Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Mil Med ; 188(3-4): e463-e467, 2023 03 20.
Article in English | MEDLINE | ID: mdl-34179992

ABSTRACT

INTRODUCTION: Staphylococcus aureus (SA) is a major human bacterial pathogen increasingly refractory to antibiotics. Given the dearth of novel antibiotics in the developmental pipeline, we require concerted efforts at optimizing novel antimicrobial approaches. One promising option is the utilization of bacteriophage (phage) therapy, which has been resurrected as a viable clinical therapeutic. Specifically, an expanded library of phages targeting SA is desired. We surmised that SA-targeting phages would be readily accessible as a major component of the cutaneous microbiome. Specifically, we sought to discern if easily accessible (convenient) and discrete anatomic locations, including the nares, axilla, fingernails, toenails, and web spaces, could provide intact phages via a noninvasive, expedient procedure involving swabbing. METHODS: One hundred subjects participated in systematic skin swab specimen collections. Pooled samples were subject to phage harvesting utilizing the soft agar overlay technique. The approval was secured from the Naval Medical Research Center Institutional Review Board (NMRC 2018.0004 FWA00000152). We utilized the same procedures from known samples containing SA-targeting phages. As another positive control, we employed the same swab and acquired samples from an active wound infection. RESULTS: As anticipated, there were no adverse events, and the procedure was successfully implemented within the projected 10-minute duration. No phages were identified exploiting this methodology. Positive controls from various environmental samples identified SA-targeting phages as did the wound effluent sample. CONCLUSIONS: Skin swabbing at multiple anatomic sites from 100 adults yielded insufficient biomass for phage recovery. The negative results provide helpful information for future phage isolation attempts. The lessons learned on why this study failed to isolate phages can be easily utilized by others. With a desire to increase our SA-targeting phage library in pursuit of future clinical trials, and acknowledging the paucity of these phages accessible via traditional recovery from environmental sources, we will next acquire large volumes of wound effluent from confirmed infected wounds with SA to optimize the biomass for phage recovery.


Subject(s)
Bacteriophages , Staphylococcal Infections , Adult , Humans , Staphylococcus aureus , Staphylococcal Infections/therapy , Anti-Bacterial Agents , Staphylococcus Phages
2.
J Infect Dis ; 226(4): 655-663, 2022 09 04.
Article in English | MEDLINE | ID: mdl-35106573

ABSTRACT

Passive antibody immunotherapeutics directed against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are promising countermeasures for protection and treatment of coronavirus disease 2019 (COVID-19). SARS-CoV-2 variants of concern (VOCs) and variants of interest (VOIs) can impact the clinical efficacy of immunotherapeutics. A fully human polyclonal antibody immunotherapeutic purified from plasma of transchromosomic (Tc) bovines hyperimmunized with SARS-CoV-2 WA-1 spike (SAB-185) is being assessed for efficacy in a phase 2/3 clinical trial when different circulating SARS-CoV-2 variants predominated. We evaluated antibody binding, avidity maturation, and SARS-CoV-2 VOCs/VOIs virus-neutralizing capacity of convalescent plasma compared with different lots of SAB-185 and individual Tc bovine sera sequentially obtained after each vaccination against Alpha, Epsilon, Iota, Gamma, Beta, Kappa, and Delta variants. In contrast to convalescent plasma, sera and SAB-185 derived from hyperimmunized Tc bovines demonstrated higher antibody avidity and more potent cross-neutralizing activity of VOCs/VOIs. Thus, SAB-185 is a potential promising therapeutic candidate for the treatment of patients infected with SARS-CoV-2 variants.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Neutralizing , Antibodies, Viral , Antibody Affinity , COVID-19/therapy , Cattle , Humans , Immunization, Passive , Immunoglobulin G , Neutralization Tests , Spike Glycoprotein, Coronavirus , COVID-19 Serotherapy
3.
Hum Vaccin Immunother ; 18(2): 1940652, 2022 04 29.
Article in English | MEDLINE | ID: mdl-34228597

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with amino-acid substitutions and deletions in spike protein (S) can reduce the effectiveness of monoclonal antibodies (mAbs) and may compromise immunity induced by vaccines. We report a polyclonal, fully human, anti-SARS-CoV-2 immunoglobulin produced in transchromosomic bovines (Tc-hIgG-SARS-CoV-2) hyperimmunized with two doses of plasmid DNA encoding the SARS-CoV-2 Wuhan strain S gene, followed by repeated immunization with S protein purified from insect cells. The resulting Tc-hIgG-SARS-CoV-2, termed SAB-185, efficiently neutralizes SARS-CoV-2, and vesicular stomatitis virus (VSV) SARS-CoV-2 chimeras in vitro. Neutralization potency was retained for S variants including S477N, E484K, and N501Y, substitutions present in recent variants of concern. In contrast to the ease of selection of escape variants with mAbs and convalescent human plasma, we were unable to isolate VSV-SARS-CoV-2 mutants resistant to Tc-hIgG-SARS-CoV-2 neutralization. This fully human immunoglobulin that potently inhibits SARS-CoV-2 infection may provide an effective therapeutic to combat COVID-19.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , Cattle , Humans , Immunoglobulin G , Neutralization Tests/methods , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus
4.
BMC Health Serv Res ; 18(1): 651, 2018 Aug 22.
Article in English | MEDLINE | ID: mdl-30134892

ABSTRACT

BACKGROUND: The hemagglutination-inhibition (HAI) assay is a critical component for measurement of immunogenicity in influenza vaccine development. It is unknown if the results can be influenced by sample type and anticoagulants. The purpose of this study was to evaluate the influence of different sample collection methods, in particular different anticoagulants, and choice of plasma or serum, on influenza virus serological assays. METHODS: Blood samples from thirty donors previously immunized against influenza viruses were collected using six different types of blood collection tubes, two of which collect serum and four of which contain various anticoagulants for collecting plasma. Serum: (1) serum separator tubes (SST); and (2) Plus Plastic serum "red-top serum" tubes. Plasma: (3) spray-coated K2 ethylenediaminetetraacetic acid (EDTA) tubes: (4) Sodium Heparin tubes; (5) Citrate tubes with 3.2% sodium citrate solution; and (6) Glass Blood Collection tubes with acid citrate dextrose. Samples were tested against three different influenza viruses (A/California/07/2009 (H1N1pdm09), A/Texas/50/2012 (H3N2), and B/Massachusetts/2/2012) for hemagglutination inhibition titer and virus neutralization titer via a microneutralization (MN) assay, and data compared to that obtained for standard serum sample collected in SST. RESULTS: HAI and MN titers against type A viruses were within two dilutions compared to SST collection method over 96% of the time irrespective of sample type or anticoagulant. However, HAI titers for type B virus were more variable across different collection methods. EDTA plasma samples were greater than two dilutions higher than SST serum samples 70% (21 of 30 samples) of the time. In contrast, MN titers were within two dilutions over 96% of the time, with the highest deviation noted in acid citrate dextrose plasma samples (3 of 30 samples tested, 10%). CONCLUSIONS: These data provide useful guidelines for sample collection and serology testing when screening: (i) influenza vaccine immunogenicity antibody response; (ii) antibody responses to newly emerging viral strains; and (iii) clinical samples for anti-influenza antibody activity.


Subject(s)
Blood Specimen Collection , Hemagglutination Inhibition Tests , Hemagglutination/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/immunology , Antibodies, Viral , Anticoagulants , Blood Specimen Collection/methods , Guidelines as Topic , Humans , Influenza Vaccines , Influenza, Human/blood , Neutralization Tests
5.
Am J Trop Med Hyg ; 98(3): 849-856, 2018 03.
Article in English | MEDLINE | ID: mdl-29363446

ABSTRACT

We conducted an open label, dose escalation Phase 1 clinical trial of a tetravalent dengue DNA vaccine (TVDV) formulated in Vaxfectin® to assess safety and immunogenicity. A total of 40 dengue- and flavivirus-naive volunteers received either low-dose (1 mg) TVDV alone (N = 10, group 1), low-dose TVDV (1 mg) formulated in Vaxfectin (N = 10, group 2), or high-dose TVDV (2 mg, group 3) formulated in Vaxfectin® (N = 20). Subjects were immunized intramuscularly with three doses on a 0-, 30-, 90-day schedule and monitored. Blood samples were obtained after each immunization and various time points thereafter to assess anti-dengue antibody and interferon gamma (IFNγ) T-cell immune responses. The most common adverse events (AEs) across all groups included mild to moderate pain and tenderness at the injection site, which typically resolved within 7 days. Common solicited signs and symptoms included fatigue (42.5%), headache (45%), and myalgias (47.5%). There were no serious AEs related to the vaccine or study procedures. No anti-dengue antibody responses were detected in group 1 subjects who received all three immunizations. There were minimal enzyme-linked immunosorbent assay and neutralizing antibody responses among groups 2 and 3 subjects who completed the immunization schedule. By contrast, IFNγ T-cell responses, regardless of serotype specificity, occurred in 70%, 50%, and 79% of subjects in groups 1, 2, and 3, respectively. The largest IFNγ T-cell responses were among group 3 subjects. We conclude that TVDV was safe and well-tolerated and elicited predominately anti-dengue T-cell IFNγ responses in a dose-related fashion.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Dengue Vaccines/administration & dosage , Dengue Virus/immunology , Dengue/prevention & control , Immunity, Cellular/drug effects , Vaccines, DNA/administration & dosage , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Adult , Dengue/immunology , Dengue/virology , Dengue Vaccines/adverse effects , Fatigue/etiology , Fatigue/physiopathology , Female , Headache/etiology , Headache/physiopathology , Humans , Immunization Schedule , Immunogenicity, Vaccine , Injections, Intramuscular , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Male , Myalgia/etiology , Myalgia/physiopathology , Patient Safety , Phosphatidylethanolamines/administration & dosage , Phosphatidylethanolamines/chemistry , Vaccination , Vaccines, DNA/adverse effects
6.
J Virol Methods ; 248: 7-18, 2017 10.
Article in English | MEDLINE | ID: mdl-28624584

ABSTRACT

This study describes an antibody-dependent NK cell degranulation assay, as a biomarker to assess antibody-dependent cellular cytotoxicity (ADCC) response in influenza plasma and for antibody therapies against influenza infection. The concentration of neutralizing antibodies (NAbs) against the hemagglutinin receptor of influenza viruses is a current determinant in protection against infection, particularly following receipt of the seasonal influenza vaccine. However, this is a limited assessment of protection, because: (i) NAb titers that incur full protection vary; and (ii) NAb titers do not account for the entire breadth of antibody responses against viral infection. Previous reports have indicated that antibodies that prime ADCC play a vital role in controlling influenza infections, and thus should be quantified for assessing protection against influenza. This report demonstrates a non-radioactive assay that assesses NK cell activation as a marker of ADCC, in which NK cells interact with opsonized viral antigen expressed on the surface of infected Raji target cells resulting in effector cell degranulation (surrogate CD107a expression). A positive correlation was determined between HAI titers and sustained NK cell activation, although NK cell activation was seen in plasma samples with HAI titers below 40 and varied amongst samples with high HAI titers. Furthermore, sustained NK cell degranulation was determined for influenza-vaccinated transchromosomic bovine intravenous immunoglobulin, indicating the potential utility of this therapy for influenza treatment. We conclude that this assay is reproducible and relevant.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Immunoassay , Immunoglobulins, Intravenous/therapeutic use , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/therapy , Killer Cells, Natural/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Biomarkers/blood , Cattle , Cell Degranulation , Cell Line , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunoglobulins, Intravenous/administration & dosage , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza, Human/blood , Influenza, Human/virology , Lymphocyte Activation , Lysosomal-Associated Membrane Protein 1/immunology
7.
Malar J ; 15(1): 377, 2016 Jul 22.
Article in English | MEDLINE | ID: mdl-27448805

ABSTRACT

BACKGROUND: In this phase 1 clinical trial, healthy adult, malaria-naïve subjects were immunized with radiation-attenuated Plasmodium falciparum sporozoites (PfRAS) by mosquito bite and then underwent controlled human malaria infection (CHMI). The PfRAS model for immunization against malaria had previously induced >90 % sterile protection against homologous CHMI. This study was to further explore the safety, tolerability and protective efficacy of the PfRAS model and to provide biological specimens to characterize protective immune responses and identify protective antigens in support of malaria vaccine development. METHODS: Fifty-seven subjects were screened, 41 enrolled and 30 received at least one immunization. The true-immunized subjects received PfRAS via mosquito bite and the mock-immunized subjects received mosquito bites from irradiated uninfected mosquitoes. Sera and peripheral blood mononuclear cells (PBMCs) were collected before and after PfRAS immunizations. RESULTS: Immunization with PfRAS was generally safe and well tolerated, and repeated immunization via mosquito bite did not appear to increase the risk or severity of AEs. Local adverse events (AEs) of true-immunized and mock-immunized groups consisted of erythaema, papules, swelling, and induration and were consistent with reactions from mosquito bites seen in nature. Two subjects, one true- and one mock-immunized, developed large local reactions that completely resolved, were likely a result of mosquito salivary antigens, and were withdrawn from further participation as a safety precaution. Systemic AEs were generally rare and mild, consisting of headache, myalgia, nausea, and low-grade fevers. Two true-immunized subjects experienced fever, malaise, myalgia, nausea, and rigours approximately 16 h after immunization. These symptoms likely resulted from pre-formed antibodies interacting with mosquito salivary antigens. Ten subjects immunized with PfRAS underwent CHMI and five subjects (50 %) were sterilely protected and there was a significant delay to parasitaemia in the other five subjects. All ten subjects developed humoral immune responses to whole sporozoites and to the circumsporozoite protein prior to CHMI, although the differences between protected and non-protected subjects were not statistically significant for this small sample size. CONCLUSIONS: The protective efficacy of this clinical trial (50 %) was notably less than previously reported (>90 %). This may be related to differences in host genetics or the inherent variability in mosquito biting behavior and numbers of sporozoites injected. Differences in trial procedures, such as the use of leukapheresis prior to CHMI and of a longer interval between the final immunization and CHMI in these subjects compared to earlier trials, may also have reduced protective efficacy. This trial has been retrospectively registered at ISRCTN ID 17372582, May 31, 2016.


Subject(s)
Antibodies, Protozoan/blood , Culicidae/physiology , Insect Bites and Stings , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Adolescent , Adult , Animals , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Humans , Malaria Vaccines/administration & dosage , Male , Middle Aged , Plasmodium falciparum/radiation effects , Sporozoites/immunology , Sporozoites/radiation effects , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology , Young Adult
8.
Sci Rep ; 6: 24897, 2016 04 25.
Article in English | MEDLINE | ID: mdl-27109916

ABSTRACT

Polyclonal antibodies, derived from humans or hyperimmunized animals, have been used prophylactically or therapeutically as countermeasures for a variety of infectious diseases. SAB Biotherapeutics has successfully developed a transchromosomic (Tc) bovine platform technology that can produce fully human immunoglobulins rapidly, and in substantial quantities, against a variety of disease targets. In this study, two Tc bovines expressing high levels of fully human IgG were hyperimmunized with a recombinant glycoprotein (GP) vaccine consisting of the 2014 Ebola virus (EBOV) Makona isolate. Serum collected from these hyperimmunized Tc bovines contained high titers of human IgG against EBOV GP as determined by GP specific ELISA, surface plasmon resonance (SPR), and virus neutralization assays. Fully human polyclonal antibodies against EBOV were purified and evaluated in a mouse challenge model using mouse adapted Ebola virus (maEBOV). Intraperitoneal administration of the purified anti-EBOV IgG (100 mg/kg) to BALB/c mice one day after lethal challenge with maEBOV resulted in 90% protection; whereas 100% of the control animals succumbed. The results show that hyperimmunization of Tc bovines with EBOV GP can elicit protective and potent neutralizing fully human IgG antibodies rapidly and in commercially viable quantities.


Subject(s)
Animals, Genetically Modified , Antibodies, Viral/blood , Antibodies, Viral/therapeutic use , Cattle , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunization, Passive , Immunoglobulin G/blood , Immunoglobulin G/therapeutic use , Mice, Inbred BALB C , Neutralization Tests , Surface Plasmon Resonance , Treatment Outcome
9.
Springerplus ; 4: 709, 2015.
Article in English | MEDLINE | ID: mdl-26618098

ABSTRACT

As of September 30, 2015, a total of 1589 laboratory-confirmed cases of infection with the Middle East respiratory syndrome coronavirus (MERS-CoV) have been reported to the World Health Organization (WHO). At present there is no effective specific therapy against MERS-CoV. The use of convalescent plasma (CP) has been suggested as a potential therapy based on existing evidence from other viral infections. We aim to study the feasibility of CP therapy as well as its safety and clinical and laboratory effects in critically ill patients with MERS-CoV infection. We will also examine the pharmacokinetics of the MERS-CoV antibody response and viral load over the course of MERS-CoV infection. This study will inform a future randomized controlled trial that will examine the efficacy of CP therapy for MERS-CoV infection. In the CP collection phase, potential donors will be tested by the enzyme linked immunosorbent assay (ELISA) and the indirect fluorescent antibody (IFA) techniques for the presence of anti-MERS-CoV antibodies. Subjects with anti-MERS-CoV IFA titer of ≥1:160 and no clinical or laboratory evidence of MERS-CoV infection will be screened for eligibility for plasma donation according to standard donation criteria. In the CP therapy phase, 20 consecutive critically ill patients admitted to intensive care unit with laboratory-confirmed MERS-CoV infection will be enrolled and each will receive 2 units of CP. Post enrollment, patients will be followed for clinical and laboratory outcomes that include anti-MERS-CoV antibodies and viral load. This protocol was developed collaboratively by King Abdullah International Medical Research Center (KAIMRC), Gulf Cooperation Council (GCC) Infection Control Center Group and the World Health Organization-International Severe Acute Respiratory and Emerging Infection Consortium (ISARIC-WHO) MERS-CoV Working Group. It was approved in June 2014 by the Ministry of the National Guard Health Affairs Institutional Review Board (IRB). A data safety monitoring board (DSMB) was formulated. The study is registered at http://www.clinicaltrials.gov (NCT02190799).

10.
Vaccine ; 31 Suppl 1: A1-20, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23587330

ABSTRACT

Both seasonal and pandemic influenza cause considerable morbidity and mortality globally. In addition, the ongoing threat of new, unpredictable influenza pandemics from emerging variant strains cannot be underestimated. Recently bioCSL (previously known as CSL Biotherapies) sponsored a symposium 'New Wisdom to Defy an Old Enemy' at the 4th Influenza Vaccines for the World Congress in Valencia, Spain. This symposium brought together a renowned faculty of experts to discuss lessons from past experience, novel influenza vaccine developments, and new methods to increase vaccine acceptance and coverage. Specific topics reviewed and discussed included new vaccine development efforts focused on improving efficacy via alternative administration routes, dose modifications, improved adjuvants, and the use of master donor viruses. Improved safety was also discussed, particularly the new finding of an excess of febrile reactions isolated to children who received the 2010 Southern Hemisphere (SH) trivalent inactivated influenza vaccine (TIV). Significant work has been done to both identify the cause and minimize the risk of febrile reactions in children. Other novel prophylactic and therapeutic advances were discussed including immunotherapy. Standard IVIg and hIVIg have been used in ferret studies and human case reports with promising results. New adjuvants, such as ISCOMATRIX™ adjuvant, were noted to provide single-dose, prolonged protection with seasonal vaccine after lethal H5N1 virus challenge in a ferret model of human influenza disease. The data suggest that adjuvanted seasonal influenza vaccines may provide broader protection than unadjuvanted vaccines. The use of an antigen-formulated vaccine to induce broad protection between pandemics that could bridge the gap between pandemic declaration and the production of a homologous vaccine was also discussed. Finally, despite the availability of effective vaccines, most current efforts to increase influenza vaccine coverage rates to higher levels (i.e., above 70-80%) have been ineffective in highly developed countries where the vaccine is used, hindered by the public's skepticism towards vaccines in general. New educational and social media methods to increase vaccine acceptance and coverage were discussed. While the first priority should be the development of improved influenza vaccines, a particular focus on the aging global population is critical. It is also important to draw lessons from other academic disciplines that can help to inform vaccine education programs, policy, and communication. By tailoring communications and patient education using an understanding of cognitive bias and the model of preferred cognitive styles, the likelihood of effecting desirable health decisions can be maximized, leading to improved vaccine coverage and control of influenza and other vaccine-preventable diseases.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Immunization, Passive/methods , Influenza Vaccines/therapeutic use , Influenza, Human/prevention & control , Adjuvants, Immunologic/pharmacology , Animals , Child , Child, Preschool , Cholesterol/administration & dosage , Congresses as Topic , Disease Models, Animal , Drug Combinations , Ferrets , Humans , Immunoglobulins, Intravenous/therapeutic use , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Influenza A Virus, H5N1 Subtype , Influenza Pandemic, 1918-1919 , Influenza Vaccines/administration & dosage , Influenza Vaccines/adverse effects , Influenza Vaccines/immunology , Influenza, Human/drug therapy , Influenza, Human/epidemiology , Influenza, Human/virology , Orthomyxoviridae Infections/prevention & control , Pandemics , Patient Acceptance of Health Care , Phospholipids/administration & dosage , Saponins/administration & dosage , Spain , Vaccination/psychology , Vaccines, Inactivated/adverse effects
11.
Hum Vaccin Immunother ; 8(11): 1564-84, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23151451

ABSTRACT

When introduced in the 1990s, immunization with DNA plasmids was considered potentially revolutionary for vaccine development, particularly for vaccines intended to induce protective CD8 T cell responses against multiple antigens. We conducted, in 1997-1998, the first clinical trial in healthy humans of a DNA vaccine, a single plasmid encoding Plasmodium falciparum circumsporozoite protein (PfCSP), as an initial step toward developing a multi-antigen malaria vaccine targeting the liver stages of the parasite. As the next step, we conducted in 2000-2001 a clinical trial of a five-plasmid mixture called MuStDO5 encoding pre-erythrocytic antigens PfCSP, PfSSP2/TRAP, PfEXP1, PfLSA1 and PfLSA3. Thirty-two, malaria-naïve, adult volunteers were enrolled sequentially into four cohorts receiving a mixture of 500 µg of each plasmid plus escalating doses (0, 20, 100 or 500 µg) of a sixth plasmid encoding human granulocyte macrophage-colony stimulating factor (hGM-CSF). Three doses of each formulation were administered intramuscularly by needle-less jet injection at 0, 4 and 8 weeks, and each cohort had controlled human malaria infection administered by five mosquito bites 18 d later. The vaccine was safe and well-tolerated, inducing moderate antigen-specific, MHC-restricted T cell interferon-γ responses but no antibodies. Although no volunteers were protected, T cell responses were boosted post malaria challenge. This trial demonstrated the MuStDO5 DNA and hGM-CSF plasmids to be safe and modestly immunogenic for T cell responses. It also laid the foundation for priming with DNA plasmids and boosting with recombinant viruses, an approach known for nearly 15 y to enhance the immunogenicity and protective efficacy of DNA vaccines.


Subject(s)
Antigens, Protozoan/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Malaria Vaccines/immunology , Malaria Vaccines/therapeutic use , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Sporozoites/immunology , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use , Adult , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Malaria Vaccines/administration & dosage , Male , Middle Aged , Plasmids/genetics , Vaccines, DNA/adverse effects , Young Adult
13.
PLoS One ; 7(12): e48229, 2012.
Article in English | MEDLINE | ID: mdl-23284613

ABSTRACT

Influenza-specific hemaggluitination inhibition (HAI) antibody titer, an indicator of immunity to influenza, is often used to measure exposure to influenza in surveillance and immunogenicity studies. Traditionally, serum has been the specimen of choice for HAI assays, but a desire to reduce the amount of blood collected during studies and the availability of plasma in archived sample collections warrant the evaluation of plasma for HAI titer. Therefore, the relationship between serum and plasma HAI titer values is of great interest. Here, we compare HAI titers determined on temporally matched serum and plasma (citrated and heparinized) using influenza A and B viruses. Bland-Altman plots, McNemar's test, and geometric coefficient of variation were used respectively for evaluating agreement, correlation and variability in the serum-plasma titer results. We observed a high degree of agreement (80.5%-98.8%) and correlation (r = 0.796-0.964) in the serum and matched plasma titer values although plasma titers were generally lower than corresponding serum titers. Calculated seropositive (HAI ≥40) rates were higher using serum titers than with plasma titers, but seroconversion rates were unaffected by sample type. Stronger agreement and decreased variability in titers were seen between serum and citrated plasma than between serum and heparinized plasma. Overall, these data suggest that serum or plasma can be used in serodiagnostic HAI assays, but seropositive rates may be underestimated using plasma HAI titers. The type of anticoagulant present in plasma may affect HAI titer values and warrants further investigation.


Subject(s)
Antibodies, Viral/blood , Hemagglutination Inhibition Tests/methods , Plasma/virology , Serum/virology , Antibodies, Viral/immunology , Anticoagulants/pharmacology , Citrates/metabolism , Heparin/metabolism , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Plasma/drug effects , Plasma/metabolism , Serum/drug effects , Sodium Citrate , Time Factors
14.
Crit Care Med ; 38(4 Suppl): e66-73, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20154602

ABSTRACT

The world is experiencing a pandemic of swine-origin influenza virus H1N1. A vaccine to prevent disease is now available, and millions have or will become ill before they can be vaccinated. The ability to use swine-origin influenza virus vaccines as a public health tool has been described as a "race against time." Oseltamivir and related drugs are being used in an effort to reduce morbidity and mortality, but their efficacy for treating severe influenza is suboptimal, and possible wide-spread emergence of oseltamivir-resistant mutants is a concern. Another approach for prevention and treatment of serious influenza is infusion of hyperimmune plasma. The United States has thousands of licensed blood product collection centers that produce millions of liters of plasma licensed by the Food and Drug Administration on an annual basis for the treatment of serious conditions. Immunotherapy using infusion of convalescent plasma (or hyperimmune intravenous immunoglobulin) has been reported to be an effective treatment for severe influenza and other virulent pathogens in animal models and humans. Plasma obtained from those that have recovered or were early recipients of vaccine offers a resource for production of an immediately available and potentially effective therapy at the local, state, and national level. Past, current, and future uses of immunotherapy and current advisory body recommendations for this approach are presented.


Subject(s)
Immune Sera/administration & dosage , Immunization, Passive/methods , Immunoglobulins, Intravenous/therapeutic use , Influenza, Human/therapy , Blood Specimen Collection/standards , Humans , Immune Sera/immunology , Immunization, Passive/standards , Influenza, Human/blood , Influenza, Human/immunology
15.
Am J Disaster Med ; 3(2): 99-107, 2008.
Article in English | MEDLINE | ID: mdl-18522251

ABSTRACT

The H5N1 influenza threat is resulting in global preparations for the next influenza pandemic. Pandemic influenza planners are prioritizing scarce vaccine, antivirals, and public health support for different segments of society. The freight, bulk goods, and energy transportation network comprise the maritime, rail, air, and trucking industries. It relies on small numbers of specialized workers who cannot be rapidly replaced if lost due to death, illness, or voluntary absenteeism. Because transportation networks link economies, provide critical infrastructures with working material, and supply citizens with necessary commodities, disrupted transportation systems can lead to cascading failures in social and economic systems. However, some pandemic influenza plans have assigned transportation workers a low priority for public health support, vaccine, and antivirals. The science of Transportation Geography demonstrates that transportation networks and workers are concentrated at, or funnel through, a small number of chokepoints and corridors. Chokepoints should be used to rapidly and efficiently vaccinate and prophylax the transportation worker cohort and to implement transmission prevention measures and thereby protect the ability to move goods. Nations, states, the transportation industry and unions, businesses, and other stakeholders must plan, resource, and exercise, and then conduct a transportation health assurance and security campaign for an influenza pandemic.


Subject(s)
Disease Outbreaks/prevention & control , Health Planning/organization & administration , Influenza A Virus, H5N1 Subtype , Influenza, Human/epidemiology , Transportation , Antiviral Agents/therapeutic use , Global Health , Humans , Influenza Vaccines/therapeutic use , Influenza, Human/virology , Mass Vaccination/organization & administration
16.
Ann Intern Med ; 145(8): 599-609, 2006 Oct 17.
Article in English | MEDLINE | ID: mdl-16940336

ABSTRACT

BACKGROUND: Studies from the Spanish influenza era reported that transfusion of influenza-convalescent human blood products reduced mortality in patients with influenza complicated by pneumonia. Treatments for H5N1 influenza are unsatisfactory, and convalescent human plasma containing H5N1 antibodies could be an effective therapy during outbreaks and pandemics. PURPOSE: To determine whether transfusion with influenza-convalescent human blood products reduced the risk for death in patients with Spanish influenza pneumonia. DATA SOURCES: Manual search of English-language journals from 1918 to 1925. Citations from retrieved studies were also searched. STUDY SELECTION: Published English-language studies that had at least 10 patients in the treatment group, used convalescent blood products to treat Spanish influenza pneumonia in a hospital setting, and reported on a control or comparison group. DATA EXTRACTION: Two investigators independently extracted data on study characteristics, outcomes, adverse events, and quality. DATA SYNTHESIS: Eight relevant studies involving 1703 patients were found. Treated patients, who were often selected because of more severe illness, were compared with untreated controls with influenza pneumonia in the same hospital or ward. The overall crude case-fatality rate was 16% (54 of 336) among treated patients and 37% (452 of 1219) among controls. The range of absolute risk differences in mortality between the treatment and control groups was 8% to 26% (pooled risk difference, 21% [95% CI, 15% to 27%]). The overall crude case-fatality rate was 19% (28 of 148) among patients who received early treatment (after <4 days of pneumonia complications) and 59% (49 of 83) among patients who received late treatment (after > or =4 days of pneumonia complications). The range of absolute risk differences in mortality between the early treatment group and the late treatment group was 26% to 50% (pooled risk difference, 41% [CI, 29% to 54%]). Adverse effects included chill reactions and possible exacerbations of symptoms in a few patients. LIMITATIONS: Studies were few and had many methodologic limitations. No study was a blinded, randomized, or placebo-controlled trial. Some pertinent studies may have been missed. CONCLUSIONS: Patients with Spanish influenza pneumonia who received influenza-convalescent human blood products may have experienced a clinically important reduction in the risk for death. Convalescent human H5N1 plasma could be an effective, timely, and widely available treatment that should be studied in clinical trials.


Subject(s)
Antibodies, Viral/therapeutic use , Immunization, Passive , Influenza A Virus, H5N1 Subtype , Influenza, Human/therapy , Orthomyxoviridae/immunology , Pneumonia, Viral/therapy , History, 20th Century , Humans , Immunization, Passive/adverse effects , Influenza, Human/history , Influenza, Human/mortality , Plasma/immunology , Pneumonia, Viral/history , Pneumonia, Viral/mortality , Research Design
17.
J Exp Biol ; 206(Pt 21): 3803-8, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14506215

ABSTRACT

Annually, malaria causes >300 million clinical cases and 1 million deaths, is responsible for the loss of >1% of gross domestic product (GDP) in Africa and is a serious concern for travelers. An effective vaccine could have a dramatic impact on the disease. For 20 years, scientists have tried to develop modern, recombinant 'subunit' malaria vaccines. This has been difficult. In fact, there is only one recombinant protein vaccine on the market for any disease, and no vaccines based on synthetic peptides, recombinant viruses, recombinant bacteria or DNA plasmids. Most vaccines are based on attenuated or inactivated whole pathogens or material derived directly from the infectious agent. It is in that context that our recent report summarizing the protection of humans with attenuated Plasmodium falciparum (Pf) sporozoites produced at four different sites over 25 years is important. In studies utilizing live mosquitoes as the vaccine delivery mechanism, there was complete protection against malaria in 93% of volunteers (13/14) and 94% of challenges (33/35). Sanaria's goal is to develop and commercialize a non-replicating, metabolically active Pf sporozoite vaccine. Three practical questions must be addressed before manufacturing for clinical trials: (1) can one administer the vaccine by a route that is clinically practical; (2) can one produce adequate quantities of sporozoites; and (3) can sporozoites be produced with the physical characteristics that meet the regulatory, potency and safety requirements of regulatory authorities? Once these questions have been answered, Sanaria will demonstrate that the vaccine protects >90% of human recipients against experimental challenge with Pf sporozoites, can be produced with an efficiency that makes it economically feasible, and protects >90% of African infants and children from infection, and has severe morbidity and mortality. By producing a vaccine for travelers, Sanaria will provide the infrastructure, regulatory foundation and funds necessary to speed licensure, manufacturing and deployment of the vaccine for the infants and children who need it most.


Subject(s)
Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Sporozoites/immunology , Vaccination/methods , Animals , Plasmodium falciparum/radiation effects , Vaccines, Attenuated/immunology , Vaccines, Attenuated/radiation effects
18.
J Infect Dis ; 185(8): 1155-64, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11930326

ABSTRACT

During 1989-1999, 11 volunteers were immunized by the bites of 1001-2927 irradiated mosquitoes harboring infectious sporozoites of Plasmodium falciparum (Pf) strain NF54 or clone 3D7/NF54. Ten volunteers were first challenged by the bites of Pf-infected mosquitoes 2-9 weeks after the last immunization, and all were protected. A volunteer challenged 10 weeks after the last immunization was not protected. Five previously protected volunteers were rechallenged 23-42 weeks after a secondary immunization, and 4 were protected. Two volunteers were protected when rechallenged with a heterologous Pf strain (7G8). In total, there was protection in 24 of 26 challenges. These results expand published findings demonstrating that immunization by exposure to thousands of mosquitoes carrying radiation-attenuated Pf sporozoites is safe and well tolerated and elicits strain-transcendent protective immunity that persists for at least 42 weeks.


Subject(s)
Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium falciparum/immunology , Adolescent , Adult , Animals , Culicidae/parasitology , Erythrocytes/parasitology , Humans , Immunization/adverse effects , Male , Middle Aged , Plasmodium falciparum/radiation effects , Vaccines, Attenuated/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...