Study design and participants
We performed this randomized, placebo-controlled, observer-blind Phase I trial in healthy young adults between 18 and 59 years old, and older adults between 65 and 85 years of age, in Taizhou, Jiangsu Province, China. Participants were in overall good health established by medical history, physical examination, and laboratory tests at the screening visit. Both males and females were included and agreed to use contraception during the trial. We excluded participants that were pregnant or breast-feeding. Participants that tested positive for SARS-CoV-2 via a commercial rapid diagnostic kit for IgM/IgG antibody to SARS-CoV-2 (manufactured by Livzon diagnostics inc., Zhuhai, China), or via testing with a pharyngeal swab nucleic acid diagnostic test (manufactured by Fosun pharma, Shanghai, China) were excluded. Imaging features of COVID-19 present in a chest CT scan was a further exclusion criterion. Participants with serious cardiovascular disease or chronic conditions such as uncontrolled diabetes and hypertension, human immunodeficiency virus, hepatitis B and hepatitis C were excluded. A complete list of the inclusion and exclusion criteria is provided in the Extended Data Set. Written informed consent was obtained from each participant before the start of the study.
The study was conducted in accordance with the Declaration of Helsinki and Good Clinical Practice. The trial protocol was reviewed and approved by the Chinese NMPA, and the institutional review board of the Jiangsu Provincial Center of Disease Control and Prevention.
This trial was registered with the Chinese Clinical Trial Registry (ChiCTR2000034825) and with clinicaltrials.gov (NCT04523571).
Randomization and blinding
Eligible participants between 18 and 55 years of age were enrolled in the younger age group, and older participants aged greater than or equal to 65 years and less than or equal to 85 years were enrolled in the older age group. Participants were randomized in a ratio of 1:1:1 to receive the low-dose BNT126b1 or high-dose BNT126b1 or placebo. Participants were stratified by gender, using a Web-based interactive response technology (IRT) system. The blocked randomization list was generated by an independent statistician using SAS software (version 9.4).
Authorized unblinded pharmacists prepared the vaccines or placebo according to the allocation of participants through the IRT system, and nurses administrated the investigational products to participants. The unblinded staff had no further involvement in the trial, and were forbidden to disclosure allocation information to others. All other investigators, participants, laboratory staff and the sponsor remained blinded throughout the trial.
Vaccine and vaccination
BNT162b1 consists of a Good Manufacturing Practice (GMP)-grade mRNA drug substance encoding the trimerized SARS-CoV-2 spike glycoprotein RBD antigen, formulated with lipids to obtain the RNA–LNP drug product. Vaccine was transported and supplied as a buffered-liquid solution for intramuscular injection, and stored at −80 °C. For details refer to (Sahin et al)13.
The low-dose and high-dose BNT126b1 contained 10 µg and 30 µg active ingredient, respectively, and the placebo was a commercial saline solution. Each participant received a prime-boost dosing regimen of vaccine candidate BNT162b1 at either 10 μg/0.5 ml or at 30 μg/0.5 ml or placebo of 0.5 ml administered into the deltoid, 21 days apart.
Monitoring of safety and immunogenicity
Each participant was asked to remain at the study site for at least six hours post vaccine administration for safety observation. Vital signs including temperature, blood pressure, pulse, and respiratory rate were measured at baseline, one hour, three hours and six hours post-vaccination. Any adverse events following the vaccination were documented by participants using diaries until Day 28 post-administration of the boost dose. Younger group participants were enrolled and received the vaccination first. Enrollment of the older age group was launched following evaluation of the preliminary safety data of the younger age group for the first 14 days post-prime vaccination. Severity of adverse events and laboratory abnormal changes are graded with both the scale issued by the China State Food and Drug Administration18 and the U.S. Food and Drug Administration (FDA)19. Blood samples for safety laboratory testing were taken at baseline, 24 hours, days 8 after the prime dose, days 8 after the boost dose and months 6 (thyroid function only).
Immunogenicity assessment was twofold. Serum were collected at baseline, days 8 and 22 after each dose, to facilitate measurement of specific IgG antibody responses to RBD-binding and spike glycoprotein S1-binding, and neutralizing antibody to SARS-CoV-2. To assess T-cell response, PBMC samples were collected at baseline, days 8 or 22 (young adult only).
All reported adverse events were reviewed by investigators. Adverse events were categorized as either possibly, probably, or definitely related to the vaccine candidate.
Human convalescent sera
A panel of 24 convalescent human serum samples were obtained from donors 18 to 70 years of age (mean age, 45.8 years) who had recovered from SARS-CoV-2 infection; samples were obtained at least 14 days after a polymerase chain reaction-confirmed diagnosis and after symptom resolution. The disease severities of these patients varied from non-symptomatic (n=3, 13%), mild (n=8, 33%), moderate (n=10, 42%), or severe (n=3, 13%).
Neutralizing GMTs in subgroups of the donors were as follows: 40.0 for the three donors with non-symptomatic infections; 226.3 for the eight donors with mild infection; 91.9 for the ten donors with moderate infection; and 160.0 in the three donors with severe infection. Each serum sample in the panel was from a different donor. Thus, most of the serum samples were obtained from persons with moderate COVID-19. The convalescent serum samples were tested side by side as comparators with the serum samples obtained from participants in this trial.
Enzyme-linked immunosorbent assay (ELISA)
Total anti-SARS-CoV-2 antibodies were determined using an indirect ELISA assay. Briefly, serum samples were diluted in a two-fold series (1:100 to 1:51200) with sample diluent (TBS buffer with 3% BSA and 0.05% Tween-20) and tested in 96 well plates coated with recombinant RBD or S1 (100 ng/100 µL, Sino Biological, China) in sodium carbonate buffer. Bound IgG was detected using an HRP-conjugated secondary antibody (Southern Biotech, USA) and TMB substrate (Surmodics, USA). Data collection was performed using a SpectraMax M5 reader (Molecular Device, USA) at OD 450 nm. To obtain sample titre, the two points adjacent to the assay cut-off value were taken for linear fitting, the sample dilution corresponding to the cut-off value was the titer value. Sample titre was set to 51200 if its OD was greater than the cutoff value at 1:51200 dilution; if sample OD was less than the cutoff at initial dilution 1:100, the sample titre was set at 50.
Microneutralization assay
SARS-CoV-2 specific neutralizing antibody titre in serum was determined by a cytopathy based microneutralization assay using SARS-CoV-2 virus strain BetaCoV/JS02/human/2020 (EPI_ISL_411952) and Vero-E6 cells (National Collection of Authenticated Cell Cultures, National Academy of Science, China). Briefly, serum samples were heat-inactivated for 30 min at 56℃ and serially diluted in a two-fold series from 1:10 to 1:5120 using Dulbecco's Modified Eagle's Medium (Thermo Fisher Scientific, USA). Serum dilutions were then mixed with the same volume of virus solution to achieve 200 TCID50 in each well. The serum-virus mixture was incubated at 37℃ for 1 hour, then added to 96 well plates containing a monolayer of Vero-E6 cells (>80% density). After culturing at 37℃ for 3 days, cytopathic effects (CPE) on VeroE6 were observed under an inverted microscope. The neutralizing titer is the reciprocal of the highest sample dilution that protects at least 50% of cells from CPE. If no neutralization reaction was observed at the initial serum dilution (1:10), and the titre would be reported as 5.
Interferon-γ enzyme-linked immunospot (IFNγ ELISpot)
SAR-CoV-2 viral antigen specific T-cell responses were assessed by an ex vivo interferon-γ (INF-γ) enzyme-linked immunospot (ELISpot) assay (ELISpotPro Kit, Mabtech AB, Sweden)20. Tests were performed in triplicates with a positive control (anti-CD3 monoclonal antibody). Per well, 1 x 105 cryopreserved (baseline samples) or freshly isolated peripheral blood mononuclear cells (PBMCs) were stimulated 21 ± 0.5 hours with different peptide pools: Sp1 overlapping peptide pool, covering the N-terminal half of spike protein, including the RBD; Sp2 overlapping peptide pool, covering the C-terminal half of spike protein (JPT Peptide Technologies, Germany)21. CEF peptide pool22 consisting of 32 MHC class I restricted viral peptides from human cytomegalovirus, epstein-barr virus and influenza virus (Mabtech AB, Sweden). Bound IFNγ was visualized using a secondary antibody directly conjugated with alkaline phosphatase followed by incubation with BCIP/NBT substrate. Plates were scanned using an AID ELISPOT Reader (AID Autoimmun Diagnostika, Germany). Spot counts were displayed as mean values of each triplicate, calculated by subtracting the mean negative control response from the mean of each peptide pool response.
Outcomes
The primary and secondary objectives of this trial were to evaluate safety and immunogenicity of the candidate vaccine BNT162b1 in healthy Chinese adults. The primary endpoints for safety evaluation were the incidence of solicited local reactions at the injection site or systemic adverse reactions within 14 days post-vaccination, and adverse events following the full immunization until 28 days after receiving the boost dose. Any clinical laboratory abnormalities from baseline to 24 hours or 7 days after vaccination, and any serious adverse event (SAE) that occurred were also recorded.
The secondary endpoints for immunogenicity were geometric mean titer (GMT), seroconversion rates and fold increase of virus-neutralizing antibody, or ELISA IgG antibodies binding to S1 or RBD measured at days 8, 22 after each vaccination. Seroconversion is defined as an increase by a factor of four or more in antibody titer over the baseline, or the lower limit value if the baseline titer is below the limit of detection. The serum dilution for ELISA started at 1:100, while that for microneutralization assay started at 1:10.
Cellular immune responses in terms of the number of positive cells with interferon gamma (IFN-γ) secretion among PBMCs at a concentration of 1×105/well at Day 8 and 22 after the boost dose were explored as an exploratory endpoint.
Statistical analysis
The total sample size in this study was 144 participants, 24 participants of each age group was included in each treatment group. The probability to observe a particular adverse event with incidence of 8% at least once in 24 participants in each dose group was 86.5%.
All randomized participants who received at least one dose of the investigational vaccine were included in the safety analysis. Safety endpoints were described as frequencies (%) with 95% confidence interval (CI) of the adverse reactions or events during the observation period. We compared the proportions of the participants with adverse reactions or events across the groups using Chi-square or Fisher exact. All participants who received at least one vaccination and had results of serologic measurements baseline or after vaccination were included in the immunogenicity analysis. The immunological endpoints were descriptively summarized at the specified time points, and compared across the groups, using ANOVA for log-transformed antibody titers, or Wilcoxon rank-sum test for non-normal data. The neutralizing antibody responses of the participants in each dose group were compared with those of patients who had PCR-confirmed SARS-CoV-2 infection. Any serologic values below the lower limit of detection were set to half of the value (1:50 for ELISA and 1:5 for microneutralization assay), while the values above the highest dilution titer were assigned values of the highest dilution for calculation. Pearson correlation analysis of the RBD-binding or S1-binding specific ELISA antibody and neutralizing antibody was performed to assess the relationship between responses on different assays.