A randomized, double-blind, placebo-controlled, dose-escalating phase I trial to evaluate safety and immunogenicity of a plant-produced, bivalent, recombinant norovirus-like particle vaccine

Isabel Leroux-Roels, Cathy Maes, Jasper Joye, Bart Jacobs, Franziska Jarczowski, André Diessner, Yorick Janssens, Gwenn Waerlop, Kirsi Tamminen, Suvi Heinimäki, Vesna Blazevic, Geert Leroux-Roels, Victor Klimyuk, Hiroshi Adachi, Kazuyuki Hiruta, Frank Thieme, Isabel Leroux-Roels, Cathy Maes, Jasper Joye, Bart Jacobs, Franziska Jarczowski, André Diessner, Yorick Janssens, Gwenn Waerlop, Kirsi Tamminen, Suvi Heinimäki, Vesna Blazevic, Geert Leroux-Roels, Victor Klimyuk, Hiroshi Adachi, Kazuyuki Hiruta, Frank Thieme

Abstract

Noroviruses (NoV) are the leading cause of epidemic acute gastroenteritis in humans worldwide and a safe and effective vaccine is needed. Here, a phase I, double-blind, placebo-controlled clinical trial was performed in 60 healthy adults, 18 to 40 years old. Safety (primary objective) and immunogenicity (secondary and exploratory objectives) of a bivalent (GI.4 and GII.4), plant-produced, virus-like particle (VLP), NoV vaccine candidate formulation were investigated at two dose levels (50 µg + 50 µg and 150 µg + 150 µg) without adjuvant. Overall, 13 subjects (65.0%) in the 50 µg group, 16 subjects (80.0%) in the 150 µg group, and 14 subjects (70.0%) in the placebo group reported at least 1 solicited local or general symptom during the 7-day post-vaccination periods following each dose. Severe solicited adverse events (AEs) were rare (2 events in the 50 µg group). A total of 8 subjects (40.0%) in each group reported at least one unsolicited AE during the 28-day post-vaccination periods. Immunogenicity was assessed on days 1, 8, 29, 57, 183 and 365. All subjects were pre-exposed to norovirus as indicated by baseline levels of the different immunological parameters examined. Vaccine-specific humoral and cellular immune responses increased after the first dose but did not rise further after the second vaccination. Increased GI.4- and GII.4-specific IgG titers persisted until day 365. The vaccine elicited cross-reactive IgG antibodies against non-vaccine NoV VLPs, which was more pronounced for NoV strains of the same genotype as the GII.4 vaccine strain than for non-vaccine genotypes. Significant blocking anti-GI.4 and anti-GII.4 VLP titers were triggered in both dose groups. Lymphoproliferation assays revealed strong cell-mediated immune responses that persisted until day 365. In conclusion, both dose levels were safe and well-tolerated, and no higher incidence of AEs was observed in the higher dose group. The data show that a single dose of the vaccine formulated at 50 µg of each VLP is sufficient to reach a peak immune response after 8 to 28 days. The results of this Phase I study warrant further evaluation of the non-adjuvanted vaccine candidate.

Clinical trial registration: https://ichgcp.net/clinical-trials-registry/NCT05508178, identifier (NCT05508178).

Keywords: adults; clinical trial; immunogenicity; norovirus; plant-produced; safety; vaccine; virus-like particle.

Conflict of interest statement

Author GL-R was retained as a consultant by Icon Genetics GmbH (Halle, Germany) to assist in the design and management of the trial, analysis of results and development of the manuscript. AD, FJ, FT, VK, HA, and KH are employees of Icon Genetics GmbH (Halle, Germany), a wholly owned subsidiary of DENKA Company, Ltd (Tokyo, Japan). The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The authors declare that this trial has received funding from DENKA Company, Ltd (Tokyo, Japan), through its wholly owned subsidiary company, Icon Genetics GmbH (Halle, Germany). The funder was involved in the study design, analysis and interpretation of data, the writing of this article, and the decision to submit it for publication.

Copyright © 2022 Leroux-Roels, Maes, Joye, Jacobs, Jarczowski, Diessner, Janssens, Waerlop, Tamminen, Heinimäki, Blazevic, Leroux-Roels, Klimyuk, Adachi, Hiruta and Thieme.

Figures

Figure 1
Figure 1
Electron micrographs of the highly purified, plant-produced, norovirus-like particles and vaccine formulations. VLPs formed by (A) GI.4 Chiba 407 (1987) capsid protein VP1 (batch IN003); (B) GII.4 Aomori 2 (2006) capsid protein VP1 (batch IN002); and formulated vaccine (1:1 mixture of both) at (C) 50 µg + 50 µg (batch IN004) and (D) 150 µg + 150 µg (batch IN005). Clinical batches were examined by transmission electron microscopy following negative staining with uranyless. The black bar corresponds to 250 nm. VLP, virus-like particle.
Figure 2
Figure 2
Subject disposition.
Figure 3
Figure 3
Solicited local and general adverse events during the 7-day post-vaccination period. Percentages of participants (with exact 95% confidence intervals) reporting solicited local adverse events (pain, redness and swelling) and general adverse events (arthralgia, fatigue, gastrointestinal symptoms, headache, myalgia, shivering and fever) possibly related to the vaccination during the 7-day post-vaccination period after each dose and overall per subject. Gastrointestinal symptoms defined as nausea, vomiting, diarrhea and/or abdominal pain. Fever defined as temperature ≥38.0°C. Error bars for zero values are omitted for the sake of clarity.
Figure 4
Figure 4
Humoral anti-GI.4 and anti-GII.4 VLP IgG responses. Humoral anti-GI.4 and anti-GII.4 VLP IgG responses by scheduled time for each treatment. OD values were processed using a 4-parameter logistic fitting algorithm to the standard curve, allowing the determination of antibody concentration in the samples, expressed as EU/mL. Anti-GI.4 (A) and anti-GII.4 (B). VLP IgG responses for rNV-2v 50 µg (grey), 150 µg (black) and placebo (white) are shown as boxplots. The horizontal line represents the median, the bottom and top of the box represent 25th and 75th percentiles of the values, respectively, and the upper and lower error bar represent 5th and 95th percentile of the values, respectively. Diamond indicates mean value. Dots represent outliers. IgG, immunoglobulin G, VLP, virus-like particle.
Figure 5
Figure 5
Humoral, cross-reactive anti-GI.3, anti-GII.4 and anti-GII.17 VLP IgG responses. Humoral, cross-reactive IgG responses against non-vaccine strain VLP scheduled time for each treatment. The cross-reactive IgG titer is defined as the reciprocal of the highest serum dilution that gives a positive result. Anti-GI.3 (2002) (A), anti-GII.4 (1999) (B), anti-GII.17 Kawasaki 308 (2015) (C) and anti-GII.4 Sydney (2012) (D) VLP IgG responses for rNV-2v 50 µg (grey), 150 µg (black) and placebo (white) are shown as boxplots. The horizontal line represents the median, the bottom and top of the box represent 25th and 75th percentiles of the values, respectively, and the upper and lower error bar represent 5th and 95th percentile of the values, respectively. IgG, immunoglobulin G, VLP virus-like particle.
Figure 6
Figure 6
Serum antibodies blocking GI.4 and GII.4 VLP binding to histo-blood group antigen. Blocking of binding of GI.4 VLP (A) and GII.4 VLP (B) to Pig Gastric Mucin by antibodies elicited by rNV-2v 50µg (gray), 150 µg (black) and placebo (white) expressed as half maximal inhibitory concentration (IC50) which represents the effective concentration (titer) of the serum where the blocking of the VLP binding is reduced by half of the maximal VLP blocking. The horizontal line represents the median, the bottom and top of the box represent 25th and 75th percentiles of the values, respectively, and the upper and lower error bar represent 5th and 95th percentile of the values, respectively. PGM, Pig Gastric Mucin, VLP, virus-like particle.
Figure 7
Figure 7
Cellular immune response. Cellular immune response measured as GI.4 and GII.4 VLP-specific lymphoproliferative responses by scheduled time for each study group. GI.4 (A) and GII.4 (B) VLP-specific responses for rNV-2v 50µg (grey), 150 µg (black) and placebo (white) are expressed as stimulation indices and shown as boxplots. The horizontal line represents the median, the bottom and top of the box represent 25th and 75th percentiles of the values, respectively, and the upper and lower error bar represent 5th and 95th percentile of the values, respectively. VLP, virus-like particle.

References

    1. Lopman BA, Steele D, Kirkwood CD, Parashar UD. The vast and varied global burden of norovirus: Prospects for prevention and control. PloS Med (2016) 13(4):e1001999. doi: 10.1371/journal.pmed.1001999
    1. Hall AJ, Lopman BA, Payne DC, Patel MM, Gastanaduy PA, Vinje J, et al. . Norovirus disease in the united states. Emerg Infect Dis (2013) 19(8):1198–205. doi: 10.3201/eid1908.130465
    1. Bartsch SM, Lopman BA, Ozawa S, Hall AJ, Lee BY. Global economic burden of norovirus gastroenteritis. PloS One (2016) 11(4):e0151219. doi: 10.1371/journal.pone.0151219
    1. Bert F, Scaioli G, Gualano MR, Passi S, Specchia ML, Cadeddu C, et al. . Norovirus outbreaks on commercial cruise ships: A systematic review and new targets for the public health agenda. Food Environ Virol (2014) 6(2):67–74. doi: 10.1007/s12560-014-9145-5
    1. Fankem SL, Boone SA, Gaither M, Gerba CP. Outbreak of norovirus illness in a college summer camp: Impact of cleaning on occurrence of norovirus on fomites. J Environ Health (2014) 76(8):20–6.
    1. Mitchell C, Meredith P, Richardson M, Greengross P, Smith GB. Reducing the number and impact of outbreaks of nosocomial viral gastroenteritis: Time-series analysis of a multidimensional quality improvement initiative. BMJ Qual Saf (2016) 25(6):466–74. doi: 10.1136/bmjqs-2015-004134
    1. Parron I, Barrabeig I, Soldevila N, Bartolome R, Guix S, Rius C, et al. . Outbreaks of gastroenteritis due to norovirus in schools and summer camps in Catalonia, 2017-2019. Microbiol Spectr (2022) 10(3):e0011922. doi: 10.1128/spectrum.00119-22
    1. Glass RI, Parashar UD, Estes MK. Norovirus gastroenteritis. N Engl J Med (2009) 361(18):1776–85. doi: 10.1056/NEJMra0804575
    1. Weber DJ, Rutala WA, Miller MB, Huslage K, Sickbert-Bennett E. Role of hospital surfaces in the transmission of emerging health care-associated pathogens: Norovirus, clostridium difficile, and acinetobacter species. Am J Infect Control (2010) 38(5 Suppl 1):S25–33. doi: 10.1016/j.ajic.2010.04.196
    1. O'Reilly KM, Sandman F, Allen D, Jarvis CI, Gimma A, Douglas A, et al. . Predicted norovirus resurgence in 2021-2022 due to the relaxation of nonpharmaceutical interventions associated with covid-19 restrictions in England: A mathematical modeling study. BMC Med (2021) 19(1):299. doi: 10.1186/s12916-021-02153-8
    1. Winkler M, Seedat J, Monning H. Epidemiologisches bulletin. Koch-Institut R, editor (2022). Available at: ()
    1. de Graaf M, van Beek J, Koopmans MP. Human norovirus transmission and evolution in a changing world. Nat Rev Microbiol (2016) 14(7):421–33. doi: 10.1038/nrmicro.2016.48
    1. Chhabra P, de Graaf M, Parra GI, Chan MC, Green K, Martella V, et al. . Updated classification of norovirus genogroups and genotypes. J Gen Virol (2019) 100(10):1393–406. doi: 10.1099/jgv.0.001318
    1. Bartsch SM, Lopman BA, Hall AJ, Parashar UD, Lee BY. The potential economic value of a human norovirus vaccine for the united states. Vaccine (2012) 30(49):7097–104. doi: 10.1016/j.vaccine.2012.09.040
    1. Esposito S, Principi N. Norovirus vaccine: Priorities for future research and development. Front Immunol (2020) 11:1383. doi: 10.3389/fimmu.2020.01383
    1. Atmar RL, Baehner F, Cramer JP, Song E, Borkowski A, Mendelman PM, et al. . Rapid responses to 2 virus-like particle norovirus vaccine candidate formulations in healthy adults: A randomized controlled trial. J Infect Dis (2016) 214(6):845–53. doi: 10.1093/infdis/jiw259
    1. Leroux-Roels G, Cramer JP, Mendelman PM, Sherwood J, Clemens R, Aerssens A, et al. . Safety and immunogenicity of different formulations of norovirus vaccine candidate in healthy adults: A randomized, controlled, double-blind clinical trial. J Infect Dis (2018) 217(4):597–607. doi: 10.1093/infdis/jix572
    1. Sherwood J, Mendelman PM, Lloyd E, Liu M, Boslego J, Borkowski A, et al. . Efficacy of an intramuscular bivalent norovirus Gi.1/Gii.4 virus-like particle vaccine candidate in healthy us adults. Vaccine (2020) 38(41):6442–9. doi: 10.1016/j.vaccine.2020.07.069
    1. Treanor JJ, Atmar RL, Frey SE, Gormley R, Chen WH, Ferreira J, et al. . A novel intramuscular bivalent norovirus virus-like particle vaccine candidate–reactogenicity, safety, and immunogenicity in a phase 1 trial in healthy adults. J Infect Dis (2014) 210(11):1763–71. doi: 10.1093/infdis/jiu337
    1. Vesikari T, Saez-Llorens X, Blazevic V, Lopez P, Lopez E, Masuda T, et al. . Immunogenicity of a bivalent virus-like particle norovirus vaccine in children from 1 to 8 years of age: A phase 2 randomized, double-blind study. Vaccine (2022) 40(26):3588–96. doi: 10.1016/j.vaccine.2022.04.089
    1. Tuse D, Malm M, Tamminen K, Diessner A, Thieme F, Jarczowski F, et al. . Safety and immunogenicity studies in animal models support clinical development of a bivalent norovirus-like particle vaccine produced in plants. Vaccine (2022) 40(7):977–87. doi: 10.1016/j.vaccine.2022.01.009
    1. Treanor J, Sherwood J, Cramer JP, Le Cam Bouveret N, Lin S, Baehner F, et al. . A phase 2 study of the bivalent vlp norovirus vaccine candidate in older adults; impact of mpl adjuvant or a second dose. Vaccine (2020) 38(36):5842–50. doi: 10.1016/j.vaccine.2020.06.011
    1. Hager KJ, Perez Marc G, Gobeil P, Diaz RS, Heizer G, Llapur C, et al. . Efficacy and safety of a recombinant plant-based adjuvanted covid-19 vaccine. N Engl J Med (2022) 386(22):2084–96. doi: 10.1056/NEJMoa2201300
    1. Maharjan PM, Choe S. Plant-based covid-19 vaccines: Current status, design, and development strategies of candidate vaccines. Vaccines (Basel) (2021) 9(9):992. doi: 10.3390/vaccines9090992
    1. Shanmugaraj B, Siriwattananon K, Malla A, Phoolcharoen W. Potential for developing plant-derived candidate vaccines and biologics against emerging coronavirus infections. Pathogens (2021) 10(8):1051. doi: 10.3390/pathogens10081051
    1. Tuse D. Safety of plant-made pharmaceuticals: Product development and regulatory considerations based on case studies of two autologous human cancer vaccines. Hum Vaccin (2011) 7(3):322–30. doi: 10.4161/hv.7.3.14213
    1. Tuse D, Tu T, McDonald KA. Manufacturing economics of plant-made biologics: Case studies in therapeutic and industrial enzymes. BioMed Res Int (2014) 2014:256135. doi: 10.1155/2014/256135
    1. Ward BJ, Gobeil P, Seguin A, Atkins J, Boulay I, Charbonneau PY, et al. . Phase 1 randomized trial of a plant-derived virus-like particle vaccine for covid-19. Nat Med (2021) 27(6):1071–8. doi: 10.1038/s41591-021-01370-1
    1. Gleba Y, Klimyuk V, Marillonnet S. Magnifection–a new platform for expressing recombinant vaccines in plants. Vaccine (2005) 23(17-18):2042–8. doi: 10.1016/j.vaccine.2005.01.006
    1. Gleba Y, Klimyuk V, Marillonnet S. Viral vectors for the expression of proteins in plants. Curr Opin Biotechnol (2007) 18(2):134–41. doi: 10.1016/j.copbio.2007.03.002
    1. Group PIW, Multi-National PIIST, Davey RT, Jr., Dodd L, Proschan MA, Neaton J, et al. . A randomized, controlled trial of zmapp for Ebola virus infection. N Engl J Med (2016) 375(15):1448–56. doi: 10.1056/NEJMoa1604330
    1. Tuse D, Ku N, Bendandi M, Becerra C, Collins R, Jr., Langford N, et al. . Clinical safety and immunogenicity of tumor-targeted, plant-made id-klh conjugate vaccines for follicular lymphoma. BioMed Res Int (2015) 2015:648143. doi: 10.1155/2015/648143
    1. Guidance for industry: Toxicity grading scale for healthy adult and adolescent volunteers enrolled in preventive vaccine clinical trials. In: Administration USDoHaHSFaD, research CfBEa. Available at:
    1. Malm M, Uusi-Kerttula H, Vesikari T, Blazevic V. High serum levels of norovirus genotype-specific blocking antibodies correlate with protection from infection in children. J Infect Dis (2014) 210(11):1755–62. doi: 10.1093/infdis/jiu361
    1. Nurminen K, Blazevic V, Huhti L, Rasanen S, Koho T, Hytonen VP, et al. . Prevalence of norovirus gii-4 antibodies in Finnish children. J Med Virol (2011) 83(3):525–31. doi: 10.1002/jmv.21990
    1. Lindesmith LC, Debbink K, Swanstrom J, Vinje J, Costantini V, Baric RS, et al. . Monoclonal antibody-based antigenic mapping of norovirus Gii.4-2002. J Virol (2012) 86(2):873–83. doi: 10.1128/JVI.06200-11
    1. Crooke SN, Ovsyannikova IG, Poland GA, Kennedy RB. Immunosenescence and human vaccine immune responses. Immun Ageing (2019) 16:25. doi: 10.1186/s12979-019-0164-9
    1. Atmar RL, Bernstein DI, Harro CD, Al-Ibrahim MS, Chen WH, Ferreira J, et al. . Norovirus vaccine against experimental human Norwalk virus illness. N Engl J Med (2011) 365(23):2178–87. doi: 10.1056/NEJMoa1101245
    1. LoBue AD, Lindesmith L, Yount B, Harrington PR, Thompson JM, Johnston RE, et al. . Multivalent norovirus vaccines induce strong mucosal and systemic blocking antibodies against multiple strains. Vaccine (2006) 24(24):5220–34. doi: 10.1016/j.vaccine.2006.03.080
    1. Cannon JL, Bonifacio J, Bucardo F, Buesa J, Bruggink L, Chan MC, et al. . Global trends in norovirus genotype distribution among children with acute gastroenteritis. Emerg Infect Dis (2021) 27(5):1438–45. doi: 10.3201/eid2705.204756
    1. Kendra JA, Tohma K, Parra GI. Global and regional circulation trends of norovirus genotypes and recombinants, 1995-2019: A comprehensive review of sequences from public databases. Rev Med Virol (2022) 32(5):e2354. doi: 10.1002/rmv.2354

Source: PubMed

3
订阅