Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Vaccines (Basel) ; 10(9)2022 Aug 25.
Article in English | MEDLINE | ID: mdl-36146462

ABSTRACT

The FDA Animal Rule was devised to facilitate approval of candidate vaccines and therapeutics using animal survival data when human efficacy studies are not practical or ethical. This regulatory pathway is critical for candidates against pathogens with high case fatality rates that prohibit human challenge trials, as well as candidates with low and sporadic incidences of outbreaks that make human field trials difficult. Important components of a vaccine development plan for Animal Rule licensure are the identification of an immune correlate of protection and immunobridging to humans. The relationship of vaccine-induced immune responses to survival after vaccination and challenge must be established in validated animal models and then used to infer predictive vaccine efficacy in humans via immunobridging. The Sabin Vaccine Institute is pursuing licensure for candidate filovirus vaccines via the Animal Rule and has convened meetings of key opinion leaders and subject matter experts to define fundamental components for vaccine licensure in the absence of human efficacy data. Here, filoviruses are used as examples to review immune correlates of protection and immunobridging. The points presented herein reflect the presentations and discussions during the second meeting held in October 2021 and are intended to address important considerations for developing immunobridging strategies.

2.
Vaccines (Basel) ; 10(3)2022 Feb 26.
Article in English | MEDLINE | ID: mdl-35335000

ABSTRACT

Clinical vaccine development and regulatory approval generally occurs in a linear, sequential manner: Phase 1: safety, immunogenicity; Phase 2: immunogenicity, safety, dose ranging, and preliminary efficacy; Phase 3: definitive efficacy, safety, lot consistency; and following regulatory approval, Phase 4: post-marketing safety and effectiveness. For candidate filovirus vaccines, where correlates of protection have not been identified, and phase 2 and 3 efficacy of disease prevention trials untenable, large and/or protracted, each trial may span decades, with full licensure expected only after several decades of development. Given the urgent unmet need for new Marburg virus and Ebola Sudan virus vaccines, the Sabin Vaccine Institute hosted a key stakeholder virtual meeting in May 2021 to explore the possibility of licensure by use of an "animal rule-like" licensure process, based on a risk/benefit assessment specific to regional needs and informed by epidemiology. This may be appropriate for diseases where there are no or limited treatment options, and those prone to sporadic outbreaks with high rates of transmission, morbidity, and mortality. The discussion focused on two contexts: licensure within the Ugandan regulatory environment, a high burden country where Ebola vaccine trials are ongoing, and licensure by the United States FDA-a well-resourced regulatory agency.

3.
Viruses ; 13(9)2021 08 27.
Article in English | MEDLINE | ID: mdl-34578289

ABSTRACT

Influenza hemagglutinin (HA) is considered a major protective antigen of seasonal influenza vaccine but antigenic drift of HA necessitates annual immunizations using new circulating HA versions. Low variation found within conserved non-HA influenza virus (INFV) antigens may maintain protection with less frequent immunizations. Conserved antigens of influenza A virus (INFV A) that can generate cross protection against multiple INFV strains were evaluated in BALB/c mice using modified Vaccinia virus Ankara (MVA)-vectored vaccines that expressed INFV A antigens hemagglutinin (HA), matrix protein 1 (M1), nucleoprotein (NP), matrix protein 2 (M2), repeats of the external portion of M2 (M2e) or as tandem repeats (METR), and M2e with transmembrane region and cytoplasmic loop (M2eTML). Protection by combinations of non-HA antigens was equivalent to that of subtype-matched HA. Combinations of NP and forms of M2e generated serum antibody responses and protected mice against lethal INFV A challenge using PR8, pandemic H1N1 A/Mexico/4108/2009 (pH1N1) or H5N1 A/Vietnam/1203/2004 (H5N1) viruses, as demonstrated by reduced lung viral burden and protection against weight loss. The highest levels of protection were obtained with NP and M2e antigens delivered as MVA inserts, resulting in broadly protective immunity in mice and enhancement of previous natural immunity to INFV A.


