Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
BMC Vet Res ; 16(1): 259, 2020 Jul 28.
Article in English | MEDLINE | ID: mdl-32723323

ABSTRACT

BACKGROUND: Bacillus anthracis is the causative agent of anthrax, a disease of both humans and various animal species, and can be used as a bioterror agent. Effective vaccines are available, but those could benefit from improvements, including increasing the immunity duration, reducing the shot frequency and adverse reactions. In addition, more sophisticated antigen delivery and potentiation systems are urgently required. The protective antigen (PA), one of three major virulence factors associated with anthrax was displayed on the surface of Bacillus subtilis spores, which is a vaccine production host and delivery vector with several advantages such as a low production cost, straightforward administration as it is safe for human consumption and the particulate adjuvanticity. Mice were immunized orally (PO), intranasally (IN), sublingually (SL) or intraperitoneally (IP) with the PA displaying probiotic spore vaccine. Clinical observation, serological analysis and challenge experiment were conducted to investigate the safety and efficacy of the vaccine. RESULTS: A/J mice immunized with the PA spore vaccine via PO, IN, SL, and IP were observed to have increased levels of active antibody titer, isotype profiles and toxin neutralizing antibody in sera, and IgA in saliva. The immunized mice were demonstrated to raise protective immunity against the challenge with lethal B. anthracis spores. CONCLUSIONS: In this study, we developed a B. subtilis spore vaccine that displays the PA on its surface and showed that the PA-displaying spore vaccine was able to confer active immunity to a murine model based on the results of antibody isotype titration, mucosal antibody identification, and a lethal challenge experiment.


Subject(s)
Anthrax Vaccines/pharmacology , Antigens, Bacterial/immunology , Bacillus subtilis/immunology , Bacterial Toxins/immunology , Animals , Anthrax/prevention & control , Anthrax Vaccines/administration & dosage , Antibodies, Neutralizing/blood , Bacillus anthracis , Immunization , Immunoglobulin A , Male , Mice , Saliva/immunology , Spores, Bacterial/immunology , Vaccines, Synthetic
2.
Int J Nanomedicine ; 15: 239-252, 2020.
Article in English | MEDLINE | ID: mdl-32021177

ABSTRACT

INTRODUCTION: Aluminum salts, although they have been used as adjuvants in many vaccine formulations since 1926, exclusively induce a Th2-biased immune response, thereby limiting their use against intracellular pathogens like Mycobacterium tuberculosis. METHODS AND RESULTS: Herein, we synthesized amorphous and crystalline forms of aluminum hydroxide nanoparticles (AH nps) of 150-200 nm size range. Using Bacillus anthracis protective antigen domain 4 (D4) as a model antigen, we demonstrated that both amorphous and crystalline forms of AH nps displayed enhanced antigen D4 uptake by THP1 cells as compared to commercial adjuvant aluminum hydroxide gel (AH gel). In a mouse model, both amorphous and crystalline AH nps triggered an enhanced D4-specific Th2- and Th1-type immune response and conferred superior protection against anthrax spore challenge as compared to AH gel. Physicochemical characterization of crystalline and amorphous AH nps revealed stronger antigen D4 binding and release than AH gel. CONCLUSION: These results demonstrate that size and crystallinity of AH nps play important roles in mediating enhanced antigen presenting cells (APCs) activation and potentiating a strong antigen-specific immune response, and are critical parameters for the rational design of alum-based Th1-type adjuvant to induce a more balanced antigen-specific immune response.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/chemistry , Anthrax/prevention & control , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Metal Nanoparticles/chemistry , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacokinetics , Aluminum Hydroxide/immunology , Aluminum Hydroxide/pharmacology , Animals , Anthrax/immunology , Anthrax Vaccines/chemistry , Anthrax Vaccines/immunology , Anthrax Vaccines/pharmacology , Cell Line , Disease Models, Animal , Dynamic Light Scattering , Female , Humans , Mice , RAW 264.7 Cells , Spectroscopy, Fourier Transform Infrared , Th1 Cells/immunology
3.
Vet Res ; 48(1): 46, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28882176

ABSTRACT

The Sterne live spore vaccine (34F2) is the most widely used veterinary vaccine against anthrax in animals. Antibody responses to several antigens of Bacillus anthracis have been described with a large focus on those against protective antigen (PA). The focus of this study was to evaluate the protective humoral immune response induced by the live spore anthrax vaccine in goats. Boer goats vaccinated twice (week 0 and week 12) with the Sterne live spore vaccine and naive goats were used to monitor the anti-PA and toxin neutralizing antibodies at week 4 and week 17 (after the second vaccine dose) post vaccination. A/J mice were passively immunized with different dilutions of sera from immune and naive goats and then challenged with spores of B. anthracis strain 34F2 to determine the protective capacity of the goat sera. The goat anti-PA ELISA titres indicated significant sero-conversion at week 17 after the second doses of vaccine (p = 0.009). Mice receiving undiluted sera from goats given two doses of vaccine (twice immunized) showed the highest protection (86%) with only 20% of mice receiving 1:1000 diluted sera surviving lethal challenge. The in vitro toxin neutralization assay (TNA) titres correlated to protection of passively immunized A/J mice against lethal infection with the vaccine strain Sterne 34F2 spores using immune goat sera up to a 1:10 dilution (rs ≥ 0.522, p = 0.046). This study suggests that the passive mouse protection model could be potentially used to evaluate the protective immune response in livestock animals vaccinated with the current live vaccine and new vaccines.


Subject(s)
Anthrax Vaccines/immunology , Goats/immunology , Immunity, Humoral , Animals , Anthrax/immunology , Anthrax/prevention & control , Anthrax/veterinary , Anthrax Vaccines/pharmacology , Bacillus anthracis/immunology , Goat Diseases/immunology , Goat Diseases/microbiology , Goat Diseases/prevention & control , Immunity, Humoral/immunology , Male , Mice
4.
Bull Math Biol ; 79(2): 303-324, 2017 02.
Article in English | MEDLINE | ID: mdl-28035484

ABSTRACT

A general mathematical model of anthrax (caused by Bacillus anthracis) transmission is formulated that includes live animals, infected carcasses and spores in the environment. The basic reproduction number [Formula: see text] is calculated, and existence of a unique endemic equilibrium is established for [Formula: see text] above the threshold value 1. Using data from the literature, elasticity indices for [Formula: see text] and type reproduction numbers are computed to quantify anthrax control measures. Including only herbivorous animals, anthrax is eradicated if [Formula: see text]. For these animals, oscillatory solutions arising from Hopf bifurcations are numerically shown to exist for certain parameter values with [Formula: see text] and to have periodicity as observed from anthrax data. Including carnivores and assuming no disease-related death, anthrax again goes extinct below the threshold. Local stability of the endemic equilibrium is established above the threshold; thus, periodic solutions are not possible for these populations. It is shown numerically that oscillations in spore growth may drive oscillations in animal populations; however, the total number of infected animals remains about the same as with constant spore growth.


Subject(s)
Anthrax/veterinary , Models, Biological , Animals , Anthrax/microbiology , Anthrax/transmission , Anthrax Vaccines/pharmacology , Bacillus anthracis/growth & development , Bacillus anthracis/pathogenicity , Basic Reproduction Number , Carnivory , Herbivory , Humans , Livestock , Mathematical Concepts , Population Dynamics , Spores, Bacterial/growth & development , Spores, Bacterial/pathogenicity
5.
Toxins (Basel) ; 7(7): 2371-84, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26120785

ABSTRACT

Protective immunity against anthrax is inferred from measurement of vaccine antigen-specific neutralizing antibody titers in serum samples. In animal models, in vivo challenges with toxin and/or spores can also be performed. However, neither of these approaches considers toxin-induced damage to specific organ systems. It is therefore important to determine to what extent anthrax vaccines and existing or candidate adjuvants can provide organ-specific protection against intoxication. We therefore compared the ability of Alum, CpG DNA and the CD1d ligand α-galactosylceramide (αGC) to enhance protective antigen-specific antibody titers, to protect mice against challenge with lethal toxin, and to block cardiotoxicity and hepatotoxicity. By measurement of serum cardiac Troponin I (cTnI), and hepatic alanine aminotransferase (ALT), and aspartate aminotransferase (AST), it was apparent that neither vaccine modality prevented hepatic intoxication, despite high Ab titers and ultimate survival of the subject. In contrast, cardiotoxicity was greatly diminished by prior immunization. This shows that a vaccine that confers survival following toxin exposure may still have an associated morbidity. We propose that organ-specific intoxication should be monitored routinely during research into new vaccine modalities.


Subject(s)
Anthrax Vaccines/pharmacology , Anthrax/complications , Antigens, Bacterial/immunology , Bacillus anthracis/immunology , Bacterial Toxins/immunology , Cardiotoxicity/prevention & control , Chemical and Drug Induced Liver Injury/prevention & control , Alanine Transaminase/blood , Animals , Anthrax/immunology , Anthrax Vaccines/immunology , Aspartate Aminotransferases/blood , Cardiotoxicity/blood , Cardiotoxicity/immunology , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/immunology , Mice, Inbred C57BL , Survival Analysis , Troponin I/blood
6.
Clin Exp Immunol ; 173(1): 84-91, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23607659

ABSTRACT

Anthrax is a toxin-mediated disease, the lethal effects of which are initiated by the binding of protective antigen (PA) with one of three reported cell surface toxin receptors (ANTXR). Receptor binding has been shown to influence host susceptibility to the toxins. Despite this crucial role for ANTXR in the outcome of disease, and the reported immunomodulatory consequence of the anthrax toxins during infection, little is known about ANTXR expression on human leucocytes. We characterized the expression levels of ANTXR1 (TEM8) on human leucocytes using flow cytometry. In order to assess the effect of prior toxin exposure on ANTXR1 expression levels, leucocytes from individuals with no known exposure, those exposed to toxin through vaccination and convalescent individuals were analysed. Donors could be defined as either 'low' or 'high' expressers based on the percentage of ANTXR1-positive monocytes detected. Previous exposure to toxins appears to modulate ANTXR1 expression, exposure through active infection being associated with lower receptor expression. A significant correlation between low receptor expression and high anthrax toxin-specific interferon (IFN)-γ responses was observed in previously infected individuals. We propose that there is an attenuation of ANTXR1 expression post-infection which may be a protective mechanism that has evolved to prevent reinfection.


Subject(s)
Anthrax/blood , Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Leukocytes, Mononuclear/drug effects , Neoplasm Proteins/biosynthesis , Receptors, Cell Surface/biosynthesis , Skin Diseases, Bacterial/blood , Anthrax/genetics , Anthrax Vaccines/pharmacology , Antigens, Bacterial/metabolism , Cohort Studies , Convalescence , Flow Cytometry , Gene Expression Regulation/drug effects , Humans , Immunization, Secondary , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Leukocytes, Mononuclear/metabolism , Microfilament Proteins , Neoplasm Proteins/genetics , Receptors, Cell Surface/genetics , Skin Diseases, Bacterial/genetics , Turkey , United Kingdom , Vaccination
7.
Int J Biostat ; 8(2)2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22499733

ABSTRACT

A major goal of vaccine development is the identification of immune responses that are responsible for vaccine efficacy. In theory, modest vaccines could be successfully improved by increasing such immune responses. And for a vaccine with a great benefit in one population, inducing such immune response in a different population could help one conclude the vaccine would have great benefit there. Such identification is tricky because the immune response to vaccination can only be measured in the vaccine group and thus immune responses might only be identifying individuals with a constitutional ability to remain uninfected, rather than being causal. Define the vaccine induced immune response as X(1). The value X(1) is a potential outcome; it is measured directly in vaccinees but unobserved in the placebo group. Our goal is to regress outcome on X(1) separately in the vaccine and placebo groups and to see if the vaccine effect varies with X(1). Put another way, our goal is to see if there is a vaccine by X(1) interaction. Regression of outcome on X(1) is easy to do in the vaccine group, but difficult in the placebo group as X(1) is not observed. In this paper we derive bounds on the regression curve in the placebo group. For a continuous endpoint these bounds can be unhelpful, or can help modestly temper our enthusiasm for a role of X(1) on the vaccine effect. For binary outcomes with 100% placebo infection the bound is very tight but unhelpful as 100% infection precludes identification of any covariate with a differential effect on placebo infection. We apply these methods to experiments of anthrax vaccine in rabbits with survival to challenge as the outcome and demonstrate how to extrapolate the model to humans.


Subject(s)
Models, Immunological , Vaccines/immunology , Vaccines/pharmacology , Animals , Anthrax/immunology , Anthrax/prevention & control , Anthrax Vaccines/immunology , Anthrax Vaccines/pharmacology , Biostatistics , Disease Models, Animal , Humans , Infection Control/methods , Infection Control/statistics & numerical data , Models, Statistical , Rabbits , Regression Analysis
8.
Biologicals ; 39(4): 236-41, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21664832

ABSTRACT

Complexities of lethal challenge animal models have prompted the investigation of immunogenicity assays as potency tests of anthrax vaccines. An ELISA was used to measure the antibody response to protective antigen (PA) in mice immunized once with a commercially available (AVA) or a recombinant PA vaccine (rPAV) formulated in-house with aluminum hydroxide. Results from the anti-PA ELISA were used to select a single dose appropriate for the development of a potency test. Immunization with 0.2 mL of AVA induced a measurable response in the majority of animals. This dose was located in the linear range of the vaccine dose-antibody response curve. In the case of rPAV, practical limitations prevented the finding of the best single dose for the potency testing of purified vaccines. In additional immunogenicity experiments neither the magnitude of the response to a single dose of vaccine, nor the estimation of the dose necessary to induce a measurable response were able to consistently detect brief exposure of vaccines to potentially damaging temperatures. However, differences detected for rPAV in the proportion of mice responding to the same dose of treated and untreated vaccine suggested that further assay development to increase the sensitivity of the latter design may be warranted.


Subject(s)
Anthrax Vaccines/pharmacology , Animals , Antibodies, Bacterial/immunology , Antibody Formation , Dose-Response Relationship, Immunologic , Enzyme-Linked Immunosorbent Assay , Feasibility Studies , Female , Mice
9.
Article in Russian | MEDLINE | ID: mdl-21446166

ABSTRACT

AIM: To study the ability of recombinant protective antigen (PA) to stimulate adaptive immune response in laboratory animals. MATERIALS AND METHODS: Vaccine, recombinant, and reference strains of Bacillus anthracis were used in the study. Laboratory animals were immunized subcutaneously with two doses of antigenic preparation or one dose of B. anthracis strain. After inoculation with reference strain of B. anthracis, measurement of LD50 as well as indexes of immunity was performed by specified methods. RESULTS: It was revealed that asporogenic recombinant strain has stable biological characteristics during passages in vitro and is effective producer of PA. Using 2-stage chromatography, highly purified protein was obtained. Experiments on different biomodels--BALB/c mice, guinea pigs, and rabbits--demonstrated high protective activity of PA obtained from asporogenic producer. Increase of immunity index was noted when EA1 protein from S-layer was added to preparation for immunization. CONCLUSION: Immunity indexes determined in experiments on laboratory animals point to high protective efficacy of recombinant PA. Further studies of its interaction with macroorganism's innate and adaptive immunity systems are promising.


Subject(s)
Anthrax Vaccines/pharmacology , Anthrax/immunology , Anthrax/prevention & control , Antigens, Bacterial/pharmacology , Bacillus anthracis/immunology , Bacterial Toxins/pharmacology , Membrane Glycoproteins/pharmacology , Adaptive Immunity/drug effects , Animals , Anthrax/genetics , Anthrax Vaccines/genetics , Anthrax Vaccines/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacillus anthracis/genetics , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Guinea Pigs , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Rabbits
10.
Vaccine ; 27 Suppl 4: D28-32, 2009 Nov 05.
Article in English | MEDLINE | ID: mdl-19837282

ABSTRACT

Anthrax, a disease of herbivores, only rarely infects humans. However, the threat of using Bacillus anthracis, the causative agent, to intentionally produce disease has been the impetus for development of next-generation vaccines. Two licensed vaccines have been available for human use for several decades. These are composed of acellular culture supernatants containing the protective antigen (PA) component of the anthrax toxins. In this review we summarize the various approaches used to develop improved vaccines. These efforts have included the use of PA with newer adjuvants and delivery systems, including bacterial and viral vectors and DNA vaccines. Attempts to broaden the protection afforded by PA-based vaccines have focused on adding other B. anthracis components, including spore and capsule antigens.


Subject(s)
Anthrax Vaccines/pharmacology , Anthrax/prevention & control , Adjuvants, Immunologic/pharmacology , Anthrax Vaccines/immunology , Antigens, Bacterial , Bacillus anthracis/immunology , Bacterial Toxins , Biological Warfare/prevention & control , Biological Warfare Agents , Humans , Vaccines, Acellular/immunology , Vaccines, Acellular/pharmacology , Vaccines, DNA/immunology , Vaccines, DNA/pharmacology
12.
J Cell Mol Med ; 13(8B): 1907-1919, 2009 Aug.
Article in English | MEDLINE | ID: mdl-18752638

ABSTRACT

Anthrax receptor (ATR) shares similarities with molecules relevant to haematopoiesis. This suggests that anthrax proteins might bind to these mimicking molecules and exert non-specific haematopoietic effects. The haematopoietic system is the site of immune cell development in the adult. As such, ATR ligand, protective antigen (PA) and the other anthrax proteins, lethal factor, edema factor, could be significant to haematopoietic responses against Bacillus anthracis infection. Because haematopoiesis is the process of immune cell development, effects by anthrax proteins could be relevant to vaccine development. Here, we report on effects of anthrax proteins and toxins on early and late haematopoiesis. Flow cytometry shows binding of PA to haematopoietic cells. This binding might be partly specific because flow cytometry and Western blots demonstrate the presence of ATR1 on haematopoietic cell subsets and the supporting stromal cells. Functional studies with long-term initiating cell and clonogenic assays determined haematopoietic suppression by anthrax toxins and stimulation by monomeric proteins. The suppressive effects were not attributed to cell death, but partly through the induction of haematopoietic suppressors, interleukin (IL)-10 and CCL3 (MIP-1alpha). In summary, anthrax proteins affect immune cell development by effects on haematopoiesis. The type of effect, stimulation or suppression, depend on whether the stimulator is a toxin or monomeric protein. The studies show effects of anthrax proteins beginning at the early stage of haematopoiesis, and also show secondary mediators such as IL-10 and CCL3. The roles of other cytokines and additional ATR are yet to be investigated.


Subject(s)
Anthrax Vaccines/pharmacology , Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Hematopoiesis/drug effects , Adult , Blotting, Western , Cells, Cultured , Flow Cytometry , Humans
13.
J Immunol ; 181(8): 5785-90, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18832738

ABSTRACT

Adding synthetic oligodeoxynucleotides containing unmethylated CpG motifs to Anthrax vaccine adsorbed (AVA, the licensed human vaccine) increases the speed and magnitude of the resultant Ab response. Ab titers persist in the protective range for >1 year, significantly longer than in animals vaccinated with AVA alone. Unexpectedly, a majority of mice immunized with CpG-adjuvanted AVA maintained resistance to anthrax infection even after their Ab titers had declined into the subprotective range. The survival of these animals was mediated by the de novo production of protective Abs by high affinity memory B cells re-stimulated immediately after challenge. Thus, a previously unrecognized benefit of CpG oligodeoxynucleotides adjuvants is their ability to expand the long-lived memory B cell population. Current findings demonstrate that CpG-adjuvanted AVA mediates protection both by stimulating a strong/persistent serum Ab response and by generating a high-affinity long-lived pool of memory B cells.


Subject(s)
Adjuvants, Immunologic/pharmacology , Anthrax Vaccines/pharmacology , Anthrax/immunology , Antibody Formation/drug effects , B-Lymphocytes/immunology , Immunologic Memory/immunology , Oligodeoxyribonucleotides/pharmacology , Animals , Anthrax/prevention & control , Antibody Formation/immunology , B-Lymphocytes/pathology , Female , Humans , Immunologic Memory/drug effects , Mice , Time Factors
14.
Clin Vaccine Immunol ; 15(4): 644-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18321882

ABSTRACT

Currently, there is no routine monitoring of an immune response to the anthrax vaccine. Simple on-site tests are needed to evaluate the antibody response of anthrax-vaccinated individuals in the Armed Forces and others at high risk. Using a prototype lateral flow assay (LFA) (R. E. Biagini, D. L. Sammons, J. P. Smith, B. A. MacKenzie, C. A. F. Striley, J. E. Snawder, S. A. Robertson, and C. P. Quinn, Clin. Vaccine Immunol. 13:541-546, 2006), we investigated the agreement between a validated anthrax protective antigen (PA) immunoglobulin G (IgG) enzyme-linked immunosorbent assay (ELISA) and the LFA for 335 unvaccinated and vaccinated subjects. We also investigated the performance of the LFA under the following conditions: thermal shock (i.e., thermal cycling between temperature extremes), high temperature/high relative humidity, high temperature/low relative humidity, and low temperature/low relative humidity. With the anti-PA ELISA used as a standard, the LFA was shown to be optimally diagnostic at 11 microg/ml anti-PA-specific IgG. At this concentration, the LFA specificity and sensitivity were 98% (95% confidence interval [CI], 97% to 100%) and 92% (CI, 88% to 97%), respectively. Receiver operating characteristic curve analysis yielded an area under the curve value of 0.988 (CI, 0.976 to 1.00), suggesting that the LFA is an extremely accurate diagnostic test. For < or = 4 or > or = 50 microg/ml PA-specific IgG, the LFA results for each environmental condition were identical to those obtained in the laboratory. These data indicate that this rapid point-of-care test would be a feasible tool in monitoring the serological antibody responses of individuals that have been vaccinated against anthrax.


Subject(s)
Anthrax Vaccines/immunology , Immunoglobulin G/blood , Point-of-Care Systems , Anthrax/prevention & control , Anthrax Vaccines/blood , Anthrax Vaccines/pharmacology , Antibody Specificity , Antigens, Bacterial/immunology , Enzyme-Linked Immunosorbent Assay , Epitopes , Humans , Immunoassay/methods , Immunoglobulin G/immunology , Sensitivity and Specificity
15.
Curr Pharm Biotechnol ; 9(1): 24-33, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18289054

ABSTRACT

Anthrax is caused by infection with Bacillus anthracis, a spore forming, rod-shaped, encapsulated gram positive bacteria. The disease manifests itself in distinct ways depending on the route of entry of infective bacterial spores: cutaneous, inhalational, and gastrointestinal. Though rare in humans, inhalational anthrax has become a major concern due to the capacity for spores to be weaponized. The limited success of antibiotic therapy has motivated investigation of complementary therapeutic strategies that target the bacteria's secreted toxin. The zinc-dependent metalloproteinase lethal factor (LF) is a critical component of anthrax toxin and an important potential target for small molecule drugs. In the past few years, a number of approaches have been taken to identify LF inhibitors, from generating conventional metal chelating substrate analogs to random screening of diverse compound libraries. These efforts have produced several different classes of specific nanomolar range inhibitors. Some compounds have fared well in animal models for anthrax toxemia and infection, and these inhibitors and their derivatives may form the basis for future therapies to treat the disease in humans.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Drug Design , Metalloendopeptidases/antagonists & inhibitors , Animals , Anthrax Vaccines/chemical synthesis , Anthrax Vaccines/pharmacology , Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Humans , Metalloendopeptidases/metabolism , Protease Inhibitors/pharmacology
17.
Vaccine ; 25(15): 2771-7, 2007 Apr 12.
Article in English | MEDLINE | ID: mdl-17240008

ABSTRACT

The serological response and efficacy of Bacillus anthracis recombinant protective antigen (rPA) vaccines formulated with aluminum hydroxide adjuvant, either with or without formaldehyde, were evaluated in rabbits. Rabbits that had been injected with a single dose of 25 microg of rPA adsorbed to 500 microg of aluminum in aluminum hydroxide gel (Alhydrogel) had a significantly higher quantitative anti-rPA IgG ELISA titers (p<0.0001) and toxin neutralizing antibody (TNA) assay titers (p<0.0001) than rabbits tested at the next lowest concentration of aluminum (158 microg). Rabbits injected with two doses of 50 microg of rPA formulated with 500 microg of aluminum also had significantly higher serological responses, as measured by a quantitative anti-rPA IgG ELISA (p<0.0001) and TNA assay (p<0.0001), than sera from rabbits injected with a rPA vaccine formulated without adjuvant. Short-term protection against an aerosol spore challenge (448 LD(50)), however, was not significantly different between the two groups (12/12 and 11/12, respectively). Rabbits injected with a single dose of 50 microg of rPA formulated with 500 microg of aluminum and 0.2% formaldehyde had significantly higher ELISA (p<0.0001) and TNA assay (p<0.0001) titers than rabbits that had been injected with a rPA vaccine formulated with adjuvant but without formaldehyde. Short-term protection against a 125 LD(50) parenteral spore challenge, however, was not significantly different between the two groups (14/24 and 9/24, respectively; p=0.2476). Under the conditions tested in the rabbit animal model, significantly higher serological responses were observed in rabbits that had been injected with rPA formulated with aluminum hydroxide gel adjuvant and formaldehyde. However, differences in short-term efficacy were not observed.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/pharmacology , Anthrax Vaccines/chemistry , Anthrax Vaccines/immunology , Formaldehyde/pharmacology , Adjuvants, Immunologic/chemistry , Aluminum Hydroxide/chemistry , Aluminum Hydroxide/immunology , Animals , Anthrax/immunology , Anthrax/prevention & control , Anthrax Vaccines/pharmacology , Anthrax Vaccines/therapeutic use , Enzyme-Linked Immunosorbent Assay , Female , Formaldehyde/chemistry , Formaldehyde/immunology , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Rabbits , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
18.
Clin Vaccine Immunol ; 13(2): 208-13, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16467327

ABSTRACT

Anthrax toxin consists of protective antigen (PA) and two toxic components, lethal factor (LF) and edema factor (EF). PA binds to mammalian cellular receptors and delivers the toxic components to the cytoplasm. PA is the primary antigenic component of the current anthrax vaccine. Immunity is due to the generation of antibodies that prevent the PA-mediated internalization of LF and EF. In this study, we characterized sera obtained from vaccinated military personnel. Anthrax vaccine is administered in a series of six injections at 0, 2, and 4 weeks and 6, 12, and 18 months, followed by annual boosters. The vaccination histories of the subjects were highly varied; many subjects had not completed the entire series, and several had not received annual boosters. We developed a simple colorimetric assay using alamarBlue dye to assess the antibody-mediated neutralization of LF-mediated toxicity to the J774A.1 murine macrophage cell line. Recently vaccinated individuals had high antibody levels and neutralizing activity. One individual who had not been boosted for 5 years had low immunoglobulin G antibody levels but a detectable neutralization activity, suggesting that this individual produced low levels of very active antibodies.


Subject(s)
Anthrax Vaccines/pharmacology , Antibodies, Bacterial/blood , Bacillus anthracis/immunology , Animals , Anthrax/immunology , Anthrax/prevention & control , Anthrax Vaccines/administration & dosage , Anthrax Vaccines/immunology , Antigens, Bacterial/immunology , Bacillus anthracis/pathogenicity , Bacterial Toxins/immunology , Cell Line , Colorimetry/methods , Humans , Immunization, Secondary , Macrophages/immunology , Mice , Military Personnel , Neutralization Tests/methods , Time Factors , United States
19.
Biologicals ; 32(2): 62-9, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15454183

ABSTRACT

A recombinant protective antigen (rPA)-based enzyme-linked immunosorbent assay (ELISA) was developed to measure the serological response of female A/J mice after inoculation with the new rPA-based anthrax vaccine. Several fundamental parameters of the ELISA were evaluated: specificity, precision, accuracy, linearity, and stability. Experimental results suggested that the quantitative anti-rPA IgG ELISA could be used to measure antibody levels in female A/J mice and may be useful as a potency assay to monitor consistency of manufacture of a rPA-based vaccine for planned clinical trials.


Subject(s)
Anthrax Vaccines/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Animals , Antibodies, Monoclonal/chemistry , Antibody Formation , Binding, Competitive , Clinical Trials as Topic , Dose-Response Relationship, Drug , Evaluation Studies as Topic , Female , Immunoglobulin G/chemistry , Linear Models , Logistic Models , Mice , Protein Binding , Sensitivity and Specificity
20.
Mol Gen Mikrobiol Virusol ; (3): 21-6, 2004.
Article in Russian | MEDLINE | ID: mdl-15354937

ABSTRACT

Anthrax belongs to highly dangerous infections of man and animals. No effective treatment methods for pulmonary types of the disease have been yet developed. The existing anthrax vaccines were designed decades ago and need improvement to fit the large-scale vaccination of population. At the same time, the immunological properties of the anthrax vaccine main component, i.e. of the protective agent, have been poorly studied. We obtained, within the present case study, a panel of mouse monoclonal antibodies to the protective agent and investigated the properties of the highest-affine panel representatives. An unusual phenomenon was detected, which is related with enhancement of the anthrax toxin action on the mouse macrophage-like cell-line in presence of the 1F2 monoclonal antibody. The remaining analyzed antibodies, i.e. 6G8 and 6G7, were found to neutralize effectively the toxin action. The enhancing and neutralizing antibodies were proven to be specific to different domains of the protective antigen and to recognize epitopes in its composition. The antibody-mediated enhancement of the anthrax lethal action is a convincing argument for further development of a new-generation anthrax vaccine. Definition of the linear antigen determinants for neutralizing antibodies in the protective antigens is an important step in the development of the next-generation anthrax vaccine.


Subject(s)
Anthrax Vaccines/immunology , Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Bacillus anthracis , Bacterial Toxins/immunology , Animals , Anthrax Vaccines/pharmacology , Antibodies, Bacterial/immunology , Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Cell Line , Epitopes/immunology , Macrophages/virology , Mice , Mice, Inbred BALB C , Neutralization Tests
SELECTION OF CITATIONS
SEARCH DETAIL