Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Infect Immun ; 88(8)2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32393506

RESUMO

Bacillus anthracis is the causative agent of anthrax disease, presents with high mortality, and has been at the center of bioweapon efforts. The only currently U.S. FDA-approved vaccine to prevent anthrax in humans is anthrax vaccine adsorbed (AVA), which is protective in several animal models and induces neutralizing antibodies against protective antigen (PA), the cell-binding component of anthrax toxin. However, AVA requires a five-course regimen to induce immunity, along with an annual booster, and is composed of undefined culture supernatants from a PA-secreting strain. In addition, it appears to be ineffective against strains that lack anthrax toxin. Here, we investigated a vaccine formulation consisting of recombinant proteins from a surface-localized heme transport system containing near-iron transporter (NEAT) domains and its efficacy as a vaccine for anthrax disease. The cocktail of five NEAT domains was protective against a lethal challenge of inhaled bacillus spores at 3 and 28 weeks after vaccination. The reduction of the formulation to three NEATs (IsdX1, IsdX2, and Bslk) was as effective as a five-NEAT domain cocktail. The adjuvant alum, approved for use in humans, was as protective as Freund's Adjuvant, and protective vaccination correlated with increased anti-NEAT antibody reactivity and reduced bacterial levels in organs. Finally, the passive transfer of anti-NEAT antisera reduced mortality and disease severity, suggesting the protective component is comprised of antibodies. Collectively, these results provide evidence that a vaccine based upon recombinant NEAT proteins should be considered in the development of a next-generation anthrax vaccine.


Assuntos
Vacinas contra Antraz/imunologia , Antraz/prevenção & controle , Anticorpos Antibacterianos/biossíntese , Anticorpos Neutralizantes/biossíntese , Antígenos de Bactérias/imunologia , Bacillus anthracis/efeitos dos fármacos , Administração por Inalação , Compostos de Alúmen/administração & dosagem , Animais , Antraz/imunologia , Antraz/microbiologia , Antraz/mortalidade , Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/genética , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/genética , Bacillus anthracis/imunologia , Bacillus anthracis/patogenicidade , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Transporte/administração & dosagem , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Complemento C5/deficiência , Feminino , Adjuvante de Freund/administração & dosagem , Humanos , Imunogenicidade da Vacina , Camundongos Knockout , Análise de Sobrevida , Vacinação/métodos
2.
Med Microbiol Immunol ; 209(2): 125-137, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31811379

RESUMO

The most promising means of controlling anthrax, a lethal zoonotic disease during the early infection stages, entail restricting the resilient infectious form, i.e., the spores from proliferating to replicating bacilli in the host. The extractible antigen (EA1), a major S-layer protein present on the vegetative cells and spores of Bacillus anthracis, is highly immunogenic and protects mice against lethal challenge upon immunization. In the present study, mice were immunized with r-EA1C, the C terminal crystallization domain of EA1, to generate a neutralizing monoclonal antibody EA752-862, that was evaluated for its anti-spore and anti-bacterial properties. The monoclonal antibody EA752-862 had a minimum inhibitory concentration of 0.08 mg/ml, was bactericidal at a concentration of 0.1 mg/ml and resulted in 100% survival of mice against challenge with B. anthracis vegetative cells. Bacterial cell lysis as observed by scanning electron microscopy and nucleic acid leakage assay could be attributed as a possible mechanism for the bactericidal property. The association of mAb EA752-862 with spores inhibits their subsequent germination to vegetative cells in vitro, enhances phagocytosis of the spores and killing of the vegetative cells within the macrophage, and subsequently resulted in 90% survival of mice upon B. anthracis Ames spore challenge. Therefore, owing to its anti-spore and bactericidal properties, the present study demonstrates mAb EA752-862 as an efficient neutralizing antibody that hinders the establishment of early infection before massive multiplication and toxin release takes place.


Assuntos
Antraz/prevenção & controle , Antibacterianos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Bacillus anthracis/imunologia , Esporos Bacterianos/imunologia , Animais , Antraz/imunologia , Antibacterianos/biossíntese , Antibacterianos/química , Antibacterianos/farmacologia , Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/química , Anticorpos Antibacterianos/isolamento & purificação , Anticorpos Antibacterianos/farmacologia , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/isolamento & purificação , Anticorpos Neutralizantes/farmacologia , Antígenos de Bactérias/imunologia , Bacillus anthracis/efeitos dos fármacos , Sítios de Ligação , Feminino , Imunização , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Varredura , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Esporos Bacterianos/efeitos dos fármacos
3.
mSphere ; 4(3)2019 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-31217301

RESUMO

Inhalational anthrax caused by Bacillus anthracis, a spore-forming Gram-positive bacterium, is a highly lethal infection. Antibodies targeting the protective antigen (PA) binding component of the toxins have recently been authorized as an adjunct to antibiotics, although no conclusive evidence demonstrates that anthrax antitoxin therapy has any significant benefit. We discuss here the rational basis of anti-PA development regarding the pathogenesis of the disease. We argue that inductive reasoning may induce therapeutic bias. We identified anthrax animal model analysis as another bias. Further studies are needed to assess the benefit of anti-PA antibodies in the treatment of inhalational anthrax, while a clearer consensus should be established around what evidence should be proven in an anthrax model.