Subject(s)
Antibodies, Viral/blood , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Nucleocapsid Proteins/immunology , Orthomyxoviridae Infections/prevention & control , Viral Matrix Proteins/immunology , Viroporin Proteins/immunology , Animals , Antigens, Viral/immunology , Cross Protection , Female , Genetic Vectors , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza Vaccines/administration & dosage , Mice, Inbred BALB C , Nucleocapsid Proteins/administration & dosage , Orthomyxoviridae Infections/immunology , Pandemics , Vaccination , Viral Matrix Proteins/administration & dosage , Viral Matrix Proteins/genetics , Viroporin Proteins/administration & dosage
4.
Vaccine ; 39(1): 1-5, 2021 01 03.
Article in English | MEDLINE | ID: mdl-33199078

ABSTRACT

The anthrax vaccine candidate AV7909 is being developed as a next-generation vaccine for post-exposure prophylaxis (PEP) against inhalational anthrax. In clinical studies, two vaccinations with AV7909 administered either two or four weeks apart induced an enhanced immune response compared to BioThrax® (Anthrax Vaccine Adsorbed) (AVA). Anthrax toxin-neutralizing antibody (TNA) levels on Day 70 following initial vaccination that were associated with protection of animals exposed to inhalational anthrax were previously reported for the 0, 4-week AV7909 vaccination regimen. The current study shows that a 0, 2-week AV7909 vaccination regimen protected guinea pigs (GPs) and nonhuman primates (NHPs) against a lethal inhalational anthrax challenge on Days 28 and 70 after the first immunization. An earlier induction of protective TNA levels using a 0, 2-week AV7909 vaccination regimen may provide benefit over the currently approved AVA PEP 0, 2, and 4-week vaccination regimen.


Subject(s)
Anthrax Vaccines , Anthrax , Bacillus anthracis , Animals , Anthrax/prevention & control , Antibodies, Bacterial , Antibodies, Neutralizing , Antigens, Bacterial , Guinea Pigs , Post-Exposure Prophylaxis , Primates
5.
Vaccine ; 38(10): 2307-2314, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32029323

ABSTRACT

A next-generation anthrax vaccine candidate, AV7909, is being developed for post-exposure prophylaxis (PEP) of inhalational anthrax in combination with the recommended course of antimicrobial therapy. Clinical efficacy studies of anthrax countermeasures in humans are not ethical or feasible, therefore, licensure of AV7909 for PEP is being pursued under the US Food and Drug Administration (FDA) Animal Rule, which requires that evidence of effectiveness be demonstrated in an animal model of anthrax, where results of studies in such a model can establish reasonable likelihood of AV7909 to produce clinical benefit in humans. Initial development of a PEP model for inhalational anthrax included evaluation of post-exposure ciprofloxacin pharmacokinetics (PK), tolerability and survival in guinea pigs treated with various ciprofloxacin dosing regimens. Three times per day (TID) intraperitoneal (IP) dosing with 7.5 mg/kg of ciprofloxacin initiated 1 day following inhalational anthrax challenge and continued for 14 days was identified as a well tolerated partially curative ciprofloxacin treatment regimen. The added benefit of AV7909 vaccination was evaluated in guinea pigs given the partially curative ciprofloxacin treatment regimen. Groups of ciprofloxacin-treated guinea pigs were vaccinated. 1 and 8 days post-challenge with serial dilutions of AV7909, a 1:16 dilution of AVA, or normal saline. A group of untreated guinea pigs was included as a positive control to confirm lethal B. anthracis exposure. Post-exposure vaccination with the AV7909 anthrax vaccine candidate administered in combination with the partially curative ciprofloxacin treatment significantly increased survival of guinea pigs compared to ciprofloxacin treatment alone. These results suggest that the developed model can be useful in demonstrating added value of the vaccine for PEP.


