Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Pathogens ; 9(11)2020 Oct 24.
Article in English | MEDLINE | ID: mdl-33114429

ABSTRACT

Bacillus anthracis spores that are re-aerosolized from surface deposits after initial contamination present significant health risks for personnel involved in decontamination. To model repeated exposure to low dose B. anthracis spores, three groups of seven rabbits were challenged with multiple low-doses of B. anthracis spores 5 days a week for 3 weeks. Mortality, body temperature, heart and respiration rates, hematology, C-reactive protein, bacteremia, and serum protective antigen were monitored for 21 days post-exposure after the last of multiple doses. All rabbits exposed to a mean daily dose of 2.91 × 102 colony forming units (CFU) survived and showed minimal physiological changes attributable to exposure. One of seven rabbits receiving a mean daily dose of 1.22 × 103 CFU died and four of seven receiving a mean daily dose of 1.17 × 104 CFU died. The LD50 was calculated to be 8.1 × 103 CFU of accumulated dose. Rabbits that succumbed to the higher dose exhibited bacteremia and increases above baseline in heart rate, respiration rate, and body temperature. Two rabbits in the mean daily dose group of 1.17 × 104 CFU exhibited clinical signs of inhalation anthrax yet survived. This study provides a description of lethality, pathophysiology, and pathology in a controlled multiple low-dose inhalation exposure study of B. anthracis in the rabbit model. The data suggest that the accumulated dose is important in survival outcome and that a subset of rabbits may show clinical signs of disease but fully recover without therapeutic intervention.

2.
Pathogens ; 9(6)2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32545184

ABSTRACT

Credible dose-response relationships are needed to more accurately assess the risk posed by exposure to low-level Bacillus anthracis contamination during or following a release. To begin to fill this knowledge gap, New Zealand White rabbits were implanted with D70-PCT telemetry transmitters and subsequently aerosol challenged with average inhaled doses of 2.86 x 102 to 2.75 x 105 colony forming units (CFU) of B. anthracis spores. Rabbits exposed to a single inhaled dose at or above 2.54 × 104 CFU succumbed with dose-dependent time to death. Death was associated with increases above baseline in heart rate, respiration rate, and body temperature and all rabbits that died exhibited bacteremia at some point prior to death. Rabbits that inhaled doses of 2.06 × 103 CFU or lower survived to the end of the study and showed no or minimal adverse changes in the measured physiological responses in response to the challenge. Moreover, no bacteremia nor toxemia were observed in rabbits that survived to the end of the study. Overall, the data indicate that challenge doses of B. anthracis below the level sufficient to establish systemic infection do not produce observable physiological responses; however, doses that triggered a response resulted in death.

3.
Article in English | MEDLINE | ID: mdl-29133571

ABSTRACT

The recommended management of inhalational anthrax, a high-priority bioterrorist threat, includes antibiotics and antitoxins. Obiltoxaximab, a chimeric monoclonal antibody against anthrax protective antigen (PA), is licensed under the U.S. Food and Drug Administration's (FDA's) Animal Rule for the treatment of inhalational anthrax. Because of spore latency, disease reemergence after treatment cessation is a concern, and there is a need to understand the development of endogenous protective immune responses following antitoxin-containing anthrax treatment regimens. Here, acquired protective immunity was examined in New Zealand White (NZW) rabbits challenged with a targeted lethal dose of Bacillus anthracis spores and treated with antibiotics, obiltoxaximab, or a combination of both. Survivors of the primary challenge were rechallenged 9 months later and monitored for survival. Survival rates after primary and rechallenge for controls and animals treated with obiltoxaximab, levofloxacin, or a combination of both were 0, 65, 100, and 95%, and 0, 100, 95, and 89%, respectively. All surviving immune animals had circulating antibodies to PA and serum toxin-neutralizing titers prior to rechallenge. Following rechallenge, systemic bacteremia and toxemia were not detected in most animals, and the levels of circulating anti-PA IgG titers increased starting at 5 days postrechallenge. We conclude that treatment with obiltoxaximab, alone or combined with antibiotics, significantly improves the survival of rabbits that received a lethal inhalation B. anthracis spore challenge dose and does not interfere with the development of immunity. Survivors of primary challenge are protected against reexposure, have rare incidents of systemic bacteremia and toxemia, and have evidence of an anamnestic response.


Subject(s)
Anthrax , Anti-Bacterial Agents , Antibodies, Monoclonal , Antitoxins , Bacillus anthracis , Levofloxacin , Respiratory Tract Infections , Spores, Bacterial , Animals , Female , Male , Rabbits , Anthrax/immunology , Anthrax/microbiology , Anthrax/mortality , Anthrax/prevention & control , Anti-Bacterial Agents/pharmacology , Antibodies, Bacterial/biosynthesis , Antibodies, Monoclonal/pharmacology , Antigens, Bacterial/blood , Antigens, Bacterial/immunology , Antitoxins/pharmacology , Bacillus anthracis/drug effects , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/blood , Bacterial Toxins/immunology , Drug Therapy, Combination , Immunization, Passive/methods , Immunoglobulin G/biosynthesis , Immunologic Memory/drug effects , Levofloxacin/pharmacology , Random Allocation , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/mortality , Respiratory Tract Infections/prevention & control , Spores, Bacterial/drug effects , Spores, Bacterial/immunology , Spores, Bacterial/pathogenicity , Survival Analysis
4.
Vaccine ; 35(37): 4952-4959, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28774566

ABSTRACT

The anthrax vaccine candidate AV7909 is being developed as a next generation vaccine for a post-exposure prophylaxis (PEP) indication against anthrax. AV7909 consists of the Anthrax Vaccine Adsorbed (AVA, BioThrax®) bulk drug substance adjuvanted with the immunostimulatory oligodeoxynucleotide (ODN) compound, CPG 7909. The addition of CPG 7909 to AVA enhances both the magnitude and the kinetics of antibody responses in animals and human subjects, making AV7909 a suitable next-generation vaccine for use in a PEP setting. The studies described here provide initial information on AV7909-induced toxin-neutralizing antibody (TNA) levels associated with the protection of animals from lethal Bacillus anthracis challenge. Guinea pigs or nonhuman primates (NHPs) were immunized on Days 0 and 28 with various dilutions of AV7909, AVA or a saline or Alhydrogel+CPG 7909 control. Animals were challenged via the inhalational route with a lethal dose of aerosolized B. anthracis (Ames strain) spores and observed for clinical signs of disease and mortality. The relationship between pre-challenge serum TNA levels and survival following challenge was determined in order to calculate a threshold TNA level associated with protection. Immunisation with AV7909 induced a rapid, highly protective TNA response in guinea pigs and NHPs. Surprisingly, the TNA threshold associated with a 70% probability of survival for AV7909 immunized animals was substantially lower than the threshold which has been established for the licensed AVA vaccine. The results of this study suggest that the TNA threshold of protection against anthrax could be modified by the addition of an immune stimulant such as CPG 7909 and that the TNA levels associated with protection may be vaccine-specific.


Subject(s)
Anthrax Vaccines/immunology , Antibodies, Neutralizing/immunology , Animals , Guinea Pigs , Post-Exposure Prophylaxis , Primates , Vaccination
5.
Antimicrob Agents Chemother ; 60(10): 5787-95, 2016 10.
Article in English | MEDLINE | ID: mdl-27431222

ABSTRACT

Inhalational anthrax has high mortality even with antibiotic treatment, and antitoxins are now recommended as an adjunct to standard antimicrobial regimens. The efficacy of obiltoxaximab, a monoclonal antibody against anthrax protective antigen (PA), was examined in multiple studies conducted in two animal models of inhalational anthrax. A single intravenous bolus of 1 to 32 mg/kg of body weight obiltoxaximab or placebo was administered to New Zealand White rabbits (two studies) and cynomolgus macaques (4 studies) at disease onset (significant body temperature increase or detection of serum PA) following lethal challenge with aerosolized Bacillus anthracis spores. The primary endpoint was survival. The relationship between efficacy and disease severity, defined by pretreatment bacteremia and toxemia levels, was explored. In rabbits, single doses of 1 to 16 mg/kg obiltoxaximab led to 17 to 93% survival. In two studies, survival following 16 mg/kg obiltoxaximab was 93% and 62% compared to 0% and 0% for placebo (P = 0.0010 and P = 0.0013, respectively). Across four macaque studies, survival was 6.3% to 78.6% following 4 to 32 mg/kg obiltoxaximab. In two macaque studies, 16 mg/kg obiltoxaximab reduced toxemia and led to survival rates of 31%, 35%, and 47% versus 0%, 0%, and 6.3% with placebo (P = 0.0085, P = 0.0053, P = 0.0068). Pretreatment bacteremia and toxemia levels inversely correlated with survival. Overall, obiltoxaximab monotherapy neutralized PA and increased survival across the range of disease severity, indicating clinical benefit of toxin neutralization with obiltoxaximab in both early and late stages of inhalational anthrax.


Subject(s)
Anthrax/drug therapy , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Antitoxins/pharmacology , Respiratory Tract Infections/drug therapy , Animals , Anthrax/etiology , Anthrax/mortality , Anti-Bacterial Agents/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Female , Macaca fascicularis , Male , Rabbits , Respiratory Tract Infections/etiology , Respiratory Tract Infections/mortality , Survival Rate , Treatment Outcome
6.
Clin Vaccine Immunol ; 22(4): 430-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25673303

ABSTRACT

AdVAV is a replication-deficient adenovirus type 5-vectored vaccine expressing the 83-kDa protective antigen (PA83) from Bacillus anthracis that is being developed for the prevention of disease caused by inhalation of aerosolized B. anthracis spores. A noninferiority study comparing the efficacy of AdVAV to the currently licensed Anthrax Vaccine Absorbed (AVA; BioThrax) was performed in New Zealand White rabbits using postchallenge survival as the study endpoint (20% noninferiority margin for survival). Three groups of 32 rabbits were vaccinated with a single intranasal dose of AdVAV (7.5 × 10(7), 1.5 × 10(9), or 3.5 × 10(10) viral particles). Three additional groups of 32 animals received two doses of either intranasal AdVAV (3.5 × 10(10) viral particles) or intramuscular AVA (diluted 1:16 or 1:64) 28 days apart. The placebo group of 16 rabbits received a single intranasal dose of AdVAV formulation buffer. All animals were challenged via the inhalation route with a targeted dose of 200 times the 50% lethal dose (LD50) of aerosolized B. anthracis Ames spores 70 days after the initial vaccination and were followed for 3 weeks. PA83 immunogenicity was evaluated by validated toxin neutralizing antibody and serum anti-PA83 IgG enzyme-linked immunosorbent assays (ELISAs). All animals in the placebo cohort died from the challenge. Three of the four AdVAV dose cohorts tested, including two single-dose cohorts, achieved statistical noninferiority relative to the AVA comparator group, with survival rates between 97% and 100%. Vaccination with AdVAV also produced antibody titers with earlier onset and greater persistence than vaccination with AVA.


Subject(s)
Anthrax Vaccines/administration & dosage , Anthrax Vaccines/immunology , Anthrax/prevention & control , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Drug Carriers , Mastadenovirus/genetics , Respiratory Tract Infections/prevention & control , Administration, Intranasal , Animals , Anthrax/immunology , Anthrax Vaccines/genetics , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Antigens, Bacterial/genetics , Antitoxins/blood , Bacterial Toxins/genetics , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Genetic Vectors , Immunoglobulin G/blood , Male , Neutralization Tests , Rabbits , Respiratory Tract Infections/immunology , Survival Analysis , Vaccination/methods , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
7.
Virology ; 452-453: 152-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24606692

ABSTRACT

Influenza infection of humans remains an important public health problem. Vaccine strategies result in a significant but only partial control (65-85%) of infection. Thus, chemotherapeutic approaches are needed to provide a solution both for vaccine failures and to limit infection in the unvaccinated population. Previously (Walsh et al., 2011; Teijaro et al., 2011) documented that sphingosine-1-phosphate 1 receptor (S1P1R) agonists significantly protected mice against pathogenic H1N1 influenza virus by limiting immunopathologic damage while allowing host control of the infection. Here we extend that observation by documenting S1P1R agonist can control pathogenic H1N1 influenza infection in ferrets. S1P1R agonist was more effective in reducing pulmonary injury than the antiviral drug oseltamivir but, importantly, combined therapy was significantly more effective than either therapy alone.


Subject(s)
Antibodies, Viral/immunology , Antiviral Agents/administration & dosage , Influenza A Virus, H1N1 Subtype/drug effects , Influenza, Human/drug therapy , Lung/pathology , Receptors, Lysosphingolipid/agonists , Animals , Disease Models, Animal , Ferrets , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/immunology , Influenza, Human/pathology , Influenza, Human/virology , Lung/immunology , Lung/virology , Male , Sphingosine-1-Phosphate Receptors
8.
Clin Vaccine Immunol ; 20(7): 1016-26, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23658392

ABSTRACT

Antimicrobials administered postexposure can reduce the incidence or progression of anthrax disease, but they do not protect against the disease resulting from the germination of spores that may remain in the body after cessation of the antimicrobial regimen. Such additional protection may be achieved by postexposure vaccination; however, no anthrax vaccine is licensed for postexposure prophylaxis (PEP). In a rabbit PEP study, animals were subjected to lethal challenge with aerosolized Bacillus anthracis spores and then were treated with levofloxacin with or without concomitant intramuscular (i.m.) vaccination with anthrax vaccine adsorbed (AVA) (BioThrax; Emergent BioDefense Operations Lansing LLC, Lansing, MI), administered twice, 1 week apart. A significant increase in survival rates was observed among vaccinated animals compared to those treated with antibiotic alone. In preexposure prophylaxis studies in rabbits and nonhuman primates (NHPs), animals received two i.m. vaccinations 1 month apart and were challenged with aerosolized anthrax spores at day 70. Prechallenge toxin-neutralizing antibody (TNA) titers correlated with animal survival postchallenge and provided the means for deriving an antibody titer associated with a specific probability of survival in animals. In a clinical immunogenicity study, 82% of the subjects met or exceeded the prechallenge TNA value that was associated with a 70% probability of survival in rabbits and 88% probability of survival in NHPs, which was estimated based on the results of animal preexposure prophylaxis studies. The animal data provide initial information on protective antibody levels for anthrax, as well as support previous findings regarding the ability of AVA to provide added protection to B. anthracis-infected animals compared to antimicrobial treatment alone.


Subject(s)
Anthrax Vaccines/administration & dosage , Anthrax Vaccines/immunology , Anthrax/prevention & control , Post-Exposure Prophylaxis/methods , Vaccination/methods , Adolescent , Adult , Aged , Animals , Anthrax Vaccines/adverse effects , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Antitoxins/blood , Disease Models, Animal , Female , Humans , Macaca fascicularis , Male , Middle Aged , Rabbits , Survival Analysis , Vaccination/adverse effects , Young Adult
9.
PLoS One ; 8(3): e58337, 2013.
Article in English | MEDLINE | ID: mdl-23472182

ABSTRACT

Influenza A viruses continue to pose a threat to human health; thus, various vaccines and prophylaxis continue to be developed. Testing of these products requires various animal models including mice, guinea pigs, and ferrets. However, because ferrets are naturally susceptible to infection with human influenza viruses and because the disease state resembles that of human influenza, these animals have been widely used as a model to study influenza virus pathogenesis. In this report, a statistical analysis was performed to evaluate data involving 269 ferrets infected with seasonal influenza, swine influenza, and highly pathogenic avian influenza (HPAI) from 16 different studies over a five year period. The aim of the analyses was to better qualify the ferret model by identifying relationships among important animal model parameters (endpoints) and variables of interest, which include survival, time-to-death, changes in body temperature and weight, and nasal wash samples containing virus, in addition to significant changes from baseline in selected hematology and clinical chemistry parameters. The results demonstrate that a disease clinical profile, consisting of various changes in the biological parameters tested, is associated with various influenza A infections in ferrets. Additionally, the analysis yielded correlates of protection associated with HPAI disease in ferrets. In all, the results from this study further validate the use of the ferret as a model to study influenza A pathology and to evaluate product efficacy.


Subject(s)
Disease Models, Animal , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/physiopathology , Animals , Body Temperature , Body Weight , Chickens , Ferrets , Hemagglutination , Hemagglutination Inhibition Tests , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Influenza A Virus, H5N1 Subtype , Time Factors
10.
Arch Virol ; 158(6): 1305-22, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23397329

ABSTRACT

Infection with pathogenic influenza viruses is associated with intense inflammatory disease. Here, we investigated the innate immune response in mice infected with H5N1 A/Vietnam/1203/04 and with reassortant human H1N1 A/Texas/36/91 viruses containing the virulence genes hemagglutinin (HA), neuraminidase (NA) and NS1 of the 1918 pandemic virus. Inclusion of the 1918 HA and NA glycoproteins rendered a seasonal H1N1 virus capable of inducing an exacerbated host innate immune response similar to that observed for highly pathogenic A/Vietnam/1203/04 virus. Infection with 1918 HA/NA:Tx/91 and A/Vietnam/1203/04 were associated with severe lung pathology, increased cytokine and chemokine production, and significant immune cell changes, including the presence of CD11b(+)Gr-1(+) cells in the blood, lung and bone marrow. Significant differential gene expression in the lung included pathways for cell death, apoptosis, production and response to reactive oxygen radicals, as well as arginine and proline metabolism and chemokines associated with monocyte and neutrophil/granulocyte accumulation and/or activation. Arginase was produced in the lung of animals infected with A/Vietnam/1204. These results demonstrate that the innate immune cell response results in the accumulation of CD11b(+)Gr-1(+) cells and products that have previously been shown to contribute to T cell suppression.


Subject(s)
Bone Marrow/immunology , CD11b Antigen/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , T-Lymphocytes/immunology , Animals , Chemokines/immunology , Female , Gene Expression Profiling , Granulocytes/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/pathogenicity , Leukocytes/immunology , Lung/cytology , Lung/metabolism , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/virology , Virulence/immunology
11.
Infect Immun ; 81(4): 1152-63, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23357384

ABSTRACT

Nonhuman primates (NHPs) and rabbits are the animal models most commonly used to evaluate the efficacy of medical countermeasures against anthrax in support of licensure under the FDA's "Animal Rule." However, a need for an alternative animal model may arise in certain cases. The development of such an alternative model requires a thorough understanding of the course and manifestation of experimental anthrax disease induced under controlled conditions in the proposed animal species. The guinea pig, which has been used extensively for anthrax pathogenesis studies and anthrax vaccine potency testing, is a good candidate for such an alternative model. This study was aimed at determining the median lethal dose (LD50) of the Bacillus anthracis Ames strain in guinea pigs and investigating the natural history, pathophysiology, and pathology of inhalational anthrax in this animal model following nose-only aerosol exposure. The inhaled LD50 of aerosolized Ames strain spores in guinea pigs was determined to be 5.0 × 10(4) spores. Aerosol challenge of guinea pigs resulted in inhalational anthrax with death occurring between 46 and 71 h postchallenge. The first clinical signs appeared as early as 36 h postchallenge. Cardiovascular function declined starting at 20 h postexposure. Hematogenous dissemination of bacteria was observed microscopically in multiple organs and tissues as early as 24 h postchallenge. Other histopathologic findings typical of disseminated anthrax included suppurative (heterophilic) inflammation, edema, fibrin, necrosis, and/or hemorrhage in the spleen, lungs, and regional lymph nodes and lymphocyte depletion and/or lymphocytolysis in the spleen and lymph nodes. This study demonstrated that the course of inhalational anthrax disease and the resulting pathology in guinea pigs are similar to those seen in rabbits and NHPs, as well as in humans.


Subject(s)
Anthrax/pathology , Anthrax/physiopathology , Bacillus anthracis/pathogenicity , Disease Models, Animal , Animals , Anthrax/mortality , Female , Guinea Pigs , Lethal Dose 50 , Male , Survival Analysis , Time Factors
12.
Sci Rep ; 2: 972, 2012.
Article in English | MEDLINE | ID: mdl-23240077

ABSTRACT

Avian influenza viruses are widespread in birds, contagious in humans, and are categorized as low pathogenicity avian influenza or highly pathogenic avian influenza. Ferrets are susceptible to infection with avian and human influenza A and B viruses and have been widely used as a model to study pathogenicity and vaccine efficacy. In this report, the natural history of the H5N1 influenza virus A/Vietnam/1203/04 influenza infection in ferrets was examined to determine clinical and laboratory parameters that may indicate (1) the onset of disease and (2) survival. In all, twenty of 24 animals infected with 7 × 10(5) TCID(50) of A/Vietnam/1203/04 succumbed. A statistical analysis identified a combination of parameters including weight loss, nasal wash TCID(50), eosinophils, and liver enzymes such as alanine amino transferase that might possibly serve as indicators of both disease onset and challenge survival.


Subject(s)
Ferrets/virology , Influenza A Virus, H5N1 Subtype/pathogenicity , Orthomyxoviridae Infections/virology , Animals , Orthomyxoviridae Infections/physiopathology
13.
Sci Transl Med ; 4(151): 151ra126, 2012 Sep 12.
Article in English | MEDLINE | ID: mdl-22972844

ABSTRACT

Because clinical trials to assess the efficacy of vaccines against anthrax are not ethical or feasible, licensure for new anthrax vaccines will likely involve the Food and Drug Administration's "Animal Rule," a set of regulations that allow approval of products based on efficacy data only in animals combined with immunogenicity and safety data in animals and humans. U.S. government-sponsored animal studies have shown anthrax vaccine efficacy in a variety of settings. We examined data from 21 of those studies to determine whether an immunological bridge based on lethal toxin neutralization activity assay (TNA) can predict survival against an inhalation anthrax challenge within and across species and genera. The 21 studies were classified into 11 different settings, each of which had the same animal species, vaccine type and formulation, vaccination schedule, time of TNA measurement, and challenge time. Logistic regression models determined the contribution of vaccine dilution dose and TNA on prediction of survival. For most settings, logistic models using only TNA explained more than 75% of the survival effect of the models with dose additionally included. Cross-species survival predictions using TNA were compared to the actual survival and shown to have good agreement (Cohen's κ ranged from 0.55 to 0.78). In one study design, cynomolgus macaque data predicted 78.6% survival in rhesus macaques (actual survival, 83.0%) and 72.6% in rabbits (actual survival, 64.6%). These data add support for the use of TNA as an immunological bridge between species to extrapolate data in animals to predict anthrax vaccine effectiveness in humans.


Subject(s)
Anthrax Vaccines/immunology , Anthrax Vaccines/therapeutic use , Anthrax/mortality , Anthrax/prevention & control , Antibodies, Bacterial/immunology , Aerosols , Animals , Anthrax/immunology , Macaca mulatta , Rabbits
14.
Clin Vaccine Immunol ; 19(11): 1765-75, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22956657

ABSTRACT

Appropriate animal models are required to test medical countermeasures to bioterrorist threats. To that end, we characterized a nonhuman primate (NHP) inhalational anthrax therapeutic model for use in testing anthrax therapeutic medical countermeasures according to the U.S. Food and Drug Administration Animal Rule. A clinical profile was recorded for each NHP exposed to a lethal dose of Bacillus anthracis Ames spores. Specific diagnostic parameters were detected relatively early in disease progression, i.e., by blood culture (∼37 h postchallenge) and the presence of circulating protective antigen (PA) detected by electrochemiluminescence (ECL) ∼38 h postchallenge, whereas nonspecific clinical signs of disease, i.e., changes in body temperature, hematologic parameters (ca. 52 to 66 h), and clinical observations, were delayed. To determine whether the presentation of antigenemia (PA in the blood) was an appropriate trigger for therapeutic intervention, a monoclonal antibody specific for PA was administered to 12 additional animals after the circulating levels of PA were detected by ECL. Seventy-five percent of the monoclonal antibody-treated animals survived compared to 17% of the untreated controls, suggesting that intervention at the onset of antigenemia is an appropriate treatment trigger for this model. Moreover, the onset of antigenemia correlated with bacteremia, and NHPs were treated in a therapeutic manner. Interestingly, brain lesions were observed by histopathology in the treated nonsurviving animals, whereas this observation was absent from 90% of the nonsurviving untreated animals. Our results support the use of the cynomolgus macaque as an appropriate therapeutic animal model for assessing the efficacy of medical countermeasures developed against anthrax when administered after a confirmation of infection.


Subject(s)
Anthrax/pathology , Anthrax/therapy , Disease Models, Animal , Primate Diseases/pathology , Primate Diseases/therapy , Respiratory Tract Infections/pathology , Respiratory Tract Infections/therapy , Animals , Anthrax/diagnosis , Antibodies, Bacterial/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antigens, Bacterial/blood , Bacterial Toxins/blood , Biomarkers/blood , Brain/pathology , Female , Guideline Adherence , Macaca fascicularis , Male , Primate Diseases/diagnosis , Respiratory Tract Infections/diagnosis , Survival Analysis , Time Factors , United States , United States Food and Drug Administration
15.
Clin Vaccine Immunol ; 19(9): 1517-25, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22837095

ABSTRACT

The development of an appropriate animal therapeutic model is essential to assess the potential efficacy of therapeutics for use in the event of a Bacillus anthracis exposure. We conducted a natural history study that showed New Zealand White rabbits exhibited a significant increase in body temperature (SIBT), changes in hematologic parameters, and increases in C-reactive protein and succumbed to disease with an average time to death of approximately 73 h following aerosol challenge with B. anthracis Ames spores. The SIBT was used as a trigger to treat with a fully human monoclonal antibody directed at protective antigen (PA). Ninety percent (9/10) of the treated rabbits survived the lethal inhalational challenge of B. anthracis. Further characterization investigated the protective window of opportunity for anti-PA antibody administration up to 12 h post-onset of SIBT. Eighty-three percent (5/6) of the rabbits treated at SIBT and 100% (6/6) of those treated at 6 h after SIBT survived challenge. Only 67% (4/6) of the rabbits treated at 12 h after SIBT survived. The increase in body temperature corresponded with both bacteremia and antigenemia (PA in the blood), indicating that SIBT is a suitable trigger to initiate treatment in a therapeutic model of inhalational anthrax.


Subject(s)
Anthrax/pathology , Anthrax/therapy , Bacillus anthracis/pathogenicity , Biomarkers , Fever/diagnosis , Animals , Antibodies, Bacterial/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antigens, Bacterial , Bacterial Toxins/antagonists & inhibitors , Blood Cells/physiology , C-Reactive Protein/analysis , Disease Models, Animal , Female , Immunotherapy/methods , Male , Rabbits , Survival Analysis , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...