Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Methods Mol Biol ; 2223: 133-149, 2021.
Article in English | MEDLINE | ID: mdl-33226592

ABSTRACT

Mouse models of allergic conjunctivitis mimic various aspects of human allergic conjunctivitis. They are useful as acute models of allergic conjunctivitis to study immunological aspects of this condition. In this chapter, we will describe ragweed-pollen-induced experimental allergic conjunctivitis (mostly driven by adaptive immunity), and papain-soaked contact lens-induced experimental allergic conjunctivitis (mostly driven by innate immunity). Giemsa staining of histological sections is used for quantification of the number of infiltrating eosinophils, which is useful to evaluate the severity of the allergic inflammation. Immunohistochemical staining and quantitative PCR are used to clarify spatiotemporal expression of proinflammatory molecules in the conjunctival tissue. Flow cytometric analysis of conjunctival tissue is used for the detection of innate lymphoid cell type 2 (ILC2) in the ocular surface tissues.


Subject(s)
Ambrosia/immunology , Conjunctiva/drug effects , Conjunctivitis, Allergic/immunology , Disease Models, Animal , Lymphocytes/drug effects , Papain/administration & dosage , Adaptive Immunity/drug effects , Adjuvants, Immunologic/administration & dosage , Allergens/administration & dosage , Aluminum Hydroxide/administration & dosage , Ambrosia/chemistry , Animals , Biomarkers/metabolism , Conjunctiva/immunology , Conjunctiva/pathology , Conjunctivitis, Allergic/chemically induced , Conjunctivitis, Allergic/genetics , Conjunctivitis, Allergic/pathology , Eosinophils/drug effects , Eosinophils/immunology , Eosinophils/pathology , Female , Flow Cytometry/methods , Gene Expression , Immunity, Innate/drug effects , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Interleukins/genetics , Interleukins/immunology , Lymphocytes/immunology , Lymphocytes/pathology , Mice , Mice, Inbred C57BL , Pollen/adverse effects , Pollen/immunology , Real-Time Polymerase Chain Reaction/methods
2.
Methods Mol Biol ; 2223: 295-335, 2021.
Article in English | MEDLINE | ID: mdl-33226602

ABSTRACT

Allergic asthma is characterized by airway hyperresponsiveness, remodeling, and reversible airway obstruction. This is associated with an eosinophilic inflammation of the airways, caused by inhaled allergens such as house dust mite or grass pollen. The inhaled allergens trigger a type-2 inflammatory response with the involvement of innate lymphoid cells (ILC2) and Th2 cells, resulting in high immunoglobulin E (IgE) antibody production by B cells and mucus production by airway epithelial cells. As a consequence of the IgE production, subsequent allergen reexposure results in a classic allergic response with distinct early and late phases, both resulting in bronchoconstriction and shortness of breath. Allergen-specific immunotherapy (AIT) is the only treatment that is capable of modifying the immunological process underlying allergic responses including allergic asthma. Both subcutaneous AIT (SCIT) as well as sublingual AIT (SLIT) have shown clinical efficacy in long-term suppression of the allergic response. Although AIT treatments are very successful for rhinitis, application in asthma is hampered by variable efficacy, long duration of treatment, and risk of severe side effects. A more profound understanding of the mechanisms by which AIT induces tolerance to allergens in sensitized individuals is needed to be able to improve its efficacy. Mouse models have been very valuable in preclinical research for characterizing the mechanisms of desensitization in AIT and evaluating novel approaches to improve its efficacy. Here, we present a rapid and reproducible mouse model for allergen-specific immunotherapy. In this model, mice are sensitized with two injections of allergen adsorbed to aluminum hydroxide, followed by subcutaneous injections (SCIT) or sublingual administrations (SLIT) of allergen extracts as an immunotherapy treatment. Finally, mice are challenged by intranasal allergen administrations. We will also describe the protocols as well as the most important readout parameters for the measurements of invasive lung function, serum immunoglobulin levels, isolation of bronchoalveolar lavage fluid (BALF), and preparation of cytospin slides. Moreover, we describe how to perform ex vivo restimulation of lung single-cell suspensions with allergens, flow cytometry for identification of relevant immune cell populations, and ELISAs and Luminex assays for assessment of the cytokine concentrations in BALF and lung tissue.


