Phase I study of safety and immunogenicity of an Escherichia coli-derived recombinant protective antigen (rPA) vaccine to prevent anthrax in adults

Bruce K Brown, Josephine Cox, Anita Gillis, Thomas C VanCott, Mary Marovich, Mark Milazzo, Tanya Santelli Antonille, Lindsay Wieczorek, Kelly T McKee Jr, Karen Metcalfe, Raburn M Mallory, Deborah Birx, Victoria R Polonis, Merlin L Robb, Bruce K Brown, Josephine Cox, Anita Gillis, Thomas C VanCott, Mary Marovich, Mark Milazzo, Tanya Santelli Antonille, Lindsay Wieczorek, Kelly T McKee Jr, Karen Metcalfe, Raburn M Mallory, Deborah Birx, Victoria R Polonis, Merlin L Robb

Abstract

Background: The fatal disease caused by Bacillus anthracis is preventable with a prophylactic vaccine. The currently available anthrax vaccine requires a lengthy immunization schedule, and simpler and more immunogenic options for protection against anthrax are a priority for development. In this report we describe a phase I clinical trial testing the safety and immunogenicity of an anthrax vaccine using recombinant Escherichia coli-derived, B. anthracis protective antigen (rPA).

Methodology/principal findings: A total of 73 healthy adults ages 18-40 were enrolled and 67 received 2 injections separated by 4 weeks of either buffered saline placebo, or rPA formulated with or without 704 µg/ml Alhydrogel® adjuvant in increasing doses (5, 25, 50, 100 µg) of rPA. Participants were followed for one year and safety and immunologic data were assessed. Tenderness and warmth were the most common post-injection site reactions. No serious adverse events related to the vaccine were observed. The most robust humoral immune responses were observed in subjects receiving 50 µg of rPA formulated with Alhydrogel® with a geometric mean concentration of anti-rPA IgG antibodies of 283 µg/ml and a toxin neutralizing geometric 50% reciprocal geometric mean titer of 1061. The highest lymphoproliferative peak cellular response (median Lymphocyte Stimulation Index of 29) was observed in the group receiving 25 µg Alhydrogel®-formulated rPA.

Conclusions/significance: The vaccine was safe, well tolerated and stimulated a robust humoral and cellular response after two doses.

Trial registration: ClinicalTrials.gov NCT00057525.

Conflict of interest statement

Competing Interests: Dr. Mallory was previously an employee of DynPort Vaccine Company LLC, a CSC company, and is currently employed by MedImmune LLC, a subsidiary of AstraZeneca. Dr. Kelly T. McKee Jr. was previously an employee of DynPort Vaccine Company LLC, a CSC company, and is currently employed by Quintiles Transnational Corporation. Dr. Josephine Cox was previously an employee of the US Military HIV Research Program and is currently employed by the International AIDS Vaccine Initiative. Dr. Thomas C. VanCott was previously an employee of the US Military HIV Research Program and is currently employed by Advanced BioScience Laboratories. Dr. Deborah Birx was previously an employee of the US Military HIV Research Program and is currently employed by the US Centers for Disease Control. DynPort Vaccine Company LLC was the study sponsor. None of the declared conflicting interests impede data sharing as described in the PLoS sharing guidelines.

Figures

Figure 1. CONSORT flowchart.
Figure 1. CONSORT flowchart.
Figure 2. Post-injection reactions (PIRs).
Figure 2. Post-injection reactions (PIRs).
The X axis displays the dosage groups along with all of the placebos, who were combined into one group. The Y axis displays the PIRs that were assessed in the study. The data displayed in the matrix represents the number of individuals who reported a given PIR and the size of the circle is proportional to the reported number. The top half of the graph consists of systemic PIRs while the bottom half of the graph consists of local PIRs.
Figure 3. Adverse events (AEs).
Figure 3. Adverse events (AEs).
The number of AEs was tallied for each dosage group, as well as all of the placebos, who were combined into one group. The empty bars represent AEs that were determined to be not related to the vaccine. The filled bars represent AEs that were determined to be related to the vaccine. The dotted line indicates the total number of volunteers in each group. All AEs that were determined to be related to the vaccine were mild in severity.
Figure 4. Geometric mean concentration of anti-rPA…
Figure 4. Geometric mean concentration of anti-rPA antibodies over time.
The concentration of anti-rPA antibodies was assessed by ELISA at weeks 0, 2, 6, 10, 26, and 52 over the course of the entire study. Arrows indicate when the two injections occurred.
Figure 5. Vaccine induced humoral immunogenicity.
Figure 5. Vaccine induced humoral immunogenicity.
(A) The geometric mean concentrations (GMC) of the anti-rPA antibodies and (B) the geometric mean reciprocal 50% titers (GMT) for the TNA were calculated for each group (horizontal bars) at study visit 8 (2 weeks post second vaccination). The scattergram of the individual responses displays participants who received rPA formulated without Alhydrogel® adjuvant (PBS) in blue and participants whom received rPA formulated with Alhydrogel® in red.
Figure 6. Correlation between the concentration of…
Figure 6. Correlation between the concentration of anti-rPA binding antibodies and the ED50 for the individual samples.
The plot depicts the relationship between the level of anti-rPA antibody binding and the neutralization ED50 in the TNA. A linear regression (grey line) indicates a strong correlation between the two parameters (R2 = 0.86). All samples from rPA recipients are included in the linear regression calculation, however, only the data that were above the limit of quantitation for both assays are displayed.
Figure 7. Vaccine induced lymphocyte proliferation.
Figure 7. Vaccine induced lymphocyte proliferation.
The LPA was performed on fresh cells obtained the day of the assay from participants at study visit 8 (2 weeks post second vaccination) incubated with 1 µg/ml of rPA. The resultant lymphocyte stimulation index (LSI) is plotted for all rPA-receiving participants. The individual data points are overlaid on box and whiskers plots, which show the median and percentiles for the data within each group. (A) Individuals whom received rPA formulated without Alhydrogel® adjuvant (PBS). (B) Individuals whom received rPA formulated with Alhydrogel®. Negative values (LSI ≤5) are denoted by open circles.

References

    1. Young JA, Collier RJ. Anthrax toxin: receptor binding, internalization, pore formation, and translocation. Annu Rev Biochem. 2007;76:243–265.
    1. Fellows PF, Linscott MK, Ivins BE, Pitt ML, Rossi CA, et al. Efficacy of a human anthrax vaccine in guinea pigs, rabbits, and rhesus macaques against challenge by Bacillus anthracis isolates of diverse geographical origin. Vaccine. 2001;19:3241–3247.
    1. Ivins BE, Fellows PF, Pitt ML, Estep JE, Welkos SL, et al. Efficacy of a standard human anthrax vaccine against Bacillus anthracis aerosol spore challenge in rhesus macaques. Salisbury Med Bull. 1996;87:125–126.
    1. Ivins BE, Pitt ML, Fellows PF, Farchaus JW, Benner GE, et al. Comparative efficacy of experimental anthrax vaccine candidates against inhalation anthrax in rhesus macaques. Vaccine. 1998;16:1141–1148.
    1. Brachman PS, Gold H, Plotkin SA, Fekety FR, Werrin M, et al. Field Evaluation of a Human Anthrax Vaccine. Am J Public Health Nations Health. 1962;52:632–645.
    1. Turnbull PC. Anthrax vaccines: past, present and future. Vaccine. 1991;9:533–539.
    1. Keitel WA. Recombinant protective antigen 102 (rPA102): profile of a second-generation anthrax vaccine. Expert Rev Vaccines. 2006;5:417–430.
    1. Anthrax Vaccine Adsorbed (Biothrax™) package insert 2008.
    1. Food and Drug Administration. Biological products, bacterial vaccines and toxoids, implementation of efficacy review, anthrax vaccine adsorbed. 2004. pp. 75180–75198. Final order, 70 F.R.
    1. Niu MT, Ball R, Woo EJ, Burwen DR, Knippen M, et al. Adverse events after anthrax vaccination reported to the Vaccine Adverse Event Reporting System (VAERS), 1990-2007. Vaccine. 2009;27:290–297.
    1. Zink TK, Gorse GJ, et al. “Immunogenicity and tolerance of ascending doses of a recombinant protective antigen (rPA102) anthrax vaccine: a randomized, double-blinded, controlled, multicenter trial” [Vaccine 24 (2006) 5950-5959]. Vaccine. 2007;25:2766–2767.
    1. Gorse GJ, Keitel W, Keyserling H, Taylor DN, Lock M, et al. Response to letter to the editor “Zink TK. Vaccine 2007;25(15):2766-7”. Vaccine. 2007;25:7285–7287.
    1. Gorse GJ, Keitel W, Keyserling H, Taylor DN, Lock M, et al. Immunogenicity and tolerance of ascending doses of a recombinant protective antigen (rPA102) anthrax vaccine: a randomized, double-blinded, controlled, multicenter trial. Vaccine. 2006;24:5950–5959.
    1. Campbell JD, Clement KH, Wasserman SS, Donegan S, Chrisley L, et al. Safety, reactogenicity and immunogenicity of a recombinant protective antigen anthrax vaccine given to healthy adults. Hum Vaccin. 2007;3:205–211.
    1. Quinn CP, Semenova VA, Elie CM, Romero-Steiner S, Greene C, et al. Specific, sensitive, and quantitative enzyme-linked immunosorbent assay for human immunoglobulin G antibodies to anthrax toxin protective antigen. Emerg Infect Dis. 2002;8:1103–1110.
    1. Quinn CP, Dull PM, Semenova V, Li H, Crotty S, et al. Immune responses to Bacillus anthracis protective antigen in patients with bioterrorism-related cutaneous or inhalation anthrax. J Infect Dis. 2004;190:1228–1236.
    1. Ratto-Kim S, Garner RP, Kim JH, Jagodzinski LL, Michael NL, et al. Prospective analyses of HIV-1-specific proliferative responses, recall antigen proliferative responses, and clinical outcomes in an HIV-1-seropositive cohort. J Infect Dis. 2004;189:1988–1995.
    1. Pittman PR, Norris SL, Barrera Oro JG, Bedwell D, Cannon TL, et al. Patterns of antibody response in humans to the anthrax vaccine adsorbed (AVA) primary (six-dose) series. Vaccine. 2006;24:3654–3660.
    1. Anthrax Vaccine Adsorbed (Biothrax™) package insert 2002.
    1. Belton FC, Darlow HM, Henderson DW. The use of anthrax antigen to immunise man and monkey. Lancet. 1956;271:476–479.
    1. Pitt ML, Little SF, Ivins BE, Fellows P, Barth J, et al. In vitro correlate of immunity in a rabbit model of inhalational anthrax. Vaccine. 2001;19:4768–4773.
    1. Little SF, Ivins BE, Fellows PF, Pitt ML, Norris SL, et al. Defining a serological correlate of protection in rabbits for a recombinant anthrax vaccine. Vaccine. 2004;22:422–430.
    1. Reuveny S, White MD, Adar YY, Kafri Y, Altboum Z, et al. Search for correlates of protective immunity conferred by anthrax vaccine. Infect Immun. 2001;69:2888–2893.
    1. Allen JS, Skowera A, Rubin GJ, Wessely S, Peakman M. Long-lasting T cell responses to biological warfare vaccines in human vaccinees. Clin Infect Dis. 2006;43:1–7.
    1. Doolan DL, Freilich DA, Brice GT, Burgess TH, Berzins MP, et al. The US capitol bioterrorism anthrax exposures: clinical epidemiological and immunological characteristics. J Infect Dis. 2007;195:174–184.

Source: PubMed

3
Iratkozz fel