Introduction

Continuously, sub-lineages of Omicron spread across the world since Omicron breakthrough in Nov. 2021, with increased transmissibility1 and resistance to humoral immunity of convalescent and vaccine2,3. BQ.1.1, XBB.1.5, and XBB.1.16 have been reported to have immune evasion greater than those of earlier sub-lineages of Omicron1,4,5. BQ.1.1 has evolved from the BA.5 sub-lineage with three additional mutations (R346T, K444T, and N460K) on the receptor-binding domain (RBD) of spike (S) protein. XBB.1.5 is a recombinant of the Omicron sub-lineages of BA.2.75 and BJ.1 with five more RBD substitutions (R346T, L368I, V445P, F486P, and F490S) than BA.2.756,7,8. XBB.1.16, another descendent lineage of XBB with two additional mutations (E180A, T478R) on the spike compared to XBB.1.5, has been spread rapidly in 31 countries9. Due to the additional RBD mutations, BQ.1.1, XBB.1.5, and XBB.1.16 became resistant to a wide range of mAbs and antibody cocktails, including those authorized for emergency clinical use, such as LY-CoV1404 (also known as bebtelovimab) and Evusheld (also known as tixagevimab and cilgavimab cocktail)2,4,10,11.

Thousands of neutralizing antibodies targeting different epitopes of the spike protein have been identified and characterized12,13,14,15,16,17,18,19. The majority of these antibodies recognize the RBD of the spike protein, while a small subset targets the NTD, SD1, SD2, or S2 stem-helix20. S3H3 antibody21 targeting SD1 as well as 12-16 and 12-19 antibodies22 targeting the NTD-SD1 were able to neutralize XBB.1.5 and BQ.1.1. Antibodies targeting RBD have proven effective in neutralizing SARS-CoV-2. However, due to the viral evolution pressure and high genetic variability of the RBD, most of the RBD antibodies failed to neutralize emerging SARS-CoV-2 variants of concern (VOCs), such as BQ.1.1, XBB.1.5, and XBB.1.16. Only the cross-neutralizing antibodies targeting the conserved epitopes of sarbecoviruses in RBD, such as S309, SA55, and S2K146, remained effective for current SARS-CoV-2 VOCs10,11. The S2 subunit is highly conserved, and the neutralization activities of S2 stem-helix antibodies were less affected by the viral escape mutation23. However, clinical application is unfortunately limited because of its low potency.

With the emergence and rapid dissemination of SARS-CoV-2 variants, the imperative for broadly neutralizing antibodies capable of pan-sarbecovirus neutralization has intensified. Antibodies possessing extensive neutralization across various sarbecoviruses are more likely to target conserved epitopes, rendering them more resilient against immune evasion by swiftly emerging SARS-CoV-2 variants. Consequently, the development of novel pan-sarbecovirus antibodies exhibiting broad and potent activity has become an urgent necessity for both the prevention and treatment of coronavirus disease 2019 (COVID-19). Moreover, considering that coronaviruses have incited two pandemics and one epidemic within the past two decades involving SARS-CoV, SARS-CoV-2, and MERS-CoV, the demand for pan-sarbecovirus antibodies becomes paramount in anticipation of potential recurrences of coronavirus pandemics. Here, we identified an RBM-binding antibody, designated 7F3, and demonstrated its unique capability to neutralize various SARS-CoV-2 VOCs, including the newly emerging Omicron subvariants BQ.1.1, XBB.1.5, and XBB.1.16. Importantly, G7-Fc, a bispecific antibody incorporating GW01 and 7F3 antibodies, showed broad neutralization of SARS-CoV-2 variants and sarbecoviruses reported. G7-Fc prevented XBB.1 infection in mice and bound two RBD sites: 7F3 bound a conserved epitope within the RBM, and GW01 bound an epitope external to the RBM. These studies highlight a conserved epitope within the RBM region targeted by 7F3 that mediated the neutralization of the highly immune-evasive Omicron variants BQ.1.1, XBB.1.5, and XBB.1.16. G7-Fc may represent a potential prophylactic countermeasure against SARS-CoV-2.

Results

Isolation of a BQ.1.1 and XBB lineages neutralizing antibody 7F3 from a COVID-19 convalescent individual

We sorted and cultured memory B cells from a patient who had recovered from COVID-19 (Supplementary Fig. S1). After culture of the B cells for two weeks, we screened the supernatants of B cells for SARS-CoV-2 neutralization. We identified a SARS-CoV-2 neutralizing antibody (NAb), designated 7F3, which belongs to the IGHV3-49 and IGKV3-11 immunoglobulin gene families. The antibody 7F3 exhibited potent binding to the RBD proteins of the SARS-CoV-2 (Wuhan-Hu-1 strain,GenBank: MN908947.3) wild-type (WT), Delta, BQ.1.1, XBB, XBB.1.5, and XBB.1.16 variants, and weak binding to the BA.1 and BA.5 RBDs, as measured by enzyme-linked immunosorbent assay (ELISA, Fig. 1a). In contrast, the control broadly neutralizing monoclonal antibody LY-CoV1404 bound robustly only to the RBDs of wild-type (WT), Delta, BA.1, and BA.5 variants but did not engage the BQ.1.1, XBB, XBB.1.5, or XBB.1.16 RBDs. We previously isolated a broadly NAb from a COVID-19 convalescent, named GW01Full size image

The binding of G7-Fc induces S trimers to the state with all three RBDs up. Due to the distance limitation of the GS linkers, 7F3 and GW01 from one arm of the G7-Fc bind to different RBDs, with 7F3 binding to RBM and GW01 binding to a non-RBM region. Two 7F3s are close to each other but do not form close interactions. The Fc regions cross-link two S trimers, forming an unsymmetrical head-to-head dimer of trimers (Fig. 4a, Supplementary Fig. S7). 7F3 did not compete with GW01 for RBD engagement (0%, Fig. 4b), implying that the 7F3 epitope likely differs from those of GW01 within the RBM. Antibodies directed towards the RBD can be classified into four general categories (classes I to IV) based on their competition with the ACE2 and their recognition of the up or down state of the three RBDs in spike27. 7F3 strongly competed with the class II antibody BD2328 for Wuhan-Hu-1 (WT) RBD engagement (100%, Fig. 4b). In contrast, CR3022 showed limited competition with 7F3 (17.4%). Moreover, CR302229, class IV, exhibited strong competition to the WT RBD compared to GW01 (96.21%, Fig. 4c), whereas 7F3 and BD23 showed no competition or limited competition with 7F3 (0% and 34.81%, respectively, Fig. 4c). These findings suggest a substantial epitope overlap exists between 7F3 and BD23, and overlap between CR3022 and GW01 epitopes. Based on the Barnes Classification of antibodies, 7F3 is categorized as a class II antibody, whereas GW01 is classified as a class IV antibody.

7F3 binds to the RBM region and shares 26 epitope residues with the ACE2 binding site (Fig. 4e, f, Supplementary Fig. S8b). The binding of 7F3 to RBD buries 976.4 Å2 surface area (Supplementary Table S3). The interactions between 7F3 and RBD are mainly contributed by CDRH2, CDRH3, and CDRL3 (Fig. 5a). Residues Y449, Y453, L455, F456, Y473, A475, G476, N477, K478, P479, A484, G485, S486, N487, C488, Y489, S490, P491, L492, Q493, S494, Y495, G496, R498, Y501, and H505 of RBD participate in the interactions with 7F3, forming 18 pairs of hydrogen bonds (Fig. 5b, Supplementary Table S4). In addition, Y229 of CDRH3 packs against a hydrophobic pocket in the interface formed by L455, F456, Y473, and Y489 of RBD (Fig. 5b). Structure comparison reveals that the binding modes of XBB S/7F3 and WT S/BD23 (PDB ID: 7BYR) are quite similar. Other than that, 7F3 was implicated in more interactions with RBD residues, and the E484A, F486S, Q498R, N501Y, and Y505H mutations may interfere with the contacts between BD23 and SARS-CoV-2 variants (Supplementary Fig. S8).

The epitope of GW01 is located outside RBM. We previously identified the epitope of GW01 on BA.1 S-RBD25. GW01 interacts with BA.1 and XBB S-RBD in a very similar way. The contacts between GW01 and the XBB S-RBD are mainly contributed by CDRH3 by forming hydrophobic interactions and 6 pairs of hydrogen bonds (Supplementary Table S5). Y226, N233, Y234, and E235 of CDRH3 interact with XBB S-RBD through hydrogen bonds. Y226, V231, F232, and Y234 of CDRH3 establish a large hydrophobic interface with Y369, F374, F375, A376, F377, V407, A435, V503, and Y508 of RBD (Fig. 5c). In addition, D155 of CDRH1 and D178 of CDRH2 are also involved in the interactions between GW01 and RBD (Fig. 5c). When compared with BA.1 S-RBD/GW01 structure, fewer residues of XBB S-RBD are involved in the GW01 interaction, resulting in a reduction of interface area by 91.7 Å2 in the XBB S-RBD/GW01 structure (Supplementary Table S3). This may explain why GW01 exhibits lower binding affinities to XBB S-RBD than BA.1 S-RBD. A comparison of XBB S/GW01 and WT S/CR3022 (PDB ID: 6W41) reveals that the CR3022 epitope, which primarily consists of L368-F392 residues, is located near the bottom of the RBD. The S371F, S375F, T376A, and R408S mutations may cause the loss of binding affinity of CR3022 to SARS-CoV-2 variants (Supplementary Fig. S9).

To characterize the key epitope of G7-Fc, we analyzed the residues involved in G7-Fc binding (Supplementary Fig. S8b, 9b) and constructed eight XBB single mutants with crucial roles in G7-Fc binding. The K378A, Y473A, Y489A, Q493A, S494A, and Y501A mutants dramatically decreased (>10 fold and <100 fold) or abolished (>100 fold) G7-Fc neutralization (Fig. 5d). The K378 forms a hydrogen bond with GW01 (Fig. 5c), whereas the Y473, Y489, Q493,S494, and Y501 residues form hydrogen bonds with 7F3. The residues involved in the G7-Fc epitope exhibit a high degree of conservation, particularly with K378, Y473, Y489, Q493, S494, and Y501, showcasing over 95% conservation in variant sequences worldwide since Jan 2023 according to the CoV-Spectrum of GISAID database (https://cov-spectrum.org/explore/World/AllSamples/AllTimes/variants?variantQuery=S%3A466R&) (Fig. 5d, Supplementary Table S6). The conserved binding epitope of 7F3 conferred notable binding breadth to a panel of 37 RBD mutant proteins with single or triple mutations (Fig. 1e). Despite the Y501A mutation leading to escape from G7-Fc neutralization, an analysis of global SARS-CoV-2 genomic databases since January 2023 revealed only 6 reported sequences containing this 501A mutation. The mutation rates of other residues in position Y501 worldwide are notably low (Supplementary Fig. S10b), indicating that escape mutations in position Y501 are uncommon. Importantly, this suggests that G7-Fc can potently neutralize the vast majority of currently circulating SARS-CoV-2 strains. Analysis of the structure of LY-CoV1404 complexed with WT RBD (PDB ID: 7MMO) showed that the epitope of LY-CoV1404 is of low conservation at R346, N440, K444-G446, L452, and Q498 (Supplementary Fig. S10a and Fig. 5e), which explains its decrease of binding affinity of SARS-CoV-2 variants. In detail, the R346T, N440K, K444T, L452R, and Q498R mutations may result in the escape of BQ.1.1 (Fig. 5f). The V445P and G446S disrupt the hydrophobic interface between V445 and L52, Y54, K60 of heavy chain and Y94 of light chain, which may contribute to the escape of XBB, XBB.1.5, and XBB.1.16 (Fig. 5g). These results indicate that G7-Fc bispecific antibody binds to two complementary epitopes across variants of the Omicron lineage of SARS-CoV-2, thereby explaining the ability of G7-Fc to neutralize even the highly antigenically evasive Omicron variants.

Neutralization mechanism of the bispecific antibody G7-Fc

To investigate the neutralization mechanism of the bispecific antibody G7-Fc, we performed a BLI competition assay and tested whether G7-Fc blocks RBD/ACE2 binding. G7-Fc prevented the RBD protein and the S trimer of XBB binding to ACE2 protein in the competition assay, while the control antibody VRC01, an HIV-1 gp120 binding antibody, did not affect the RBD/ACE2 interaction (Fig. 4d). Structural alignment of the G7-Fc/RBD complex with the ACE2/RBD complex indicated that both 7F3 and GW01 were able to compete with ACE2 when binding to the RBD (Fig. 4f), which was consistent with the competition assay (Fig. 4d).

These findings suggest that G7-Fc inhibits SARS-CoV-2 XBB variant infection by synergistically inducing the formation of trimer-dimers. Both 7F3 and GW01 bind to conserved epitopes in XBB, enlarging the interface area, thereby improving the affinity between the RBD and single scFv and blocking the RBD from interacting with the ACE2 receptor. The structural arrangement of the bispecific antibody, particularly the Fc region, plays a critical role in facilitating G7-Fc binding to the trimer.