Assuntos
Antraz/imunologia , Antraz/terapia , Anticorpos Antibacterianos/uso terapêutico , Bacillus anthracis/imunologia , Imunoterapia , Infecções Respiratórias/imunologia , Infecções Respiratórias/terapia , Animais , Anticorpos Monoclonais/uso terapêutico , Antígenos de Bactérias/imunologia , Antitoxinas/uso terapêutico , Toxinas Bacterianas/imunologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Humanos
4.
Artigo em Inglês | MEDLINE | ID: mdl-28348035

RESUMO

Animal models have played a pivotal role in all stages of vaccine development. Their predictive value for vaccine effectiveness depends on the pathogen, the robustness of the animal challenge model, and the correlates of protection (if known). This article will cover key questions regarding bridging animal studies to efficacy trials in humans. Examples include human papillomavirus (HPV) vaccine in which animal protection after vaccination with heterologous prototype virus-like particles (VLPs) predicted successful efficacy trials in humans, and a recent approval of anthrax vaccine in accordance with the "Animal Rule." The establishment of animal models predictive of vaccine effectiveness in humans has been fraught with difficulties with low success rate to date. Challenges facing the use of animal models for vaccine development against Ebola and HIV will be discussed.


Assuntos
Modelos Animais de Doenças , Vacinas/uso terapêutico , Vacinas contra a AIDS/efeitos adversos , Vacinas contra a AIDS/uso terapêutico , Animais , Antraz/imunologia , Vacinas contra Antraz/efeitos adversos , Vacinas contra Antraz/uso terapêutico , Aprovação de Drogas , Avaliação Pré-Clínica de Medicamentos , Vacinas contra Ebola/efeitos adversos , Vacinas contra Ebola/uso terapêutico , Humanos , Papillomaviridae/imunologia , Vacinas contra Papillomavirus/efeitos adversos , Vacinas contra Papillomavirus/uso terapêutico , Estudo de Prova de Conceito , Especificidade da Espécie
5.
Clin Pharmacol Drug Dev ; 7(6): 652-660, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29125719

RESUMO

Inhalational anthrax is a highly lethal infection caused by Bacillus anthracis and a serious bioterrorism threat. Protective antigen (PA) is a critical component required for the virulence of Bacillus anthracis. Obiltoxaximab, a high-affinity monoclonal antibody that neutralizes PA, is approved in the United States for intravenous use for the treatment of inhalational anthrax in combination with appropriate antibacterial drugs and for prophylaxis of inhalational anthrax when alternative therapies are not available or appropriate. Here, we explored the safety, pharmacokinetics (PK), and immunogenicity of obiltoxaximab administered by intramuscular injection at doses of 4, 8, 16, 20, and 24 mg/kg in healthy humans. Systemic exposures were approximately dose proportional, maximum serum concentrations were observed after 6-9 days, and terminal half-life ranged from 16 to 23 days. Average absolute intramuscular bioavailability was 64%. Obiltoxaximab was well tolerated, and local tolerability was acceptable up to 24 mg/kg intramuscularly, up to 6 injections per dose, and up to 5 mL per injection. No injection-site abscesses or hypersensitivity reactions occurred; no subjects developed treatment-emergent antitherapeutic antibodies over the study period of 71 days.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacocinética , Antitoxinas/efeitos adversos , Hipersensibilidade a Drogas/imunologia , Antraz/tratamento farmacológico , Antraz/imunologia , Anticorpos Monoclonais/administração & dosagem , Antígenos de Bactérias/imunologia , Antitoxinas/administração & dosagem , Área Sob a Curva , Bacillus anthracis/efeitos dos fármacos , Bacillus anthracis/imunologia , Toxinas Bacterianas/imunologia , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Meia-Vida , Voluntários Saudáveis , Humanos , Injeções Intramusculares , Masculino , Pessoa de Meia-Idade , Infecções Respiratórias/tratamento farmacológico , Infecções Respiratórias/imunologia
6.
Eur J Pharm Biopharm ; 93: 149-64, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25818119

RESUMO

In a time in which mucosal vaccines development has been delayed by the lack of safe and effective mucosal adjuvants, the combination of adjuvants has started to be explored as a strategy to obtain potent vaccine formulations. This study describes a novel adjuvant combination as an effective approach for a nasal vaccine - the association of the mast cell activator compound 48/80 with chitosan based nanoparticles. It was hypothesized that mucoadhesive nanoparticles would promote the cellular uptake and prolong the antigen residence time on nasal cavity. Simultaneously, mast cell activation would promote a local microenvironment favorable to the development of an immune response. To test this hypothesis, two different C48/80 loaded nanoparticles (NPs) were prepared: Chitosan-C48/80 NP (Chi-C48/80 NP) and Chitosan/Alginate-C48/80 NP (Chi/Alg-C48/80 NP). The potential as a vaccine adjuvant of the two delivery systems was evaluated and directly compared. Both formulations had a mean size near 500nm and a positive charge; however, Chi-C48/80 NP was a more effective adjuvant delivery system when compared with Chi/Alg-C48/80 NP or C48/80 alone. Chi-C48/80 NP activated mast cells at a greater extent, were better internalized by antigen presenting cells than Chi/Alg-C48/80 NP and successfully enhanced the nasal residence time of a model antigen. Superiority of Chi-C48/80 NP as adjuvant was also observed in vivo. Therefore, nasal immunization of mice with Bacillus anthracis protective antigen (PA) adsorbed on Chi-C48/80 NP elicited high levels of serum anti-PA neutralizing antibodies and a more balanced Th1/Th2 profile than C48/80 in solution or Chi/Alg-C48/80 NP. The incorporation of C48/80 within Chi NP also promoted a mucosal immunity greater than all the other adjuvanted groups tested, showing that the combination of a mast cell activator and chitosan NP could be a promising strategy for nasal immunization.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas contra Antraz/administração & dosagem , Antraz/prevenção & controle , Antígenos de Bactérias/administração & dosagem , Toxinas Bacterianas/administração & dosagem , Quitosana/administração & dosagem , Portadores de Fármacos , Imunidade nas Mucosas/efeitos dos fármacos , Nanopartículas , Mucosa Nasal/efeitos dos fármacos , p-Metoxi-N-metilfenetilamina/administração & dosagem , Adjuvantes Imunológicos/química , Administração Intranasal , Alginatos/administração & dosagem , Alginatos/química , Animais , Antraz/sangue , Antraz/imunologia , Antraz/microbiologia , Vacinas contra Antraz/química , Vacinas contra Antraz/imunologia , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/química , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/química , Toxinas Bacterianas/imunologia , Biomarcadores/sangue , Química Farmacêutica , Quitosana/química , Quitosana/imunologia , Relação Dose-Resposta a Droga , Feminino , Ácido Glucurônico/administração & dosagem , Ácido Glucurônico/química , Ácidos Hexurônicos/administração & dosagem , Ácidos Hexurônicos/química , Humanos , Imunização , Mastócitos/efeitos dos fármacos , Mastócitos/imunologia , Mastócitos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Nanomedicina , Mucosa Nasal/imunologia , Tamanho da Partícula , Células RAW 264.7 , Propriedades de Superfície , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/microbiologia , Tecnologia Farmacêutica/métodos , Fatores de Tempo , p-Metoxi-N-metilfenetilamina/química , p-Metoxi-N-metilfenetilamina/imunologia
7.
Artif Cells Nanomed Biotechnol ; 42(1): 47-57, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23452384

RESUMO

The aim of present study was to evaluate the potential of mucoadhesive alginate-coated chitosan microparticles (A-CHMp) for oral vaccine against anthrax. The zeta potential of A-CHMp was -29.7 mV, and alginate coating could prevent the burst release of antigen in simulated gastric fluid. The results indicated that A-CHMp was mucoadhesive in nature and transported it to the peyer's patch upon oral delivery. The immunization studies indicated that A-CHMp resulted in the induction of potent systemic and mucosal immune responses, whereas alum-adjuvanted rPA could induce only systemic immune response. Thus, A-CHMp represents a promising acid carrier adjuvant for oral immunization against anthrax.


Assuntos
Vacinas contra Antraz/administração & dosagem , Antraz/prevenção & controle , Anticorpos Antibacterianos/biossíntese , Antígenos de Bactérias/imunologia , Bacillus anthracis/imunologia , Toxinas Bacterianas/antagonistas & inibidores , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/química , Administração Oral , Alginatos/química , Animais , Antraz/sangue , Antraz/imunologia , Antraz/microbiologia , Vacinas contra Antraz/imunologia , Anticorpos Antibacterianos/sangue , Bacillus anthracis/química , Toxinas Bacterianas/imunologia , Materiais Biomiméticos/química , Quitosana/química , Feminino , Suco Gástrico/química , Imunidade Inata , Imunidade nas Mucosas , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Nódulos Linfáticos Agregados , Vacinação
8.
Curr Med Chem ; 18(33): 5083-94, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22050756

RESUMO

Bacillus anthracis, the causative agent responsible for anthrax infections, poses a significant biodefense threat. There is a high mortality rate associated with untreated anthrax infections; specifically, inhalation anthrax is a particularly virulent form of infection with mortality rates close to 100%, even with aggressive treatment. Currently, a vaccine is not available to the general public and few antibiotics have been approved by the FDA for the treatment of inhalation anthrax. With the threat of natural or engineered bacterial resistance to antibiotics and the limited population for whom the current drugs are approved, there is a clear need for more effective treatments against this deadly infection. A comprehensive review of current research in drug discovery is presented in this article, including efforts to improve the purity and stability of vaccines, design inhibitors targeting the anthrax toxins, and identify inhibitors of novel enzyme targets. High resolution structural information for the anthrax toxins and several essential metabolic enzymes has played a significant role in aiding the structure-based design of potent and selective antibiotics.


Assuntos
Vacinas contra Antraz/uso terapêutico , Antraz/tratamento farmacológico , Antibacterianos/química , Bacillus anthracis/metabolismo , Toxinas Bacterianas/antagonistas & inibidores , Antraz/imunologia , Antraz/prevenção & controle , Vacinas contra Antraz/imunologia , Antibacterianos/uso terapêutico , Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Simulação por Computador , Avaliação Pré-Clínica de Medicamentos , Humanos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/uso terapêutico
9.
N Engl J Med ; 361(2): 135-44, 2009 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-19587338

RESUMO

BACKGROUND: Inhalational anthrax caused by Bacillus anthracis is associated with high mortality primarily due to toxin-mediated injury. Raxibacumab is a human IgG1lambda monoclonal antibody directed against protective antigen, a component of the anthrax toxin. METHODS: We evaluated the efficacy of raxibacumab as a prophylactic agent and after disease onset in a total of four randomized, placebo-controlled studies conducted in rabbits and monkeys. Animals were exposed to an aerosolized target exposure of B. anthracis spores that was approximately 100 times (in the prophylactic studies) and 200 times (in the therapeutic-intervention studies) the median lethal dose. In the therapeutic-intervention studies, animals were monitored for the onset of symptoms. Animals with detectable protective antigen in serum, a significant increase in temperature, or both received a single intravenous bolus of placebo or raxibacumab at a dose of either 20 mg per kilogram of body weight or 40 mg per kilogram. The primary end point was survival at day 14 (in rabbits) or at day 28 (in monkeys). Safety studies were conducted with intravenous raxibacumab (40 mg per kilogram) in 333 healthy human volunteers. RESULTS: In both rabbits and monkeys, the time to detection of protective antigen correlated with the time to bacteremia (r=0.9, P<0.001). In the therapeutic-intervention studies, the survival rate was significantly higher among rabbits that received raxibacumab at a dose of 40 mg per kilogram (44% [8 of 18]) than among rabbits that received placebo (0% [0 of 18]; P=0.003). Raxibacumab treatment also significantly increased survival in monkeys (64% [9 of 14], vs. 0% [0 of 12] with placebo; P<0.001). In human subjects, intravenous raxibacumab at a dose of 40 mg per kilogram had a half-life of 20 to 22 days and provided a maximum concentration of the drug in excess of levels that are protective in animals. Concentrations of raxibacumab provide a surrogate end point that should be predictive of clinical benefit. CONCLUSIONS: A single dose of raxibacumab improved survival in rabbits and monkeys with symptomatic inhalational anthrax. (ClinicalTrials.gov number, NCT00639678.)


Assuntos
Antraz/tratamento farmacológico , Anti-Infecciosos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Bacillus anthracis , Infecções Respiratórias/tratamento farmacológico , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antraz/imunologia , Anti-Infecciosos/efeitos adversos , Anticorpos Antibacterianos/sangue , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Humanizados , Bacteriemia , Ciprofloxacina/efeitos adversos , Ciprofloxacina/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Quimioterapia Combinada , Feminino , Humanos , Imunoglobulina G , Macaca fascicularis , Masculino , Pessoa de Meia-Idade , Coelhos , Distribuição Aleatória , Análise de Sobrevida , Adulto Jovem
10.
Hum Vaccin ; 5(8): 536-44, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19458488