Subject(s)
Allergens/administration & dosage , Asthma/therapy , Disease Models, Animal , Pollen/immunology , Pyroglyphidae/immunology , Sublingual Immunotherapy/methods , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Allergens/immunology , Aluminum Hydroxide/administration & dosage , Animals , Asthma/immunology , Asthma/pathology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Complex Mixtures/administration & dosage , Complex Mixtures/immunology , Cytokines/genetics , Cytokines/immunology , Ear , Eosinophils/immunology , Eosinophils/pathology , Female , Humans , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Injections, Subcutaneous , Lung/drug effects , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutrophils/immunology , Neutrophils/pathology , Pollen/chemistry , Pyroglyphidae/chemistry , Single-Cell Analysis/methods
3.
Int Arch Allergy Immunol ; 181(2): 94-102, 2020.
Article in English | MEDLINE | ID: mdl-31865326

ABSTRACT

Only few data on safety during high-dose, accelerated escalation schedules during subcutaneous allergen immunotherapy (AIT) are available. The aim of this study was to assess the safety and tolerability of an accelerated dose escalation schedule of AIT in adult patients with moderate to severe seasonal rhinoconjunctivitis in a multicenter, open-label, randomized phase II trial. The dose escalation scheme for patients in Group I (1 strength) included 3 injections with 1 strength, B (10,000 TU/mL), whereas the dose escalation scheme for Group II (standard) included 7 injections with 2 strengths, A (1,000 TU/mL) and B (10,000 TU/mL), of an aluminum hydroxide-adsorbed allergoid grass pollen preparation. Overall, 72 of 87 randomized patients (83.7%) reported at least 1 treatment-emergent adverse event (TEAE; 82.2 [Group I] vs. 85.4% [Group II]); 58.8% of all reported TEAEs were assessed as being related to AIT (60.0 vs. 48.8%). The most frequently reported AIT-related TEAEs were swelling (46.7 vs. 34.1%), erythema (28.9 vs. 36.6%), and pruritus (31.1 vs. 17.1%) at the site of the injection. Systemic allergic reactions occurred in 5 (5.8%) patients overall, with more being reported in the 1-strength group (4 [8.9%] vs. 1 [2.4%]). All systemic allergic reactions were classified as World Allergy Organization (WAO) Grade 1 or Grade 2 reactions. Accelerated high-dose escalation with an aluminum hydroxide-adsorbed grass pollen allergoid can be initiated with a safety and tolerability profile comparable to the standard dose escalation schedule in patients with allergic rhinitis with or without asthma.


Subject(s)
Allergoids/chemistry , Allergoids/immunology , Aluminum Hydroxide/chemistry , Poaceae/immunology , Rhinitis, Allergic, Seasonal/immunology , Rhinitis, Allergic/immunology , Adult , Allergens/immunology , Allergoids/administration & dosage , Aluminum Hydroxide/administration & dosage , Aluminum Hydroxide/immunology , Antigens, Plant/immunology , Asthma/immunology , Conjunctivitis, Allergic/immunology , Desensitization, Immunologic/methods , Female , Humans , Male , Pollen/immunology
4.
Arch Toxicol ; 93(10): 2787-2796, 2019 10.
Article in English | MEDLINE | ID: mdl-31522239

ABSTRACT

Aluminium (Al) toxicokinetics after intramuscular (IM) injection of Al-adjuvanted vaccines is unknown. Since animal data are required for modeling and extrapolation, a rat study was conducted measuring Al in plasma and tissues after IM injection of either plain Al-hydroxide (pAH) or Al-phosphate (pAP) adjuvant (Al dose 1.25 mg), single human doses of three Al-adjuvanted vaccines (V1, V2, and V3; Al doses 0.5-0.82 mg), or vehicle (saline). A significant increase in Al plasma levels compared to controls was observed after pAP (AUC(0-80 d), mean ± SD: 2424 ± 496 vs. 1744 ± 508 µg/L*d). Percentage of Al dose released from injected muscle until day 80 was higher after pAP (66.9%) and AP-adjuvanted V3 (85.5%) than after pAH and AH-adjuvanted V1 (0 and 22.3%, resp.). Estimated absolute Al release was highest for pAP (836.8 µg per rat). Al concentration in humerus bone was increased in all groups, again strongest in the pAP group [3.35 ± 0.39 vs. 0.05 ± 0.06 µg/g wet weight (ww)]. Extrapolated amounts in whole skeleton corresponded to 5-12% of the released Al dose. Very low brain Al concentrations were observed in all groups (adjuvant group means 0.14-0.29 µg/g ww; control 0.13 ± 0.04 µg/g ww). The results demonstrate systemically available Al from marketed vaccines in rats being mainly detectable in bone. Al release appears to be faster from AP- than AH-adjuvants. Dose scaling to human adults suggests that increase of Al in plasma and tissues after single vaccinations will be indistinguishable from baseline levels.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum Compounds/administration & dosage , Aluminum Hydroxide/administration & dosage , Phosphates/administration & dosage , Vaccines/administration & dosage , Adjuvants, Immunologic/pharmacokinetics , Aluminum Compounds/pharmacokinetics , Aluminum Hydroxide/pharmacokinetics , Animals , Area Under Curve , Humans , Injections, Intramuscular , Male , Phosphates/pharmacokinetics , Rats , Rats, Wistar , Tissue Distribution , Vaccines/pharmacokinetics
5.
J Gastroenterol Hepatol ; 34(12): 2077-2085, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31117149

