Skip to main content

This is a preprint.

It has not yet been peer reviewed by a journal.

The National Library of Medicine is running a pilot to include preprints that result from research funded by NIH in PMC and PubMed.

bioRxiv logoLink to bioRxiv
[Preprint]. 2021 Sep 2:2021.09.02.458740. [Version 1] doi: 10.1101/2021.09.02.458740

Distinct neutralizing kinetics and magnitudes elicited by different SARS-CoV-2 variant spikes

Yang Liu 1,*, Jianying Liu 2,*, Jing Zou 1,*, Ping Ren 3, Scott C Weaver 2,4,5, Xuping Xie 1,#, Pei-Yong Shi 1,4,5,6,7,#
PMCID: PMC8423216  PMID: 34494020

Abstract

The rapid evolution of SARS-CoV-2 mandates a better understanding of cross-protection between variants after vaccination or infection, but studies directly evaluating such cross-protection are lacking. Here we report that immunization with different variant spikes elicits distinct neutralizing kinetics and magnitudes against other SARS-CoV-2 variants. After immunizing hamsters with wild-type or mutant SARS-CoV-2 bearing variant spikes from Alpha, Beta, Gamma, or Epsilon, the animals developed faster and greater neutralization activities against homologous SARS-CoV-2 variants than heterologous variants, including Delta. The rank of neutralizing titers against different heterologous variants varied, depending on the immunized variant spikes. The differences in neutralizing titers between homologous and heterologous variants were as large as 62-, 15-, and 9.7-fold at days 14, 28, and 45 post-immunization, respectively. Nevertheless, all immunized hamsters were protected from challenges with all SARS-CoV-2 variants, including those exhibiting the lowest neutralizing antibody titers. The results provide insights into the COVID-19 vaccine booster strategies.

Introduction

The global pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused >213 million infections and >4.4 million deaths (as of August 25, 2021 per https://coronavirus.jhu.edu/). Despite the unprecedented success of vaccine development for coronavirus disease 2019 (COVID-19),1 global control of the pandemic remains challenging because of insufficient vaccine production and vaccine hesitancy, as well as the emergence of new, more transmissible variants. Although coronaviruses have an intrinsic proofreading mechanism to maintain their long RNA genomes,2 SARS-CoV-2 continues to evolve, leading to the emergence of variants. Since viral spike protein is responsible for binding to the cellular receptor angiotensin-converting enzyme 2 (ACE2), SARS-CoV-2 variants have accumulated many of their mutations in the spike gene. Such spike mutations can alter transmission efficiency and/or immune escape. The first prevalent substitution that underwent a selective sweep, D614G, is located at the spike protein that enhances spike/ACE2 binding, making the virus more transmissible.37 Other substitutions, such as L452R and E484K in the spike receptor-binding domain (RBD), confer resistance of SARS-CoV-2 variants to therapeutic antibodies.8,9 Among the emerged variants, Beta (B.1.351) and Kappa (B.1.617.1) exhibit the least sensitivity to neutralization by immune sera from vaccinated people,8,1013 whereas Alpha (B.1.1.7) and Delta (B.1.617.2) were associated with increased viral transmissibility.14,15 These observations have prompted the desire to modify the vaccine sequence to match variants of concern, such as Beta because of its reduced neutralization sensitivity to the current vaccine sera.8,10 However, one critical question about this modified vaccine approach is whether the new vaccine elicits potent neutralizing activities against other co-circulating variants. Along the same line, cross-protection among different variants after natural infection remains to be studied in unvaccinated populations.

Results

To examine cross-protection among different variant spikes, we prepared a panel of four chimeric SARS-CoV-2 (Extended Data Fig. 1a), each bearing a distinct variant spike gene from Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), or Epsilon (B.1.429) in the backbone of an early virus strain USA-WA1/2020 [isolated in January 2020 and defined as wild-type (WT)]. The four variants were selected based on their high prevalence at the onset of the project. Each variant spike contained a distinct set of mutations (Fig. 1a). An additional substitution E484K was added to the original Alpha variant (Alpha+E484K) as this mutation occurred in many clinical isolates.16 The spike genes from all recombinant viruses were sequenced to ensure no aberrant mutations. Comparable ratios of viral RNA copies versus plaque-forming units (RNA/PFU) were found for both WT and chimeric viruses when produced and analyzed on Vero E6 cells (Extended Data Fig. 1b), suggesting equivalent specific infectivity of the viral stocks.

Figure 1. Variant spikes elicit neutralizing antibodies that cross-protect hamsters from challenges with SARS-CoV-2 variants.

Figure 1.

a, Amino acid substitutions in the spike protein among SARS-CoV-2 variants. The sequence of the spike from USA-WA1/2020 strain was used as a reference. NTD, N-terminal domain; RBD, Receptor binding domain. b, Experimental scheme of immunization and challenge in hamsters. The hamsters (n=4 per group) were intranasally immunized with 106 PFU of WT or variant-spike SARS-CoV-2. Serum specimens were measured for FFRNT50 values on days 14, 28, and 45 post-immunization. On day 49 post-immunization, the hamsters were intranasally challenged by the indicated variant-spike SARS-CoV-2 (104 PFU). The nasal washes were quantified for viral titers on days 1 and 2 post-challenge. All the hamsters were sacrificed on day 2 post-challenge for viral titers detection. c, Neutralization titers of hamster sera against SARS-CoV-2 spike variants on days 14, 28, and 45 post-immunization. Means ± standard errors of the mean are shown. d-g, Protection of immunized hamsters from the challenge of SARS-CoV-2 spike variants. The immunized hamsters and age-matched non-immunized hamsters (Mock) were challenged with selected variant viruses exhibiting the lowest neutralizing titers. The viral loads in the nasal wash (NW, days 1 and 2), lung, and trachea (day 2) were detected by plaque assays. The numbers above individual columns indicate the fold decrease in viral loads by comparing the means from the immunized group with that from the non-immunized mock group. Means ± standard errors of the mean are shown. The assay limit is 10 PFU/ml.

To analyze the immunogenicity of different variant spikes, we intranasally immunized hamsters with 106 PFU of recombinant WT or variant-spike virus (Fig. 1b). The immunized animals developed different degrees of weight loss in the order of Alpha+E484K-spike > Beta-spike ≈ Gamma-spike > WT ≈ Epsilon-spike (Extended Data Fig. 2a). The weight loss results were consistent with the clinical scores, with the Alpha+E484K-spike virus causing the most severe disease (Extended Data figure 2b). These results suggest that Alpha+E484K-spike is the most pathogenic virus in the hamster model. Sera were collected on days 14, 28, and 45 post-immunization and measured for neutralizing titers against homologous and heterologous variant-spike viruses, including the currently prevalent Delta-spike virus (Extended Data Fig. 1). To increase assay throughput, we developed a “fluorescent foci” reduction neutralization test (FFRNT) by using mNeonGreen (mNG) reporter viruses (Extended Data Fig. 3a). The mNG gene was engineered into the open-reading-frame-7 (ORF7) of the viral genome.17 The protocols for the conventional plaque reduction neutralization test (PRNT) and FFRNT (Extended Data Fig. 3b) were similar except that the latter quantifies “fluorescent Foci” using a high-content imager in a high-throughput manner (Extended Data Fig. 3c). The two assays yielded comparable neutralizing titers for the same set of BNT162b2-vaccinated human sera (Extended Data Fig. 3d,e), validating the utility of FFRNT for neutralization test.

FFRNT analysis of immunized hamster sera showed distinct neutralizing profiles against homologous and heterologous SARS-CoV-2 variants (Summary in Fig. 1c and details in Extended Data Fig. 4 and Extended Data Tables 13). (i) Each variant spike elicited faster and higher neutralizing titers against its homologous SARS-CoV-2 variant than heterologous variants; (ii) The magnitudes and ranks of neutralizing titers against different heterologous variants varied depending on the immunized variant spikes; (iii) Unlike other variant spike-immunized groups, the Alpha-spike-immunized animals did not seem to increase the neutralizing titers against heterologous variants from days 14 to 45 post-immunization. It is notable that from days 14 to 45 post-immunization, homologous neutralization titers increased by ≤ 2.32-fold, whereas heterologous neutralization titers could increase up to 22-fold when Gamma-spike-immunized sera were tested against epsilon-spike SARS-CoV-2 (Fig. 1c). On days 14, 28, and 45 post-immunization, the differences in neutralizing titers between homologous and heterologous variants could be as large as 62-, 15-, 9.7-fold, respectively (Fig. 1c). Collectively, the results demonstrate that vaccination of hamsters with different variant spikes elicits distinct kinetics, magnitudes, and ranks of neutralizing titers against homologous and heterologous SARS-CoV-2 variants.

To directly evaluate cross-protection, we selected variant viruses exhibiting the lowest neutralizing titers for each immunized group to challenge the hamsters on day 49 post-immunization. Specifically, animals immunized with WT, Alpha-, Beta-, Gamma-, or Epsilon-spike were challenged with 104 PFU of Beta-, Delta-, Epsilon-, Epsilon-, and Gamma-spike SARS-CoV-2, respectively. Compared with PBS-immunized, challenged animals, all variant spike-immunized hamsters were protected from the challenge and developed significantly lower viral loads in nasal washes (82- to 10,112-fold), tracheas (955- to 120,000-fold), and lungs (57,000- to 490,000-fold) (Fig. 1d).

Discussion

Our study provided experimental evidence against the need to modify vaccine sequences to match the currently circulating SARS-CoV-2 variants of concern. Our results showed distinct cross-neutralizing profiles elicited by different variant spikes, underscoring the heterogenicity in neutralization titers against different variants for any modified spike vaccines. Such modified vaccines may pose logistic challenges for vaccine implementation because (i) multiple variants often cocirculate and (ii) the constellation of variants may differ at different geographic regions and change rapidly over time, such as the recent replacement of the Alpha by the Delta variant in many regions; these replacements have generally not been predictable. Although different vaccine choices should be prescribed depending on the prevalence of specific variants, the prescribed vaccine should also be effective against other co-circulating variants. Our results, together with the observation that BNT612b2-immunized sera remained active in neutralizing all tested variants,1012 support the strategy to continue the currently approved BNT612b2 vaccine for global immunization. This strategy is further bolstered by the real-world effectiveness of two BNT612b2 doses at efficacy rates of 89.5%, 75%, and 88% against Alpha, Beta, and Delta variants, respectively18,19. When protective immunity wanes over time, a third BNT612b2 booster could be administered to enhance the overall neutralizing titers to prevent infection and disease due to new variants. However, this strategy is contingent on the sensitivity of future variants to the immunity elicited by the current vaccine. As herd immunity continues to increase through natural infection and vaccination, selective pressures for the evasion of immunity may rise. The long-term strategy should include (i) surveillance of immune escape of new variants and (ii) preparedness for changes to vaccine strains with immune escape capability.

A limitation of this study is the use of chimeric viruses rather than the use of clinically approved vaccine platforms for expressing variant spikes or clinical variant isolates for the challenge. The neutralizing profile elicited by chimeric viruses may differ from that elicited by the clinically approved vaccine platforms. In chimeric virus-immunized hamsters, immune responses to non-spike viral proteins may provide added protection when compared with animals immunized with spike-alone vaccines such as mRNA and adenovirus-expression platforms. Despite this limitation, it is conceivable that the relative rank of neutralizing levels would be preserved against different SARS-CoV-2 variants.

In summary, increasing global immunization with the currently available safe and effective vaccines, together with boosters when needed, is the strategy to end the COVID-19 pandemic. The design of the booster vaccines depends on whether the newly emerged variants can escape the immunity generated by the current vaccines or natural infections. Potential immune escape of any new variants should be closely monitored by laboratory studies and real-world breakthroughs in vaccinated and infected individuals.

Methods

Ethics statement.

Hamster studies were performed under the guidance of the Care and Use of Laboratory Animals of the University of Texas Medical Branch (UTMB). The protocol was approved by the Institutional Animal Care and Use Committee (IACUC) at UTMB. All the hamster operations were performed under anesthesia by isoflurane to minimize animal suffering.

Animals and Cells.

The Syrian golden hamsters (HsdHan:AURA strain) were purchased from Envigo (Indianapolis, IN). Vero E6 cells, an African green monkey kidney epithelial cell line (ATCC, Manassas, VA, USA), were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Gibco/Thermo Fisher, Waltham, MA, USA) with 10% fetal bovine serum (FBS; HyClone Laboratories, South Logan, UT) plus 1% ampicillin/streptomycin (Gibco). The authenticity of Vero E6 cells was verified using Short Tandem Repeat profiling by ATCC. The cells were tested negative for mycoplasma.

Construction of chimeric SARS-CoV-2s with variant spikes and mNeonGreen (mNG) reporter viruses.

All spike mutations from different variants were engineered into an infectious cDNA clone of an early SARS-CoV-2 isolate USA-WA1/2020 using a standard PCR-based mutagenesis method. The protocol for the construction of recombinant SARS-CoV-2 was reported previously.17,20 To construct the mNG reporter viruses with variant spikes, the mNG gene was engineered into the open-reading-frame-7 (ORF7) of the viral genome. The full-length cDNAs of the viral genome containing the variant spike mutations were assembled by in vitro ligation. The resulting genome-length cDNAs served as templates for in vitro transcription of full-length viral RNAs. The full-length viral RNA transcripts were electroporated into Vero E6 cells. On day 2 post electroporation (when the electroporated cells developed cytopathic effects due to recombinant virus production and replication), the original viral stocks (P0) were harvested from the culture medium. The P0 viruses were amplified on Vero E6 cells for another round to produce working viral stocks (P1). The complete spike genes from the P1 viruses were sequenced to ensure no undesired mutations. The P1 viruses were used for the following study.

Plaque assay.

Approximately 1.2×106 Vero E6 cells were seeded to each well of 6-well plates and cultured at 37°C, 5% CO2 for 16 h. The virus was serially diluted in DMEM with 2% FBS and 200 μl diluted viruses were transferred onto the monolayer of Vero E6 cells. The viruses were incubated with the cells at 37°C with 5% CO2 for 1 h. After the incubation, 2 ml of overlay medium (DMEM medium supplemented with 1% agar) was added to the infected cells per well. The overlay medium contained DMEM with 2% FBS, 1% penicillin/streptomycin, and 1% sea-plaque agarose (Lonza, Walkersville, MD). After a 2-day incubation, plates were stained with neutral red (Sigma-Aldrich, St. Louis, MO) and plaques were counted on a lightbox.

Quantitative real-time RT-PCR assays.

RNA copies of SARS-CoV-2 samples were detected by quantitative real-time RT-PCR (RT-qPCR) assays were performed using the iTaq SYBR Green One-Step Kit (Bio-Rad) on the LightCycler 480 system (Roche, Indianapolis, IN) following the manufacturer’s protocols. The absolute quantification of viral RNA was determined by a standard curve method using an RNA standard (in vitro transcribed 3,480 bp containing genomic nucleotide positions 26,044 to 29,883 of SARS-CoV-2 genome).

Hamster infections.

Four- to six-week-old male golden Syrian hamsters, strain HsdHan:AURA (Envigo, Indianapolis, IN), were intranasally immunized with 106 PFU recombinant WT or variant spike virus on day 0. The immunized animals were weighed and monitored for signs of illness daily. Sera were collected on days 14, 28, and 45 post-immunization and measured for neutralizing titers against homologous and heterologous variant-spike viruses. On day 49, animals from each immunized group were challenged with 104 PFU of selected variant viruses exhibiting the lowest neutralizing titers. Specifically, animals immunized with WT, Alpha-, Beta-, Gamma-, or Epsilon-spike were challenged with the Beta-, Delta-, Epsilon-, Epsilon-, and Gamma-spike SARS-CoV-2, respectively. Nasal washes were collected in 400 μl sterile DPBS at indicated time points. Animals were humanely euthanized for organ collections after 2 days of the challenge. The harvested tracheae and lungs were placed in a 2-ml homogenizer tube containing 1 ml of maintenance media (DMEM supplemented with 2% FBS and 1% penicillin/streptomycin) and stored at −80°C. Samples were subsequently thawed, lung or tracheae were homogenized using TissueLyser II (Qiagen, Hilden, Germany) for 1 min at 26 sec-1, and debris was pelleted by centrifugation for 5 min at 16,100×g. Infectious titers were determined by plaque assay.

Human serum specimens.

The research protocol regarding the use of human serum specimens was reviewed and approved by the University of Texas Medical Branch (UTMB) Institutional Review Board. The approved IRB protocol number is 20–0070. All human serum specimens were obtained from the vaccinated subjects at the UTMB. All specimens were deidentified from patient information.

Fluorescent foci reduction neutralization assay.

Neutralization titers of human and hamster sera were measured by fluorescent foci reduction neutralization assay (FFRNT) using the mNG SARS-CoV-2. Briefly, Vero E6 cells (2.5 × 104) were seeded in each well of black CLEAR flat-bottom 96-well plate (Greiner Bio-one). The cells were incubated overnight at 37°C with 5% CO2. On the following day, each serum was 2-fold serially diluted in the culture medium with the first dilution of 1:10. The diluted serum was incubated with 100 PFU of mNG SARS-CoV-2 at 37 °C for 1 h (final dilution range of 1:20 to 1:5120), after which the serum-virus mixtures were inoculated onto Vero E6 cell monolayer in 96-well plates. After 1 h of infection, the inoculum was removed and 100 μl of overlay medium (DMEM supplemented with 0.8% methylcellulose, 2% FBS, and 1% P/S) was added to each well. The plates were incubated at 37°C for 20 h. The raw images were acquired using Cytation 7 (BioTek) armed with 2.5× objective and processed using the default software setting. The foci in each well were counted and normalized to the non-serum-treated controls to calculate the relative infectivities. The curves of the relative infectivity versus the serum dilutions (log10 values) were plotted using Prism 9 (GraphPad). A nonlinear regression method was used to determine the dilution fold that neutralized 50% of mNG SARS-CoV-2 (defined as FFRNT). Each serum was tested in duplicates.

Plaque reduction neutralization test (PRNT).

A conventional 50% plaque-reduction neutralization test (PRNT50) was performed to measure the serum-mediated virus suppression as reported previously21. Individual sera were 2-fold serially diluted in culture medium with a starting dilution of 1:40 (dilution range of 1:40 to 1:1280). The diluted sera were incubated with 100 PFU of USA-WA1/2020 (WT) or mutant SARS-CoV-2. After 1 h incubation at 37°C, the serum-virus mixtures were inoculated onto 6-well plates with a monolayer of Vero E6 cells preseeded on the previous day. The minimal serum dilution that suppressed >50% of viral plaques is defined as PRNT50.

Extended Data

Extended Data Figure 1. The RNA/PFU ratios of different SARS-CoV-2 variants.

Extended Data Figure 1.

a, Diagram of SARS-CoV-2 spike variants. The spike genes from Alpha, Beta, Gamma, Epsilon, and Delta variants of SARS-CoV-2 were introduced into USA-WA1/2020 backbone. b, Ratios of viral genomic RNA versus plaque-forming unit (RNA/PFU) of SARS-CoV-2 spike variants. The genomic RNA and PFU of individual viral stocks were measured by RT-qPCR and plaque assay, respectively. The USA-WA1/2020 strain served as a control. Dots represent individual biological replicates from 4 aliquots of viruses. The means with 95% confidence intervals are shown. A non-parametric Mann-Whitney test was used to determine significant differences between USA-WA1/2020 and other variants. P values were adjusted using the Bonferroni correction to account for multiple comparisons. Differences were considered significant if P < 0.05; n.s., no statistical difference.

Extended Data Figure 2. Morbidity of hamsters after immunized with variant-spike SARS-CoV-2.

Extended Data Figure 2.

a, Hamster body weight loss after immunized with variant-spike SARS-CoV-2. The hamsters (n=4) were intranasally infected with 106 PFU viruses. The body weights were measured daily from day 0 to day 14 days post-immunization. The weight loss data are shown as mean ± standard deviation and statistically analyzed using two-way ANOVA Turkey’s multiple comparisons. The red stars show the statistical significance (*** P < 0.001) between USA-WA1/2020-immunized hamsters and Alpha+E484K-spike-immunized hamsters. b, Percentages of hamsters with or without diseases (including ruffled fur, lethargic, hunched, and reluctance to move when stimulated) from day 1 to day 14 post-immunization.

Extended Data Figure 3. Correlation between FFRNT50 and PRNT50.

Extended Data Figure 3.

a, Diagram of mNG USA-WA1/2020 and spike variants. mNG, mNeongreen fluorescence protein gene. b, Workflow of fluorescent foci reduction neutralization (FFRNT) assay. The details of the FFRNT assay were described in the Methods. c, Representative images of foci formed in a 96-well plate after 20 h of infection. d, FFRNT50, and PRNT50 values for four human sera. The FFRNT50 values are shaded in green. e, Correlation of FFRNT50 and PRNT50. The Pearson’s correlation coefficients and P values (two-tailed) are indicated.

Extended Data Figure 4. FFRNT50s of hamster sera against mNG SARS-CoV-2 spike variants on days 14, 28, and 45 post-immunization.

Extended Data Figure 4.

a-e, Hamster (n=4 per group) were immunized with WT USA-WA1/2020 (a), Alpha+E484K-spike virus (b), Beta-spike virus (c), Gamma-spike virus (d), Epsilon-spike virus (e). Sera were collected on days 14, 28, and 45 post-immunization and tested for neutralizing activities against the indicated mNG viruses by FFRNT. The original FFRNT50 values are presented in Extended Data Tables 13.

Extended Data Table 1.

FFRNT50s of twenty hamster sera on day 14 post-immunization.

Virus for infection Hamster ID FFRNT50s against SARS-CoV-2 spike variants
USA-WA1/2020 mNG Alpha+E484K-spike mNG Beta-spike mNG Gamma-spike mNG Epsilon-spike mNG
USA-WA1/2020 1 1048 320 194 446 1113
2 1875 1158 498 785 1916
3 1643 242 143 413 1304
4 1426 528 149 637 1099
Mean 1498 562 246 570 1358
Alpha+E484 K-spike 5 689 5561 1789 1660 1111
6 669 5526 2214 2543 864
7 1431 2981 3122 2967 762
8 714 3087 2175 2318 1246
Mean 876 4289 2325 2372 996
Beta-spike 9 461 1079 1610 2226 296
10 188 1482 2537 2520 161
11 51 1082 1947 1486 40
12 144 1113 1881 1802 208
Mean 211 1189 1994 2009 176
Gamma-spike 13 108 1215 2525 2833 49
14 125 583 1006 1346 <10
15 155 967 1075 2166 14
16 82 1617 1275 3092 52
Mean 118 1096 1470 2359 38
Epsilon-spike 17 966 337 156 194 2346
18 2602 502 358 583 5297
19 1535 457 175 179 2977
20 1552 2637 1572 1321 3085
Mean 1664 983 565 569 3426

Extended Data Table 2.

FFRNT50s of twenty hamster sera on day 28 post-immunization.

Virus for infection Hamster ID FFRNT50s against SARS-CoV-2 spike variants
USA-WA1/2020 mNG Alpha+E484K-spike mNG Beta-spike mNG Gamma-spike mNG Epsilon-spike mNG Delta-spike mNG
USA-WA1/2020 1 1226 632 714 1601 1167 1278
2 2140 1807 1749 1442 2646 2691
3 1137 141 93 392 920 713
4 1336 321 197 442 594 1068
Mean 1460 725 688 969 1332 1438
Alpha+E484 K-spike 5 445 3007 963 1169 288 430
6 1239 4511 1453 1920 950 1044
7 792 3174 1248 1507 569 867
8 291 3787 2405 2425 349 343
Mean 692 3620 1517 1755 539 671
Beta-spike 9 1511 3184 3494 4027 1850 2068
10 241 1671 2048 2209 230 293
11 1144 2253 2848 3473 336 349
12 290 931 1059 1932 261 299
Mean 797 2010 2362 2910 669 752
Gamma-spike 13 914 3874 5009 4685 564 480
14 1837 4197 4954 8391 498 535
15 247 707 1282 2090 73 85
16 150 1068 2555 3644 137 153
Mean 787 2462 3450 4703 318 313
Epsilon-spike 17 1961 1484 1080 770 3683 5442
18 3347 1153 1432 1373 4163 3634
19 1654 662 455 562 2132 1868
20 2298 3833 2251 1545 4182 3605
Mean 2315 1783 1305 1063 3540 3637

Extended Data Table 3.

FFRNT50s of twenty hamster sera on day 45 post-immunization.

Virus for infection Hamster ID FFRNT50s against SARS-CoV-2 spike variants
USA-WA1/2020 mNG Alpha+E484K-spike mNG Beta-spike mNG Gamma-spike mNG Epsilon-spike mNG Delta-spike mNG
USA-WA1/2020 1 4574 3509 3590 4437 4843 6410
2 2299 2042 2094 1850 2207 2337
3 953 510 370 807 1125 1612
4 2205 1682 723 1707 871 1232
Mean 2508 1936 1694 2200 2262 2898
Alpha+E484 K-spike 5 718 3794 2628 3627 601 462
6 1326 5262 2624 3211 1248 719
7 1237 4554 2115 3225 844 477
8 405 4930 2181 2210 372 248
Mean 922 4635 2387 3068 766 477
Beta-spike 9 1881 3305 4574 3994 2047 2005
10 582 1852 2857 2527 545 584
11 1184 2374 3746 3139 906 988
12 920 1692 1554 2068 688 680
Mean 1142 2306 3183 2932 1047 1064
Gamma-spike 13 1391 3638 7481 7345 1272 1505
14 2251 5095 7255 7892 1282 1566
15 656 1638 1274 2734 203 268
16 828 2521 3882 3986 514 633
Mean 1282 3223 4973 5489 818 993
Epsilon-spike 17 1982 1315 2236 1548 3379 2149
18 2205 1991 2742 3084 4212 4069
19 1928 1182 1158 1326 2419 2208
20 2930 4189 3004 2268 4641 4016
Mean 2261 2169 2285 2057 3663 3111

Acknowledgments

We thank Phillip R. Dormitzer for his helpful discussions during the study. P.-Y.S. was supported by NIH grants HHSN272201600013C, AI134907, AI145617, and UL1TR001439, and awards from the Sealy & Smith Foundation, the Kleberg Foundation, the John S. Dunn Foundation, the Amon G. Carter Foundation, the Gilson Longenbaugh Foundation, and the Summerfield Robert Foundation. S.C.W. was supported by NIH grant R24 AI120942. P.R. and X.X. were partially supported by the Sealy & Smith Foundation. J.L. was supported by James W. McLaughlin Fellowship Fund.

Footnotes

Competing financial interests

X.X. and P.-Y.S. have filed a patent on the reverse genetic system. Other authors declare no competing interests.

Data availability

The data that support the findings of this study are available from the corresponding authors upon reasonable request.

References

  • 1.Walsh E. E. et al. Safety and Immunogenicity of Two RNA-Based Covid-19 Vaccine Candidates. N Engl J Med 383, 2439–2450, doi: 10.1056/NEJMoa2027906 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Smith E. C., Blanc H., Surdel M. C., Vignuzzi M. & Denison M. R. Coronaviruses lacking exoribonuclease activity are susceptible to lethal mutagenesis: evidence for proofreading and potential therapeutics. PLoS Pathog 9, e1003565, doi: 10.1371/journal.ppat.1003565 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Korber B. et al. Tracking Changes in SARS-CoV-2 Spike: Evidence that D614G Increases Infectivity of the COVID-19 Virus. Cell, doi: 10.1016/j.cell.2020.06.043 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Yurkovetskiy L. et al. Structural and Functional Analysis of the D614G SARS-CoV-2 Spike Protein Variant. Cell 183, 739–751, doi: 10.1016/j.cell.2020.09.032 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Plante J. A. et al. Spike mutation D614G alters SARS-CoV-2 fitness. Nature 592, 116–121, doi: 10.1038/s41586-020-2895-3 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hou Y. J. et al. SARS-CoV-2 D614G variant exhibits efficient replication ex vivo and transmission in vivo. Science, doi: 10.1126/science.abe8499 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zhou B. et al. SARS-CoV-2 spike D614G change enhances replication and transmission. Nature 592, 122–127, doi: 10.1038/s41586-021-03361-1 (2021). [DOI] [PubMed] [Google Scholar]
  • 8.Chen R. E. et al. Resistance of SARS-CoV-2 variants to neutralization by monoclonal and serum-derived polyclonal antibodies. Nat Med 27, 717–726, doi: 10.1038/s41591-021-01294-w (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Xie X. et al. Neutralization of SARS-CoV-2 spike 69/70 deletion, E484K and N501Y variants by BNT162b2 vaccine-elicited sera. Nat Med 27, 620–621, doi: 10.1038/s41591-021-01270-4 (2021). [DOI] [PubMed] [Google Scholar]
  • 10.Liu Y. et al. Neutralizing Activity of BNT162b2-Elicited Serum. N Engl J Med 384, 1466–1468, doi: 10.1056/NEJMc2102017 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Liu Y. et al. BNT162b2-Elicited Neutralization against New SARS-CoV-2 Spike Variants. N Engl J Med, doi: 10.1056/NEJMc2106083 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Liu J. et al. BNT162b2-elicited neutralization of B.1.617 and other SARS-CoV-2 variants. Nature, doi: 10.1038/s41586-021-03693-y (2021). [DOI] [PubMed] [Google Scholar]
  • 13.Edara V. V. et al. Infection and Vaccine-Induced Neutralizing-Antibody Responses to the SARS-CoV-2 B.1.617 Variants. N Engl J Med, doi: 10.1056/NEJMc2107799 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Liu Y. et al. The N501Y spike substitution enhances SARS-CoV-2 transmission. bioRxiv, doi: 10.1101/2021.03.08.434499 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hanada K., Suzuki Y. & Gojobori T. A large variation in the rates of synonymous substitution for RNA viruses and its relationship to a diversity of viral infection and transmission modes. Mol Biol Evol 21, 1074–1080, doi: 10.1093/molbev/msh109 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Moustafa A. M. et al. Comparative Analysis of Emerging B.1.1.7+E484K SARS-CoV-2 Isolates. Open Forum Infect Dis 8, ofab300, doi: 10.1093/ofid/ofab300 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Xie X. et al. An Infectious cDNA Clone of SARS-CoV-2. Cell Host Microbe 27, 841–848 e843, doi: 10.1016/j.chom.2020.04.004 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Abu-Raddad L. J., Chemaitelly H., Butt A. A. & National Study Group for, C.-V. Effectiveness of the BNT162b2 Covid-19 Vaccine against the B.1.1.7 and B.1.351 Variants. N Engl J Med 385, 187–189, doi: 10.1056/NEJMc2104974 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lopez Bernal J. et al. Effectiveness of Covid-19 Vaccines against the B.1.617.2 (Delta) Variant. N Engl J Med 385, 585–594, doi: 10.1056/NEJMoa2108891 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Xie X. et al. Engineering SARS-CoV-2 using a reverse genetic system. Nature Protocols 16, 1761–1784, doi: 10.1038/s41596-021-00491-8 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Muruato A. E. et al. A high-throughput neutralizing antibody assay for COVID-19 diagnosis and vaccine evaluation. Nature Communications 11, 4059, doi: 10.1038/s41467-020-17892-0 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The data that support the findings of this study are available from the corresponding authors upon reasonable request.


Articles from bioRxiv are provided here courtesy of Cold Spring Harbor Laboratory Preprints

RESOURCES