RESUMO

VCL-AB01, a cationic lipid-formulated plasmid DNA (pDNA)-based vaccine that contains genes encoding genetically detoxified Bacillus anthracis protective antigen (PA) and lethal factor (LF), was assessed in a Phase 1, dose-escalating clinical trial in healthy adults for safety and immunogenicity, and in nonhuman primates for immunogenicity and efficacy against challenge with a lethal dose of B. anthracis spores. Healthy 18-45 year old subjects were randomly assigned to receive either the investigational vaccine containing 0.2 mg, 0.6 mg, or 2 mg of total pDNA per dose, or saline placebo, administered at 0, 1 and 2 months. The 0.2 mg and 0.6 mg dose levels were generally well tolerated; however, dose-limiting reactogenicity was observed among subjects given the first 2 mg dose and the remaining two injections in the 2 mg group were reduced to 0.6 mg. Dose-related increases in seroconversion frequencies were observed. Overall, 10%, 33.3% and 80% of subjects in the 0.2, 0.6 and 2 mg groups, respectively, developed antibodies to PA and/or LF as measured by ELISA; however, antibodies with toxin neutralizing activity (TNA) were detected in only one subject. In monkeys that received a 0.6 mg dose three times at 2 week intervals, low levels of antibodies were detected by ELISA but not by the TNA assay in all animals just prior to challenge. Despite the absence of TNA, 75% animals survived the lethal challenge. In summary, VCL-AB01 was generally well tolerated in humans at a dose that provided immunity in monkeys despite the lack of robust TNA titers in either species.


Assuntos
Vacinas contra Antraz/imunologia , Antraz/imunologia , Antraz/prevenção & controle , Bacillus anthracis/imunologia , Vacinas de DNA/imunologia , Adolescente , Adulto , Animais , Antraz/sangue , Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/efeitos adversos , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Esquema de Medicação , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Injeções Intramusculares , Macaca fascicularis , Masculino , Pessoa de Meia-Idade , New York , Coelhos , Texas , Vacinação , Vacinas de DNA/administração & dosagem , Vacinas de DNA/efeitos adversos
11.
Antimicrob Agents Chemother ; 51(4): 1373-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17296745

RESUMO

An anthrax spore aerosol infection mouse model was developed as a first test of in vivo efficacy of antibiotics identified as active against Bacillus anthracis. Whole-body, 50% lethal dose (LD50) aerosol challenge doses in a range of 1.9x10(3) to 3.4x10(4) CFU with spores of the fully virulent Ames strain were established for three inbred and one outbred mouse strain (A/J, BALB/c, C57BL, and Swiss Webster). The BALB/c strain was further developed as a model for antibiotic efficacy. Time course microbiological examinations of tissue burdens in mice after challenge showed that spores could remain dormant in the lungs while vegetative cells disseminated to the mediastinal lymph nodes and then to the spleen, accompanied by bacteremia. For antibiotic efficacy studies, BALB/c mice were challenged with 50 to 100 LD50 of spores followed by intraperitoneal injection of either ciprofloxacin at 30 mg/kg of body weight (every 12 h [q12h]) or doxycycline at 40 mg/kg (q6h). A control group was treated with phosphate-buffered saline (PBS) q6h. Treatment was begun 24 h after challenge with groups of 10 mice for 14 or 21 days. The PBS-treated control mice all succumbed (10/10) to inhalation anthrax infection within 72 h. Sixty-day survival rates for ciprofloxacin and doxycycline-treated groups were 8/10 and 9/10, respectively, for 14-day treatment and 10/10 and 7/10 for 21-day treatment. Delayed treatment with ciprofloxacin initiated 36 and 48 h postexposure resulted in 80% survival and was statistically no different than early (24 h) postexposure treatment. Results using this mouse model correlate closely with clinical observations of inhalational anthrax in humans and with earlier antibiotic studies in the nonhuman primate inhalational anthrax model.


Assuntos
Antraz/prevenção & controle , Antibacterianos/uso terapêutico , Antibioticoprofilaxia , Ciprofloxacina/uso terapêutico , Ofloxacino/uso terapêutico , Administração por Inalação , Aerossóis , Animais , Antraz/imunologia , Bacillus anthracis/metabolismo , Ciprofloxacina/administração & dosagem , Ciprofloxacina/farmacocinética , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos BALB C , Ofloxacino/administração & dosagem , Ofloxacino/farmacocinética , Esporos Bacterianos/efeitos dos fármacos
12.
Proc Natl Acad Sci U S A ; 103(20): 7813-6, 2006 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-16672361

RESUMO

Prevention of inhalational anthrax after Bacillus anthracis spore exposure requires a prolonged course of antibiotic prophylaxis. In response to the 2001 anthrax attack in the United States, approximately 10,000 people were offered 60 days of antibiotic prophylaxis to prevent inhalational anthrax, but adherence to this regimen was poor. We sought to determine whether a short course of antibiotic prophylaxis after exposure could protect non-human primates from a high-dose spore challenge if vaccination was combined with antibiotics. Two groups of 10 rhesus macaques were exposed to approximately 1,600 LD50 of spores by aerosol. Both groups were given ciprofloxacin by orogastric tube twice daily for 14 days, beginning 1-2 h after exposure. One group also received three doses of the licensed human anthrax vaccine (anthrax vaccine adsorbed) after exposure. In the ciprofloxacin-only group, four of nine monkeys (44%) survived the challenge. In contrast, all 10 monkeys that received 14 days of antibiotic plus anthrax vaccine adsorbed survived (P = 0.011). Thus postexposure vaccination enhanced the protection afforded by 14 days of antibiotic prophylaxis alone and completely protected animals against inhalational anthrax. These data provide evidence that postexposure vaccination can shorten the duration of antibiotic prophylaxis required to protect against inhalational anthrax and may impact public health management of a bioterrorism event.


Assuntos
Administração por Inalação , Antraz/prevenção & controle , Antibacterianos/uso terapêutico , Antibioticoprofilaxia , Ciprofloxacina/uso terapêutico , Vacinação , Animais , Antraz/imunologia , Vacinas contra Antraz , Bacillus anthracis/metabolismo , Bioterrorismo , Sinergismo Farmacológico , Humanos , Macaca mulatta , Testes de Sensibilidade Microbiana , Distribuição Aleatória , Esporos Bacterianos , Taxa de Sobrevida , Fatores de Tempo
13.
J Pharm Pharmacol ; 58(4): 439-47, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16597361

RESUMO

To better protect against inhalational anthrax infection, a nasal anthrax vaccine based on the protective antigen (PA) protein of Bacillus anthracis could be an attractive alternative to the current Anthrax-Vaccine-Adsorbed (AVA), which was licensed for cutaneous anthrax prevention. Previously, we have demonstrated that an anti-PA immune response comparable with that in mice subcutaneously immunized with PA protein adjuvanted with aluminium hydroxide was induced in both the systemic compartment and the mucosal secretions of the nose and lung of anaesthetized mice when they were nasally immunized with PA protein incorporated into previously reported LPD (Liposome-Protamine-DNA) particles. In this study, we evaluated the anti-PA immune response induced by the nasal PA/LPD particles in non-anaesthetized mice and compared it with that in anaesthetized mice. Our data showed that the anti-PA antibody response and the anthrax lethal toxin-neutralization activity induced by the nasal PA/LPD in non-anaesthetized mice was relatively weaker than that in anaesthetized mice. However, the splenocytes isolated from the nasally immunized mice, anaesthetized and non-anaesthetized, proliferated comparably after in-vitro re-stimulation. By evaluating the uptake of fluorescence-labelled LPD particles by phagocytes in the nasal and broncho-alveolar lavages of mice after the nasal administration, we concluded that the relatively weaker anti-PA immune response in the non-anaesthetized mice might be partially attributed to the reduced retention of the PA/LPD particles in the nasal cavity of the non-anaesthetized mice. Data collected in this study are expected to be useful for future anthrax nasal vaccine studies when mice are used as a model.


Assuntos
Anestesia , Vacinas contra Antraz/imunologia , Antraz/imunologia , Anticorpos Antibacterianos/biossíntese , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/administração & dosagem , Toxinas Bacterianas/imunologia , Administração Intranasal , Anestesia/métodos , Animais , Antraz/prevenção & controle , Vacinas contra Antraz/administração & dosagem , Bacillus anthracis/efeitos dos fármacos , Bacillus anthracis/imunologia , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Camundongos , Camundongos Endogâmicos BALB C
14.
Hum Gene Ther ; 16(10): 1151-6, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16218776

RESUMO

Several formulated plasmid DNA (pDNA)-based vaccines are being evaluated for safety and efficacy in healthy human subjects. A safety concern for any vaccine that contains genetic material, be it whole organism, live-attenuated, or gene-based, is the potential for integration into genomic DNA (gDNA). To address this concern, a preclinical pDNA persistence/integration study was conducted in rabbits to determine the level of pDNA in muscle 2, 28, and 64 days after intramuscular injection of DMRIE:DOPE-formulated pDNAs encoding Bacillus anthracis detoxified LF and PA proteins (VCL-AB01 vaccine). Total DNA was extracted from day 64 muscle tissue and fractionated by column agarose gel electrophoresis (CAGE). Plasmid copy number (PCN) in muscle 64 days after injection (geometric mean, 2808 PCN/microg of total DNA or 150,000 diploid genomes) was determined by quantitative polymerase chain reaction. Analysis of total DNA from five VCLAB01- injected rabbits revealed that two of five samples had no detectable PCN in the high molecular weight fraction after one round of CAGE, two samples had PCN under the lower limit of quantitation, and the remaining sample had 123 PCN/microg. All PCN in the latter sample cleared after an additional round of CAGE. It appears, therefore, that persisting PCN fractionate as low molecular weight material and are most likely not integrated into gDNA. Even if the worst-case assumption is made that the highest PCN found associated with gDNA represented covalently integrated pDNA inserts, the frequency of mutation would still be 500-fold lower than the autosomal spontaneous mutation rate.


Assuntos
Vacinas contra Antraz/farmacocinética , Bacillus anthracis , Lipídeos/farmacocinética , Fosfatidiletanolaminas/farmacocinética , Plasmídeos/farmacocinética , Compostos de Amônio Quaternário/farmacocinética , Vacinas de DNA/farmacocinética , Animais , Antraz/genética , Antraz/imunologia , Antraz/prevenção & controle , Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/genética , Vacinas contra Antraz/imunologia , Bacillus anthracis/genética , Bacillus anthracis/imunologia , Avaliação Pré-Clínica de Medicamentos , Humanos , Injeções Intramusculares , Lipídeos/imunologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Fosfatidiletanolaminas/imunologia , Plasmídeos/genética , Plasmídeos/imunologia , Compostos de Amônio Quaternário/imunologia , Coelhos , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas de DNA/imunologia
15.
Vaccine ; 23(16): 1993-9, 2005 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-15734073

RESUMO

The only US-licensed anthrax vaccine for human use, as well as several experimental vaccines containing solely purified recombinant protective antigen (rPA), are formulated using aluminum hydroxide (Al(OH)3) as an adjuvant. It has been suggested that effective adjuvanticity of aluminum salts for protein antigens depends, at least partially, on the degree of adsorption of the antigen to the adjuvant. On the other hand, the ease of antigen desorption from the adjuvant in a quantitative fashion may facilitate the assessment of vaccine characteristics in the laboratory. In this regard, aluminum phosphate (AlPO4), although deemed a "weaker" adjuvant than Al(OH)3, appears superior to the latter. To investigate the possibility of formulating rPA vaccines with AlPO4, as well as the significance of the adsorption of this antigen to the aluminum salt for adjuvanticity, we studied the effect of AlPO4 and Al(OH)3 on the induction of anti-rPA antibodies in mice. In a first immunization experiment the adjuvanticity of AlPO4 combined with rPA was examined. Antibodies against rPA were measured using an ELISA. Results indicated that AlPO4 is able to significantly increase the antibody response to rPA, irrespective of its degree of adsorption to the adjuvant. Based on these results, in a second experiment mice were immunized twice, with different formulations of rPA containing either AlPO4 or Al(OH)3, and rPA-antibodies were measured using ELISA and an in vitro toxin neutralization assay. Comparable immune responses to rPA were obtained with both aluminum salts. Additionally, results with AlPO4 as adjuvant confirmed that, in this mouse model, binding of the protein to the adjuvant is not essential for adjuvanticity, whereas the amount of adjuvant has an influence on the antibody response induced.


Assuntos
Adjuvantes Imunológicos/farmacologia , Alumínio/farmacologia , Vacinas contra Antraz/imunologia , Antraz/prevenção & controle , Compostos de Alumínio/farmacologia , Hidróxido de Alumínio/farmacologia , Animais , Antraz/imunologia , Bacillus anthracis/imunologia , Química Farmacêutica , Ensaio de Imunoadsorção Enzimática , Esquemas de Imunização , Focalização Isoelétrica , Camundongos , Fosfatos/farmacologia , Solubilidade , Vacinas Sintéticas/imunologia
16.
Pharmacoepidemiol Drug Saf ; 14(6): 393-401, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15717323

RESUMO

PURPOSE: The CDC's Anthrax Vaccine and Antibiotic Availability Program was implemented under an Investigational New Drug (IND) application to provide additional post-exposure prophylaxis for individuals potentially exposed to Bacillus anthracis in the fall of 2001. Participants were provided with two options: (1) 40 additional days of antimicrobial prophylaxis (i.e., ciprofloxacin, doxycycline, or amoxicillin); or (2) 40 additional days of antimicrobial prophylaxis plus three doses of anthrax vaccine adsorbed (AVA). METHODS: Participants were monitored for adverse events (AEs). Participants were asked to complete 2-week AE diaries for 6 weeks post-enrollment, and approximately 2 months after enrollment, active surveillance was conducted through telephone interviews with 1113 (64%) participants. RESULTS: A total of 1727 of approximately 10 000 previously prophylaxed persons enrolled to receive 40 additional days of antibiotics. Of these, 199 opted at enrollment to receive three doses of AVA in addition to the additional 40 days of antibiotic. Overall, 28% of participants reported at least one AE on their diaries. Results varied by surveillance mechanism, the diary data indicated differences in the proportion reporting AEs between participants receiving antibiotic only and participants receiving antibiotic and AVA. However, during the active 2-month telephone follow-up, the rates of AEs reported for both the antibiotic only and antibiotic plus AVA treatment regimens were similar. Additionally, ciprofloxacin and doxycycline had similar AE profiles, with only rigors reported significantly more often among ciprofloxacin recipients. CONCLUSIONS: Overall, the rates of AEs experienced by all participants were acceptable given the seriousness of potential B. anthracis exposure.