Subject(s)
Anthrax Vaccines/administration & dosage , Anthrax , Disease Models, Animal , Post-Exposure Prophylaxis , Respiratory Tract Infections , Animals , Anthrax/prevention & control , Anti-Bacterial Agents/pharmacokinetics , Ciprofloxacin/pharmacokinetics , Guinea Pigs , Respiratory Tract Infections/prevention & control
6.
Int J Toxicol ; 38(3): 163-172, 2019.
Article in English | MEDLINE | ID: mdl-31179828

ABSTRACT

A recombinant protective antigen (rPA) anthrax vaccine candidate (rPA7909) was developed as a next-generation vaccine indicated for postexposure prophylaxis of disease resulting from suspected or confirmed Bacillus anthracis exposure. The lyophilized form of rPA7909-vaccinated candidate contains 75 µg purified rPA, 750 µg aluminum (as Alhydrogel adjuvant), and 250 µg of an immunostimulatory Toll-like receptor 9 agonist oligodeoxynucleotide CpG 7909 in a 0.5 mL phosphate-buffered suspension. General toxicity and local reactogenicity were evaluated in Sprague Dawley rats vaccinated with the full human dose of rPA7909 by intramuscular injection. Animals were immunized on study days 1, 15, and 29. Control groups were administered diluent only or adjuvant control (excipients, CpG 7909, and Alhydrogel adjuvant in diluent) intramuscularly at the same dose volume and according to the same schedule used for rPA7909. Toxicity was assessed based on the results of clinical observations, physical examinations, body weights, injection site reactogenicity, ophthalmology, clinical pathology (hematology, coagulation, and serum chemistry), organ weights, and macroscopic and microscopic pathology evaluation. The immune response to rPA7909 vaccination was confirmed by measuring serum anti-PA immunoglobulin G levels. The rPA7909 vaccine produced no apparent systemic toxicity and only transient reactogenicity at the injection site. The injection site reaction from animals receiving the adjuvant control was very similar to those receiving rPA7909 with respect to the inflammation. The inflammatory response observed in the injection site and the draining lymph nodes was consistent with expected immune stimulation. The overall results indicated a favorable safety profile for rPA7909.


Subject(s)
Adjuvants, Immunologic/toxicity , Anthrax Vaccines/toxicity , Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Oligodeoxyribonucleotides/toxicity , Animals , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Female , Freeze Drying , Immunoglobulin G/blood , Male , Rats, Sprague-Dawley , Recombinant Proteins/toxicity
7.
Chem Biol Interact ; 296: 34-42, 2018 Dec 25.
Article in English | MEDLINE | ID: mdl-30217478

ABSTRACT

The efficacy and pharmacokinetics of the aqueous co-formulation contents of the Trobigard™ (atropine sulfate, obidoxime chloride) auto-injector were evaluated in a sarin exposed guinea pig model. Two subcutaneous (sc) sarin challenge doses were evaluated in guinea pigs instrumented with brain and heart electrodes for electroencephalogram (EEG) and electrocardiogram (ECG). Sarin challenge doses were chosen to reflect exposure subclasses with sublethal (moderate to severe clinical signs) and lethal consequences. The level of protection of intramuscular human equivalent doses of the co-formulation was defined by (1) the mitigation of signs and symptoms at a sublethal level and (2) the increase of survival time at the supralethal sarin dose levels. Pharmacokinetics of both atropine sulfate and obidoxime were proportional at 1 and 3 human equivalent doses, and only a small increase in heart rate was observed briefly as a side effect. At both sarin challenge doses, 54 µg/kg and 84 µg/kg, the co-formulation treatment was effective against sarin-induced effects. Survival rates were improved at both sarin challenge levels, whereas clinical signs and changes in EEG activity could not in all cases be effectively mitigated, in particular at the supralethal sarin challenge dose level. Reactivation of sarin inhibited cholinesterase was observed in blood, and higher brain cholinesterase activity levels were associated with a better clinical condition of the co-formulation treated animals. Although the results cannot be directly extrapolated to the human situation, pharmacokinetics and the effects over time related to plasma levels of therapeutics in a freely moving guinea pig could aid translational models and possibly improve prediction of efficacy in humans.


Subject(s)
Atropine/pharmacology , Obidoxime Chloride/pharmacology , Sarin/antagonists & inhibitors , Animals , Atropine/administration & dosage , Atropine/chemistry , Atropine/pharmacokinetics , Cholinesterase Reactivators/administration & dosage , Cholinesterase Reactivators/chemistry , Cholinesterase Reactivators/pharmacokinetics , Cholinesterase Reactivators/pharmacology , Cholinesterases/metabolism , Dose-Response Relationship, Drug , Drug Compounding , Electroencephalography , Guinea Pigs , Injections, Subcutaneous , Male , Obidoxime Chloride/administration & dosage , Obidoxime Chloride/chemistry , Obidoxime Chloride/pharmacokinetics , Sarin/pharmacology , Structure-Activity Relationship , Survival Rate
8.
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
9.
Expert Rev Vaccines ; 15(12): 1467-1479, 2016 12.
Article in English | MEDLINE | ID: mdl-27792416

ABSTRACT

INTRODUCTION: The availability of a licensed anthrax vaccine that is safe, effective, and easy to administer for both pre- and post-exposure prophylaxis is critical to successfully manage and prevent potential anthrax attacks. BioThrax® (Anthrax Vaccine Adsorbed; AVA) is the only licensed anthrax vaccine in the US. Areas covered: Recent licensed improvements to BioThrax vaccine for pre-exposure prophylaxis (PrEP) have included an intramuscular (IM) five-dose schedule (in 2008) and a three-dose IM primary series at 0, 1 and 6 months (in 2012). Post-exposure prophylaxis (PEP) - three doses given subcutaneously (SC) at 0, 2, and 4 weeks - was licensed in 2015. We review the anthrax disease and vaccine literature that supported these licensure efforts. Expert commentary: This PEP licensure is the first time the FDA's Animal Rule has been used to license a vaccine. Additional improvements such as fewer vaccine doses and reduced time to protection are desirable for a PEP vaccine and are being pursued with next generation vaccine candidates.


Subject(s)
Anthrax Vaccines/administration & dosage , Anthrax Vaccines/immunology , Anthrax/prevention & control , Drug Approval , Post-Exposure Prophylaxis/methods , Animals , Humans , Immunization Schedule , Injections, Intramuscular , Injections, Subcutaneous , United States , United States Food and Drug Administration
10.
Antimicrob Agents Chemother ; 58(7): 3618-25, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24733473

ABSTRACT

Anthrax is an acute infectious disease caused by the spore-forming bacterium Bacillus anthracis. Timely administration of antibiotics approved for the treatment of anthrax disease may prevent associated morbidity and mortality. However, any delay in initiating antimicrobial therapy may result in increased mortality, as inhalational anthrax progresses rapidly to the toxemic phase of disease. An anthrax antitoxin, AVP-21D9, also known as Thravixa (fully human anthrax monoclonal antibody), is being developed as a therapeutic agent against anthrax toxemia. The efficacy of AVP-21D9 in B. anthracis-infected New Zealand White rabbits and in cynomolgus macaques was evaluated, and its safety and pharmacokinetics were assessed in healthy human volunteers. The estimated mean elimination half-life values of AVP-21D9 in surviving anthrax-challenged rabbits and nonhuman primates (NHPs) ranged from approximately 2 to 4 days and 6 to 11 days, respectively. In healthy humans, the mean elimination half-life was in the range of 20 to 27 days. Dose proportionality was observed for the maximum serum concentration (Cmax) of AVP-21D9 and the area under the concentration-time curve (AUC). In therapeutic efficacy animal models, treatment with AVP-21D9 resulted in survival of up to 92% of the rabbits and up to 67% of the macaques. Single infusions of AVP-21D9 were well tolerated in healthy adult volunteers across all doses evaluated, and no serious adverse events were reported. (This study has been registered at ClinicalTrials.gov under registration no. NCT01202695.).


Subject(s)
Anthrax/drug therapy , Anthrax/immunology , Anti-Bacterial Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Adolescent , Adult , Animals , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/adverse effects , Antibodies, Neutralizing/pharmacology , Antigens, Bacterial/blood , Bacteremia/blood , Bacteremia/drug therapy , Broadly Neutralizing Antibodies , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Female , Humans , Macaca fascicularis , Male , Middle Aged , Rabbits , Young Adult
11.
Antimicrob Agents Chemother ; 57(11): 5684-92, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23979731

ABSTRACT

Bacillus anthracis toxins can be neutralized by antibodies against protective antigen (PA), a component of anthrax toxins. Anthrivig (human anthrax immunoglobulin), also known as AIGIV, derived from plasma of humans immunized with BioThrax (anthrax vaccine adsorbed), is under development for the treatment of toxemia following exposure to anthrax spores. The pharmacokinetics (PK) of AIGIV was assessed in naive animals and healthy human volunteers, and the efficacy of AIGIV was assessed in animals exposed via inhalation to aerosolized B. anthracis spores. In the clinical study, safety, tolerability, and PK were evaluated in three dose cohorts (3.5, 7.1, and 14.2 mg/kg of body weight of anti-PA IgG) with 30 volunteers per cohort. The elimination half-life of AIGIV in rabbits, nonhuman primates (NHPs), and humans following intravenous infusion was estimated to be approximately 4, 12, and 24 days, respectively, and dose proportionality was observed. In a time-based treatment study, AIGIV protected 89 to 100% of animals when administered 12 h postexposure; however, a lower survival rate of 39% was observed when animals were treated 24 h postexposure, underscoring the need for early intervention. In a separate set of studies, animals were treated on an individual basis upon detection of a clinical sign or biomarker of disease, namely, a significant increase in body temperature (SIBT) in rabbits and presence of PA in the serum of NHPs. In these trigger-based intervention studies, AIGIV induced up to 75% survival in rabbits depending on the dose and severity of toxemia at the time of treatment. In NHPs, up to 33% survival was observed in AIGIV-treated animals. (The clinical study has been registered at ClinicalTrials.gov under registration no. NCT00845650.).


Subject(s)
Anthrax Vaccines/administration & dosage , Anthrax/prevention & control , Antibodies, Bacterial/administration & dosage , Bacillus anthracis/drug effects , Immunoglobulins, Intravenous/pharmacokinetics , Respiratory Tract Infections/prevention & control , Spores, Bacterial/drug effects , Animals , Anthrax/immunology , Anthrax/microbiology , Anthrax/mortality , Anthrax Vaccines/immunology , Antibodies, Bacterial/immunology , Antibodies, Bacterial/isolation & purification , Antigens, Bacterial/blood , Antigens, Bacterial/immunology , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacterial Toxins/blood , Bacterial Toxins/immunology , Biomarkers/analysis , Double-Blind Method , Female , Half-Life , Humans , Immunoglobulins, Intravenous/immunology , Immunoglobulins, Intravenous/isolation & purification , Infusions, Intravenous , Macaca fascicularis , Male , Rabbits , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/mortality , Spores, Bacterial/immunology , Spores, Bacterial/pathogenicity , Survival Analysis , Time Factors , Vaccination
12.
Antimicrob Agents Chemother ; 57(11): 5693-6, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23979740

ABSTRACT

Development of anthrax countermeasures that may be used concomitantly in a postexposure setting requires an understanding of the interaction between these products. Anthrax immune globulin intravenous (AIGIV) is a candidate immunotherapeutic that contains neutralizing antibodies against protective antigen (PA), a component of anthrax toxins. We evaluated the interaction between AIGIV and BioThrax (anthrax vaccine adsorbed) in rabbits. While pharmacokinetics of AIGIV were not altered by vaccination, the vaccine-induced immune response was abrogated in AIGIV-treated animals.


Subject(s)
Anthrax Vaccines/administration & dosage , Antibodies, Bacterial/administration & dosage , Immunoglobulins, Intravenous/pharmacokinetics , Animals , Anthrax/immunology , Anthrax/microbiology , Anthrax/prevention & control , Anthrax Vaccines/immunology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Area Under Curve , Bacillus anthracis/immunology , Drug Antagonism , Female , Half-Life , Humans , Immunoglobulins, Intravenous/blood , Immunoglobulins, Intravenous/immunology , Infusions, Intravenous , Male , Rabbits , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/prevention & control , Vaccination
13.
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
14.
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
15.
Virology ; 406(1): 65-79, 2010 Oct 10.
Article in English | MEDLINE | ID: mdl-20667570

ABSTRACT

The HPIV2 V protein inhibits type I interferon (IFN) induction and signaling. To manipulate the V protein, whose coding sequence overlaps that of the polymerase-associated phosphoprotein (P), without altering the P protein, we generated an HPIV2 virus in which P and V are expressed from separate genes (rHPIV2-P+V). rHPIV2-P+V replicated like HPIV2-WT in vitro and in non-human primates. HPIV2-P+V was modified by introducing two separate mutations into the V protein to create rHPIV2-L101E/L102E and rHPIV2-Delta122-127. In contrast to HPIV2-WT, both mutant viruses were unable to degrade STAT2, leaving virus-infected cells susceptible to IFN. Neither mutant, nor HPIV2-WT, induced significant amounts of IFN-beta in infected cells. Surprisingly, neither rHPIV2-L101E/L102E nor rHPIV2-Delta122-127 was attenuated in two species of non-human primates. This indicates that loss of HPIV2's ability to inhibit IFN signaling is insufficient to attenuate virus replication in vivo as long as IFN induction is still inhibited.


Subject(s)
Interferons/physiology , Parainfluenza Virus 2, Human/genetics , Primates/virology , Viral Proteins/genetics , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , DNA, Viral/genetics , DNA-Binding Proteins/metabolism , Genes, Viral , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Interferon Type I/pharmacology , Macaca mulatta , Mutation , Open Reading Frames , Parainfluenza Virus 2, Human/immunology , Parainfluenza Virus 2, Human/pathogenicity , Parainfluenza Virus 2, Human/physiology , Phosphoproteins/genetics , Primates/immunology , Recombinant Proteins , Signal Transduction , Species Specificity , Vero Cells , Viral Proteins/immunology , Virus Replication/drug effects , Virus Replication/genetics , Virus Replication/immunology
16.
Vaccine ; 28(3): 767-79, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-19857454

ABSTRACT

A novel recombinant human parainfluenza virus type 1 (rHPIV1), rHPIV1-C+P, in which the overlapping open reading frames of the C and P genes were separated in order to introduce mutations into the C gene without affecting P, was generated. Infectious rHPIV1-C+P was readily recovered and replicated as efficiently as HPIV1 wild type (wt) in vitro and in African green monkeys (AGMs). rHPIV1-C+P expressed increased levels of C protein and, surprisingly, activated the type I IFN and apoptosis responses more strongly than HPIV1 wt. rHPIV1-C+P provided a useful backbone for recovering an attenuated P/C gene mutation (Delta 84-85), which was previously unrecoverable, likely due to detrimental effects of the deletion on the P protein. rHPIV1-C(Delta 84-85)+P and an additional mutant, rHPIV1-C(Delta 169-170)+P, were found to replicate to similar titers in vitro and to activate the type I IFN and apoptosis responses to a similar degree as rHPIV1-C+P. rHPIV1-C(Delta 84-85)+P was found to be highly attenuated in AGMs, and all viruses were immunogenic and effective in protecting AGMs against challenge with HPIV1 wt. rHPIV1-C(Delta 84-85)+P will be investigated as a potential live-attenuated vaccine candidate for HPIV1.


Subject(s)
Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 1, Human/pathogenicity , Phosphoproteins/genetics , Sequence Deletion , Viral Proteins/genetics , Animals , Antibodies, Viral/blood , Apoptosis , Base Sequence , Cell Line , Chlorocebus aethiops , Humans , Interferon Type I/biosynthesis , Molecular Sequence Data , Parainfluenza Vaccines/genetics , Parainfluenza Virus 1, Human/genetics , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Load , Virulence , Virus Replication
17.
PLoS Biol ; 7(7): e1000155, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19621064

