Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 255
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Viral Immunol ; 37(7): 324-336, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39172659

ABSTRACT

The widespread use of efficient vaccines against infectious diseases is regarded as one of the most significant advancements in public health and techniques for preventing and protecting against infectious diseases and cancer. Because the purpose of vaccination is to elicit an appropriate, powerful, and long-lasting immune response against the pathogen, compounds such as adjuvants must be used to enhance these responses. Adjuvants have been widely used since their discovery to boost immune responses, prevent diseases, and activate protective immunity. Today, several types of adjuvants with varying properties are available for specific applications. Adjuvants are supramolecular substances or complexes that strengthen and prolong the immune response to antigens. These compounds have long-term immunological effects and are low in toxicity. They also lower the amount of antigen or the number of immunogenic reactions needed to improve vaccine efficacy and are used in specific populations. This article provides an overview of the adjuvants commonly used in the vaccination industry, their respective mechanisms of action, and discusses how they function to stimulate the immune system. Understanding the mechanisms of action of adjuvants is crucial for the development of effective and safe vaccines.


Subject(s)
Adjuvants, Immunologic , Vaccination , Humans , Adjuvants, Immunologic/administration & dosage , Animals , Vaccines/immunology , Vaccines/administration & dosage , Adjuvants, Vaccine/administration & dosage
8.
Virulence ; 15(1): 2387181, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39101682

ABSTRACT

Infectious bursal disease (IBD) is a widespread problem in the poultry industry, and vaccination is the primary preventive method. However, moderately virulent vaccines may damage the bursa, necessitating the development of a safe and effective vaccine. The Newcastle disease virus (NDV) has been explored as a vector for vaccine development. In this study, reverse genetic technology was used to obtain three recombinant viruses, namely, rClone30-VP2L (P/M)-chGM-CSF (NP), rClone30-chGM-CSF (P/M)-VP2L (NP), and rClone30-VP2L-chGM-CSF (P/M). Animal experiments showed that the three biological adjuvant bivalent vaccines effectively increased anti-NDV and anti-infectious bursal disease virus (IBDV) titres, enhancing both humoral and cellular immune responses in chickens without leading to any harm. Amongst the three biological adjuvant bivalent vaccines, the rClone30-chGM-CSF (P/M)-VP2L (NP) group had higher levels of anti-NDV antibodies at 14 days after the first immunization and stimulated a greater humoral immune response in 7-10 days. While, the rClone30-VP2L (P/M)-chGM-CSF (NP) group was the most effective in producing a higher level of IBDV antibody response. In conclusion, these three vaccines can induce immune responses more rapidly and effectively, streamline production processes, be cost-effective, and provide a new avenue for the development of Newcastle disease (ND) and IBD bivalent vaccines.


Subject(s)
Antibodies, Viral , Birnaviridae Infections , Chickens , Infectious bursal disease virus , Newcastle Disease , Newcastle disease virus , Poultry Diseases , Viral Vaccines , Animals , Viral Vaccines/immunology , Poultry Diseases/prevention & control , Poultry Diseases/virology , Poultry Diseases/immunology , Birnaviridae Infections/prevention & control , Birnaviridae Infections/immunology , Birnaviridae Infections/veterinary , Newcastle disease virus/immunology , Newcastle disease virus/genetics , Infectious bursal disease virus/immunology , Infectious bursal disease virus/genetics , Newcastle Disease/prevention & control , Newcastle Disease/immunology , Antibodies, Viral/blood , Immunity, Humoral , Adjuvants, Immunologic/administration & dosage , Adjuvants, Vaccine , Immunity, Cellular , Vaccination
9.
J Nanobiotechnology ; 22(1): 480, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39135073

ABSTRACT

Biomaterials are substances that can be injected, implanted, or applied to the surface of tissues in biomedical applications and have the ability to interact with biological systems to initiate therapeutic responses. Biomaterial-based vaccine delivery systems possess robust packaging capabilities, enabling sustained and localized drug release at the target site. Throughout the vaccine delivery process, they can contribute to protecting, stabilizing, and guiding the immunogen while also serving as adjuvants to enhance vaccine efficacy. In this article, we provide a comprehensive review of the contributions of biomaterials to the advancement of vaccine development. We begin by categorizing biomaterial types and properties, detailing their reprocessing strategies, and exploring several common delivery systems, such as polymeric nanoparticles, lipid nanoparticles, hydrogels, and microneedles. Additionally, we investigated how the physicochemical properties and delivery routes of biomaterials influence immune responses. Notably, we delve into the design considerations of biomaterials as vaccine adjuvants, showcasing their application in vaccine development for cancer, acquired immunodeficiency syndrome, influenza, corona virus disease 2019 (COVID-19), tuberculosis, malaria, and hepatitis B. Throughout this review, we highlight successful instances where biomaterials have enhanced vaccine efficacy and discuss the limitations and future directions of biomaterials in vaccine delivery and immunotherapy. This review aims to offer researchers a comprehensive understanding of the application of biomaterials in vaccine development and stimulate further progress in related fields.


Subject(s)
Biocompatible Materials , Drug Delivery Systems , Vaccines , Biocompatible Materials/chemistry , Humans , Animals , Drug Delivery Systems/methods , Nanoparticles/chemistry , Hydrogels/chemistry , Vaccine Development , COVID-19/prevention & control , Adjuvants, Vaccine/chemistry
10.
Acta Parasitol ; 69(3): 1613-1620, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39164549

ABSTRACT

Visceral Leishmaniasis is a serious public health problem caused by Leishmania species parasites. Approximately 500 thousand people get Visceral Leishmaniasis (VL) every year. An effective and reliable vaccine against the disease has still not been formulated. Choosing the right adjuvant is important to increase immunogenicity in vaccines prepared with total antigens. In this study, we investigate the ideal adjuvant for use in vaccine formulations against VL. For this purpose, Leishmania antigens (FTLA) obtained from L. infantum parasites by the freeze-thaw method and three different adjuvants (alum-saponin and calcium phosphate) were used. The effectiveness of the formulations was investigated in vitro by cell viability analysis and determination of nitric oxide and cytokine production abilities in J774 macrophage cells. According to the study results, it was determined that formulations prepared with calcium phosphate produced 72% more NO and approximately 7.2 times more IL-12 cytokine. The results obtained showed that calcium phosphate salts can be used as ideal adjuvants in vaccine research against leishmaniasis.


Subject(s)
Antigens, Protozoan , Leishmania infantum , Leishmaniasis Vaccines , Animals , Mice , Leishmaniasis Vaccines/immunology , Leishmania infantum/immunology , Antigens, Protozoan/immunology , Cell Line , Macrophages/immunology , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Nitric Oxide/metabolism , Calcium Phosphates , Cytokines/metabolism , Adjuvants, Vaccine , Leishmaniasis, Visceral/prevention & control , Leishmaniasis, Visceral/immunology , Saponins/pharmacology , Alum Compounds/administration & dosage , Adjuvants, Immunologic/administration & dosage , Cell Survival/drug effects
12.
Front Immunol ; 15: 1412732, 2024.
Article in English | MEDLINE | ID: mdl-39206189

ABSTRACT

Background: Antibody-mediated protection can depend on mechanisms varying from neutralization to Fc-dependent innate immune-cell recruitment. Adjuvanted vaccine development relies on a holistic understanding of how adjuvants modulate the quantity/titer and quality of the antibody response. Methods: A Phase 2 trial (ClinicalTrials.gov: NCT00805389) evaluated hepatitis B vaccines formulated with licensed adjuvants (AS01B, AS01E, AS03, AS04 or Alum) in antigen-naïve adults. The trial investigated the role of adjuvants in shaping antibody-effector functions, and identified an innate transcriptional response shared by AS01B, AS01E and AS03. We integrated previously reported data on the innate response (gene expression, cytokine/C-reactive protein levels) and on quantitative/qualitative features of the mature antibody response (Fc-related parameters, immunoglobulin titers, avidity). Associations between the innate and humoral parameters were explored using systems vaccinology and a machine-learning framework. Results: A dichotomy in responses between AS01/AS03 and AS04/Alum (with the former two contributing most to the association with the humoral response) was observed across all timepoints of this longitudinal study. The consistent patterns over time suggested a similarity in the impacts of the two-dose immunization regimen, year-long interval, and non-adjuvanted antigenic challenge given one year later. An innate signature characterized by interferon pathway-related gene expression and secreted interferon-γ-induced protein 10 and C-reactive protein, which was shared by AS01 and AS03, consistently predicted both the qualitative antibody response features and the titers. The signature also predicted from the antibody response quality, the group of adjuvants from which the administered vaccine was derived. Conclusion: An innate signature induced by AS01- or AS03-adjuvanted vaccines predicts the antibody response magnitude and quality consistently over time.


Subject(s)
Hepatitis B Vaccines , Immunity, Innate , Humans , Immunity, Innate/drug effects , Adult , Hepatitis B Vaccines/immunology , Hepatitis B Vaccines/administration & dosage , Female , Adjuvants, Vaccine/administration & dosage , Adjuvants, Immunologic/administration & dosage , Male , Antibody Formation/immunology , Drug Combinations , Hepatitis B Antibodies/blood , Hepatitis B Antibodies/immunology , Squalene/administration & dosage , Squalene/immunology , Polysorbates/administration & dosage , Hepatitis B/prevention & control , Hepatitis B/immunology , Immunogenicity, Vaccine , Lipid A/analogs & derivatives , Saponins , alpha-Tocopherol
13.
Vaccine ; 42(25): 126213, 2024 Nov 14.
Article in English | MEDLINE | ID: mdl-39138071

ABSTRACT

Zika virus (ZIKV) infection remains a global public health problem. After the "Public Health Emergencies of International Concern" declared in February 2016, the incidence of new infections by this pathogen has been decreasing in many areas. However, there is still a likely risk that ZIKV will spread to more countries. To date, there is no vaccine or antiviral drug available to prevent or treat Zika virus infection. In the Zika vaccine development, those based on protein subunits are attractive as a non-replicable platform due to their potentially enhanced safety profile to be used in all populations. However, these vaccines frequently require multiple doses and adjuvants to achieve protective immunity. In this study we show the immunological evaluation of new formulations of the recombinant protein ZEC, which combines regions of domain III of the envelope and the capsid from ZIKV. Two nucleotide-based adjuvants were used to enhance the immunity elicited by the vaccine candidate ZEC. ODN 39M or c-di-AMP was incorporated as immunomodulator into the formulations combined with aluminum hydroxide. Following immunizations in immunocompetent BALB/c mice, the formulations stimulated high IgG antibodies. Although the IgG subtypes suggested a predominantly Th1-biased immune response by the formulation including the ODN 39M, cellular immune responses measured by IFNγ secretion from spleen cells after in vitro stimulations were induced by both immunomodulators. These results demonstrate the capacity of both immunomodulators to enhance the immunogenicity of the recombinant subunit ZEC as a vaccine candidate against ZIKV.


Subject(s)
Adjuvants, Immunologic , Antibodies, Viral , Mice, Inbred BALB C , Vaccines, Subunit , Vaccines, Synthetic , Zika Virus Infection , Zika Virus , Animals , Zika Virus/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Zika Virus Infection/prevention & control , Zika Virus Infection/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Mice , Female , Adjuvants, Immunologic/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunogenicity, Vaccine , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Adjuvants, Vaccine , Immunity, Cellular , Viral Envelope Proteins/immunology , Capsid Proteins/immunology , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology
14.
Microb Pathog ; 195: 106871, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39163919

ABSTRACT

The H9N2 avian influenza virus (AIV) is spreading worldwide. Presence of H9N2 virus tends to increase the chances of infection with other pathogens which can lead to more serious economic losses. In a previous study, a regulated delayed lysis Salmonella vector was used to deliver a DNA vaccine named pYL233 encoding M1 protein, mosaic HA protein and chicken GM-CSF adjuvant. To further increase its efficiency, chitosan as a natural adjuvant was applied in this study. The purified plasmid pYL233 was coated with chitosan to form a DNA containing nanoparticles (named CS233) by ionic gel method and immunized by intranasal boost immunization in birds primed by oral administration with Salmonella strain. The CS233 DNA nanoparticle has a particle size of about 150 nm, with an encapsulation efficiency of 93.2 ± 0.12 % which protected the DNA plasmid from DNase I digestion and could be stable for a period of time at 37°. After intranasal boost immunization, the CS233 immunized chickens elicited higher antibody response, elevated CD4+ T cells and CD8+ T cells activation and increased T-lymphocyte proliferation, as well as increased productions of IL-4 and IFN-γ. After challenge, chickens immunized with CS233 resulted in the lowest levels of pulmonary virus titer and viral shedding as compared to the other challenge groups. The results showed that the combination of intranasal immunization with chitosan-coated DNA vaccine and oral immunization with regulatory delayed lytic Salmonella strain could enhance the immune response and able to provide protection against H9N2 challenge.


Subject(s)
Administration, Intranasal , Antibodies, Viral , Chickens , Chitosan , Immunity, Cellular , Influenza A Virus, H9N2 Subtype , Influenza Vaccines , Influenza in Birds , Plasmids , Vaccines, DNA , Virus Shedding , Animals , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/genetics , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Influenza in Birds/prevention & control , Influenza in Birds/immunology , Chickens/immunology , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Antibodies, Viral/blood , Plasmids/genetics , Nanoparticles , Immunization, Secondary , CD8-Positive T-Lymphocytes/immunology , Adjuvants, Immunologic/administration & dosage , Interferon-gamma , Interleukin-4 , Adjuvants, Vaccine , Poultry Diseases/prevention & control , Poultry Diseases/immunology , Poultry Diseases/virology , CD4-Positive T-Lymphocytes/immunology , Salmonella/immunology , Salmonella/genetics
15.
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
16.
Hum Vaccin Immunother ; 20(1): 2374147, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39090779

ABSTRACT

Entamoeba histolytica, the causative agent of amebiasis, is one of the top three parasitic causes of mortality worldwide. However, no vaccine exists against amebiasis. Using a lead candidate vaccine containing the LecA fragment of Gal-lectin and GLA-3M-052 liposome adjuvant, we immunized rhesus macaques via intranasal or intramuscular routes. The vaccine elicited high-avidity functional humoral responses as seen by the inhibition of amebic attachment to mammalian target cells by plasma and stool antibodies. Importantly, antigen-specific IFN-γ-secreting peripheral blood mononuclear cells (PBMCs) and IgG/IgA memory B cells (BMEM) were detected in immunized animals. Furthermore, antigen-specific antibody and cellular responses were maintained for at least 8 months after the final immunization as observed by robust LecA-specific BMEM as well as IFN-γ+ PBMC responses. Overall, both intranasal and intramuscular immunizations elicited a durable and functional response in systemic and mucosal compartments, which supports advancing the LecA+GLA-3M-052 liposome vaccine candidate to clinical testing.


Subject(s)
Administration, Intranasal , Antibodies, Protozoan , Entamoeba histolytica , Entamoebiasis , Interferon-gamma , Leukocytes, Mononuclear , Liposomes , Macaca mulatta , Protozoan Vaccines , Animals , Entamoeba histolytica/immunology , Liposomes/immunology , Liposomes/administration & dosage , Protozoan Vaccines/immunology , Protozoan Vaccines/administration & dosage , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Leukocytes, Mononuclear/immunology , Entamoebiasis/prevention & control , Entamoebiasis/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Injections, Intramuscular , Immunogenicity, Vaccine , Adjuvants, Vaccine/administration & dosage , Adjuvants, Immunologic/administration & dosage , B-Lymphocytes/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin A/immunology , Immunoglobulin A/blood , Antigens, Protozoan/immunology , Immunity, Humoral , Immunologic Memory , Protozoan Proteins/immunology
17.
Vaccine ; 42(25): 126151, 2024 Nov 14.
Article in English | MEDLINE | ID: mdl-39089961

ABSTRACT

Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), remains one of the top three causes of death. Currently, the only licensed vaccine against TB is the bacillus Calmette-Guerin (BCG), which lacks efficacy in preventing and controlling pulmonary TB in adults. We aimed to evaluate a nasal TB vaccine formulation composed of the Mtb-specific vaccine antigen ESAT-6, an Mtb-associated protein that can trigger protective immune responses, and S100A4, a recently characterized novel mucosal adjuvant. Mice were intranasally given recombinant ESAT-6 in the presence or absence of S100A4 as an adjuvant. We have provided experimental evidence demonstrating that S100A4 admixed to ESAT-6 could induce Mtb-specific adaptive immune responses after intranasal immunization. S100A4 remarkably augmented the levels of anti-ESAT-6 IgG in serum and IgA in mucosal sites, including lung exudates, bronchoalveolar lavage fluid (BALF) and nasal lavage. Furthermore, in both lung and spleen tissues, S100A4 strongly promoted ESAT-6-specific expansion of CD4 T cells. Both CD4 and CD8 T cells from these tissues expressed increased levels of IFN-γ, TNF-α, and IL-17, cytokines critical for antimicrobial activity. Antigen-reencounter-induced T cell proliferative responses, a key vaccine performance indicator, were augmented in the spleen of S100A4-adjuvanted mice. Furthermore, CD8 T cells from the spleen and lung tissues of these mice expressed higher levels of granzyme B upon antigen re-stimulation. S100A4-adjuvanted immunization may predict good mucosal protection against TB.


Subject(s)
Adjuvants, Immunologic , Administration, Intranasal , Antigens, Bacterial , Bacterial Proteins , Mycobacterium tuberculosis , S100 Calcium-Binding Protein A4 , Tuberculosis Vaccines , Animals , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/administration & dosage , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Mice , Mycobacterium tuberculosis/immunology , Female , S100 Calcium-Binding Protein A4/immunology , CD4-Positive T-Lymphocytes/immunology , Tuberculosis/prevention & control , Tuberculosis/immunology , Adjuvants, Vaccine/administration & dosage , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Lung/immunology , Lung/microbiology , CD8-Positive T-Lymphocytes/immunology , Immunity, Mucosal , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Cytokines/metabolism , Mice, Inbred C57BL , Spleen/immunology , Bronchoalveolar Lavage Fluid/immunology , Mice, Inbred BALB C
18.
J Control Release ; 374: 39-49, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39111597

ABSTRACT

Immunological adjuvants are vaccine components that enhance long-lasting adaptive immune responses to weakly immunogenic antigens. Monophosphoryl lipid A (MPLA) is a potent and safe vaccine adjuvant that initiates an early innate immune response by binding to the Toll-like receptor 4 (TLR4). Importantly, the binding and recognition process is highly dependent on the monomeric state of MPLA. However, current vaccine delivery systems often prioritize improving the loading efficiency of MPLA, while neglecting the need to maintain its monomeric form for optimal immune activation. Here, we introduce a Pickering emulsion-guided MPLA monomeric delivery system (PMMS), which embed MPLA into the oil-water interface to achieve the monomeric loading of MPLA. During interactions with antigen-presenting cells, PMMS functions as a chaperone for MPLA, facilitating efficient recognition by TLR4 regardless of the presence of lipopolysaccharide-binding proteins. At the injection site, PMMS efficiently elicited local immune responses, subsequently promoting the migration of antigen-internalized dendritic cells to the lymph nodes. Within the draining lymph nodes, PMMS enhanced antigen presentation and maturation of dendritic cells. In C57BL/6 mice models, PMMS vaccination provoked potent antigen-specific CD8+ T cell-based immune responses. Additionally, PMMS demonstrated strong anti-tumor effects against E.G7-OVA lymphoma. These data indicate that PMMS provides a straightforward and efficient strategy for delivering monomeric MPLA to achieve robust cellular immune responses and effective cancer immunotherapy.


Subject(s)
Adjuvants, Immunologic , Dendritic Cells , Emulsions , Lipid A , Mice, Inbred C57BL , Toll-Like Receptor 4 , Animals , Lipid A/analogs & derivatives , Lipid A/administration & dosage , Lipid A/chemistry , Dendritic Cells/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Vaccination/methods , Female , Mice , Drug Delivery Systems , Adjuvants, Vaccine/administration & dosage , Adjuvants, Vaccine/chemistry , Antigen Presentation , Ovalbumin/administration & dosage , Ovalbumin/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology
19.
Sci Adv ; 10(32): eadn7187, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39110802

ABSTRACT

Over the past few decades, the development of potent and safe immune-activating adjuvant technologies has become the heart of intensive research in the constant fight against highly mutative and immune evasive viruses such as influenza, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and human immunodeficiency virus (HIV). Herein, we developed a highly modular saponin-based nanoparticle platform incorporating Toll-like receptor agonists (TLRas) including TLR1/2a, TLR4a, and TLR7/8a adjuvants and their mixtures. These various TLRa-saponin nanoparticle adjuvant constructs induce unique acute cytokine and immune-signaling profiles, leading to specific T helper responses that could be of interest depending on the target disease for prevention. In a murine vaccine study, the adjuvants greatly improved the potency, durability, breadth, and neutralization of both COVID-19 and HIV vaccine candidates, suggesting the potential broad application of these adjuvant constructs to a range of different antigens. Overall, this work demonstrates a modular TLRa-SNP adjuvant platform that could improve the design of vaccines and affect modern vaccine development.


Subject(s)
Adjuvants, Immunologic , COVID-19 Vaccines , Nanoparticles , SARS-CoV-2 , Saponins , Toll-Like Receptor Agonists , Animals , Humans , Mice , Adjuvants, Immunologic/pharmacology , Adjuvants, Vaccine/chemistry , AIDS Vaccines/immunology , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , COVID-19 Vaccines/immunology , Cytokines/metabolism , Nanoparticles/chemistry , Saponins/pharmacology , Saponins/chemistry , Saponins/immunology , SARS-CoV-2/immunology
20.
Colloids Surf B Biointerfaces ; 244: 114144, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39116600

ABSTRACT

Traditional Alum adjuvants mainly elicit a Th2 humoral immune response, but fail to generate a robust Th1 cellular immune response. However, the cellular immune response is essential for vaccination against cancer and a number of chronic infectious diseases, including human immunodeficiency virus infection and tuberculosis. In our previous study, we demonstrated that the polysaccharide from Poria cocos (PCP) has the potential to serve as an immunologic stimulant, enhancing both humoral and cellular immune responses. However, this effect was only observed at high concentrations. In this study, to enhance the immune-stimulation effect of PCP and modify the type of immune response elicited by Alum adjuvant, we successfully developed a Pickering emulsion delivery system (PCP-Al-Pickering) using PCP-loaded Alhydrogel particles as the stabilizer. After optimization, the Pickering emulsion exhibited excellent storage capacity and effectively adsorbed the PCP and antigen. As an adjuvant delivery system, the PCP-Al-Pickering emulsion facilitated the antigen uptake by macrophages, increased the recruitment of cells at injection sites, improved the activation of dendritic cells in draining lymph nodes, elicited a potent and durable antibody response, and promoted the activation of CD4+ and CD8+ T cells. Importantly, the PCP-Al-Pickering emulsion adjuvant elicited a balanced Th1 and Th2 immune response, in comparison to Alum adjuvant. The PCP-Al-Pickering emulsion may serve as a safe and promising adjuvant delivery system to enhance immune responses.


Subject(s)
Adjuvants, Immunologic , Alum Compounds , Emulsions , Polysaccharides , Wolfiporia , Emulsions/chemistry , Animals , Alum Compounds/chemistry , Polysaccharides/chemistry , Polysaccharides/pharmacology , Mice , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Wolfiporia/chemistry , Mice, Inbred BALB C , Female , Adjuvants, Vaccine/chemistry , Immunity, Cellular/drug effects , Th1 Cells/immunology , Particle Size , Immunity, Humoral/drug effects , Th2 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL