relies significantly on the utilization of potent adjuvants, particularly those capable of triggering multiple immune pathways. To achieve synergistic immune augmentation by Toll-like receptor 4 agonist (TLR4a) and nucleotide-binding oligomerization-domain-containing protein 2 agonist (NOD2a), in this work, we conjugated RC529 (TLR4a) and MDP (NOD2a) to give RC529-MDP, and evaluated its adjuvanticity for OVA antigen. Compared to the unconjugated RC529+MDP, RC529-MDP remarkably enhanced innate immune responses with 6.8-fold increase in IL-6 cytokine, and promoted the maturation of antigen-presenting cells (APCs), possibly because of the conjugation of multiple agonists ensuring their delivery to the same cell and activation of various signaling pathways within that cell. Furthermore, RC529-MDP improved OVA-specific antibody response, T cells response and the memory T cells ratio relative to the unconjugated mixture. Therefore, covalently conjugating TLR4 agonist and NOD2 agonist was an effective strategy to enhance immune responses, providing the potential to design and develop more effective vaccines.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine , Adjuvants, Immunologic , Nod2 Signaling Adaptor Protein , Toll-Like Receptor 4 , Nod2 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/metabolism , Toll-Like Receptor 4/agonists , Animals , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemistry , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Acetylmuramyl-Alanyl-Isoglutamine/chemistry , Mice , Ovalbumin/immunology , Humans , Mice, Inbred C57BL , Molecular Structure , Dose-Response Relationship, Drug , Structure-Activity Relationship
7.
Vaccine ; 42(24): 126251, 2024 Oct 24.
Article in English | MEDLINE | ID: mdl-39226786

ABSTRACT

This phase 1 trial assessed the safety and immunogenicity of an investigational tetanus/diphtheria/acellular pertussis vaccine combined with CpG 1018 adjuvant 1500 µg (Tdap-1018 1500 µg) or 3000 µg (Tdap-1018 3000 µg) in adults and adolescents. In this randomized, active-controlled, multicenter, dose-escalation trial, healthy participants aged 10 to 22 years received 1 dose of Tdap-1018 1500 µg, Tdap-1018 3000 µg, or Boostrix. Geometric mean concentrations (GMCs) and booster response rates (BRRs) for antibodies against pertussis (pertussis toxin, filamentous hemagglutinin, pertactin), tetanus, and diphtheria antigens, and neutralizing antibodies against pertussis toxin were assessed 4 weeks after vaccination. Safety and tolerability were assessed for solicited post-injection reactions within 7 days after vaccination and unsolicited adverse events up to 12 weeks after vaccination. Of 117 enrolled participants, 80 adults (92%) and 30 adolescents (100%) completed the study. Both Tdap-1018 formulations were generally well tolerated, with no vaccine-related serious adverse events. Frequency and severity in post-injection reactions after Tdap-1018 administration were similar to Boostrix except for higher proportions of moderate pain for Tdap-1018. In adults at week 4, ratio of GMCs and BRRs for all antigens in the 3000-µg group were similar to or higher than Boostrix, with significantly higher GMC ratios for anti-pertussis toxin (2.1 [1.5-3.0]) and anti-tetanus (1.8 [1.1-2.9]) and significantly higher BRRs for anti-pertussis toxin (difference [95% CI]: 34.5% [13.4-54.6]), anti-pertactin (19.2% [4.4-38.1]), and anti-tetanus (30.0% [3.6-52.7]) antibodies. For adolescents, in the 3000-µg group, ratio of GMCs and BRRs were similar to or higher than Boostrix for all antigens. Both Tdap-1018 formulations showed acceptable safety and tolerability profiles. Tdap-1018 3000 µg induced similar or higher immune responses than Boostrix. ACTRN12620001177943 (Australian New Zealand Clinical Trials Registry; https://anzctr.org.au/Trial/Registration/TrialReview.aspx?ACTRN=ACTRN12620001177943p).


Subject(s)
Adjuvants, Immunologic , Antibodies, Bacterial , Diphtheria-Tetanus-acellular Pertussis Vaccines , Immunization, Secondary , Oligodeoxyribonucleotides , Whooping Cough , Humans , Adolescent , Female , Male , Antibodies, Bacterial/blood , Immunization, Secondary/methods , Adult , Young Adult , Diphtheria-Tetanus-acellular Pertussis Vaccines/immunology , Diphtheria-Tetanus-acellular Pertussis Vaccines/administration & dosage , Diphtheria-Tetanus-acellular Pertussis Vaccines/adverse effects , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Child , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Whooping Cough/prevention & control , Whooping Cough/immunology , Antibodies, Neutralizing/blood , Diphtheria-Tetanus-Pertussis Vaccine/immunology , Diphtheria-Tetanus-Pertussis Vaccine/administration & dosage , Diphtheria-Tetanus-Pertussis Vaccine/adverse effects , Tetanus/prevention & control , Tetanus/immunology , Healthy Volunteers , Immunogenicity, Vaccine
8.
Proc Natl Acad Sci U S A ; 121(37): e2320482121, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39226349

ABSTRACT

Oral delivery of proteins faces challenges due to the harsh conditions of the gastrointestinal (GI) tract, including gastric acid and intestinal enzyme degradation. Permeation enhancers are limited in their ability to deliver proteins with high molecular weight and can potentially cause toxicity by opening tight junctions. To overcome these challenges, we propose the use of montmorillonite (MMT) as an adjuvant that possesses both inflammation-oriented abilities and the ability to regulate gut microbiota. This adjuvant can be used as a universal protein oral delivery technology by fusing with advantageous binding amino acid sequences. We demonstrated that anti-TNF-α nanobody (VII) can be intercalated into the MMT interlayer space. The carboxylate groups (-COOH) of aspartic acid (D) and glutamic acid (E) interact with the MMT surface through electrostatic interactions with sodium ions (Na+). The amino groups (NH2) of asparagine (N) and glutamine (Q) are primarily attracted to the MMT layers through hydrogen bonding with oxygen atoms on the surface. This binding mechanism protects VII from degradation and ensures its release in the intestinal tract, as well as retaining biological activity, leading to significantly enhanced therapeutic effects on colitis. Furthermore, VII@MMT increases the abundance of short-chain fatty acids (SCFAs)-producing strains, including Clostridia, Prevotellaceae, Alloprevotella, Oscillospiraceae, Clostridia_vadinBB60_group, and Ruminococcaceae, therefore enhance the production of SCFAs and butyrate, inducing regulatory T cells (Tregs) production to modulate local and systemic immune homeostasis. Overall, the MMT adjuvant provides a promising universal strategy for protein oral delivery by rational designed protein.


Subject(s)
Bentonite , Gastrointestinal Microbiome , Tumor Necrosis Factor-alpha , Bentonite/chemistry , Animals , Administration, Oral , Tumor Necrosis Factor-alpha/metabolism , Mice , Gastrointestinal Microbiome/drug effects , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Single-Domain Antibodies/administration & dosage , Single-Domain Antibodies/immunology , Single-Domain Antibodies/pharmacology , Humans , Inflammation/drug therapy , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology
9.
Dev Comp Immunol ; 161: 105260, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39237067

ABSTRACT

This study investigates the prolonged effect of immune disease resistance in Litopenaeus vannamei through the administration of tyramine (TA) formulated with polyethylene glycol (PEG). Facing the challenges of intensive farming, environmental stress, and global climate changes, innovative approaches to improve shrimp health are essential. The research focuses on the role of biogenic amines in stress response and immune regulation, demonstrating that TA, especially when combined with PEG, significantly prolongs immunity and resistance against Vibrio alginolyticus. The experimental design included administering TA, PEG, and TA-PEG, followed by evaluations of immunity, lactate and glucose levels, and immune-related gene expressions. Results showed notable prolonged effects in total hemocyte count, phenoloxidase activity, and phagocytic activity in the TA-PEG group, indicating enhanced immune activation period. Additionally, the expression of prophenoloxidase system-related genes was significantly upregulated in the TA-PEG group. Furthermore, the TA-PEG group exhibited a significantly higher survival rate in a susceptibility test against V. alginolyticus. The results of this study confirm that the combined use of PEG can effectively extend the immunostimulatory duration of TA.


Subject(s)
Disease Resistance , Hemocytes , Penaeidae , Polyethylene Glycols , Tyramine , Vibrio alginolyticus , Animals , Penaeidae/immunology , Polyethylene Glycols/chemistry , Polyethylene Glycols/administration & dosage , Vibrio alginolyticus/immunology , Vibrio alginolyticus/physiology , Disease Resistance/immunology , Disease Resistance/genetics , Hemocytes/immunology , Catechol Oxidase/metabolism , Immunity, Innate , Vibrio Infections/immunology , Enzyme Precursors/metabolism , Enzyme Precursors/genetics , Phagocytosis , Arthropod Proteins/genetics , Arthropod Proteins/metabolism , Arthropod Proteins/immunology , Adjuvants, Immunologic/administration & dosage
10.
J Exp Med ; 221(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39235529

ABSTRACT

Stabilized trimers preserving the native-like HIV envelope structure may be key components of a preventive HIV vaccine regimen to induce broadly neutralizing antibodies (bnAbs). We evaluated trimeric BG505 SOSIP.664 gp140 formulated with a novel TLR7/8 signaling adjuvant, 3M-052-AF/Alum, for safety, adjuvant dose-finding, and immunogenicity in a first-in-healthy adult (n = 17), randomized, and placebo-controlled trial (HVTN 137A). The vaccine regimen appeared safe. Robust, trimer-specific antibody, and B cell and CD4+ T cell responses emerged after vaccination. Five vaccinees developed serum autologous tier 2 nAbs (ID50 titer, 1:28-1:8647) after two to three doses targeting C3/V5 and/or V1/V2/V3 Env regions by electron microscopy and mutated pseudovirus-based neutralization analyses. Trimer-specific, B cell-derived monoclonal antibody activities confirmed these results and showed weak heterologous neutralization in the strongest responder. Our findings demonstrate the clinical utility of the 3M-052-AF/Alum adjuvant and support further improvements of trimer-based Env immunogens to focus responses on multiple broad nAb epitopes.


Subject(s)
AIDS Vaccines , Adjuvants, Immunologic , Alum Compounds , Antibodies, Neutralizing , env Gene Products, Human Immunodeficiency Virus , Humans , Antibodies, Neutralizing/immunology , AIDS Vaccines/immunology , AIDS Vaccines/administration & dosage , Alum Compounds/administration & dosage , Adult , Adjuvants, Immunologic/administration & dosage , env Gene Products, Human Immunodeficiency Virus/immunology , HIV Antibodies/immunology , Female , HIV-1/immunology , Male , HIV Infections/immunology , HIV Infections/prevention & control , B-Lymphocytes/immunology , Adjuvants, Vaccine , Middle Aged , Young Adult , CD4-Positive T-Lymphocytes/immunology
11.
Open Vet J ; 14(8): 1794-1800, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39308706

ABSTRACT

Background: Natural product active ingredients are currently being studied rigorously worldwide and offer a viable substitute for traditional immunotherapy for various medical disorders. Aim: The objective of the study was to investigate the immunostimulatory properties of fucoidan in albino Wistar rats. Methods: For the current study, forty rats were divided into five groups of rats that were used in good condition. In-vivo experiments of fucoidan were carried out in Wistar albino rats, such as the cyclophosphamide-caused myelosuppression, the delayed-type hypersensitivity (DTH) response, the phagocytic activity, the haemagglutinating antibody (HA) titer, and the neutrophil adhesion test. Results: The phagocytic index increased significantly in response to Fucoidan in a dose-dependent manner, as well as enhanced DTH reaction, and HA titer caused by sheep red blood cells sheep red blood cells. Additionally, fucoidan decreased myelosuppression in rats after cyclophosphamide treatment and enhanced neutrophil adhesion with nylon fiber. Conclusion: These findings imply that fucoidan has immunostimulant properties and could potentially utilised to treat immune-depression diseases.


Subject(s)
Adjuvants, Immunologic , Cyclophosphamide , Immunity, Cellular , Immunity, Humoral , Polysaccharides , Rats, Wistar , Animals , Polysaccharides/pharmacology , Polysaccharides/administration & dosage , Rats , Immunity, Humoral/drug effects , Cyclophosphamide/pharmacology , Immunity, Cellular/drug effects , Adjuvants, Immunologic/pharmacology , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/drug therapy , Phagocytosis/drug effects , Male , Neutrophils/drug effects , Neutrophils/immunology
12.
Hum Vaccin Immunother ; 20(1): 2395081, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39278862

ABSTRACT

Archaeosomes are liposomes traditionally comprised of total polar lipids or semi-synthetic glycerolipids of ether-linked isoprenoid phytanyl cores with varied glycol- and amino-head groups. We have developed a semi-synthetic archaeosome formulation based on sulfated lactosylarchaeol (SLA) that can be readily synthesized and easily formulated to induce robust humoral and cell-mediated immunity following systemic immunization, enhancing protection in models of infectious disease and cancer. Liposomes composed of SLA have been shown to be a safe and effective vaccine adjuvant to a multitude of antigens in preclinical studies including hepatitis C virus E1/E2 glycoproteins, hepatitis B surface antigen, influenza hemagglutinin, Rabbit Hemorrhagic Disease Virus antigens, and SARS-CoV-2 Spike antigens based on the ancestral strain as well as multiple variants of concern. With the COVID-19 pandemic highlighting the need for new vaccine technologies including adjuvants, this review outlines the studies conducted to date to support the development of SLA archaeosomes as a vaccine adjuvant.


Subject(s)
COVID-19 Vaccines , COVID-19 , Liposomes , Humans , Animals , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , COVID-19/immunology , Adjuvants, Vaccine , SARS-CoV-2/immunology , Glyceryl Ethers , Adjuvants, Immunologic/administration & dosage , Glycolipids/immunology , Glycolipids/chemistry
13.
Front Immunol ; 15: 1347736, 2024.
Article in English | MEDLINE | ID: mdl-39286241

ABSTRACT

Introduction: This study aimed to evaluate the efficiency of tea polyphenols (TP) and medicinal plant mixtures (Astragalus membranaceus + Lonicera japonica, Rheum officinale Bail + Scutellaria baicalensis + Platycladus orientalis) combined with astaxanthin (AST), benzoic acid (BA), and yeast complex on the health status of Eriocheir sinensis. Method: A total of 630 crabs (male crabs: 41.51 ± 1.63 g; female crabs: 47.27 ± 0.79 g) were randomly distributed into seven groups with three replicates (male: female, 1:1). These crabs were fed as follows for 8 weeks: basal diet (M1), M2 (M1 + 100 mg/kg TP), M3 (M1 + 2.0 g/kg A. membranaceus + 20 g/kg L. japonica), M4 (M1 + 2.5 g/kg R. officinale Bail + 1.5 g/kg S. baicalensis + 1.0 g/kg P. orientalis), and M5, M6, M7 (M2, M3 and M4 with 600 mg/kg AST +1.0 g/kg BA + 20 mg/kg yeast complex added, respectively). Results and discussion: The results showed that the activities of acid phosphatase (ACP), alkaline phosphatase (AKP), and lysosome (LZM) in the hemolymph were significantly increased in M5, M6, and M7 (P < 0.05), and the highest phagocytosis index (PI) and LZM activity were observed in M7 of female crabs. Moreover, the antioxidant indicators superoxide dismutase (SOD), glutathione (GSH), glutathione peroxidase (GPx), and catalase (CAT) of hepatopancreas were also significantly improved in M5, M6, and M7 (P < 0.05), while the malondialdehyde (MDA) contents showed an opposite trend. Furthermore, a morphological examination also showed the improved histological structure of hepatopancreas in M7, especially as seen in the clear lumens, no vacuolation, and integrity of the basal membrane of the hepatopancreatic tubule. Taken together, these results suggested that 2.5 g/kg R. officinale Bail, 1.5 g/kg S. baicalensis, and 1.0 g/kg P. orientalis in combination with 600 mg/kg AST, 1.0 g/kg BA, and 20 mg/kg yeast complex could improve the non-specific immunity, antioxidant capacity, and hepatopancreatic health of E. sinensis.


Subject(s)
Antioxidants , Brachyura , Dietary Supplements , Hepatopancreas , Plants, Medicinal , Animals , Brachyura/immunology , Antioxidants/pharmacology , Male , Female , Plants, Medicinal/chemistry , Hepatopancreas/drug effects , Hepatopancreas/immunology , Hepatopancreas/metabolism , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Animal Feed/analysis
14.
Virol J ; 21(1): 220, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285440

ABSTRACT

BACKGROUND: Human adenovirus type 55 (hAd55) infection can lead to acute respiratory diseases that often present with severe symptoms. Despite its persistent prevalence in military camps and communities, there are no commercially available vaccines or vaccine candidates undergoing clinical evaluation; therefore, there is an urgent need to address this. In this study, we evaluated the immunogenicity of inactivated hAd55 isolates and investigated the effects of adjuvants and various immunization intervals. METHODS AND RESULTS: To select a vaccine candidate, four hAd55 strains (6-9, 6-15 (AFMRI 41014), 28-48 (AFMRI 41013), and 12-164 (AFMRI 41012)) were isolated from infected patients in military camps. Sequence analysis revealed no variation in the coding regions of structural proteins, including pentons, hexons, and fibers. Immunization with inactivated hAd55 isolates elicited robust hAd55-specific binding and neutralizing antibody responses in mice, with adjuvants, particularly alum hydroxide (AH), enhancing antibody titers. Co-immunization with AH also induced hAd14-specific neutralizing antibody responses but did not induce hAd11-specific neutralizing antibody responses. Notably, booster immunization administered at a four-week interval resulted in superior immune responses compared with shorter immunization intervals. CONCLUSIONS: Prime-boost immunization with the inactivated hAd55 isolate and an AH adjuvant shows promise as a potential approach for preventing hAd55-induced respiratory disease. Further research is needed to evaluate the efficacy and safety of these vaccine candidates in preventing hAd55-associated respiratory illnesses.


Subject(s)
Adenoviruses, Human , Adjuvants, Immunologic , Antibodies, Neutralizing , Antibodies, Viral , Immunization, Secondary , Vaccines, Inactivated , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Mice , Antibodies, Viral/blood , Antibodies, Viral/immunology , Humans , Adenoviruses, Human/immunology , Adenoviruses, Human/genetics , Adjuvants, Immunologic/administration & dosage , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Female , Adenovirus Vaccines/immunology , Adenovirus Vaccines/administration & dosage , Mice, Inbred BALB C , Adjuvants, Vaccine/administration & dosage , Adenovirus Infections, Human/immunology , Adenovirus Infections, Human/prevention & control , Adenovirus Infections, Human/virology
15.
BMC Vet Res ; 20(1): 407, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261948

ABSTRACT

BACKGROUND: Rabbit hemorrhagic disease (RHD) is an acute infectious disease that damages the rabbit industry by producing significant mortality rates in young and adult rabbits. RHD is better controlled by vaccination. OBJECTIVE: The current study's goal was to prepare and evaluate the immuno-enhancing effect of montanide ISA70 and aluminum hydroxide (Al(OH)3) gel incorporated within the inactivated RHDV2 vaccine and assess the vaccine's protective efficacy against the homologous and heterologous local RHDV2 strains in rabbits. METHODS: Inactivated RHDV vaccines were prepared using Montanide ISA70 oil or Al(OH)3 gel adjuvants and submitted to sterility, safety, and potency tests. 200 rabbits were equally divided into 4 groups: G1 (control), G2 (vaccinated with gel-incorporated vaccine), G3 (vaccinated with montanide-incorporated vaccine), and G4 (vaccinated with gel- and montanide-incorporated vaccines). Individual blood samples were collected from one week to six months from all groups. The vaccine's potency was measured by the HI test and protection percentage post challenge. RESULTS: Data revealed slightly increasing HI titer means reaching the 1st peak at 4 weeks post-vaccination (7.33, 7.67, and 7.33 log2 in the 2nd, 3rd, and 4th groups, respectively), then slightly decreasing and peaked again, giving 9.33 log2 for the2nd group at 3 months post-vaccination (MPV), 10.67 log2 for 3rd the group, and 10.33 log2 for the 4th group at 5 months post-vaccination. Titer gradually decreased but remained protective. The protection rate ranged from 80-100% and 80-90% for homologous and heterologous local RHDV2 vaccines, respectively, within 3 weeks and 6 months post-challenge. The montanide oil RHDV2 vaccine induced better protection than the aluminum gel RHDV2 vaccine. CONCLUSION: The results demonstrated evidence of cross-protection between RHDV2 strains. The oil emulsion vaccine induced higher and longer-lasting antibody titers than those obtained with the RHDV2 aluminum gel vaccine.


Subject(s)
Aluminum Hydroxide , Caliciviridae Infections , Hemorrhagic Disease Virus, Rabbit , Viral Vaccines , Animals , Rabbits , Aluminum Hydroxide/pharmacology , Aluminum Hydroxide/administration & dosage , Hemorrhagic Disease Virus, Rabbit/immunology , Viral Vaccines/immunology , Caliciviridae Infections/veterinary , Caliciviridae Infections/prevention & control , Gels , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Vaccines, Inactivated/immunology , Oleic Acids/pharmacology , Oleic Acids/administration & dosage
16.
Front Immunol ; 15: 1439418, 2024.
Article in English | MEDLINE | ID: mdl-39267766

ABSTRACT

Introduction: Adjuvants added to subunit vaccines augment antigen-specific immune responses. One mechanism of adjuvant action is activation of pattern recognition receptors (PRRs) on innate immune cells. Bordetella colonization factor A (BcfA); an outer membrane protein with adjuvant function, activates TH1/TH17-polarized immune responses to protein antigens from Bordetella pertussis and SARS CoV-2. Unlike other adjuvants, BcfA does not elicit a TH2 response. Methods: To understand the mechanism of BcfA-driven TH1/TH17 vs. TH2 activation, we screened PRRs to identify pathways activated by BcfA. We then tested the role of this receptor in the BcfA-mediated activation of bone marrow-derived dendritic cells (BMDCs) using mice with germline deletion of TLR4 to quantify upregulation of costimulatory molecule expression and cytokine production in vitro and in vivo. Activity was also tested on human PBMCs. Results: PRR screening showed that BcfA activates antigen presenting cells through murine TLR4. BcfA-treated WT BMDCs upregulated expression of the costimulatory molecules CD40, CD80, and CD86 and produced IL-6, IL-12/23 p40, and TNF-α while TLR4 KO BMDCs were not activated. Furthermore, human PBMCs stimulated with BcfA produced IL-6. BcfA-stimulated murine BMDCs also exhibited increased uptake of the antigen DQ-OVA, supporting a role for BcfA in improving antigen presentation to T cells. BcfA further activated APCs in murine lungs. Using an in vitro TH cell polarization system, we found that BcfA-stimulated BMDC supernatant supported TFH and TH1 while suppressing TH2 gene programming. Conclusions: Overall, these data provide mechanistic understanding of how this novel adjuvant activates immune responses.


Subject(s)
Adjuvants, Immunologic , Th1 Cells , Th2 Cells , Toll-Like Receptor 4 , Animals , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Mice , Th1 Cells/immunology , Th2 Cells/immunology , Adjuvants, Immunologic/pharmacology , Humans , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Mice, Knockout , Dendritic Cells/immunology , Mice, Inbred C57BL , T Follicular Helper Cells/immunology , Cytokines/metabolism , Lymphocyte Activation/immunology
17.
Hum Vaccin Immunother ; 20(1): 2397872, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39222955

ABSTRACT

HepB-CpG is a licensed adjuvanted two-dose hepatitis B vaccine for adults, with limited data on exposure during pregnancy. We assessed the risk of pregnancy outcomes among individuals who received HepB-CpG or the 3-dose HepB-alum vaccine ≤28 d prior to conception or during pregnancy at Kaiser Permanente Southern California (KPSC). The pregnancy cohort included KPSC members aged ≥18 y who received ≥1 dose of hepatitis B vaccine (HepB-CpG or HepB-alum) at KPSC outpatient family or internal medicine departments from August 2018 to November 2020. We followed these individuals through electronic health records from the vaccination date until the end of pregnancy, KPSC health plan disenrollment, or death, whichever came first. Among 81 and 125 eligible individuals who received HepB-CpG and HepB-alum, respectively, live births occurred in 84% and 74%, spontaneous abortion occurred in 7% and 17% (adjusted relative risk [aRR] 0.40, 95% CI: 0.16-1.00), and preterm birth occurred in 15% and 14% of liveborn infants (aRR 0.97, 95% CI 0.47-1.99). No major birth defects were identified through 6 months of age. The study found no evidence of adverse pregnancy outcomes for recipients of HepB-CpG in comparison to HepB-alum.


Subject(s)
Hepatitis B Vaccines , Hepatitis B , Pregnancy Outcome , Product Surveillance, Postmarketing , Humans , Pregnancy , Female , Adult , Hepatitis B Vaccines/administration & dosage , Hepatitis B Vaccines/adverse effects , Product Surveillance, Postmarketing/statistics & numerical data , Young Adult , Hepatitis B/prevention & control , Adolescent , California/epidemiology , Infant, Newborn , Vaccination/adverse effects , Vaccination/statistics & numerical data , Premature Birth/epidemiology , Abortion, Spontaneous/epidemiology , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/administration & dosage , Live Birth/epidemiology
18.
Sci Transl Med ; 16(763): eabq7378, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39231242

ABSTRACT

Elucidating optimal vaccine adjuvants for harnessing age-specific immune pathways to enhance magnitude, breadth, and durability of immunogenicity remains a key gap area in pediatric vaccine design. A better understanding of age-specific adjuvants will inform precision discovery and development of safe and effective vaccines for protecting children from preventable infectious diseases.


Subject(s)
Precision Medicine , Vaccines , Humans , Child , Vaccines/immunology , Adjuvants, Immunologic , Adjuvants, Vaccine , Pediatrics
20.
ACS Infect Dis ; 10(9): 3419-3429, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39196071

ABSTRACT

Despite concerted efforts to tackle the COVID-19 pandemic, the persistent transmission of SARS-CoV-2 demands continued research into novel vaccination strategies to combat the virus. In light of this, intranasally administered peptide vaccines, particularly those conjugated to an immune adjuvant to afford so-called "self-adjuvanted vaccines", remain underexplored. Here, we describe the synthesis and immunological evaluation of self-adjuvanting peptide vaccines derived from epitopes of the spike glycoprotein of SARS-CoV-2 covalently fused to the potent adjuvant, Pam2Cys, that targets toll-like receptor 2 (TLR2). When administered intranasally, these vaccines elicited a strong antigen-specific CD4+ and CD8+ T-cell response in the lungs as well as high titers of IgG and IgA specific to the native spike protein of SARS-CoV-2. Unfortunately, serum and lung fluid from mice immunized with these vaccines failed to inhibit viral entry in spike-expressing pseudovirus assays. Following this, we designed and synthesized fusion vaccines composed of the T-cell epitope discovered in this work, covalently fused to epitopes of the receptor-binding domain of the spike protein reported to be neutralizing. While antibodies elicited against these fusion vaccines were not neutralizing, the T-cell epitope retained its ability to stimulate strong antigen-specific CD4+ lymphocyte responses within the lungs. Given the Spike(883-909) region is still completely conserved in SARS-CoV-2 variants of concern and variants of interest, we envision the self-adjuvanting vaccine platform reported here may inform future vaccine efforts.


Subject(s)
Adjuvants, Immunologic , Administration, Intranasal , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Lipopeptides , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , SARS-CoV-2/immunology , Mice , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Spike Glycoprotein, Coronavirus/immunology , COVID-19/prevention & control , COVID-19/immunology , Lipopeptides/immunology , Lipopeptides/administration & dosage , Antibodies, Viral/immunology , Antibodies, Viral/blood , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Female , Humans , Mice, Inbred BALB C , Adjuvants, Vaccine/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Immunity, Cellular , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , CD8-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL