Safety and immunogenicity of SARS-CoV-2 recombinant protein vaccine formulations in healthy adults: a randomised, placebo-controlled, dose-ranging study ======================================================================================================================================================== * Paul A Goepfert * Bo Fu * Anne-Laure Chabanon * Matthew I Bonaparte * Matthew G Davis * Brandon J Essink * Ian Frank * Owen Haney * Helene Janosczyk * Michael C Keefer * Marguerite Koutsoukos * Murray A Kimmel * Roger Masotti * Stephen J Savarino * Lode Schuerman * Howard Schwartz * Lawrence D Sher * Jon Smith * Fernanda Tavares-Da-Silva * Sanjay Gurunathan * Carlos A DiazGranados * Guy De Bruyn ## Abstract **Background** Effective vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are urgently needed. CoV2 preS dTM is a stabilised pre-fusion S protein vaccine produced in a baculovirus expression system. We present interim safety and immunogenicity results of the first-in-human study of the CoV2 preS dTM vaccine with two different adjuvant formulations. **Methods** This Phase I/II, randomised, double-blind study ([NCT04537208](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT04537208&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F20%2F2021.01.19.20248611.atom)) is being conducted in healthy, SARS-CoV-2-seronegative adults in the USA. Participants were stratified by age (18–49 and ≥50 years) and randomised to receive one (on Day[D]1) or two doses (D1, D22) of placebo or candidate vaccine, containing: low-dose (LD, effective dose 1.3 µg) or high-dose (HD, 2.6 µg) antigen with adjuvant AF03 (Sanofi Pasteur) or AS03 (GlaxoSmithKline); or unadjuvanted HD (18–49 years only). Safety was assessed up to D43. SARS-CoV-2 neutralising and binding antibody profiles were assessed in D1, D22 and D36 serum samples. **Findings** The interim safety analyses included 439/441 randomised participants. There were no related unsolicited immediate AEs, serious AEs, medically attended AEs classified as severe, or AE of special interest. More grade 3 solicited reactions were reported than expected after the second dose in the adjuvanted vaccine groups. Neutralising and binding antibody responses after two vaccine doses were higher in adjuvanted versus unadjuvanted groups, in AS03-versus AF03-adjuvanted groups, in HD versus LD groups, and in younger versus older age strata. **Interpretation** The lower than expected immune responses, especially in the older age stratum, and the higher than anticipated reactogenicity post dose 2 were likely due to a higher than anticipated host cell protein content and lower than planned antigen dose in the clinical material. Further development of the AS03-adjuvanted candidate vaccine will focus on identifying the optimal antigen formulation and dose. ## Introduction Coronavirus disease of 2019 (Covid-19), caused by the novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), emerged in December 2019 in Wuhan City, China, and spread rapidly to become a Public Health Emergency of International Concern in January 2020 1; a global pandemic was declared in March 2020. The ongoing pandemic has had, and continues to have, catastrophic consequences with over 1.8 million deaths and over 87.3 million confirmed cases to date worldwide (as of 07 January 2021).2 Interventions to reduce transmission (including the isolation of affected individuals and those deemed at high-risk of severe outcomes, the use of face masks and limiting face-to-face interactions between individuals from separate households) have been undertaken on an unprecedented worldwide scale and have had far reaching socio-economic impact.3 The dominant Covid-19 disease manifestations are fever, fatigue and dry cough.4,5 While most young people and children tend to have only mild symptoms or are asymptomatic, adults over age 50 years and individuals with chronic medical conditions are at increased risk of severe outcomes and death.4,6 Vaccination against SARS-CoV-2 will likely provide the most effective interventional long-term means of preventing and controlling SARS-CoV-2 infection. The development of safe, effective vaccines against SARS-CoV-2 is therefore an urgent global priority. Of over 60 vaccines currently in clinical development, several have reached Phase III testing,7 with interim efficacy results already available for a number of these through peer-reviewed publications8,9 or public statements.10-12 At the time of writing, the mRNA-based vaccine BNT162b2 (Pfizer, BioNTech) has received conditional marketing authorization, emergency use authorization or a temporary authorization in a number of regions including the USA, EU, UK, Mexico, and Canada (for use in relation to the Covid-19 pandemic); another mRNA vaccine mRNA-1273 (Moderna, Inc.) also received approval for emergency use in the USA and in Europe; and the chimpanzee adenovirus vectored vaccine ChAdOx1 nCoV-19 (AstraZeneca, Oxford University) was approved for emergency use in the UK. The SARS-CoV-2 spike (S) glycoprotein on the virion surface mediates host cell entry, making the S protein a key target in vaccine development. Previous work on the S protein of the closely-related Middle East respiratory syndrome coronavirus (MERS-CoV), showed that the introduction of double proline substitutions at the beginning of the central helix in the S2 subunit stabilised the protein in a prefusion conformation, preventing the major conformational changes that occur during fusion of the viral and host cell membranes.13 This prefusion S protein elicited potent neutralising antibody responses in mice.13 Wrapp and colleagues successfully applied this stabilising strategy to the SARS-CoV-2 S protein.14 Sanofi Pasteur has developed a candidate SARS-CoV-2 recombinant protein vaccine containing the stabilised SARS-CoV-2 prefusion S protein as the vaccine target, as have other vaccine developers. Recombinant protein vaccines offer the advantages of fewer potential safety concerns and lower production costs over other traditional vaccines, which rely mostly on attenuation or inactivation of the pathogen.15 However, they often require the use of an adjuvant to enhance the magnitude, quality, and persistence of the immune response.15 The antigen dose-sparing qualities of a formulation containing an adjuvant allows a reduced quantity of vaccine antigen to achieve a robust immune response compared to antigen alone. This may be of particular importance in a pandemic situation, where there may be potential constraints in antigen supply. We tested two different oil-in-water emulsions as vaccine adjuvant components in the current Phase I/II trial: the AF03 adjuvant (Sanofi Pasteur, France)16 and the AS03 Adjuvant System (GlaxoSmithKline, Wavre, Belgium).17 This first-in-human study evaluated the safety and immunogenicity, including binding and neutralising antibody responses and cell-mediated immunity, of the candidate vaccine with the goal of informing selection of an adjuvant formulation, antigen dose, and immunisation schedule to proceed to further clinical development. ## Methods ### Study design and participants This is a Phase I/II, randomised, modified double-blind (observer-blind), first-in-human, parallel group, placebo-controlled, and dose-ranging study ([NCT04537208](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT04537208&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F20%2F2021.01.19.20248611.atom)), with a sentinel safety cohort and early safety data review. The study is conducted across 10 centres in the USA, with a planned duration of approximately 12 months following the last study injection. Here, we present interim safety and immunogenicity data up to 43 days (D43) after first vaccination with the stabilised pre-fusion S (preS) protein vaccine, CoV2 preS dTM. The study was undertaken in compliance with the International Conference on Harmonisation (ICH) guidelines for Good Clinical Practice and the principles of the Declaration of Helsinki. The protocol and amendments were approved by applicable Independent Ethics Committees/Institutional Review Boards and the regulatory agency as per local regulations. Informed consent was obtained from the participants before any study procedures were performed. Healthy adults aged 18 years and older were eligible for inclusion. A lateral flow immunochromatographic assay (COVID-19 IgG/IgM Rapid Test Cassette, Healgen Scientific, MD, USA) was used to identify those with recent or prior SARS-CoV-2 infection; the test was performed at the clinical site by trained personnel, according to the manufacturer’s instructions. Individuals testing negative for SARS-CoV-2 antibodies were included in the study. Exclusion criteria included chronic illness or medical conditions considered to potentially increase the risk for severe Covid-19 illness; women who were pregnant or lactating; women of childbearing potential who were not using an effective method of contraception or abstinence from at least 4 weeks prior to the first vaccination until at least 12 weeks after the last vaccination; participation, or planned participation, in another clinical trial during the study period; receipt or planned receipt of any vaccine in the 30 days before the first or up to 30 days following the last study vaccination (except for influenza vaccination, which may be received at least 2 weeks before or after study vaccines); receipt of immunoglobulins, blood or blood-derived products in the past 3 months; and active or prior documented autoimmune disease. Participants were stratified by age (18–49 years and ≥50 years) and randomised to 11 different treatment groups to receive one of five candidate vaccine formulations or placebo, as a single-dose or two-dose schedule. The first injection was on D1 and the second on D22 (**Supplementary Figure S1**). The candidate vaccine formulations were: low-dose (LD) antigen with AF03 or AS03 adjuvant, high-dose (HD) antigen with AF03 or AS03 adjuvant, or unadjuvanted HD antigen (**Supplementary Table S1**). No participants aged 50 years and older were allocated to the unadjuvanted HD antigen group, as older adults are less likely to respond without the presence of an adjuvant and to minimise the theoretical risk of vaccine enhanced disease. Groups were randomly assigned using an interactive response technology system. Block randomisation was used, with blocks of varying sizes, whereby greater numbers of participants were allocated to the two-dose, AS03-adjuvanted groups (**Supplementary Table S1**). Only the study site staff who prepared and administered the vaccine knew which vaccine was administered, and they were not involved in assessment of adverse events or of the study data. Initially, 30 participants aged 18–49 years were enrolled into a safety sentinel cohort and received a single dose of the intervention to which they were randomised. A review of the safety data up to nine days after the first dose, unblinded to treatment group, was carried out by the Sanofi Pasteur internal safety committee. Only upon demonstration of acceptable safety were the remaining participants enrolled. A participant subset from the two-dose cohort, across both age strata, was randomly assigned for evaluation of cell-mediated immunity (CMI). ### Vaccines and vaccination The targeted quantities of the SARS-CoV-2 preS antigen per vaccine dose were 5 µg for the low-dose formulation and 15 µg for the high-dose formulation. However, during characterisation studies on the final bulk drug substance, a key polyclonal antibody reagent used to detect the SARS-CoV-2 preS protein was found to also recognise glycosylated host cell proteins (HCP). As a result, the purity and HCP levels reported for the Phase I/II clinical trial materials were inaccurate and the concentration of SARS-CoV-2 preS protein in the formulated vaccine product was significantly lower (approximately 4–6 fold) than planned. Upon re-calculation, the effective dose levels administered in a 0.5 mL vaccine dose in this study were 1.3 µg (LD) and 2.6 µg (HD) of functional SARS-CoV-2 preS protein. The underdosing of the vaccine formulation was discovered after the study was fully enrolled and all participants had received at least one dose of their assigned product. The differences between the targeted and the effective dose levels correspond to an excess HCP content in the clinical materials (recalculated HCP content, 3.7 µg and 12.4 µg, respectively). The AF03 (Sanofi Pasteur) and AS03 (GlaxoSmithKline) adjuvants are oil-in-water emulsions. One dose of the AF03 adjuvant emulsion16 contained 12.5 mg squalene, 1.85 mg sorbitan monooleate (Dehymuls S SMO™), 2.38 mg Macrogol cetostearyl ether (Kolliphor CS12™) and 2.31 mg mannitol in phosphate-buffered saline (PBS), and was presented in a 0.7 mL monodose vial (single dose, 0.25mL per dose). One dose of the AS03 Adjuvant System17 contained 11.86 mg α-tocopherol, 10.69 mg squalene and 4.86 mg polysorbate-80 (Tween®80) in PBS, and was presented in a 3.15 mL multidose vial (10 doses, 0.25 mL per dose). Vaccine formulations were supplied in two separate vials, one vial containing antigen suspension and another containing the adjuvant emulsion or PBS diluent. These were mixed prior to injection to give a final dose volume for injection of 0.5 mL, containing 0.25mL antigen and 0.25mL adjuvant emulsion or PBS diluent. Placebo recipients received 0.5 mL 150 mM NaCl. Vaccine formulations and placebo were prepared by qualified and trained study personnel and administered into the deltoid region of the upper arm by intramuscular injection. ### Safety The primary objective was to describe the safety profile of the candidate vaccine formulations in all participants. Safety endpoints included immediate unsolicited systemic AEs (occurring within 30 minutes after each dose); solicited injection site reactions (pain, erythema and swelling) and solicited systemic reactions (fever, headache, malaise and myalgia) up to 7 days after each dose; clinical safety laboratory measures; unsolicited AEs up to 21 days after each dose; and medically attended adverse events (MAAEs), serious adverse events (SAEs) and adverse events of special interest (AESIs) are documented throughout the study. AESIs included anaphylactic reactions and potential immune-mediated diseases (pIMDs). pIMDs are a subset of AEs that include autoimmune diseases and other inflammatory and/or neurologic disorders of interest which may or may not have an autoimmune aetiology.18 AEs were assessed for intensity (grade 1 to grade 3) and their relationship to the study intervention by the Investigator. Participants were instructed to contact the site if they experienced symptoms of a Covid-19-like illness, defined by specified clinical symptoms and signs, at any time during the study (see **Supplementary Material** for the pre-defined list of signs and symptoms). Nasopharyngeal swabs were collected as soon as possible after the date of first clinical manifestation to test for the presence of SARS-CoV-2 by nucleic acid amplification test (Abbott RealTime SARS-CoV-2 RT-PCR, USA; available under EUA), in which RNA from the samples was extracted and purified and SARS-CoV-2 specific primers were used. Clinical safety laboratory parameters were measured eight days after the last dose (on D9 for the single-dose cohort and D30 for the two-dose cohort). Laboratory assessments included serum biochemistry tests, haematology (platelet count, haemoglobin, haematocrit, differential white blood cell count [neutrophils, lymphocytes, monocytes, eosinophils and basophils]) and urine analyses. ### Immunogenicity assessment The primary immunogenicity objective was to describe the neutralising capacity of vaccine-induced antibodies at D1, D22 and D36 for each study group. SARS-CoV-2 neutralising antibodies were measured in serum samples by microneutralisation assay performed at Sanofi Pasteur Global Clinical Immunology (GCI) Swiftwater, PA, USA, using the SARS-CoV-2 USA-WA1/2020 strain (BEI Resources; catalog# NR-52281). The reduction in SARS-CoV-2 infectivity, compared to that in the control wells, indicated the presence of neutralising antibodies in the serum sample. The 50% neutralisation titre was recorded19 (**Supplementary Methods**). Secondary objectives for immunogenicity included a description of the binding antibody profile on D1, D22 and D36 for each study group measured using indirect enzyme-linked immunosorbent assay performed at Nexelis, Laval, Quebec, Canada (ELISA; **Supplementary Methods**), in which the reference standard was human serum with known concentration of anti-S protein IgG antibodies; quantitative results were reported in ELISA Units (EU)/mL. In an exploratory analysis, neutralising antibody titres were measured in a panel of human convalescent serum (Sanguine Biobank, iSpecimen and PPD). Convalescent samples were obtained from 93 donors between 17 and 47 days following PCR-positive diagnosis of Covid-19. Donors had recovered (with clinical severity ranging from mild to severe), and were asymptomatic at time of sample collection. ### Cell-mediated immunity Th1/Th2 CMI responses were measured from blood samples obtained at D1, D22, and D36, following *ex vivo* stimulation, using the TruCulture® system (Myriad Biosciences, Austin, TX). Blood samples (1 mL) were drawn directly into the TruCulture tubes, containing 2 mL of buffered media without stimulation (negative control), SARS-CoV-2 S protein for specific stimulation (S 2P-GCN4, GeneArt); or Staphylococcal enterotoxin B plus anti-cluster of differentiation [CD]28 for unspecific stimulation (positive control) (**Supplementary Methods**). Validated cytokine profiling panels were used to evaluate relevant cytokine concentrations: IFN-gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), interleukin (IL)-2, IL-4, IL-5 and IL-13) (**Supplementary Methods**). ### Statistical Analysis All analyses were descriptive; there was no hypothesis testing. No sample size calculations were performed. Approximately 440 participants were planned to be enrolled in this study (**Supplementary Table S1**), with 300 participants aged 18–49 years (N=20 in each group, except AS03-adjuvanted groups in the two-dose cohort, with 60 participants in each group) and 140 participants aged 50 years or older (N=10 in each group, except in AS03-adjuvanted groups in the two-dose cohort, with 30 participants in each group). Safety objectives were assessed in the safety analysis set (SafAS), defined as randomised participants who received at least one dose; participants were analysed according to the study treatment received. The full analysis set (FAS) included all randomised participants who received at least one dose; participants were analysed according to the treatment group to which they were randomised. The per-protocol analysis set (PPAS) was defined as the subset of participants from the FAS who met all inclusion/exclusion criteria, who had no protocol deviation and who had negative results in the ELISA and/or neutralisation test at baseline; those who had at least one valid post-dose serology sample within the pre-defined time window were included in the PPAS for immunogenicity (PPAS-IAS). CMI analyses were carried out in the subset of PPAS participants who provided at least one CMI sample within the pre-defined time window (PPAS-CMI). Primary safety endpoints were summarised by study intervention group. Neutralising antibody profiles were described based on geometric mean antibody titres (GMTs) and 95% confidence intervals (CI). Fold-rises in serum antibody neutralisation titre post-vaccination relative to D1 were calculated, whereby pre-vaccination titres below the lower limit of quantification (LLOQ) were converted to LLOQ/2. The percentage of participants with a 4-fold rise in serum neutralisation titre relative to D1 at D22 and D36 (post-/pre-dose) are presented. Neutralising antibody seroconversion was defined as baseline values below the LLOQ with detectable neutralisation titres above assay LLOQ at D22 and D36. Binding antibody profiles were described based on S-specific antibody geometric mean concentrations (GMCs) measured at D22 and D36. Antibody concentrations 3.0.CO;2-E&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9595616&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F20%2F2021.01.19.20248611.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000073221800005&link_type=ISI) 21. 21.Diez-Domingo J, Baldo JM, Planelles-Catarino MV, et al. Phase II, randomized, open, controlled study of AS03-adjuvanted H5N1 pre-pandemic influenza vaccine in children aged 3 to 9 years: follow-up of safety and immunogenicity persistence at 24 months post-vaccination. Influenza Other Respir Viruses 2015; 9(2): 68–77. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/irv.12295&link_type=DOI) 22. 22.Leroux-Roels G, Marchant A, Levy J, et al. Impact of adjuvants on CD4(+) T cell and B cell responses to a protein antigen vaccine: Results from a phase II, randomized, multicenter trial. Clin Immunol 2016; 169: 16–27. 23. 23.Moris P, van der Most R, Leroux-Roels I, et al. H5N1 influenza vaccine formulated with AS03 A induces strong cross-reactive and polyfunctional CD4 T-cell responses. J Clin Immunol 2011; 31(3): 443–54. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s10875-010-9490-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21174144&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F20%2F2021.01.19.20248611.atom) 24. 24.Ward BJ, Gobeil P, Séguin A, et al. Phase 1 trial of a Candidate Recombinant Virus-Like Particle Vaccine for Covid-19 Disease Produced in Plants. medRxiv 2020: 2020.11.04.20226282. 25. 25.Callow KA. Effect of specific humoral immunity and some non-specific factors on resistance of volunteers to respiratory coronavirus infection. J Hyg (Lond) 1985; 95(1): 173–89. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1017/S0022172400062410&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2991366&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F20%2F2021.01.19.20248611.atom) 26. 26.Reed SE. The behaviour of recent isolates of human respiratory coronavirus in vitro and in volunteers: evidence of heterogeneity among 229E-related strains. J Med Virol 1984; 13(2): 179–92. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/jmv.1890130208&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=6319590&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F20%2F2021.01.19.20248611.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1984SD61300007&link_type=ISI) 27. 27.McMahan K, Yu J, Mercado NB, et al. Correlates of protection against SARS-CoV-2 in rhesus macaques. Nature 2020. 28. 28.Cohet C, van der Most R, Bauchau V, et al. Safety of AS03-adjuvanted influenza vaccines: A review of the evidence. Vaccine 2019; 37(23): 3006–21. 29. 29.Jackson LA, Campbell JD, Frey SE, et al. Effect of Varying Doses of a Monovalent H7N9 Influenza Vaccine With and Without AS03 and MF59 Adjuvants on Immune Response: A Randomized Clinical Trial. Jama 2015; 314(3): 237–46. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2015.7916&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26197184&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F20%2F2021.01.19.20248611.atom) 30. 30.Levie K, Leroux-Roels I, Hoppenbrouwers K, et al. An adjuvanted, low-dose, pandemic influenza A (H5N1) vaccine candidate is safe, immunogenic, and induces cross-reactive immune responses in healthy adults. The Journal of infectious diseases 2008; 198(5): 642–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/590913&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18576945&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F20%2F2021.01.19.20248611.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000258363700004&link_type=ISI) 31. 31.Vesikari T, Pepin S, Kusters I, Hoffenbach A, Denis M. Assessment of squalene adjuvanted and non-adjuvanted vaccines against pandemic H1N1 influenza in children 6 months to 17 years of age. Hum Vaccin Immunother 2012; 8(9): 1283–92. 32. 32.Smatti MK, Al Thani AA, Yassine HM. Viral-Induced Enhanced Disease Illness. Front Microbiol 2018; 9: 2991. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fmicb.2018.02991&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30568643&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F20%2F2021.01.19.20248611.atom) 33. 33.Czub M, Weingartl H, Czub S, He R, Cao J. Evaluation of modified vaccinia virus Ankara based recombinant SARS vaccine in ferrets. Vaccine 2005; 23(17-18): 2273–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.vaccine.2005.01.033&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15755610&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F20%2F2021.01.19.20248611.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000227898700048&link_type=ISI) 34. 34.Lambert P-H, Ambrosino DM, Andersen SR, et al. Consensus summary report for CEPI/BC March 12-13, 2020 meeting: Assessment of risk of disease enhancement with COVID-19 vaccines. Vaccine 2020; 38(31): 4783–91. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.vaccine.2020.05.064&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32507409&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F20%2F2021.01.19.20248611.atom) 35. 35.Tseng CT, Sbrana E, Iwata-Yoshikawa N, et al. Immunization with SARS coronavirus vaccines leads to pulmonary immunopathology on challenge with the SARS virus. PLoS One 2012; 7(4): e35421. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0035421&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22536382&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F20%2F2021.01.19.20248611.atom) 36. 36.Yasui F, Kai C, Kitabatake M, et al. Prior immunization with severe acute respiratory syndrome (SARS)-associated coronavirus (SARS-CoV) nucleocapsid protein causes severe pneumonia in mice infected with SARS-CoV. Journal of immunology (Baltimore, Md : 1950) 2008; 181(9): 6337–48. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjE4MS85LzYzMzciO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8wMS8yMC8yMDIxLjAxLjE5LjIwMjQ4NjExLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==)