Using a Human Challenge Model of Infection to Measure Vaccine Efficacy: A Randomised, Controlled Trial Comparing the Typhoid Vaccines M01ZH09 with Placebo and Ty21a

Thomas C Darton, Claire Jones, Christoph J Blohmke, Claire S Waddington, Liqing Zhou, Anna Peters, Kathryn Haworth, Rebecca Sie, Christopher A Green, Catherine A Jeppesen, Maria Moore, Ben A V Thompson, Tessa John, Robert A Kingsley, Ly-Mee Yu, Merryn Voysey, Zoe Hindle, Stephen Lockhart, Marcelo B Sztein, Gordon Dougan, Brian Angus, Myron M Levine, Andrew J Pollard, Thomas C Darton, Claire Jones, Christoph J Blohmke, Claire S Waddington, Liqing Zhou, Anna Peters, Kathryn Haworth, Rebecca Sie, Christopher A Green, Catherine A Jeppesen, Maria Moore, Ben A V Thompson, Tessa John, Robert A Kingsley, Ly-Mee Yu, Merryn Voysey, Zoe Hindle, Stephen Lockhart, Marcelo B Sztein, Gordon Dougan, Brian Angus, Myron M Levine, Andrew J Pollard

Abstract

Background: Typhoid persists as a major cause of global morbidity. While several licensed vaccines to prevent typhoid are available, they are of only moderate efficacy and unsuitable for use in children less than two years of age. Development of new efficacious vaccines is complicated by the human host-restriction of Salmonella enterica serovar Typhi (S. Typhi) and lack of clear correlates of protection. In this study, we aimed to evaluate the protective efficacy of a single dose of the oral vaccine candidate, M01ZH09, in susceptible volunteers by direct typhoid challenge.

Methods and findings: We performed a randomised, double-blind, placebo-controlled trial in healthy adult participants at a single centre in Oxford (UK). Participants were allocated to receive one dose of double-blinded M01ZH09 or placebo or 3-doses of open-label Ty21a. Twenty-eight days after vaccination, participants were challenged with 104CFU S. Typhi Quailes strain. The efficacy of M01ZH09 compared with placebo (primary outcome) was assessed as the percentage of participants reaching pre-defined endpoints constituting typhoid diagnosis (fever and/or bacteraemia) during the 14 days after challenge. Ninety-nine participants were randomised to receive M01ZH09 (n = 33), placebo (n = 33) or 3-doses of Ty21a (n = 33). After challenge, typhoid was diagnosed in 18/31 (58.1% [95% CI 39.1 to 75.5]) M01ZH09, 20/30 (66.7% [47.2 to 87.2]) placebo, and 13/30 (43.3% [25.5 to 62.6]) Ty21a vaccine recipients. Vaccine efficacy (VE) for one dose of M01ZH09 was 13% [95% CI -29 to 41] and 35% [-5 to 60] for 3-doses of Ty21a. Retrospective multivariable analyses demonstrated that pre-existing anti-Vi antibody significantly reduced susceptibility to infection after challenge; a 1 log increase in anti-Vi IgG resulting in a 71% decrease in the hazard ratio of typhoid diagnosis ([95% CI 30 to 88%], p = 0.006) during the 14 day challenge period. Limitations to the study included the requirement to limit the challenge period prior to treatment to 2 weeks, the intensity of the study procedures and the high challenge dose used resulting in a stringent model.

Conclusions: Despite successfully demonstrating the use of a human challenge study to directly evaluate vaccine efficacy, a single-dose M01ZH09 failed to demonstrate significant protection after challenge with virulent Salmonella Typhi in this model. Anti-Vi antibody detected prior to vaccination played a major role in outcome after challenge.

Trial registration: ClinicalTrials.gov (NCT01405521) and EudraCT (number 2011-000381-35).

Conflict of interest statement

I have read the journal's policy and the authors of this manuscript have the following competing interests: ZH is a named inventor of a patent (US7887816 B2) now owned by Prokarium Ltd, UK. ZH and SL were paid employees of Emergent BioSolutions at the time this trial was performed. All other authors have declared that no competing interests exist.

Figures

Fig 1. Study profile.
Fig 1. Study profile.
During the screening process, participant exclusions were made either during initial telephone screening or at/after the study centre screening visit. ‘Other’ reasons for exclusions were: contact with young children (3), contact with vulnerable individuals (9), food-related occupation (5), previously resident in typhoid-endemic area for >6 months (15), unable to contact (6) & unknown (8).
Fig 2. Cumulative incidence of typhoid infection…
Fig 2. Cumulative incidence of typhoid infection after S. Typhi challenge at Time = 0.
Time to infection, measured from challenge agent ingestion to development of first fever ≥38°C or first positive blood culture sampling. Non-diagnosed participants censored at 348 hours (dashed line). P value from log-rank test comparing all three groups.
Fig 3. Cumulative incidence of bacteraemia or…
Fig 3. Cumulative incidence of bacteraemia or fever after S. Typhi challenge at Time = 0.
(A) Time to bacteraemia, measured from challenge agent ingestion to time of first positive blood culture sampling. Non-bacteraemic participants censored at time of diagnosis or at 348 hours (dashed line). P value from log-rank test comparing all three groups and comparing M01ZH09 and Ty21a to placebo, respectively. (B) Time to fever, measured from challenge agent ingestion to first recording of fever (oral temperature ≥38°C). Afebrile participants censored at time of diagnosis or at 336 hours (dashed line). P value from log-rank test comparing all three groups.
Fig 4. Changes in physiological signs in…
Fig 4. Changes in physiological signs in those participants developing typhoid by time after diagnosis according to vaccine group allocation.
(A) Temperature, (B) Heart rate and (C) Systolic blood pressure. Mean change from baseline and 95% confidence interval. Dashed black vertical line marks point of typhoid diagnosis; grey horizontal bar indicates all participant mean (95% CI) values pre-vaccination.
Fig 5. Proportion of participants reporting each…
Fig 5. Proportion of participants reporting each solicited symptom during 14 days after challenge according to vaccine group allocation.
(A) Placebo, (B) M01ZH09, and (C) Ty21a vaccine recipient groups. Maximum severity score per participant for each symptom was used and graded according to criteria detailed in the study protocol: fever thresholds are Grade 1: 38.0–38.4°C; Grade 2: 38.5–38.9°C; Grade 3: 39.0–40.0°C; Grade 4: >40.0°C.
Fig 6. Group mean changes (95% confidence…
Fig 6. Group mean changes (95% confidence intervals) in haematological blood parameters compared to pre-challenge measurements according to vaccine allocation and challenge outcome.
(A) Haemoglobin, (B) platelets, (C) total white cell count, (D) neutrophils, (E) lymphocytes, and (F) eosinophils. TD, typhoid diagnosis; nTD, non-typhoid diagnosis.
Fig 7. Group mean changes (95% confidence…
Fig 7. Group mean changes (95% confidence intervals) in biochemistry blood parameters compared to individual baseline measurements according to vaccine allocation and challenge outcome.
(A) Sodium, (B) potassium, (C) urea, (D) creatinine, (E) Albumin, (F) amylase, (G) alkaline phosphatase, (H) alanine aminotransferase, (I) bilirubin, and (J) C-reactive protein. TD, typhoid diagnosis; nTD, non-typhoid diagnosis.
Fig 8. Blood quantification of Salmonella Typhi…
Fig 8. Blood quantification of Salmonella Typhi bacteria present at point of typhoid diagnosis by vaccine group.
10mL peripheral blood was collected at typhoid diagnosis (prior to antibiotic treatment) into an Isolator 10 tube (Wampole Laboratories). Lysis centrifugation was performed (30min x 3000G without brake) after which the deposit was plated to XLD and incubated aerobically at 37°C for 24 hours. After incubation, colony counts and slide agglutination tests were performed. P values calculated using Mann Whitney U test. Median [IQR] bacterial loads in CFU/mL were: M01ZH09 (n = 14): 0.13 [0.05–0.80]; Placebo (n = 15): 1.30 [0.30–5.40]; Ty21a (n = 12): 0.05 [0.05–0.99]. Lower limit of detection, 0.1 CFU/mL; zero values were substituted with LOD/2, i.e. 0.05 CFU/mL.
Fig 9. Proportion of participants reporting each…
Fig 9. Proportion of participants reporting each solicited symptom during the 7-days after receipt of (first) vaccine dose.
(A) Placebo, (B) M01ZH09, and (C) Ty21a vaccine recipient groups. Maximum severity score per participant for each symptom was used; grade 1: symptom reported but no interference with daily activity; grade 2: some interference with normal daily activities; grade 3: significant symptoms preventing normal daily activity; grade 4: potentially life-threatening (see S1 Protocol).
Fig 10. Immunoglobulin G ASC and antibody…
Fig 10. Immunoglobulin G ASC and antibody responses to S. Typhi LPS and flagellin before (Day -28) and after vaccination (either Day -21 or Day 0 for ASC and ELISA assays, respectively).
(A) LPS and (B) flagellin specific IgG antibody secreting cell responses, respectively, measured pre-vaccination (Day -28) and 7 days later (Day -21). (C) Anti-LPS and (D) anti-flagellin antibody titres, respectively, measured pre-vaccination (Day -28) and 28 days later (Day 0, i.e. prior to S. Typhi challenge). Data are grouped according to vaccine allocation.
Fig 11. Pre-vaccination (Day -28) and pre-challenge…
Fig 11. Pre-vaccination (Day -28) and pre-challenge (Day 0) IgG antibody titres according to outcome after challenge.
(A) Anti-LPS IgG, (B) anti-H IgG, and (C) anti-Vi IgG. Clear circles, non-typhoid diagnosed participants; black circles, typhoid diagnosed participants. Difference between Day -28 and Day 0 anti-LPS and anti-H antibodies analysed by ANCOVA adjusted for vaccine group and baseline titre; P values for anti-Vi antibody calculated by Wilcoxon Rank Sum. Lower limit-of Vi-antibody detection, 7.4EU/mL.

References

    1. Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380(9859):2095–128. Epub 2012/12/19. 10.1016/S0140-6736(12)61728-0
    1. Rahman MM, Gilmour S, Saito E, Sultana P, Shibuya K. Self-reported illness and household strategies for coping with health-care payments in Bangladesh. Bulletin of the World Health Organization. 2013;91(6):449–58. 10.2471/BLT.12.115428
    1. Ochiai RL, Acosta CJ, Danovaro-Holliday MC, Baiqing D, Bhattacharya SK, Agtini MD, et al. A study of typhoid fever in five Asian countries: disease burden and implications for controls. Bulletin of the World Health Organization. 2008;86(4):260–8.
    1. Crump JA, Luby SP, Mintz ED. The global burden of typhoid fever. Bulletin of the World Health Organization. 2004;82(5):346–53. Epub 2004/08/10.
    1. World Health Organisation. Meeting of the Strategic Advisory Group of Experts on Immunization, November 2010—summary, conclusions and recommendations. Weekly Epidemiological Record. 2011;86(1–2):1–16.
    1. Anwar E, Goldberg E, Fraser A, Acosta CJ, Paul M, Leibovici L. Vaccines for preventing typhoid fever. The Cochrane database of systematic reviews. 2014;1:Cd001261 Epub 2014/01/05. 10.1002/14651858.CD001261.pub3
    1. World Health Organisation. Typhoid vaccines: WHO position paper. Weekly Epidemiological Record. 2008;6(83):49–60.
    1. Darton TC, Blohmke CJ, Moorthy VS, Altmann DM, Hayden FG, Clutterbuck EA, et al. Design, recruitment, and microbiological considerations in human challenge studies. The Lancet infectious diseases. 2015;15(7):840–51. 10.1016/S1473-3099(15)00068-7
    1. Hindle Z, Chatfield SN, Phillimore J, Bentley M, Johnson J, Cosgrove CA, et al. Characterization of Salmonella enterica derivatives harboring defined aroC and Salmonella pathogenicity island 2 type III secretion system (ssaV) mutations by immunization of healthy volunteers. Infection and immunity. 2002;70(7):3457–67. Epub 2002/06/18.
    1. Kirkpatrick BD, McKenzie R, O'Neill JP, Larsson CJ, Bourgeois AL, Shimko J, et al. Evaluation of Salmonella enterica serovar Typhi (Ty2 aroC-ssaV-) M01ZH09, with a defined mutation in the Salmonella pathogenicity island 2, as a live, oral typhoid vaccine in human volunteers. Vaccine. 2006;24(2):116–23. Epub 2005/09/06.
    1. Kirkpatrick BD, Tenney KM, Larsson CJ, O’Neill JP, Ventrone C, Bentley M, et al. The novel oral typhoid vaccine M01ZH09 is well tolerated and highly immunogenic in 2 vaccine presentations. The Journal of infectious diseases. 2005;192(3):360–6.
    1. Tran TH, Nguyen TD, Nguyen TT, Ninh TT, Tran NB, Nguyen VM, et al. A randomised trial evaluating the safety and immunogenicity of the novel single oral dose typhoid vaccine M01ZH09 in healthy Vietnamese children. PloS one. 2010;5(7):e11778 Epub 2010/07/30. 10.1371/journal.pone.0011778
    1. Lyon CE, Sadigh KS, Carmolli MP, Harro C, Sheldon E, Lindow JC, et al. In a randomized, double-blinded, placebo-controlled trial, the single oral dose typhoid vaccine, M01ZH09, is safe and immunogenic at doses up to 1.7 x 10(10) colony-forming units. Vaccine. 2010;28(20):3602–8. Epub 2010/03/02. 10.1016/j.vaccine.2010.02.017
    1. Levine MM, Ferreccio C, Black RE, Tacket CO, Germanier R. Progress in vaccines against typhoid fever. Reviews of infectious diseases. 1989;11 Suppl 3:S552–67.
    1. Lindow JC, Fimlaid KA, Bunn JY, Kirkpatrick BD. Antibodies in action: role of human opsonins in killing Salmonella enterica serovar Typhi. Infection and immunity. 2011;79(8):3188–94. 10.1128/IAI.05081-11
    1. Gilman RH, Hornick RB, Woodard WE, DuPont HL, Snyder MJ, Levine MM, et al. Evaluation of a UDP-glucose-4-epimeraseless mutant of Salmonella Typhi as a liver oral vaccine. The Journal of infectious diseases. 1977;136(6):717–23.
    1. Hornick RB, Greisman SE, Woodward TE, DuPont HL, Dawkins AT, Snyder MJ. Typhoid fever: pathogenesis and immunologic control. 2. The New England journal of medicine. 1970;283(14):739–46. Epub 1970/10/01.
    1. Levine MM, Tacket CO, Sztein MB. Host-Salmonella interaction: human trials. Microbes and infection / Institut Pasteur. 2001;3(14-15):1271–9. Epub 2002/01/05. 10.1016/S1286-4579(01)01487-3 .
    1. Waddington CS, Darton TC, Jones C, Haworth K, Peters A, John T, et al. An outpatient, ambulant-design, controlled human infection model using escalating doses of Salmonella Typhi challenge delivered in sodium bicarbonate solution. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2014;58(9):1230–40. Epub 2014/02/13. 10.1093/cid/ciu078 .
    1. Electronic Medicines Compendium. Summary of Product Characteristics: Vivotif: Datapharm Commnuications Ltd; 2013. [updated 01/04/2012; cited 2013 18 Jun 2013]. Available from: .
    1. Bale JA, de Pinna EM, Threlfall E, Ward LR, Centre for Infections (Health Protection Agency). Kauffmann-White scheme - 2007 : salmonella identification : serotypes and antigen formulae London: Centre for Infections, Health Protection Agency; 2007.
    1. Health Protection Agency. Identification of Salmonella species: UK Standards for Microbiology Investigations; 2011. Available from: .
    1. Health Protection Agency. Investigation of faecal specimens for enteric pathogens: UK Standards for Microbiology Investigations; 2013. Available from: .
    1. Health Protection Agency. Investigation of blood cultures (for organisms other than Mycobacterium species): UK Standards for Microbiological Investigations; 2013. Available from: .
    1. UK Health Research Authority. The Over Volunteering Prevention System 2013 [updated 26 Mar 201330 Jan 2014]. Available from: .
    1. Waddington CS, Darton TC, Woodward WE, Angus B, Levine MM, Pollard AJ. Advancing the management and control of typhoid fever: A review of the historical role of human challenge studies. The Journal of infection. 2014;68:405–18. Epub 2014/02/05. 10.1016/j.jinf.2014.01.006
    1. Kantele A, Pakkanen SH, Karttunen R, Kantele JM. Head-to-head comparison of humoral immune responses to Vi capsular polysaccharide and Salmonella Typhi Ty21a typhoid vaccines—a randomized trial. PloS one. 2013;8(4):e60583 Epub 2013/04/18. 10.1371/journal.pone.0060583
    1. Simanjuntak CH, Paleologo FP, Punjabi NH, Darmowigoto R, Soeprawoto, Totosudirjo H, et al. Oral immunisation against typhoid fever in Indonesia with Ty21a vaccine. Lancet. 1991;338(8774):1055–9. Epub 1991/10/26.
    1. Colwell DE, Michalek SM, Briles DE, Jirillo E, McGhee JR. Monoclonal antibodies to Salmonella lipopolysaccharide: anti-O-polysaccharide antibodies protect C3H mice against challenge with virulent Salmonella typhimurium. The Journal of Immunology. 1984;133(2):950–7.
    1. Salerno-Goncalves R, Wahid R, Sztein MB. Ex Vivo kinetics of early and long-term multifunctional human leukocyte antigen E-specific CD8+ cells in volunteers immunized with the Ty21a typhoid vaccine. Clinical and vaccine immunology: CVI. 2010;17(9):1305–14. Epub 2010/07/28. 10.1128/CVI.00234-10
    1. Salerno-Goncalves R, Rezwan T, Sztein MB. B Cells Modulate Mucosal Associated Invariant T Cell Immune Responses. Frontiers in Immunology. 2013;4:511 10.3389/fimmu.2013.00511 .
    1. McArthur MA, Fresnay S, Magder LS, Darton TC, Jones C, Waddington CS, et al. Activation of Salmonella Typhi-specific regulatory T cells in typhoid disease in a wild-type S. Typhi challenge model. PLoS pathogens. 2015;11(5):e1004914 10.1371/journal.ppat.1004914
    1. Toapanta FR, Bernal PJ, Fresnay S, Darton TC, Jones C, Waddington CS, et al. Oral Wild-Type Salmonella Typhi Challenge Induces Activation of Circulating Monocytes and Dendritic Cells in Individuals Who Develop Typhoid Disease. PLoS neglected tropical diseases. 2015;9(6):e0003837 10.1371/journal.pntd.0003837
    1. Sztein MB, Salerno-Goncalves R, McArthur MA. Complex adaptive immunity to enteric fevers in humans: lessons learned and the path forward. Front Immunol. 2014;5:516 10.3389/fimmu.2014.00516
    1. Boyd MA, Tennant SM, Saague VA, Simon R, Muhsen K, Ramachandran G, et al. Serum bactericidal assays to evaluate typhoidal and nontyphoidal Salmonella vaccines. Clinical and vaccine immunology: CVI. 2014;21(5):712–21. Epub 2014/03/14. 10.1128/CVI.00115-14

Source: PubMed

3
Abonnieren