ABSTRACT

Dysfunction of CFTR in cystic fibrosis (CF) airway epithelium perturbs the normal regulation of ion transport, leading to a reduced volume of airway surface liquid (ASL), mucus dehydration, decreased mucus transport, and mucus plugging of the airways. CFTR is normally expressed in ciliated epithelial cells of the surface and submucosal gland ductal epithelium and submucosal gland acinar cells. Critical questions for the development of gene transfer strategies for CF airway disease are what airway regions require CFTR function and how many epithelial cells require CFTR expression to restore normal ASL volume regulation and mucus transport to CF airway epithelium? An in vitro model of human CF ciliated surface airway epithelium (CF HAE) was used to test whether a human parainfluenza virus (PIV) vector engineered to express CFTR (PIVCFTR) could deliver sufficient CFTR to CF HAE to restore mucus transport, thus correcting the CF phenotype. PIVCFTR delivered CFTR to >60% of airway surface epithelial cells and expressed CFTR protein in CF HAE approximately 100-fold over endogenous levels in non-CF HAE. This efficiency of CFTR delivery fully corrected the basic bioelectric defects of Cl(-) and Na(+) epithelial ion transport and restored ASL volume regulation and mucus transport to levels approaching those of non-CF HAE. To determine the numbers of CF HAE surface epithelial cells required to express CFTR for restoration of mucus transport to normal levels, different amounts of PIVCFTR were used to express CFTR in 3%-65% of the surface epithelial cells of CF HAE and correlated to increasing ASL volumes and mucus transport rates. These data demonstrate for the first time, to our knowledge, that restoration of normal mucus transport rates in CF HAE was achieved after CFTR delivery to 25% of surface epithelial cells. In vivo experimentation in appropriate models will be required to determine what level of mucus transport will afford clinical benefit to CF patients, but we predict that a future goal for corrective gene transfer to the CF human airways in vivo would attempt to target at least 25% of surface epithelial cells to achieve mucus transport rates comparable to those in non-CF airways.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Cystic Fibrosis/metabolism , Epithelial Cells/metabolism , Mucus/metabolism , Respiratory Mucosa/metabolism , Analysis of Variance , Biological Transport/physiology , Cells, Cultured , Chlorides/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Gene Expression , Gene Transfer Techniques , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Microscopy, Fluorescence , Parainfluenza Virus 1, Human/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Respiratory Mucosa/pathology , Reverse Transcriptase Polymerase Chain Reaction , Sodium/metabolism
18.
J Virol ; 82(16): 8059-70, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18524813

ABSTRACT

Human parainfluenza virus type 1 (HPIV1) is a significant cause of pediatric respiratory disease in the upper and lower airways. An in vitro model of human ciliated airway epithelium (HAE), a useful tool for studying respiratory virus-host interactions, was used in this study to show that HPIV1 selectively infects ciliated cells within the HAE and that progeny virus is released from the apical surface with little apparent gross cytopathology. In HAE, type I interferon (IFN) is induced following infection with an HPIV1 mutant expressing defective C proteins with an F170S amino acid substitution, rHPIV1-C(F170S), but not following infection with wild-type HPIV1. IFN induction coincided with a 100- to 1,000-fold reduction in virus titer, supporting the hypothesis that the HPIV1 C proteins are critical for the inhibition of the innate immune response. Two recently characterized live attenuated HPIV1 vaccine candidates expressing mutant C proteins were also evaluated in HAE. The vaccine candidates, rHPIV1-C(R84G/Delta170)HN(T553A)L(Y942A) and rHPIV1-C(R84G/Delta170)HN(T553A)L(Delta1710-11), which contain temperature-sensitive (ts) attenuating (att) and non-ts att mutations, were highly restricted in growth in HAE at permissive (32 degrees C) and restrictive (37 degrees C) temperatures. The viruses grew slightly better at 37 degrees C than at 32 degrees C, and rHPIV1-C(R84G/Delta170)HN(T553A)L(Y942A) was less attenuated than rHPIV1-C(R84G/Delta170)HN(T553A)L(Delta1710-11). The level of replication in HAE correlated with that previously observed for African green monkeys, suggesting that the HAE model has potential as a tool for the preclinical evaluation of HPIV1 vaccines, although how these in vitro data will correlate with vaccine virus replication in seronegative human subjects remains to be seen.


Subject(s)
Bronchi/virology , Cilia/virology , Interferons/metabolism , Mutation , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/metabolism , Trachea/virology , Bronchi/metabolism , Humans , Interferon-alpha/metabolism , Interferon-beta/metabolism , Microscopy, Confocal , Models, Biological , Phenotype , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Temperature , Trachea/metabolism
19.
Vaccine ; 25(34): 6409-22, 2007 Aug 21.
Article in English | MEDLINE | ID: mdl-17658669

ABSTRACT

Previously, we identified several attenuating mutations in the L polymerase protein of human parainfluenza virus type 2 (HPIV2) and genetically stabilized those mutations using reverse genetics [Nolan SM, Surman S, Amaro-Carambot E, Collins PL, Murphy BR, Skiadopoulos MH. Live-attenuated intranasal parainfluenza virus type 2 vaccine candidates developed by reverse genetics containing L polymerase protein mutations imported from heterologous paramyxoviruses. Vaccine 2005;39(23):4765-74]. Here we describe the discovery of an attenuating mutation at nucleotide 15 (15(T-->C)) in the 3' genomic promoter that was also present in the previously characterized mutants. We evaluated the properties of this promoter mutation alone and in various combinations with the L polymerase mutations. Amino acid substitutions at L protein positions 460 (460A or 460P) or 948 (948L), or deletion of amino acids 1724 and 1725 (Delta1724), each conferred a temperature sensitivity (ts) phenotype whereas the 15(T-->C) mutation did not. The 460A and 948L mutations each contributed to restricted replication in the lower respiratory tract of African green monkeys, but the Delta1724 mutation increased attenuation only in certain combinations with other mutations. We constructed two highly attenuated viruses, rV94(15C)/460A/948L and rV94(15C)/948L/Delta1724, that were immunogenic and protective against challenge with wild-type HPIV2 in African green monkeys and, therefore, appear to be suitable for evaluation in humans.


Subject(s)
Mutation , Parainfluenza Vaccines/immunology , Parainfluenza Virus 2, Human/immunology , Promoter Regions, Genetic , Vaccines, Synthetic/immunology , Viral Proteins/genetics , Animals , Cell Line , Cricetinae , Humans , Macaca mulatta , Mesocricetus , Parainfluenza Virus 2, Human/genetics , Parainfluenza Virus 2, Human/physiology , Respiratory System/virology , Temperature , Vaccines, Attenuated/immunology , Virus Replication
20.
Virol J ; 4: 67, 2007 Jul 02.
Article in English | MEDLINE | ID: mdl-17605811

ABSTRACT

BACKGROUND: Two recombinant, live attenuated human parainfluenza virus type 1 (rHPIV1) mutant viruses have been developed, using a reverse genetics system, for evaluation as potential intranasal vaccine candidates. These rHPIV1 vaccine candidates have two non-temperature sensitive (non-ts) attenuating (att) mutations primarily in the P/C gene, namely CR84GHNT553A (two point mutations used together as a set) and CDelta170 (a short deletion mutation), and two ts att mutations in the L gene, namely LY942A (a point mutation), and LDelta1710-11 (a short deletion), the last of which has not been previously described. The latter three mutations were specifically designed for increased genetic and phenotypic stability. These mutations were evaluated on the HPIV1 backbone, both individually and in combination, for attenuation, immunogenicity, and protective efficacy in African green monkeys (AGMs). RESULTS: The rHPIV1 mutant bearing the novel LDelta1710-11 mutation was highly ts and attenuated in AGMs and was immunogenic and efficacious against HPIV1 wt challenge. The rHPIV1-CR84G/Delta170HNT553ALY942A and rHPIV1-CR84G/Delta170HNT553ALDelta1710-11 vaccine candidates were highly ts, with shut-off temperatures of 38 degrees C and 35 degrees C, respectively, and were highly attenuated in AGMs. Immunization with rHPIV1-CR84G/Delta170HNT553ALY942A protected against HPIV1 wt challenge in both the upper and lower respiratory tracts. In contrast, rHPIV1-CR84G/Delta170HNT553ALDelta1710-11 was not protective in AGMs due to over-attenuation, but it is expected to replicate more efficiently and be more immunogenic in the natural human host. CONCLUSION: The rHPIV1-CR84G/Delta170HNT553ALY942A and rHPIV1-CR84G/Delta170HNT553ALDelta1710-11 vaccine candidates are clearly highly attenuated in AGMs and clinical trials are planned to address safety and immunogenicity in humans.


Subject(s)
Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/immunology , Vaccines, DNA/immunology , Viral Proteins/genetics , Administration, Intranasal , Animals , Attachment Sites, Microbiological/genetics , Base Sequence , Cell Line , Chlorocebus aethiops , Humans , Molecular Sequence Data , Mutation , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/genetics , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/physiology , Phosphoproteins/genetics , Phosphoproteins/immunology , Respirovirus Infections/immunology , Respirovirus Infections/prevention & control , Respirovirus Infections/virology , Treatment Outcome , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vero Cells , Viral Proteins/immunology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...