Assuntos
Sistemas de Notificação de Reações Adversas a Medicamentos , Vacinas contra Antraz/efeitos adversos , Antraz/prevenção & controle , Antibacterianos/uso terapêutico , Bacillus anthracis/efeitos dos fármacos , Experimentação Humana/estatística & dados numéricos , Amoxicilina/uso terapêutico , Antraz/tratamento farmacológico , Antraz/imunologia , Vacinas contra Antraz/administração & dosagem , Bacillus anthracis/imunologia , Bioterrorismo/prevenção & controle , Centers for Disease Control and Prevention, U.S. , Quimioprevenção/efeitos adversos , Quimioprevenção/métodos , Ciprofloxacina/uso terapêutico , Estudos de Coortes , Coleta de Dados , Doxiciclina/uso terapêutico , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Inquéritos e Questionários , Fatores de Tempo , Estados Unidos
17.
Cell Biol Toxicol ; 16(3): 165-74, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11032360

RESUMO

The lethal toxin of Bacillus anthracis, which is composed of two separate proteinaceous exotoxins, namely protective antigen and lethal factor, is central to the pathogenesis of anthrax. Low levels of this toxin are known to induce release of cytokines such as tumor necrosis factor alpha (TNF-alpha). In the present study we investigated the effect of dehydroepiandrosterone (DHEA), melatonin (MLT), or DHEA + MLT on production of lethal toxin-induced TNF-alpha in mouse peritoneal macrophages. We found that treatment with DHEA significantly inhibited the TNF-alpha production caused by anthrax lethal toxin. Exposure of MLT to anthrax lethal toxin-treated macrophages also decreased the release of TNF-alpha to the extracellular medium as compared to the control. However, combined use of DHEA and MLT also inhibited TNF-alpha release, but not more than single therapies. These results suggest that DHEA and MLT may have a therapeutic role in reducing the increased cytokine production induced by anthrax lethal toxin.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antraz/tratamento farmacológico , Antígenos de Bactérias , Bacillus anthracis , Toxinas Bacterianas/imunologia , Desidroepiandrosterona/farmacologia , Macrófagos/imunologia , Melatonina/farmacologia , Fator de Necrose Tumoral alfa/biossíntese , Animais , Antraz/imunologia , Antraz/metabolismo , Toxinas Bacterianas/farmacologia , Células Cultivadas , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos ICR
18.
Mol Microbiol ; 5(6): 1501-6, 1991 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-1787799

RESUMO

To probe the role of the protective antigen (PA) component of anthrax toxin in toxin entry into animals cells, we examined the membrane channel-forming properties and hydrophobicity of intact and trypsin-cleaved forms of the protein at various pH values. At neutral pH neither form caused release of entrapped K+ from unilamellar lipid vesicles. At pH values below 6.0, however, K+ was rapidly released upon addition of either the nicked PA (PAN) or the 63 kDa tryptic fragment of PA (PA63), which has been implicated in the toxin entry process. Under the same conditions intact PA exhibited only weak channel-forming activity, and PA20, the complementary tryptic fragment, showed no such activity. Both PA and PA63 exhibited enhanced hydrophobicity at acidic pH values, but the enhancement was greater and the pH threshold higher with PA63. Our findings indicate that proteolytic removal of PA20 from intact PA enables the residual protein, PA63, to adopt a conformation at mildly acidic pH values that permits it to insert readily and form channels in membranes. Thus acidic conditions within endocytic vesicles may trigger membrane insertion of PA63, which in turn promotes translocation of ligated effector moieties, edema factor or lethal factor, across the vesicle membrane into the cytosol.


Assuntos
Antígenos de Bactérias , Bacillus anthracis/química , Toxinas Bacterianas/química , Antraz/imunologia , Antraz/metabolismo , Bacillus anthracis/imunologia , Bacillus anthracis/metabolismo , Toxinas Bacterianas/imunologia , Toxinas Bacterianas/metabolismo , Fluoresceínas/metabolismo , Fluorescência , Concentração de Íons de Hidrogênio , Cinética , Lipossomos/metabolismo , Substâncias Macromoleculares , Fosfatidilcolinas , Fosfolipídeos/metabolismo , Potássio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA