Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 346
Filter
1.
Lancet Microbe ; 5(6): e581-e593, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38761816

ABSTRACT

BACKGROUND: A self-assembling SARS-CoV-2 WA-1 recombinant spike ferritin nanoparticle (SpFN) vaccine co-formulated with Army Liposomal Formulation (ALFQ) adjuvant containing monophosphoryl lipid A and QS-21 (SpFN/ALFQ) has shown protective efficacy in animal challenge models. This trial aims to assess the safety and immunogenicity of SpFN/ALFQ in a first-in-human clinical trial. METHODS: In this phase 1, randomised, double-blind, placebo-controlled, first-in-human clinical trial, adults were randomly assigned (5:5:2) to receive 25 µg or 50 µg of SpFN/ALFQ or saline placebo intramuscularly at day 1 and day 29, with an optional open-label third vaccination at day 181. Enrolment and randomisation occurred sequentially by group; randomisation was done by an interactive web-based randomisation system and only designated unmasked study personnel had access to the randomisation code. Adults were required to be seronegative and unvaccinated for inclusion. Local and systemic reactogenicity, adverse events, binding and neutralising antibodies, and antigen-specific T-cell responses were quantified. For safety analyses, exact 95% Clopper-Pearson CIs for the probability of any incidence of an unsolicited adverse event was computed for each group. For immunogenicity results, CIs for binary variables were computed using the exact Clopper-Pearson methodology, while CIs for geometric mean titres were based on 10 000 empirical bootstrap samples. Post-hoc, paired one-sample t tests were used to assess the increase in mean log-10 neutralising antibody titres between day 29 and day 43 (after the second vaccination) for the primary SARS-CoV-2 targets of interest. This trial is registered at ClinicalTrials.gov, NCT04784767, and is closed to new participants. FINDINGS: Between April 7, and June 29, 2021, 29 participants were enrolled in the study. 20 individuals were assigned to receive 25 µg SpFN/ALFQ, four to 50 µg SpFN/ALFQ, and five to placebo. Neutralising antibody responses peaked at day 43, 2 weeks after the second dose. Neutralisation activity against multiple omicron subvariants decayed more slowly than against the D614G or beta variants until 5 months after second vaccination for both dose groups. CD4+ T-cell responses were elicited 4 weeks after the first dose and were boosted after a second dose of SpFN/ALFQ for both dose groups. Neutralising antibody titres against early omicron subvariants and clade 1 sarbecoviruses were detectable after two immunisations and peaked after the third immunisation for both dose groups. Neutralising antibody titres against XBB.1.5 were detected after three vaccinations. Passive IgG transfer from vaccinated volunteers into Syrian golden hamsters controlled replication of SARS-CoV-1 after challenge. INTERPRETATION: SpFN/ALFQ was well tolerated and elicited robust and durable binding antibody and neutralising antibody titres against a broad panel of SARS-CoV-2 variants and other sarbecoviruses. FUNDING: US Department of Defense, Defense Health Agency.


Subject(s)
COVID-19 Vaccines , COVID-19 , Ferritins , Lipid A , Liposomes , Nanoparticles , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/adverse effects , Double-Blind Method , Adult , Male , Female , COVID-19/prevention & control , COVID-19/immunology , SARS-CoV-2/immunology , Nanoparticles/administration & dosage , Lipid A/analogs & derivatives , Lipid A/administration & dosage , Lipid A/pharmacology , Lipid A/immunology , Liposomes/administration & dosage , Spike Glycoprotein, Coronavirus/immunology , Saponins/administration & dosage , Saponins/immunology , Saponins/pharmacology , Saponins/adverse effects , Antibodies, Viral/blood , Middle Aged , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Adjuvants, Vaccine/administration & dosage , Antibodies, Neutralizing/blood , Young Adult , Nanovaccines
2.
Vet Immunol Immunopathol ; 271: 110743, 2024 May.
Article in English | MEDLINE | ID: mdl-38522410

ABSTRACT

Equine influenza is a contagious respiratory disease caused by H3N8 type A influenza virus. Vaccination against equine influenza is conducted regularly; however, infection still occurs globally because of the short immunity duration and suboptimal efficacy of current vaccines. Hence the objective of this study was to investigate whether an adjuvant combination can improve immune responses to equine influenza virus (EIV) vaccines. Seventy-two mice were immunized with an EIV vaccine only or with monophosphoryl lipid A (MPL), polyinosinic-polycytidylic acid (Poly I:C), or MPL + Poly I:C. Prime immunization was followed by boost immunization after 2 weeks. Mice were euthanized at 4, 8, and 32 weeks post-prime immunization, respectively. Sera were collected to determine humoral response. Bone marrow, spleen, and lung samples were harvested to determine memory cell responses, antigen-specific T-cell proliferation, and lung viral titers. MPL + Poly I:C resulted in the highest IgG, IgG1, and IgG2a antibodies and hemagglutination inhibition titers among the groups and sustained their levels until 32 weeks post-prime immunization. The combination enhanced memory B cell responses in the bone marrow and spleen. At 8 weeks post-prime immunization, the combination induced higher CD8+ central memory T cell frequencies in the lungs and CD8+ central memory T cells in the spleen. In addition, the combination group exhibited enhanced antigen-specific T cell proliferation, except for CD4+ T cells in the lungs. Our results demonstrated improved immune responses when using MPL + Poly I:C in EIV vaccines by inducing enhanced humoral responses, memory cell responses, and antigen-specific T cell proliferation.


Subject(s)
Adjuvants, Immunologic , Influenza A Virus, H3N8 Subtype , Influenza Vaccines , Lipid A , Lipid A/analogs & derivatives , Orthomyxoviridae Infections , Poly I-C , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Poly I-C/pharmacology , Poly I-C/administration & dosage , Lipid A/pharmacology , Lipid A/administration & dosage , Lipid A/immunology , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/veterinary , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Female , Influenza A Virus, H3N8 Subtype/immunology , Antibodies, Viral/blood , Horses/immunology , Horse Diseases/immunology , Horse Diseases/prevention & control , Horse Diseases/virology , Immunoglobulin G/blood , Immunologic Memory
3.
Immunol Res ; 72(3): 490-502, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38383811

ABSTRACT

To increase the effectiveness of methicillin-resistant Staphylococcus aureus vaccines (MRSA), a new generation of immune system stimulating adjuvants is necessary, along with other adjuvants. In some vaccines, monophosphoryl lipid A (MPLA) as a toll-like receptor 4 agonist is currently used as an adjuvant or co-adjuvant. MPLA could increase the immune response and vaccine immunogenicity. The current investigation assessed the immunogenicity and anti-MRSA efficacy of recombinant autolysin formulated in MPLA and Alum as co-adjuvant/adjuvant. r-Autolysin was expressed and purified by Ni-NTA affinity chromatography and characterized by SDS-PAGE. Then, the vaccine candidate formulation in MPLAs and Alum was prepared. To investigate the immunogenic responses, total IgG, isotype (IgG1 and IgG2a) levels, and cytokines (IL-4, IL-12, TNF-α, and IFN-γ) profiles were evaluated by ELISA. Also, the bacterial burden in internal organs, opsonophagocytosis, survival rate, and pathobiology changes was compared among the groups. Results demonstrated that mice immunized with the r-Autolysin + Alum + MPLA Synthetic and r-Autolysin + Alum + MPLA Biologic led to increased levels of opsonic antibodies, IgG1, IgG2a isotype as well as increased levels of cytokines profiles, as compared with other experimental groups. More importantly, mice immunized with MPLA and r-Autolysin exhibited a decrease in mortality and bacterial burden, as compared with the control group. The highest level of survival was seen in the r-Autolysin + Alum + MPLA Synthetic group. We concluded that both MPLA forms, synthetic and biological, are reliable candidates for immune response improvement against MRSA infection.


Subject(s)
Adjuvants, Immunologic , Antibodies, Bacterial , Disease Models, Animal , Immunoglobulin G , Lipid A , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Staphylococcal Vaccines , Animals , Lipid A/analogs & derivatives , Lipid A/immunology , Lipid A/administration & dosage , Lipid A/pharmacology , Mice , Methicillin-Resistant Staphylococcus aureus/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Staphylococcal Vaccines/administration & dosage , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Adjuvants, Immunologic/administration & dosage , Female , Cytokines/metabolism , N-Acetylmuramoyl-L-alanine Amidase/immunology , Vaccine Development , Alum Compounds/administration & dosage , Mice, Inbred BALB C , Adjuvants, Vaccine , Humans
4.
J Infect Dis ; 229(6): 1883-1893, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38330357

ABSTRACT

BACKGROUND: Malaria is preventable yet causes >600 000 deaths annually. RTS,S, the first marketed malaria vaccine, has modest efficacy, but improvements are needed for eradication. METHODS: We conducted an open-label, dose escalation phase 1 study of a full-length recombinant circumsporozoite protein vaccine (rCSP) administered with adjuvant glucopyranosyl lipid A-liposome Quillaja saponaria 21 formulation (GLA-LSQ) on days 1, 29, and 85 or 1 and 490 to healthy, malaria-naive adults. The primary end points were safety and reactogenicity. The secondary end points were antibody responses and Plasmodium falciparum parasitemia after homologous controlled human malaria infection. RESULTS: Participants were enrolled into 4 groups receiving rCSP/GLA-LSQ: 10 µg × 3 (n = 20), 30 µg × 3 (n = 10), 60 µg × 3 (n = 10), or 60 µg × 2 (n = 9); 10 participants received 30 µg rCSP alone × 3, and there were 6 infectivity controls. Participants experienced no serious adverse events. Rates of solicited and unsolicited adverse events were similar among groups. All 26 participants who underwent controlled human malaria infection 28 days after final vaccinations developed malaria. Increasing vaccine doses induced higher immunoglobulin G titers but did not achieve previously established RTS,S benchmarks. CONCLUSIONS: rCSP/GLA-LSQ had favorable safety results. However, tested regimens did not induce protective immunity. Further investigation could assess whether adjuvant or schedule adjustments improve efficacy. CLINICAL TRIALS REGISTRATION: NCT03589794.


Subject(s)
Adjuvants, Immunologic , Antibodies, Protozoan , Lipid A , Liposomes , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Protozoan Proteins , Humans , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Malaria Vaccines/adverse effects , Malaria, Falciparum/prevention & control , Malaria, Falciparum/immunology , Adult , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Female , Male , Adjuvants, Immunologic/administration & dosage , Young Adult , Lipid A/analogs & derivatives , Lipid A/administration & dosage , Lipid A/immunology , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Quillaja/chemistry , Adolescent , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Middle Aged , Glucosides
5.
Virology ; 566: 56-59, 2022 01.
Article in English | MEDLINE | ID: mdl-34864488

ABSTRACT

BACKGROUND: Recombinant protein subunit vaccination is considered to be a safe, fast and reliable technique when combating emerging and re-emerging diseases such as coronavirus disease 2019 (COVID-19). Typically, such subunit vaccines require the addition of adjuvants to attain adequate immunogenicity. AS01, which contains adjuvants MPL and saponin QS21, is a liposome-based vaccine adjuvant system that is one of the leading candidates. However, the adjuvant effect of AS01 in COVID-19 vaccines is not well described yet. METHODS: In this study, we utilized a mixture of AS01 as the adjuvant for an S1 protein-based COVID-19 vaccine. RESULTS: The adjuvanted vaccine induced robust immunoglobulin G (IgG) binding antibody and virus-neutralizing antibody responses. Importantly, two doses induced similar levels of IgG binding antibody and neutralizing antibody responses compared with three doses and the antibody responses weakened only slightly over time up to six weeks after immunization. CONCLUSION: These results suggested that two doses may be enough for a clinical vaccine strategy design using MPL & QS21 adjuvanted recombinant protein, especially in consideration of the limited production capacity of COVID-19 vaccine in a public health emergency.


Subject(s)
Antigens, Viral/immunology , COVID-19 Vaccines/immunology , COVID-19/immunology , Lipid A/analogs & derivatives , SARS-CoV-2/immunology , Saponins/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Subunit/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Vaccine/administration & dosage , Animals , Antibodies, Neutralizing , Antibodies, Viral/metabolism , Antibody Formation , COVID-19/virology , Dose-Response Relationship, Immunologic , Drug Combinations , Female , HEK293 Cells , Humans , Immunization , Immunogenicity, Vaccine , Lipid A/administration & dosage , Lipid A/immunology , Mice, Inbred BALB C , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Saponins/administration & dosage
6.
Front Immunol ; 12: 692151, 2021.
Article in English | MEDLINE | ID: mdl-34335601

ABSTRACT

Combining variant antigens into a multivalent vaccine is a traditional approach used to provide broad coverage against antigenically variable pathogens, such as polio, human papilloma and influenza viruses. However, strategies for increasing the breadth of antibody coverage beyond the vaccine are not well understood, but may provide more anticipatory protection. Influenza virus hemagglutinin (HA) is a prototypic variant antigen. Vaccines that induce HA-specific neutralizing antibodies lose efficacy as amino acid substitutions accumulate in neutralizing epitopes during influenza virus evolution. Here we studied the effect of a potent combination adjuvant (CpG/MPLA/squalene-in-water emulsion) on the breadth and maturation of the antibody response to a representative variant of HA subtypes H1, H5 and H7. Using HA protein microarrays and antigen-specific B cell labelling, we show when administered individually, each HA elicits a cross-reactive antibody profile for multiple variants within the same subtype and other closely-related subtypes (homosubtypic and heterosubtypic cross-reactivity, respectively). Despite a capacity for each subtype to induce heterosubtypic cross-reactivity, broader coverage was elicited by simply combining the subtypes into a multivalent vaccine. Importantly, multiplexing did not compromise antibody avidity or affinity maturation to the individual HA constituents. The use of adjuvants to increase the breadth of antibody coverage beyond the vaccine antigens may help future-proof vaccines against newly-emerging variants.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Viral/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinins/immunology , Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Vaccines, Combined/administration & dosage , Animals , Antibodies, Viral/blood , CpG Islands , Dogs , Female , Lipid A/administration & dosage , Lipid A/analogs & derivatives , Madin Darby Canine Kidney Cells , Mice, Inbred C57BL , Oligodeoxyribonucleotides/administration & dosage , Orthomyxoviridae Infections/prevention & control , Squalene/administration & dosage , Vaccines, Synthetic/administration & dosage
7.
Inflammation ; 44(5): 2132-2141, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34080091

ABSTRACT

Nitrogen-containing bisphosphonates (NBPs), such as alendronate (ALN), are anti-bone-resorptive drugs that have inflammatory side effects. We previously reported that ALN augmented lipid A-induced interleukin (IL)-1ß production and NOD-like receptor pyrin domain-containing-3 (NLRP3)/apoptosis-associated speck-like protein containing a CARD (ASC)-dependent cell death. The present study aimed to examine whether ALN augments lipid A-induced IL-1α release and necroptosis, which is induced by the activation of receptor-interacting protein kinase (RIPK) 3. Treatment of J774.1 cells with ALN augmented lipid A-induced IL-1α release, which was not inhibited by Ac-IETD-CHO, a caspase-8 inhibitor. ALN also activated mixed lineage kinase domain-like (MLKL), a key mediator of the necroptosis pathway, and upregulated the expression of caspase-11, a lipid A receptor. GSK'872, a RIPK3 inhibitor, suppressed the ALN-upregulated expression of caspase-11 and augmented lipid A-induced caspase-8 activation. Moreover, ALN induced the release of NLRP3 and ASC into culture supernatants. GSK'872, but not Ac-IETD-CHO, reduced the ALN-induced release of NLRP3, but not ASC, into culture supernatants, and reduced ALN-induced cell death, but not ALN-induced LDH release. Antibodies against NLRP3 and ASC upregulated caspase-11 expression in the cytosol by inhibiting ALN-induced cell death. However, pretreating cells with an antibody against ASC, but not NLRP3, before ALN addition also inhibited lipid A-induced IL-1α release. Pretreating cells with an antibody against caspase-11 before the addition of ALN or lipid A did not downregulate lipid A-induced production of IL-1α. Taken together, our findings suggest that ALN augments lipid A-induced IL-1α release via activation of ASC, but not caspase-11.


Subject(s)
Alendronate/administration & dosage , CARD Signaling Adaptor Proteins/metabolism , Caspases, Initiator , Interleukin-1alpha/metabolism , Lipid A/administration & dosage , Macrophages/metabolism , Animals , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Synergism , Macrophages/drug effects
8.
Cell Rep Med ; 2(3): 100207, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33763653

ABSTRACT

Interactions between B cells and CD4+ T follicular helper (Tfh) cells are key determinants of humoral responses. Using samples from clinical trials performed with the malaria vaccine candidate antigen Plasmodium falciparum merozoite protein (PfRH5), we compare the frequency, phenotype, and gene expression profiles of PfRH5-specific circulating Tfh (cTfh) cells elicited by two leading human vaccine delivery platforms: heterologous viral vector prime boost and protein with AS01B adjuvant. We demonstrate that the protein/AS01B platform induces a higher-magnitude antigen-specific cTfh cell response and that this correlates with peak anti-PfRH5 IgG concentrations, frequency of PfRH5-specific memory B cells, and antibody functionality. Furthermore, our data indicate a greater Th2/Tfh2 skew within the polyfunctional response elicited following vaccination with protein/AS01B as compared to a Th1/Tfh1 skew with viral vectors. These data highlight the impact of vaccine platform on the cTfh cell response driving humoral immunity, associating a high-magnitude, Th2-biased cTfh response with potent antibody production.


Subject(s)
Antibodies, Protozoan/biosynthesis , Carrier Proteins/immunology , Immunity, Humoral/drug effects , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Adolescent , Adult , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Carrier Proteins/administration & dosage , Carrier Proteins/genetics , Female , Gene Expression Profiling , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/immunology , Humans , Immunogenicity, Vaccine , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-5/genetics , Interleukin-5/immunology , Lipid A/administration & dosage , Lipid A/analogs & derivatives , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Male , Middle Aged , Receptors, CXCR5/genetics , Receptors, CXCR5/immunology , Saponins/administration & dosage , T Follicular Helper Cells/cytology , T Follicular Helper Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology , Vaccination , Vaccines, Subunit , Vaccinia virus/genetics , Vaccinia virus/immunology
9.
Lancet Respir Med ; 9(4): 373-386, 2021 04.
Article in English | MEDLINE | ID: mdl-33306991

ABSTRACT

BACKGROUND: A therapeutic vaccine that prevents recurrent tuberculosis would be a major advance in the development of shorter treatment regimens. We aimed to assess the safety and immunogenicity of the ID93 + GLA-SE vaccine at various doses and injection schedules in patients with previously treated tuberculosis. METHODS: This randomised, double-blind, placebo-controlled, phase 2a trial was conducted at three clinical sites near Cape Town, South Africa. Patients were recruited at local clinics after receiving 4 months of tuberculosis treatment, and screened for eligibility after providing written informed consent. Participants were aged 18-60 years, BCG-vaccinated, HIV-uninfected, and diagnosed with drug-sensitive pulmonary tuberculosis. Eligible patients had completed standard treatment for pulmonary tuberculosis in the past 28 days. Participants were enrolled after completing standard treatment and randomly assigned sequentially to receive vaccine or placebo in three cohorts: 2 µg intramuscular ID93 + 2 µg GLA-SE on days 0 and 56 (cohort 1); 10 µg ID93 + 2 µg GLA-SE on days 0 and 56 (cohort 2); 2 µg ID93 + 5 µg GLA-SE on days 0 and 56 and placebo on day 28 (cohort 3); 2 µg ID93 + 5 µg GLA-SE on days 0, 28, and 56 (cohort 3); or placebo on days 0 and 56 (cohorts 1 and 2), with the placebo group for cohort 3 receiving an additional injection on day 28. Randomisation was in a ratio of 3:1 for ID93 + GLA-SE and saline placebo in cohorts 1 and 2, and in a ratio of 3:3:1 for (2 ×) ID93 + GLA-SE, (3 ×) ID93 + GLA-SE, and placebo in cohort 3. The primary outcomes were safety and immunogenicity (vaccine-specific antibody response and T-cell response). For the safety outcome, participants were observed for 30 min after each injection, injection site reactions and systemic adverse events were monitored until day 84, and serious adverse events and adverse events of special interest were monitored for 6 months after the last injection. Vaccine-specific antibody responses were measured by serum ELISA, and T-cell responses after stimulation with vaccine antigens were measured in cryopreserved peripheral blood mononuclear cells specimens using intracellular cytokine staining followed by flow cytometry. This study is registered with ClinicalTrials.gov, number NCT02465216. FINDINGS: Between June 17, 2015, and May 30, 2016, we assessed 177 patients for inclusion. 61 eligible patients were randomly assigned to receive: saline placebo (n=5) or (2 ×) 2 µg ID93 + 2 µg GLA-SE (n=15) on days 0 and 56 (cohort 1); saline placebo (n=2) or (2 ×) 10 µg ID93 + 2 µg GLA-SE (n=5) on days 0 and 56 (cohort 2); saline placebo (n=5) on days 0, 28 and 56, or 2 µg ID93 + 5 µg GLA-SE (n=15) on days 0 and 56 and placebo injection on day 28, or (3 ×) 2 µg ID93 + 5 µg GLA-SE (n=14) on days 0, 28, and 56 (cohort 3). ID93 + GLA-SE induced robust and durable antibody responses and specific, polyfunctional CD4 T-cell responses to vaccine antigens. Two injections of the 2 µg ID93 + 5 µg GLA-SE dose induced antigen-specific IgG and CD4 T-cell responses that were significantly higher than those with placebo and persisted for the 6-month study duration. Mild to moderate injection site pain was reported after vaccination across all dose combinations, and induration and erythema in patients given 2 µg ID93 + 5 µg GLA-SE in two or three doses. One participant had grade 3 erythema and induration at the injection site. No vaccine-related serious adverse events were observed. INTERPRETATION: Vaccination with ID93 + GLA-SE was safe and immunogenic for all tested regimens. These data support further evaluation of ID93 + GLA-SE in therapeutic vaccination strategies to improve tuberculosis treatment outcomes. FUNDING: Wellcome Trust (102028/Z/13/Z).


Subject(s)
Immunogenicity, Vaccine , Secondary Prevention/methods , Tuberculosis Vaccines/adverse effects , Tuberculosis, Multidrug-Resistant/therapy , Tuberculosis, Pulmonary/therapy , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Adolescent , Adult , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Dose-Response Relationship, Immunologic , Double-Blind Method , Female , Glucosides/administration & dosage , Glucosides/adverse effects , Glucosides/immunology , Humans , Lipid A/administration & dosage , Lipid A/adverse effects , Lipid A/immunology , Male , Middle Aged , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/immunology , Recurrence , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/immunology , Tuberculosis, Multidrug-Resistant/blood , Tuberculosis, Multidrug-Resistant/immunology , Tuberculosis, Multidrug-Resistant/microbiology , Tuberculosis, Pulmonary/blood , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Young Adult
10.
Malar J ; 19(1): 309, 2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32859199

ABSTRACT

BACKGROUND: The Plasmodium falciparum sexual-stage surface proteins Pfs25 and Pfs230 are antigen candidates for a malaria transmission-blocking vaccine (TBV), and have been widely investigated as such. It is not clear whether simultaneously presenting these two antigens in a particulate vaccine would enhance the transmission reducing activity (TRA) of induced antibodies. To assess this, immunization was carried out with liposomes containing synthetic lipid adjuvant monophosphoryl lipid A (MPLA), and cobalt-porphyrin-phospholipid (CoPoP), which rapidly converts recombinant, his-tagged antigens into particles. METHODS: His-tagged, recombinant Pfs25 and Pfs230C1 were mixed with CoPoP liposomes to form a bivalent vaccine. Antigens were fluorescently labelled to infer duplex particleization serum-stability and binding kinetics using fluorescence resonance energy transfer. Mice and rabbits were immunized with individual or duplexed particleized Pfs25 and Pfs230C1, at fixed total antigen doses. The resulting antibody responses were assessed for magnitude and TRA. RESULTS: Pfs230C1 and Pfs25 rapidly bound CoPoP liposomes to form a serum-stable, bivalent particle vaccine. In mice, immunization with 5 ng of total antigen (individual antigen or duplexed) elicited functional antibodies against Pfs25 and Pfs230. Compared to immunization with the individual antigen, Pfs25 antibody production was moderately lower for the bivalent CoPoP vaccine, whereas Pfs230C1 antibody production was not impacted. All antibodies demonstrated at least 92% inhibition in oocyst density at 750 µg/mL purified mouse IgG in the standard membrane feeding assay (SMFA). At lower IgG concentrations, the bivalent vaccine did not improve TRA; antibodies induced by particleized Pfs25 alone showed stronger function in these conditions. In rabbits, immunization with a 20 µg total antigen dose with the duplexed antigens yielded similar antibody production against Pfs25 and Pfs230 compared to immunization with a 20 µg dose of individual antigens. However, no enhanced TRA was observed with duplexing. CONCLUSIONS: Pfs25, Pfs230 or the duplexed combination can readily be prepared as particulate vaccines by mixing CoPoP liposomes with soluble, recombinant antigens. This approach induces potent transmission-reducing antibodies following immunization in mice and rabbits. Immunization with bivalent, particleized, Pfs230 and Pfs25 did not yield antibodies with superior TRA compared to immunization with particleized Pfs25 as a single antigen. Altogether, duplexing antigens is straightforward and effective using CoPoP liposomes, but is likely to be more useful for targeting distinct parasite life stages.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Immunization , Lipid A/analogs & derivatives , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Animals , Female , Injections, Intramuscular , Lipid A/administration & dosage , Lipid A/immunology , Liposomes/administration & dosage , Liposomes/immunology , Mice , Rabbits
11.
Immunotherapy ; 12(13): 983-995, 2020 09.
Article in English | MEDLINE | ID: mdl-32752904

ABSTRACT

Aim: We tested the safety and immunogenicity of a novel vaccine in patients with resected high-risk melanoma. Patients & methods: HLA-A2-positive patients with resected Stage II-IV melanoma were randomized to receive up to three vaccinations of melanoma-associated peptide (MART-1a) combined with a stable oil-in-water emulsion (SE) either with the Toll-like receptor 4 agonist glucopyranosyl lipid A (GLA-SE-Schedule 1) or alone (SE-Schedule 2). Safety and immunogenicity of the vaccines were monitored. Results: A total of 23 patients were registered. No treatment-related grade 3 or higher adverse events were observed. Increases in MART-1a-specific T cells were seen in 70 and 63% of Schedule 1 and Schedule 2 patients, respectively. Conclusion: Both vaccine schedules were well-tolerated and resulted in an increase in MART-1a-specific T cells. Clinical Trial registration: NCT02320305 (ClinicalTrials.gov).


Subject(s)
Glucosides/therapeutic use , Lipid A/therapeutic use , Melanoma/immunology , Vaccines, Subunit/therapeutic use , Adult , Aged , Aged, 80 and over , Emulsions/administration & dosage , Emulsions/therapeutic use , Female , Glucosides/administration & dosage , Humans , Lipid A/administration & dosage , Male , Middle Aged , Water
12.
Front Immunol ; 11: 1264, 2020.
Article in English | MEDLINE | ID: mdl-32714323

ABSTRACT

Subunit vaccines are theoretically safe and easy to manufacture but require effective adjuvants and delivery systems to yield protective immunity, particularly at critical mucosal sites such as the lung. We investigated nanolipoprotein particles (NLPs) containing the Toll-like receptor 4 agonist monophosphoryl lipid A (MPLA) as a platform for intranasal vaccination against Bacillus anthracis. Modified lipids enabled attachment of disparate spore and toxin protein antigens. Intranasal vaccination of mice with B. anthracis antigen-MPLA-NLP constructs induced robust IgG and IgA responses in serum and in bronchoalveolar and nasal lavage. Typically, a single dose sufficed to induce sustained antibody titers over time. When multiple immunizations were required for sustained titers, specific antibodies were detected earlier in the boost schedule with MPLA-NLP-mediated delivery than with free MPLA. Administering combinations of constructs induced responses to multiple antigens, indicating potential for a multivalent vaccine preparation. No off-target responses to the NLP scaffold protein were detected. In summary, the NLP platform enhances humoral and mucosal responses to intranasal immunization, indicating promise for NLPs as a flexible, robust vaccine platform against B. anthracis and potentially other inhalational pathogens.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/prevention & control , Bacillus anthracis/immunology , Nanoparticles , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Anthrax Vaccines/administration & dosage , Antibodies, Bacterial/immunology , Female , Lipid A/administration & dosage , Lipid A/analogs & derivatives , Lipid A/immunology , Mice , Mice, Inbred BALB C , Spores, Bacterial/immunology , Vaccines, Subunit/immunology
13.
Cancer Immunol Immunother ; 69(10): 2113-2124, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32448982

ABSTRACT

Metastatic cancer involving spread to the peritoneal cavity is referred to as peritoneal carcinomatosis and has a very poor prognosis. Our previous studies demonstrated a toll-like receptor 4 (TLR4) and C-type lectin receptor (CLR; Mincle/MCL) agonist pairing of monophosphoryl lipid A (MPL) and trehalose-6,6'-dicorynomycolate (TDCM) effectively inhibits peritoneal tumor growth and ascites development through a mechanism dependent upon B1a cell-produced natural IgM, complement, and phagocytes. In the current study, we investigated the requirement for TLR4 and Fc receptor common γ chain (FcRγ), required for Mincle/MCL signaling, in the MPL/TDCM-elicited response. MPL/TDCM significantly increased macrophages and Ly6Chi monocytes in the peritoneal cavity of both TLR4-/- and FcRγ-/- mice, suggesting redundancy in the signals required for monocyte/macrophage recruitment. However, B1 cell activation, antibody secreting cell differentiation, and tumor-reactive IgM production were defective in TLR4-/-, but not FcRγ-/- mice. TRIF was required for production of IgM reactive against tumor- and mucin-related antigens, but not phosphorylcholine, whereas TLR4 was required for production of both types of reactivities. Consistent with this, B1 cells lacking TLR4 or TRIF did not proliferate or differentiate into tumor-reactive IgM-producing cells in vitro and did not reconstitute MPL/TDCM-dependent protection against peritoneal carcinomatosis in CD19-/- mice. Our results indicate a TLR4/TRIF-dependent pathway is required by B1 cells for MPL/TDCM-elicited production of protective tumor-reactive natural IgM. The dependency on TRIF signaling for tumor-reactive, but not phosphorylcholine-reactive, IgM production reveals unexpected heterogeneity in TLR4-dependent regulation of natural IgM production, thereby highlighting important differences to consider when designing vaccines or therapies targeting these specificities.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , B-Lymphocyte Subsets/immunology , Cord Factors/administration & dosage , Immunoglobulin M/immunology , Lipid A/analogs & derivatives , Peritoneal Neoplasms/immunology , Toll-Like Receptor 4/physiology , Adjuvants, Immunologic/administration & dosage , Animals , B-Lymphocyte Subsets/drug effects , Lipid A/administration & dosage , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/physiology , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology
14.
Acta Parasitol ; 65(3): 652-660, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32240492

ABSTRACT

PURPOSE: Schistosomiasis is a disease that afflicts over 220 million people worldwide. To date, there is no vaccine against schistosomiasis and chemotherapy relies basically on a single drug, praziquantel. The current study was undertaken to investigate the therapeutic effects of monophosphoryl lipid A (MPLA) as an adjuvant in soluble egg antigen (SEA)-vaccinated and Schistosoma mansoni-infected mice. METHODS: Mice were divided into two groups of uninfected and Schistosoma mansoni infected. The two groups were treated differently with MPLA, SEA and praziquantel. Study parameters included parasitological, immunological and biochemical parameters. RESULTS: Parasitological parameters revealed that intraperitoneal injection of MPLA into SEA-vaccinated and S. mansoni-infected mice was effective in reducing the worm and egg burden, granuloma count and diameter as well as the total area of infection in their livers versus SEA-untreated but infected ones. In addition, MPLA showed ameliorative action on the elevated liver oxidative stress marker, including malondialdehyde (MDA) and a decrease in the level of the antioxidant enzymes, reduced glutathione (GSH) and catalase (CAT) which may have a role in the liver damage and fibrosis due to S. mansoni infection. CONCLUSION: Treatment with MPLA has multi-functions in attenuating the deleterious impacts of S. mansoni infection in mice livers. Its effects are mediated through a reduction of ova count, worm burden, granuloma diameter and amelioration of antioxidant defense systems, and liver function biomarkers.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Lipid A/analogs & derivatives , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/prevention & control , Toll-Like Receptors/agonists , Animals , Anthelmintics/therapeutic use , Antigens, Helminth/administration & dosage , Antigens, Helminth/immunology , Helminth Proteins/administration & dosage , Helminth Proteins/immunology , Lipid A/administration & dosage , Lipid A/immunology , Male , Mice , Praziquantel/therapeutic use , Schistosoma mansoni , Vaccination
15.
Cancer Immunol Immunother ; 69(5): 745-758, 2020 May.
Article in English | MEDLINE | ID: mdl-32047957

ABSTRACT

BACKGROUND: Osteosarcoma (OS) is the most common malignant bone tumor and the prognosis of advanced cases is still poor. Recently, there have been several reports suggesting the relationship between innate immunity and OS, but the detailed mechanism is unknown. We demonstrate the relationship between OS and Toll-like receptor 4 (TLR4) which is one of the most important factors in innate immunity. METHODS: We established a syngenic mouse tumor model using C3H/HeN, C3H/HeJ mouse and a highly metastatic OS cell line, LM8. TLR4 activation with lipopolysaccharide (LPS) was performed on both mice and its influence on the progression of OS was evaluated. We also performed CD8 + cells depletion to examine the influence on TLR4 activation effects. RESULTS: Tumor volume of C3H/HeN mice was significantly smaller and overall survival of C3H/HeN mice was significantly longer than C3H/HeJ mice. We found more CD8+ cells infiltrating in lung metastases of C3H/HeN mice and depletion of CD8+ cells canceled the antitumor effects of LPS. CONCLUSION: TLR4 activation by LPS increased CD8+ cells infiltrating into lung metastases and suppressed OS progression in the mouse model. TLR4 activation may suppress the progression of OS via stimulating CD8+ cells and can be expected as a novel treatment for OS.


Subject(s)
Bone Neoplasms/immunology , Lung Neoplasms/immunology , Osteosarcoma/immunology , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 4/metabolism , Adjuvants, Immunologic/administration & dosage , Adolescent , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Cell Line, Tumor/transplantation , Chemotherapy, Adjuvant , Child , Child, Preschool , Disease Models, Animal , Disease Progression , Female , Humans , Kaplan-Meier Estimate , Lipid A/administration & dosage , Lipid A/analogs & derivatives , Lipopolysaccharides/administration & dosage , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Lymphocyte Depletion , Male , Mice , Middle Aged , Osteosarcoma/mortality , Osteosarcoma/secondary , Osteosarcoma/therapy , Prognosis , Progression-Free Survival , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/immunology , Tumor Burden/immunology , Young Adult
16.
Biomater Sci ; 8(4): 1106-1116, 2020 Feb 21.
Article in English | MEDLINE | ID: mdl-31994549

ABSTRACT

In this study, we sought to design a bionanomaterial that could exert anticancer effects against primary tumors and protect against rechallenged tumors via photodynamic immunotherapy. As a biomaterial, we used an amphiphilic phenylalanine derivative of poly-gamma glutamic acid, which forms nanoparticles by self-assembly. For anticancer effects, we co-entrapped hydrophobic chlorin e6 and monophosphoryl lipid A in the core of the plain amphiphilic phenylalanine nanoparticles (AN), to generate M/C/AN. For comparison, we used plain AN and chlorin e6-loaded AN (C/AN). In vitro studies showed that B16F10 cancer cells treated with C/AN or M/C/AN generated reactive oxygen species and exhibited an enhanced surface display of calreticulin upon exposure to 660 nm light irradiation. C/AN and M/C/AN exerted similar photodynamic anticancer effects; however, M/C/AN, but not C/AN, induced in vitro dendritic cell maturation. Our biodistribution study revealed that C/AN and M/C/AN showed higher accumulation at the tumor tissues compared to that seen in the free chlorin e6-treated group. In B16F10 tumor-bearing mice, the intravenous injection of C/AN or M/C/AN showed similar photodynamic anticancer effects against primary tumors. However, the growth of rechallenged tumors was more significantly inhibited in the M/C/AN group compared to the C/AN group. At day 40 after inoculation of the primary tumor, M/C/AN-treated mice showed 100% survival, whereas the other groups showed 0% survival. In the tumor microenvironment, higher infiltration of CD8+ T cells was observed in the M/C/AN group compared to the other groups. Our results suggest that AN co-loaded with a photosensitizer and an immune stimulant may hold great potential for use in photodynamic immunotherapy to inhibit both primary and metastatic tumors.


Subject(s)
Biomimetics/methods , Lipid A/analogs & derivatives , Melanoma, Experimental/drug therapy , Porphyrins/administration & dosage , Administration, Intravenous , Animals , Capsules , Cell Line, Tumor , Cell Survival/drug effects , Chlorophyllides , Immunotherapy , Lipid A/administration & dosage , Lipid A/chemistry , Lipid A/pharmacokinetics , Lipid A/pharmacology , Melanoma, Experimental/immunology , Mice , Nanoparticles , Photochemotherapy , Polyglutamic Acid/analogs & derivatives , Polyglutamic Acid/chemistry , Polymers/chemistry , Porphyrins/chemistry , Porphyrins/pharmacokinetics , Porphyrins/pharmacology , Tissue Distribution , Tumor Microenvironment/drug effects
17.
Int J Cancer ; 147(1): 170-174, 2020 07 01.
Article in English | MEDLINE | ID: mdl-31736068

ABSTRACT

We studied effectiveness of the AS04-adjuvanted HPV-16/18 (AS04-HPV-16/18) vaccine against human papillomavirus (HPV) oropharyngeal infections associated with the increase of head/neck cancers in western countries. All 38,631 resident adolescents from 1994 to 1995 birth cohorts of 33 Finnish communities were invited in this community-randomized trial (NCT00534638). During 2008-2009, 11,275 girls and 6,129 boys were enrolled in three arms of 11 communities each. In Arm A, 90% of vaccinated girls/boys, and in Arm B, 90% of vaccinated girls received AS04-HPV-16/18 vaccine. Other Arm A/B and all Arm C vaccinated participants received control vaccine. All Arm A participants and Arm B female participants were blinded to vaccine allocation. Oropharyngeal samples were analyzed from 4,871 18.5-year-old females who attended follow-up visit 3-6 years postvaccination. HPV DNA prevalence was determined by SPF-10 LiPA and Multiplex type-specific PCR. Total vaccine effectiveness (VE) was defined as relative reduction of oropharyngeal HPV prevalence in pooled Arms A/B HPV-vaccinated females vs. all Arm C females. VE against oropharyngeal HPV-16/18, HPV-31/45 and HPV-31/33/45 infections were 82.4% (95% confidence intervals [CI]: 47.3-94.1), 75.3% (95%CI: 12.7-93.0) and 69.9% (95% CI: 29.6-87.1), respectively. In conclusion, the AS04-HPV-16/18 vaccine showed effectiveness against vaccine and nonvaccine HPV-types oropharyngeal infections in adolescent females up to 6 years postvaccination.


Subject(s)
Human papillomavirus 16/immunology , Human papillomavirus 18/immunology , Oropharynx/virology , Papillomavirus Infections/epidemiology , Papillomavirus Vaccines/administration & dosage , Pharyngeal Diseases/epidemiology , Adolescent , Adult , Aluminum Hydroxide/administration & dosage , Female , Finland/epidemiology , Humans , Lipid A/administration & dosage , Lipid A/analogs & derivatives , Oropharyngeal Neoplasms/epidemiology , Oropharyngeal Neoplasms/immunology , Oropharyngeal Neoplasms/prevention & control , Oropharyngeal Neoplasms/virology , Oropharynx/immunology , Papillomavirus Infections/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/immunology , Pharyngeal Diseases/immunology , Pharyngeal Diseases/prevention & control , Pharyngeal Diseases/virology , Seroepidemiologic Studies , Young Adult
18.
Oncol Rep ; 43(1): 270-281, 2020 01.
Article in English | MEDLINE | ID: mdl-31746432

ABSTRACT

Lipopolysaccharides are the main surface antigens and virulence factors of gram­negative bacteria. Removal of four ester­bound fatty acid residues from hexaacyl lipid A of Escherichia coli lipooligosaccharide (LOS) resulted in the de­O­acylated derivative E. coli LOS­OH (LOS­OH). This procedure caused a significant reduction in the toxicity of this compound compared to the native molecule. We investigated the effect of such a structural LOS modification on its biological activity using in vitro assays with monocytic cells of the RAW264.7 line, dendritic cells of the JAWS II line, bone marrow­derived dendritic cells (BM­DCs), and spleen cells. Furthermore, in in vivo experiments with a melanoma B16 metastasis model, the anti­metastatic activity of the compounds and spleen cell reactivity mediated by them representing a systemic response were analyzed. The results revealed that LOS­OH demonstrated weaker ability than LOS to stimulate and polarize an immune response both in vitro and in vivo. It induced lower cytokine production by cells of myeloid lines. Multiple applications of LOS­OH into mice injected intravenously with B16 cells significantly (P<0.05; P<0.01) reduced the number of metastatic foci in the lungs, presumably via silencing of myeloid cell reactivity as well as the inability to stimulate lymphoid cells both directly and indirectly. These findings suggest that LOS­OH maintained in the body of metastasis­bearing mice appears to modulate or downregulate the innate response, leading to the inability of blood myeloid cells to support the migration of melanoma cells to lung tissue.


Subject(s)
Escherichia coli/metabolism , Lipid A/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Melanoma, Experimental/drug therapy , Animals , Cell Line, Tumor , Escherichia coli Proteins/administration & dosage , Escherichia coli Proteins/pharmacology , Female , Humans , Injections, Intravenous , Lipid A/chemistry , Lipid A/pharmacology , Lung Neoplasms/immunology , Melanoma, Experimental/immunology , Mice , RAW 264.7 Cells , Tumor Escape/drug effects , Xenograft Model Antitumor Assays
19.
Nanomedicine ; 22: 102092, 2019 11.
Article in English | MEDLINE | ID: mdl-31593795

ABSTRACT

Dendritic cell (DC)-targeted vaccines based on nanotechnology are a promising strategy to efficiently induce potent immune responses. We synthesized and manufactured a mannose-modified poly (ß-amino ester) (PBAE) nano-vaccines with easily tuneable and pH-sensitive characteristics to co-deliver the tumor-associated antigen polypeptide Trp-2 and the TLR4 agonist monophosphoryl lipid A (MPLA). To reduce immunosuppression in the tumor microenvironment, an immune checkpoint inhibitor, PD-L1 antagonist, was administrated along with PBAE nano-vaccines to delay melanoma development. We found that mannosylated Trp-2 and MPLA-loaded PBAE nano-vaccines can target and mature DCs, consequently boosting antigen-specific cytotoxic T lymphocyte activity against melanoma. The prophylactic study indicates that combination therapy with PD-L1 antagonist further enhanced anti-tumor efficacy by 3.7-fold and prolonged median survival time by 1.6-fold more than free Trp-2/MPLA inoculation. DC-targeting PBAE polymers have a great potential as a nanotechnology platform to design vaccines and achieve synergistic anti-tumor effects with immune checkpoint therapy.


Subject(s)
Cancer Vaccines/immunology , Drug Delivery Systems , Lipid A/analogs & derivatives , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Nanoparticles/chemistry , Peptides/administration & dosage , Polymers/chemistry , Animals , Cancer Vaccines/adverse effects , Cytokines/metabolism , Dendritic Cells/immunology , Female , Hydrogen-Ion Concentration , Immunization , Lipid A/administration & dosage , Lymph Nodes/metabolism , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Nanoparticles/ultrastructure , Treatment Outcome
20.
Drug Alcohol Depend ; 204: 107484, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31521953

ABSTRACT

BACKGROUND: Methamphetamine use disorder continues to be inadequately treated, but improvements are being made in the field of immunotherapeutics, including vaccines, which could provide new options for treatment. Cocaine and nicotine vaccines have been tested clinically, but have yet to elicit the necessary antibody concentrations required to be effective. Methamphetamine vaccines have been tested in multiple nonclinical models and appear promising. Improved adjuvants have the potential to further stimulate the immune system to reach effective levels of antibodies. Previously, the methamphetamine vaccine IXT-v100 was administered with GLA-SE, a toll-like receptor 4 agonist, in mice to produce higher levels of antibodies than when it was administered with two other widely used adjuvants, Alhydrogel and Sigma Adjuvant System. METHODS: The purpose of this research was to evaluate IXT-v100, given in combination with the adjuvant GLA-SE, to determine its efficacy in antagonizing methamphetamine disposition in a rat pharmacokinetic study. Additional rat studies were conducted to compare the ability of IXT-v100 manufactured with greater hapten densities to elicit higher antibody levels. RESULTS: As expected based on prior studies with anti-methamphetamine monoclonal antibodies, the antibodies resulting from vaccination with IXT-v100 altered methamphetamine pharmacokinetics by increasing serum concentrations and extending the half-life. Furthermore, intentional variations in the ratio of components during manufacturing led to production of vaccines with higher hapten densities. The higher hapten densities resulted in production of antibodies that maintained the ability to bind methamphetamine with high affinity. CONCLUSIONS: The results support continued development of IXT-v100 for the treatment of methamphetamine use disorder.


Subject(s)
Antibody Formation/drug effects , Central Nervous System Stimulants/blood , Glucosides/administration & dosage , Lipid A/administration & dosage , Methamphetamine/blood , Vaccination/trends , Adjuvants, Immunologic/administration & dosage , Amphetamine-Related Disorders/blood , Amphetamine-Related Disorders/drug therapy , Animals , Antibody Formation/physiology , Central Nervous System Stimulants/administration & dosage , Central Nervous System Stimulants/pharmacokinetics , Dose-Response Relationship, Drug , Male , Methamphetamine/administration & dosage , Methamphetamine/pharmacokinetics , Rats , Rats, Sprague-Dawley , Vaccines/administration & dosage , Vaccines/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...