ABSTRACT

BACKGROUND AND AIM: Treatment options for functional dyspepsia (FD) refractory to pharmacological treatments are limited but the effectiveness of electroacupuncture (EA) is uncertain. We assessed the effectiveness of EA combined with on-demand gastrocaine. METHODS: We conducted a single-center, assessor-blind, randomized parallel-group 2-arm trial on Helicobacter pylori negative FD patients of the postprandial distress syndrome subtype refractory to proton pump inhibitor, prokinetics, or H2 antagonists. Enrolled participants were block randomized in a 1:1 ratio, with concealed random sequence. The treatment and control groups both received on-demand gastrocaine for 12 weeks, but only those in treatment group were offered 20 sessions of EA over 10 weeks. The primary endpoint was the between-group difference in proportion of patients achieving adequate relief of symptoms at week 12. RESULTS: Of 132 participants randomly assigned to EA plus on-demand gastrocaine (n = 66) or on-demand gastrocaine alone (n = 66), 125 (94.7%) completed all follow-up at 12 weeks. The EA group had a compliance rate 97.7%. They had a significantly higher likelihood in achieving adequate symptom relief at 12 weeks, with a clinically relevant number needed to treat (NNT) value of 2.36 (95% CI: 1.74, 3.64). Among secondary outcomes, statistically and clinically significant improvements were observed among global symptom (NNT = 3.85 [95% CI: 2.63, 7.69]); postprandial fullness and early satiation (NNT = 5.00 [95% CI: 2.86, 25.00]); as well as epigastric pain, epigastric burning, and postprandial nausea (NNT = 4.17 [95% CI: 2.56, 11.11]). Adverse events were minimal and nonsignificant. CONCLUSION: For refractory FD, EA provides significant, clinically relevant symptom relief when added to on-demand gastrocaine (ChiCTR-IPC-15007109).


Subject(s)
Aluminum Hydroxide/therapeutic use , Aminobenzoates/therapeutic use , Atropine/therapeutic use , Dyspepsia/drug therapy , Electroacupuncture/methods , Magnesium Compounds/therapeutic use , Adult , Aluminum Hydroxide/administration & dosage , Aminobenzoates/administration & dosage , Atropine/administration & dosage , Combined Modality Therapy , Drug Administration Schedule , Drug Combinations , Electroacupuncture/adverse effects , Female , Humans , Magnesium Compounds/administration & dosage , Male , Middle Aged , Prospective Studies , Single-Blind Method , Treatment Outcome
6.
Fish Shellfish Immunol ; 85: 44-51, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29017943

ABSTRACT

Flavobacterium psychrophilum is the causative agent of Rainbow Trout Fry Syndrome which has had a major impact on global salmonid aquaculture. Recent outbreaks in Atlantic salmon in Scotland and Chile have added to the need for a vaccine to protect both salmon and trout. At present no licensed vaccines are available in Europe, leaving antibiotics as the only course of action to contain disease outbreaks. Outbreaks generally occur in fry at temperatures between 10 and 15 °C. Recently outbreaks in larger fish have given added impetus to the development of a vaccine which can provide long term protection from this highly heterogeneous pathogen. Most fish injectable vaccines are formulated with oil emulsion adjuvants to induce strong and long lasting immunity, but which are known to cause side effects. Alternative adjuvants are currently sought to minimise these adverse effects. The current study was performed to assess the efficacy of a polyvalent, whole cell vaccine containing formalin-inactivated F. psychrophilum to induce protective immunity in Atlantic salmon. The vaccine was formulated with an adjuvant containing squalene and aluminium hydroxide, and was compared to a vaccine formulated with a traditional oil adjuvant, Montanide ISA 760VG, and a non-adjuvanted vaccine. Duplicate groups of salmon (23.5 ± 6.8 g) were vaccinated with each of the vaccine formulations or phosphate buffered saline by intraperitoneal injection. Fish were challenged by intramuscular injection with F. psychrophilum six weeks post-vaccination to test the efficacy of the vaccines. Cumulative mortality reached 70% in the control salmon, while the groups of salmon that received vaccine had significantly lower mortality than the controls (p = 0.0001), with no significant difference in survival between vaccinated groups. The squalene/alum adjuvant was safe, more readily metabolised by the fish and induced less histopathological changes than the traditional oil adjuvant.


Subject(s)
Adjuvants, Immunologic/pharmacology , Bacterial Vaccines/pharmacology , Fish Diseases/prevention & control , Flavobacteriaceae Infections/veterinary , Flavobacterium/immunology , Salmo salar/immunology , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Aluminum Hydroxide/pharmacology , Animals , Bacterial Vaccines/administration & dosage , Fish Diseases/immunology , Fish Diseases/microbiology , Flavobacteriaceae Infections/immunology , Flavobacteriaceae Infections/microbiology , Flavobacteriaceae Infections/prevention & control , Random Allocation , Squalene/administration & dosage , Squalene/pharmacology
7.
J Control Release ; 292: 111-118, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30339906

ABSTRACT

Intranasal vaccination using dry powder vaccine formulation represents an attractive, non-invasive vaccination modality with better storage stability and added protection at the mucosal surfaces. Herein we report that it is feasible to induce specific mucosal and systemic antibody responses by intranasal immunization with a dry powder vaccine adjuvanted with an insoluble aluminum salt. The dry powder vaccine was prepared by thin-film freeze-drying of a model antigen, ovalbumin, adsorbed on aluminum (oxy)hydroxide as an adjuvant. Special emphasis was placed on the characterization of the dry powder vaccine formulation that can be realistically used in humans by a nasal dry powder delivery device. The vaccine powder was found to have "passable" to "good" flow properties, and the vaccine was uniformly distributed in the dry powder. An in vitro nasal deposition study using nasal casts of adult humans showed that around 90% of the powder was deposited in the nasal cavity. Intranasal immunization of rats with the dry powder vaccine elicited a specific serum antibody response as well as specific IgA responses in the nose and lung secretions of the rats. This study demonstrates the generation of systemic and mucosal immune responses by intranasal immunization using a dry powder vaccine adjuvanted with an aluminum salt.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Aluminum Oxide/administration & dosage , Vaccines/administration & dosage , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacokinetics , Administration, Intranasal , Aluminum Hydroxide/chemistry , Aluminum Hydroxide/pharmacokinetics , Aluminum Oxide/chemistry , Aluminum Oxide/pharmacokinetics , Animals , Antigens/administration & dosage , Antigens/chemistry , Antigens/immunology , Brain/metabolism , Bronchoalveolar Lavage Fluid/immunology , Female , Immunization , Immunoglobulin A/immunology , Immunoglobulin G/blood , Nasal Lavage Fluid/immunology , Ovalbumin/administration & dosage , Ovalbumin/chemistry , Ovalbumin/immunology , Powders , Rats, Sprague-Dawley , Vaccines/chemistry , Vaccines/pharmacokinetics
8.
Eur Arch Otorhinolaryngol ; 275(10): 2515-2521, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30062580

ABSTRACT

OBJECTIVES: Management of laryngopharyngeal reflux (LPR) typically comprises alginates and proton pump inhibitors (PPIs) alone or in combination, yet evidence to support any particular treatment regimen is lacking. We sought to evaluate the efficacy of Gaviscon® Advance alone versus co-prescription with a PPI in treating LPR. METHODS: One hundred consecutive LPR patients with a reflux symptom index (RSI) score > 10 attending our joint voice clinic (JVC) were studied prospectively. All were treated with Gaviscon® Advance four times daily. If patients had been started on a PPI prior to their JVC attendance, this was optimised to a twice-daily dosing regimen and continued. RSI scores were recorded at first attendance and 3 months post-treatment via postal questionnaire. Scores were analysed using t tests and Levene's test for equality of variances. RESULTS: Follow-up RSI scores were returned by 72 patients, 39 of whom were treated with Gaviscon® Advance only (group A) and 33 with Gaviscon® Advance + PPI (group B). Mean pre-treatment RSI scores were similar between groups [group A: 19.2, 95% confidence interval (CI) ± 2.4; group B: 21.3, 95% CI ± 3.2 (p = 0.65)]. No significant differences were observed with respect to 3-month post-treatment RSI scores [group A: 9.9, 95% CI ± 2.8; group B: 12.6, 95% CI ± 4.2 (p = 0.82)] and change in RSI scores [group A: 9.3, 95% CI ± 3.0; group B: 8.7, 95% CI ± 2.9 [p = 0.75]). CONCLUSIONS: Gaviscon® Advance alone is effective in treating symptoms of LPR, while co-prescription with a high-dose PPI offers no additional benefit.


Subject(s)
Alginates/administration & dosage , Aluminum Hydroxide/administration & dosage , Laryngopharyngeal Reflux/drug therapy , Silicic Acid/administration & dosage , Sodium Bicarbonate/administration & dosage , Adult , Aged , Aged, 80 and over , Antacids/administration & dosage , Dose-Response Relationship, Drug , Drug Combinations , Female , Humans , Laryngopharyngeal Reflux/diagnosis , Male , Middle Aged , Surveys and Questionnaires , Treatment Outcome , Voice/drug effects
9.
Toxicol In Vitro ; 43: 58-61, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28599844

ABSTRACT

Vaccines against human papilloma virus (HPV) have been demonstrated to be very effective to prevent infection-related neoplasms. However, several reports describing heterogeneous post-vaccination phenomena have been published in last few years. The spectrum of these disorders includes both immune-mediated neurological diseases and neuropsychiatric functional disorders. Some researchers speculated about a genetic predisposition, but others hypothesized a role of adjuvants, including some metals and, particularly, aluminum. Here, we tested sixteen young girls developing somatoform and neurocognitive syndromes after the HPV immunization, through MELISA® test, detecting cell-mediated hypersensitivity to several metals. We found no association between these neurocognitive disorders and the results provided by this test; importantly, no patients showed hypersensitivity to aluminum, which is the inorganic adjuvant included in HPV vaccines. Thus, if aluminum played a role in the pathophysiology of musculoskeletal and neurocognitive disturbances occurring in some young girls after HPV immunization, that should recognize other mechanisms than the activation of aluminum-specific lymphocytes.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Hypersensitivity/etiology , Papillomavirus Vaccines/adverse effects , Phosphates/administration & dosage , Adolescent , Adult , Child , Female , Humans , Hypersensitivity/blood , Hypersensitivity/immunology , Lymphocytes/immunology , Metals/administration & dosage , Metals/adverse effects , Neurocognitive Disorders/blood , Neurocognitive Disorders/etiology , Neurocognitive Disorders/immunology , Papillomavirus Vaccines/administration & dosage , Somatoform Disorders/blood , Somatoform Disorders/etiology , Somatoform Disorders/immunology , Young Adult
11.
BMC Infect Dis ; 17(1): 232, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28347293

ABSTRACT

BACKGROUND: Vaccination against seasonal influenza strains is recommended for "high risk" patient groups such as infants, elderly and those with respiratory or circulatory diseases. However, efficacy of the trivalent influenza vaccine (TIV) is poor in many cases and in the event of an influenza pandemic, mono-valent vaccines have been rapidly developed and deployed. One of the main issues with use of vaccine in pandemic situations is the lack of a suitable quantity of vaccine early enough during the pandemic to exert a major influence on the transmission of virus and disease outcome. One approach is to use a dose-sparing regimen which inevitably involves enhancing the efficacy using adjuvants. METHODS: In this study we compare the use of a novel microcrystalline tyrosine (MCT) adjuvant, which is currently used in a niche area of allergy immunotherapy, for its ability to enhance the efficacy of a seasonal TIV preparation. The efficacy of the MCT adjuvant formulation was compared to alum adjuvanted TIV and to TIV administered without adjuvant using a ferret challenge model to determine vaccine efficacy. RESULTS: The MCT was found to possess high protein-binding capacity. In the two groups where TIV was formulated with adjuvant, the immune response was found to be higher (as determined by HAI titre) than vaccine administered without adjuvant and especially so after challenge with a live influenza virus. Vaccinated animals exhibited lower viral loads (as determined using RT-PCR) than control animals where no vaccine was administered. CONCLUSIONS: The attributes of each adjuvant in stimulating single-dose protection against a poorly immunogenic vaccine was demonstrated. The properties of MCT that lead to the reported effectiveness warrants further exploration in this and other vaccine targets - particularly where appropriate immunogenic, biodegradable and stable alternative adjuvants are sought.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Influenza, Human/prevention & control , Orthomyxoviridae Infections/prevention & control , Tyrosine/administration & dosage , Vaccination/methods , Animals , Crystallization , Dogs , Drug Compounding , Drug Synergism , Ferrets , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Madin Darby Canine Kidney Cells , Microspheres , Seasons , Tyrosine/chemistry
12.
J Appl Toxicol ; 37(2): 222-230, 2017 02.
Article in English | MEDLINE | ID: mdl-27241723

ABSTRACT

A recombinant vaccine composed of a fusion protein formulated with aluminum hydroxide adjuvant is under development for protection against diseases caused by Streptococcus pyogenes. The safety and local reactogenicity of the vaccine was assessed by a comprehensive series of clinical, pathologic and immunologic tests in preclinical experiments. Outbred mice received three intramuscular injections of 1/5th of the human dose (0.1 ml) and rabbits received two injections of the full human dose. Control groups received adjuvant or protein antigen. The vaccine did not cause clinical evidence of systemic toxicity in mice or rabbits. There was a transient increase of peripheral blood neutrophils after the third vaccination of mice. In addition, the concentration of acute phase proteins serum amyloid A and haptoglobin was significantly increased 1 day after injection of the vaccine in mice. There was mild transient swelling and erythema of the injection site in both mice and rabbits. Treatment-related pathology was limited to inflammation at the injection site and accumulation of adjuvant-containing macrophages in the draining lymph nodes. In conclusion, the absence of clinical toxicity in two animal species suggest that the vaccine is safe for use in a phase I human clinical trial. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Adjuvants, Immunologic/adverse effects , Aluminum Hydroxide/adverse effects , Bacterial Proteins/immunology , Exotoxins/immunology , Streptococcal Vaccines/adverse effects , Streptococcus pyogenes/immunology , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Aluminum Hydroxide/immunology , Animals , Antibodies, Bacterial/blood , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Injection Site Reaction , Male , Mice, Inbred Strains , Rabbits , Recombinant Fusion Proteins , Streptococcal Vaccines/administration & dosage , Streptococcal Vaccines/immunology
13.
Hum Vaccin Immunother ; 12(9): 2341-50, 2016 09.
Article in English | MEDLINE | ID: mdl-27104338

ABSTRACT

Chlamydia trachomatis is one of the most common sexually transmitted pathogens and the development of an effective vaccine is highly desirable. The Major Outer Membrane Protein (MOMP) is one of the most abundant and immunogenic chlamydial proteins. Here we investigated the effects of phosphate substitution on the physicochemical and immunochemical properties of an experimental vaccine composed of serovar E recombinant MOMP (rMOMP) and a proprietary adjuvant system SPA08, consisting of aluminum oxyhydroxide (AlOOH) containing the TLR4 agonist E6020. An increase in phosphate substitution in the AlOOH component of the adjuvant markedly decreased the adsorptive coefficient and adsorptive capacity for both Ser E rMOMP and E6020. In vaccine formulations used for immunizations, phosphate substitution induced a decrease in the % adsorption of Ser E rMOMP without affecting the % adsorption of E6020. Immunogenicity studies in CD1 mice showed that an increase in phosphate substitution of the SPA08 adjuvant resulted in an increase in Ser E rMOMP-specific serum total IgG and IgG1 but not IgG2a titers. The degree of phosphate substitution in SPA08 also significantly increased in vitro neutralization concomitant with a decrease in proinflammatory cytokines secreted by Ser E rMOMP-restimulated splenocytes. Taken together, the results of these studies suggest that the degree of phosphate substitution in AlOOH greatly affects the adsorption of E6020 and Ser E rMOMP to AlOOH resulting in significant effects on vaccine-induced cellular and humoral responses.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Aluminum Oxide/administration & dosage , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Chlamydia trachomatis/immunology , Phosphates/administration & dosage , Toll-Like Receptor 4/administration & dosage , Animals , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Female , Immunoglobulin G/blood , Mice , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
14.
Vaccine ; 33(50): 7126-34, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26458804

ABSTRACT

This review focuses on a dengue virus (DENV) vaccine candidate based on a recombinant subunit approach which targets the DENV envelope glycoprotein (E). Truncated versions of E consisting of the N-terminal portion of E (DEN-80E) have been expressed recombinantly in the Drosophila S2 expression system and shown to have native-like conformation. Preclinical studies demonstrate that formulations containing tetravalent DEN-80E adjuvanted with ISCOMATRIX™ adjuvant induce high titer virus neutralizing antibodies and IFN-γ producing T cells in flavivirus-naïve non-human primates. The preclinical data further suggest that administration of such formulations on a 0, 1, 6 month schedule may result in higher maximum virus neutralizing antibody titers and better durability of those titers compared to administration on a 0, 1, 2 month schedule. In addition, the virus neutralizing antibody titers induced by adjuvanted tetravalent DEN-80E compare favorably to the titers induced by a tetravalent live virus comparator. Furthermore, DEN-80E was demonstrated to be able to boost virus neutralizing antibody titers in macaques that have had a prior DENV exposure. A monovalent version of the vaccine candidate, DEN1-80E, was formulated with Alhydrogel™ and studied in a proof-of-principle Phase I clinical trial by Hawaii Biotech, Inc. (NCT00936429). The clinical trial results demonstrate that both the 10 µg and 50 µg formulations of DEN1-80E with 1.25 mg of elemental aluminum were immunogenic when administered in a 3-injection series (0, 1, 2 months) to healthy, flavivirus-naïve adults. The vaccine formulations induced DENV-1 neutralizing antibodies in the majority of subjects, although the titers in most subjects were modest and waned over time. Both the 10 µg DEN1-80E and the 50 µg DEN1-80E formulations with Alhydrogel™ were generally well tolerated.


Subject(s)
Clinical Trials, Phase I as Topic , Dengue Vaccines/administration & dosage , Dengue Vaccines/immunology , Dengue/prevention & control , Drug Evaluation, Preclinical , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cholesterol/administration & dosage , Dengue/epidemiology , Dengue Vaccines/genetics , Dengue Vaccines/isolation & purification , Drug Combinations , Humans , Immunization Schedule , Interferon-gamma/metabolism , Macaca , Phospholipids/administration & dosage , Saponins/administration & dosage , T-Lymphocytes/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Subunit/isolation & purification , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification
15.
J Pharm Sci ; 104(11): 3681-3690, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26190624

ABSTRACT

Subvisible particles in a therapeutic protein product may act as adjuvants to promote unwanted immune responses against the protein. Silicone oil is used as a lubricant in prefilled syringes, and microdroplets of silicone oil are often detected in protein formulations expelled from prefilled syringes. In order to test the adjuvant potency of silicone oil microdroplets, antibody responses in mice to subcutaneous injections of formulations of ovalbumin (OVA) that contained silicone oil microdroplets were measured. These responses were compared against responses to oil-free OVA formulations and to OVA formulations that contained microparticulate aluminum hydroxide ("alum"), the common vaccine adjuvant. When administered with high concentrations of silicone oil microdroplets, OVA formulations elicited strong anti-OVA IgG1 and IgG2a antibody responses. These responses were equivalent to those observed when alum microparticles were added to OVA formulations, suggesting that silicone oil can act as a potent adjuvant. However, when OVA formulations were prepared with lower levels of silicone oil that had been obtained directly from commercial siliconized syringes, the anti-OVA antibody response was not enhanced significantly compared with responses against OVA alone.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibody Formation/drug effects , Ovalbumin/immunology , Silicone Oils/pharmacology , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Aluminum Hydroxide/pharmacology , Animals , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/administration & dosage , Silicone Oils/administration & dosage , Syringes
16.
J Appl Toxicol ; 35(12): 1577-85, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25752809

ABSTRACT

The human papillomavirus (HPV)-16/18 vaccine (Cervarix®) is a prophylactic vaccine for the prevention of cervical cancer. The vaccine contains recombinant virus-like particles assembled from the L1 major capsid proteins of the cervical cancer-causing viral types HPV-16 and HPV-18, and Adjuvant System 04 (AS04), which contains the immunostimulant MPL and aluminium salt. To evaluate potential local and systemic toxic effects of the HPV-16/18 vaccine or AS04 alone, three repeated-dose studies were performed in rabbits and rats. One rabbit study also included a single-dose evaluation. In rabbits (~2.5 kg), the full human dose (HD) of the vaccine was evaluated (0.5 ml per injection site), and in rats (~250 g), 1/5 HD of vaccine was evaluated, corresponding to ≥ 12 times the dosage in humans relative to body weight. In both animal models, the treatment-related changes included a slight transient increase in the number of circulating neutrophils as well as a local inflammatory reaction at the injection site. These treatment-related changes were less pronounced after four doses of AS04 alone than after four doses of the HPV-16/18 vaccine. Additional treatment-related changes in the rat included lower albumin/globulin ratios and microscopic signs of inflammation in the popliteal lymph nodes. In both animal models, 13 weeks after the fourth dose, recovery was nearly complete, although at the injection site in some animals there were signs of discoloration, muscle-fibre regeneration and focal points of macrophage infiltration. Therefore, in these non-clinical models, the single and repeated dose administrations of the HPV-16/18 vaccine or AS04 alone were safe and well tolerated.


Subject(s)
Aluminum Hydroxide/toxicity , Human papillomavirus 16/immunology , Human papillomavirus 18/immunology , Lipid A/analogs & derivatives , Papillomavirus Vaccines/toxicity , Aluminum Hydroxide/administration & dosage , Aluminum Hydroxide/immunology , Animals , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Injections, Intramuscular , Lipid A/administration & dosage , Lipid A/immunology , Lipid A/toxicity , Papillomavirus Infections/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/immunology , Rabbits , Rats , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/prevention & control
17.
Vaccine ; 32(29): 3664-9, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24793947

ABSTRACT

Nanotechnology promises a revolution in medicine including through new vaccine approaches. The use of nanoparticles in vaccination has, to date, focused on attaching antigen directly to or within nanoparticle structures to enhance antigen uptake by immune cells. Here we question whether antigen incorporation with the nanoparticle is actually necessary to boost vaccine effectiveness. We show that the immunogenicity of a sub-unit protein antigen was significantly boosted by formulation with silica nanoparticles even without specific conjugation of antigen to the nanoparticle. We further show that this effect was observed only for virus-sized nanoparticles (50 nm) but not for larger (1,000 nm) particles, demonstrating a pronounced effect of nanoparticle size. This non-attachment approach has potential to radically simplify the development and application of nanoparticle-based formulations, leading to safer and simpler nanoparticle applications in vaccine development.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens/immunology , Influenza Vaccines/immunology , Nanoparticles/administration & dosage , Aluminum Hydroxide/administration & dosage , Animals , Antibodies, Viral/blood , Escherichia coli/metabolism , Genetic Vectors , Immunoglobulin G/blood , Mice, Inbred BALB C , Particle Size , Silicon Dioxide/administration & dosage , Viral Matrix Proteins/immunology
18.
J Immunotoxicol ; 11(1): 44-9, 2014.
Article in English | MEDLINE | ID: mdl-23506358

ABSTRACT

The adjuvanticity of aluminum hydroxide and calcium phosphate on the antibody response in mice towards the venom of the snake Bothrops asper was studied. It was found that, in vitro, most of the venom proteins are similarly adsorbed by both mineral salts, with the exception of some basic phospholipases A2, which are better adsorbed by calcium phosphate. After injection, the adjuvants promoted a slow release of the venom, as judged by the lack of acute toxicity when lethal doses of venom were administered to mice. Leukocyte recruitment induced by the venom was enhanced when it was adsorbed on both mineral salts; however, venom adsorbed on calcium phosphate induced a higher antibody response towards all tested HPLC fractions of the venom. On the other hand, co-precipitation of venom with calcium phosphate was the best strategy for increasing: (1) the capacity of the salt to couple venom proteins in vitro; (2) the venom ability to induce leukocyte recruitment; (3) phagocytosis by macrophages; and (4) a host antibody response. These findings suggest that the chemical nature is not the only one determining factor of the adjuvant activity of mineral salts.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Bothrops/immunology , Calcium Phosphates/administration & dosage , Crotalid Venoms/metabolism , Leukocytes/immunology , Macrophages/immunology , Aluminum Hydroxide/chemistry , Animals , Antibody Formation/drug effects , Antivenins/biosynthesis , Calcium Phosphates/chemistry , Cell Movement , Cells, Cultured , Crotalid Venoms/immunology , Male , Mice , Mice, Inbred Strains , Phagocytosis , Protein Binding
19.
Clin Vaccine Immunol ; 20(11): 1659-68, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23986317

ABSTRACT

Aluminum salts are the most widely used vaccine adjuvants, and phosphate is known to modulate antigen-adjuvant interactions. Here we report an unexpected role for phosphate buffer in an anthrax vaccine (SparVax) containing recombinant protective antigen (rPA) and aluminum oxyhydroxide (AlOH) adjuvant (Alhydrogel). Phosphate ions bind to AlOH to produce an aluminum phosphate surface with a reduced rPA adsorption coefficient and binding capacity. However, these effects continued to increase as the free phosphate concentration increased, and the binding of rPA changed from endothermic to exothermic. Crucially, phosphate restored the thermostability of bound rPA so that it resembled the soluble form, even though it remained tightly bound to the surface. Batches of vaccine with either 0.25 mM (subsaturated) or 4 mM (saturated) phosphate were tested in a disease model at batch release, which showed that the latter was significantly more potent. Both formulations retained their potency for 3 years. The strongest aluminum adjuvant effects are thus likely to be via weakly attached or easily released native-state antigen proteins.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Anthrax Vaccines/administration & dosage , Anthrax Vaccines/immunology , Anthrax/prevention & control , Adjuvants, Immunologic/metabolism , Aluminum Hydroxide/metabolism , Animals , Anthrax Vaccines/chemistry , Antigens, Bacterial/metabolism , Buffers , Chemistry, Pharmaceutical , Disease Models, Animal , Drug Stability , Female , Mice , Phosphates/metabolism , Protein Binding
20.
Expert Rev Clin Immunol ; 9(4): 361-73, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23557271

ABSTRACT

An adjuvant is a substance that enhances the antigen-specific immune response, induces the release of inflammatory cytokines, and interacts with Toll-like receptors and the NALP3 inflammasome. The immunological consequence of these actions is to stimulate the innate and adaptive immune response. The activation of the immune system by adjuvants, a desirable effect, could trigger manifestations of autoimmunity or autoimmune disease. Recently, a new syndrome was introduced, autoimmune/inflammatory syndrome induced by adjuvants (ASIA), that includes postvaccination phenomena, macrophagic myofasciitis, Gulf War syndrome and siliconosis. This syndrome is characterized by nonspecific and specific manifestations of autoimmune disease. The main substances associated with ASIA are squalene (Gulf War syndrome), aluminum hydroxide (postvaccination phenomena, macrophagic myofasciitis) and silicone with siliconosis. Mineral oil, guaiacol and iodine gadital are also associated with ASIA. The following review describes the wide clinical spectrum and pathogenesis of ASIA including defined autoimmune diseases and nonspecific autoimmune manifestations, as well as the outlook of future research in this field.


Subject(s)
Adjuvants, Immunologic/adverse effects , Autoimmune Diseases/immunology , Fasciitis/immunology , Inflammation/immunology , Myositis/immunology , Persian Gulf Syndrome/immunology , Silicosis/immunology , Adaptive Immunity , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Aluminum Hydroxide/adverse effects , Animals , Autoimmune Diseases/chemically induced , Fasciitis/chemically induced , Humans , Immunity, Innate , Inflammasomes/metabolism , Inflammation/chemically induced , Myositis/chemically induced , Persian Gulf Syndrome/chemically induced , Silicones/administration & dosage , Silicones/adverse effects , Squalene/administration & dosage , Squalene/adverse effects , Syndrome , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL