Open Access

Current development of severe acute respiratory syndrome coronavirus 2 neutralizing antibodies (Review)

  • Authors:
    • Tong Zhang
    • Di Yang
    • Liang Tang
    • Yu Hu
  • View Affiliations

  • Published online on: June 26, 2024     https://doi.org/10.3892/mmr.2024.13272
  • Article Number: 148
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The coronavirus disease 2019 pandemic due to severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) seriously affected global public health security. Studies on vaccines, neutralizing antibodies (NAbs) and small molecule antiviral drugs are currently ongoing. In particular, NAbs have emerged as promising therapeutic agents due to their well‑defined mechanism, high specificity, superior safety profile, ease of large‑scale production and simultaneous application for both prevention and treatment of viral infection. Numerous NAb therapeutics have entered the clinical research stages, demonstrating promising therapeutic and preventive effects. These agents have been used for outbreak prevention and control under urgent authorization processes. The present review summarizes the molecular targets of SARS‑CoV‑2‑associated NAbs and screening and identification techniques for NAb development. Moreover, the current shortcomings and challenges that persist with the use of NAbs are discussed. The aim of the present review is to offer a reference for the development of NAbs for any future emergent infectious diseases, including SARS‑CoV‑2.

Introduction

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in a global pandemic of coronavirus disease 2019 (COVID-19). Although infection typically starts with flu-like symptoms (1), patients can also be asymptomatic and may go on to have a mild to severe disease course (2). Mechanistically, COVID-19 is characterized by an important burden of inflammation mainly in the respiratory system, although it can also affect other organ systems (3,4). Previous studies have assessed the association between hemogram indices and COVID-19 pathogenesis, improving the prognostic judgement and patient management of this disease (5,6). Nevertheless, a requirement for safe prevention and treatment methods for COVID-19 remains. Efforts for this are currently ongoing, including the development of numerous types of vaccines, attempts to establish active herd immunity, exploration of the efficacy of existing drugs and the development of novel small-molecule drugs targeting viral or host proteins to inhibit the replication of viruses (79). The majority of patients with SARS-CoV-2 infections typically exhibit an antibody response 5–15 days after the occurrence of symptoms, peaking at 21–28 days, before declining (10,11). Given the time it takes to develop immunity, this is a dangerous period for individuals who are at high risk of exposure, who are immunocompromised, or who are unable to acquire antibodies from active immunization. Thus, prompt passive immunization is urgently required (12).

The earliest attempt at passive immunization was through convalescent plasma infusion, which provides neutralizing antibodies (NAbs), which can alleviate the inflammatory burden (13). Convalescent plasma is plasma donated by individuals previously infected with infectious diseases, who have produced protective NAbs 14–28 days after infection (14). Although early clinical trials on convalescent plasma transfusion reported promising results, its shortcomings were not negligible. For the treatment to work effectively, patients should ideally receive an infusion during the early stages of infection, whilst being monitored to avoid risks associated with blood transfusions, such as hyperinflammatory immune reactions and transfusion-associated circulatory overload (15). Moreover, the screening and monitoring of plasma donors is labor and resource-intensive. There is also a lack of a universally accepted standard for the infusion volume. Recent clinical trials have reported that convalescent plasma transfusion does not significantly reduce mortality, as its neutralization capacity declines after viral mutations (16,17). However, the total antibody titer following human COVID-19 immunoglobulin intravenous injection was previously reported to be three times higher compared with that of convalescent plasma, suggesting this treatment to be more effective. pH4 is manufactured from already inactivated, filtered and purified convalescent plasma of patients infected with SARS-CoV-2. It contains high purity and high titer SARS-CoV-2 NAbs, which have been reported to effectively neutralize SARS-CoV-2 in vivo. A number of clinical trials have reported that the intravenous injection of human immunoglobulin can abridge the duration of positive PCR confirmation and inflammation, as evidenced by computed tomography data in patients (18,19). According to the diagnostic and treatment guidelines in China, intravenous immunoglobulins can be used in an emergency for patients with rapid severe disease progression (20). Furthermore, another study has previously proposed the potential application of pH4 for patients with SARS-CoV-2 infection with low immunoglobin M levels (21).

However, one limitation of convalescent plasma and intravenous immunoglobulin is the requirement to recruit blood donors with high titers of NAbs against the pathogens of interest to maintain a stable, sufficient supply. Identification of single B cells that produce virus-specific neutralizing monoclonal antibodies (mAbs) from these donors can potentially be used to circumvent this limitation. The immunoglobulin gene expressed in B cells can be cloned and expressed to produce high titers of neutralizing monoclonal antibodies (22). Large-scale preparations of specifically targeting mAbs that are of high purity with potent neutralizing activity can render them powerful tools for the prevention and treatment of infectious diseases (23). For SARS-CoV-2, specific B cells have been obtained from patients recovered from COVID-19 to produce large quantities of humanized mAbs through numerous methods, including mouse hybridoma fusion, single B cell sorting, phage display and transgenic mice and antibody screening technologies (2426). A number of NAbs against SARS-CoV-2 have been developed. According to randomized double-blinded controlled clinical trials, the Food and Drug Administration has previously approved the emergency use of certain NAb drugs for the treatment of SARS-CoV-2 such as bamlanivimab, etesevimab, casirivmab and imdevimab (27,28). Furthermore, The State Drug Administration of China has authorized Amubarvimab and Romlusevimab cocktail therapies, which were designed for treating adults or young individuals (12–17 years old, weight ≥40 kg) with mild and common types of SARS-CoV-2 infection who are at high risk of developing severe type infections (29). This treatment strategy is now serving a pivotal role in the clinical treatment process in Chinese hospitals to reduce incidence of serious adverse events (30). Compared with convalescent plasma, these types of NAbs confer a number of distinct advantages. The final selection of the most effective candidate antibody can be evaluated, with the IC50 value typically on nanomolar or picomolar levels, and the dose can be evaluated more accurately (31). Furthermore, the probability of the antibody-dependent enhancement (ADE) phenomenon associated with this type of NAbs, wherein non-neutralizing antibodies facilitate the entry or replication of a pathogen, thereby potentiating infection rather than providing protection, is considerably lower (32).

New variants continue to emerge, such as the ο variants, which are notably more transmissible compared with those of the original strain, and have negatively impacted the health and normal life of the global population (33). Mutations that produce mutant forms of the spike (S) protein have resulted in the reduction or disappearance of the efficacy of vaccines and NAbs, increasing the demand for the development of next generation vaccines and antibody drugs (34). Nanobodies (Nbs) are heavy chain antibodies, which have been previously reported to be found in camelids and cartilaginous fish, such as sharks (35). They form the smallest known complete functional structure that can target virus antigens. Numerous characteristics of Nbs, such as their small sizes, high specificity, stability, ease of production, potent penetration and low immunogenicity, render them able to recognize antibody epitopes that cannot be readily recognized by conventional antibodies (36). This would in turn increase the diversity of potential targets and binding ability of antibodies, providing a broader and more targeted choice for the research and development (R&D) of next generation antibody drugs with wide clinical implications.

In the present review (Fig. 1), the current application status of NAbs is discussed from the perspective of the epitope features in SARS-CoV-2, the current preparation techniques available and the latest research status of traditional NAbs and Nbs, including persisting challenges and future prospects.

Structural basis and process of SARS-CoV-2 infection

Structure of SARS-CoV-2 transcriptome

There are currently four known genera of coronaviruses, specifically α, β, γ and δ. β coronaviruses can be classified into subgenera A, B, C and D (37). SARS-CoV-2 is a member of the B subgenus of β coronaviruses. SARS-CoV-2 is a single-stranded, positive-sense RNA virus that can directly guide protein synthesis upon entering the cell to replicate itself, by generating negative strands using RNA polymerase (38). The RNA sequence of SARS-CoV-2 contains 29,891 nucleotides, encoding 9,860 amino acids with a GC percentage of ~38%. Furthermore, there is a 5′ cap-like structure and a 3′ poly-A tail on its genome (Fig. 2) (39). In total, two overlapping open reading frames (ORFs), namely ORF1a and ORF1b, encode 16 non-structural proteins (40). The −1 frameshift between ORF1a and ORF1b contributes to the production of polypeptide 1a and a larger polypeptide 1ab. For genome amplification, SARS-CoV-2 viruses generate antisense RNAs as templates for creating the sense genomic RNA (gRNA) and subgenomic RNA (sgRNA). gRNA together with the structural proteins expressed contributes to producing the viral offspring. The shorter sgRNA has the role of expressing the four types of conserved structural proteins in coronaviruses, namely S protein, membrane protein, envelope protein and nucleocapsid protein, as well as six auxiliary proteins (3, 6, 7a, 7b, 8 and 10) (41).

S proteins

The S protein is a type I fusion protein that mediates viral entry into targeted cells. It is a major target of post-infection NAbs and the main focus of numerous therapy and vaccine studies. The S protein forms a surface-exposed trimer on the viral particles and consists of a long extracellular region, a transmembrane region and an intramembrane region (42). On the surfaces of targeted cells, the S protein binds with angiotensin converting enzyme 2 (ACE2) before undergoing structural transformations to induce the fusion of the viral and cell membranes. Each S protein contains 1,273 amino acids, including an N-terminal signal peptide, a receptor-binding part S1 and a fusion part S2. The S protein has been previously studied using cryoelectron microscopy (Cryo-EM), which reported two structural states (43,44). While in the closed state, three receptor binding domains (RBDs) of the S protein are locked in the ‘down’ conformation, whereas during the open state, there is one RBD in the ‘up’ conformation. The RBDs form the key region necessary for SARS-CoV-2 interactions with ACE2, where its open state is the prerequisite for virus-cell membrane fusion (44,45).

S1 subunit

The S1 subunit has four domains, namely A, B, C and D, with domains A and B being responsible for receptor binding. The structure of domain A consists of a galectin-like β-fold, whereas domain B has a reverse parallel β-fold structure. There is an extended loop of domain B at the end of the virion that is structurally different depending on the species of the β-coronavirus, known as the hypervariable region (46). Domains C and D on the C-terminus constitute discrete segments of the primary protein sequence and are directly linked to the stem core of the S2 subunit to form a β-fold structure. The entire S1 subunit is connected by a ring covering the surface of S2 (47).

From the perspective of its linear peptide structure, S1 consists of the N-terminal domain (NTD), RBD and the C-terminal domain (CTD). Due to the swing of the S protein on the viral membrane with a 40 main angle of inclination, the perceived primary point of interaction with the epithelium is the NTD, which can be targeted by numerous powerful Nabs (4850). The NTD of the SARS-CoV-2 S protein can form a ribbon structure that is analogous to that of human galectin and is located at residues 14–305. This mediates weak and reversible interactions with superficial glycans, such as sialic acid, through low-affinity hydrogen bonds, is crucial for the virus to attach to and navigate along the cell surface, a process referred to as ‘viral surfing’. Subsequently, numerous critical residues of the NTD combine with sialic acid (51,52), forming a flat surface to strengthen the primary interaction between SARS-CoV-2 and targeted cells, consolidating the infection (53). The RBD is located at residues 336–525 of the S protein, which has two domains: A central structure consisting of five parallel β-sheets and an extended loop known as the receptor-binding motif (RBM) at residues 437–508. This extended loop serves to surround the edge of the central structure and interacts with ACE2 (31). In the down state, the receptor-binding motif (RBM) is partially obstructed, limiting its engagement with ACE2. Upon RBD's transition to the up state, the RBM becomes accessible to interact with ACE2, facilitating viral entry (54,55). At residues 528–685 of the S protein is the CTD, which mainly consists of a β-structure. CTD1 serves to ‘sense’ changes in its neighboring sites, whilst CTD2 is vital for membrane fusion with the entire rearranged S protein (46,56).

S2 subunit

The S2 subunit promotes membrane fusion and binds the S protein onto the host cell membrane. It is highly conserved among coronaviruses and contains important regions for promoting fusion with target cells (45). Upon RBD's engagement with the receptor, the fusion peptide (FP) is inserted into the cell membrane, which then triggers the unfolding of the heptad repeat 1 (HR1) domain and the folding back of the HR2 domain. This sequence of events leads to the domains coming together, causing the membranes to bend towards each other and facilitating membrane fusion, thus enabling viral entry (57). In particular, the FP proximal region in S2 appears to serve a supporting role in clamping the RBD and stabilizing the closed conformation of the S protein (46).

Viral entry into the cell

For all enveloped viruses, membrane fusion is the critical initial phase for entry into the targeted cell and the establishment of infection. There is a high dynamic barrier when the two membranes approach each other, where the free energy required to overcome the kinetic barrier comes from the rearrangement of the fusion protein encoded by the virus, specifically by changing from the basal variable conformational state to the stable state upon fusion. Subsequently, through two protein cleavage events, the S protein transforms into a state that can readily transition into a low-energy state. The first cleavage occurs at the boundary between S1 and S2, commonly known as the S1/S2 site. A four-amino acid residue Arg-Arg-Ala-Arg sequence is present at the S1/S2 site, which is cleaved by the protease furin (58). The prefusion S protein trimer fluctuates between closed and open conformations. It has been previously hypothesized that the near-universal expression of furin-like protease may have a role in enhancing the cellular and tissue tropism of SARS-CoV-2, thus enhancing its infectivity and/or modifying its pathogenicity. The second cleavage event occurs at the S2′ site. This cleavage site is only accessible after the initial S1/S2 cleavage and RBD-ACE2 binding. The differential access route to SARS-CoV-2 results in the S2′ site being cleaved by distinct proteases. The cleavage of S2′ site occurs on the cell surface and is mediated by transmembrane protease serine 2 (TMPRSS2). In the absence of TMPRSS2 or when the likelihood of encountering TMPRSS2 diminishes on the cell surface, the virus-ACE2 complex will be internalized by lectin-mediated endocytosis into the endolysosome, where the S2′ site is cleaved by cathepsins, particularly cathepsin L (59). In both of these access routes, S2′ cleavage releases the structural constraints on the FP, whilst the dissociation of S1 from S2 results in a drastic change in the conformation of the S2 subunit, particularly in HR1, driving the FP into the cell membrane. This forms a fusion pore through which viral RNA is delivered into the cytosol of the target cell.

Moreover, SARS-CoV-2 can be cleaved by serine endonuclease protein convertase 1, trypsin and trypsin-like integral membrane serine peptidase, all of which can readily recognize and cleave the S1/S2 site. The S protein is cleavable by a broader range of proteases compared with the SARS-CoV-related viruses, which is a critical contributor to its ease of entrance into targeted cells through the ACE2 pathway and infectiousness (60).

Current status of preparation methods and development of NAbs

Polyclonal antibodies (pAbs)

pAbs are prepared by the direct injection of antigens into animals for immunization, followed by serum collection and purification (61). Compared with monoclonal antibodies, they possess a higher affinity for target antigens as they can target multiple binding sites on a single antigen. Moreover, due to the inherent diversity of pAbs, they tend to be more resistant to the polymorphism of target antigens, retaining activity despite antigen glycosylation or other post-translational modifications (62). pAb therapeutics are associated with abbreviated production timelines and reduced manufacturing expenses relative to monoclonal antibody counterparts (63). However, due to the large divergence among batches of pAb therapeutics and the rapid development of more effective vaccines and monoclonal antibody therapy, the popularity of pAb drugs has gradually waned. At present, to the best of our knowledge, there have only been a small number of studies on the use of pAb against COVID-19.

A horse pAb therapy was previously reported to be safe and potent for treating SARS-CoV-2 (64). The efficiency of this antibody against RBD was reported to be ~50 times greater than that of normal convalescent plasma, and the resultant drug INM 005 (COVID-19) derived from this horse pAb has been approved for SARS-CoV-2 treatment in Argentina (65). Moreover, another previous study used the RBD region of the S protein to immunize pigs, which then produced polyclonal NAbs that do not interact with human Fc receptors, avoiding potential ADE effects (66). A purified polyclonal IgG fraction drug, XAV-19, was also reported to be able to neutralize the original Wuhan strain and subsequent variants, including the γ and δ variants. XAV-19 was also well tolerated in hospitalized patients with moderate COVID-associated pneumonia who required low-flow oxygenation, according to results from clinical trials (67,68).

mAbs
Production technologies

Commonly used monoclonal antibody screening technologies include traditional murine hybridoma fusion, transgenic mouse technology, single B cell isolation, cloning and phage display (69). The conventional murine hybridoma method yields antibodies derived from mice, which are susceptible to human anti-mouse antibody responses, limiting their clinical applicability. Advances in transgenic mouse technology have enabled the direct production of fully humanized antibodies without the need for subsequent humanization steps (70). This method entails genetically integrating human antibody genes into the mouse genome, followed by immunization of these transgenic mice to elicit fully human antibodies. These antibodies mature in vivo, exhibiting high affinity and specificity, thus establishing this platform as the preferred and auspicious route for antibody-based pharmaceutical development (71). Single B-cell sorting is a technique used to isolate individual B cells capable of producing specific NAbs from recovered individuals. The genes encoding the antibody are sequenced to allow the recombinant expression and purification of the required mAbs. This method has emerged as a cornerstone for the expedited development of anti-SARS-CoV-2 NAbs, as it allows for the rapid and high-throughput generation of human antibodies from peripheral blood mononuclear cells, while maintaining the native pairing of heavy and light chains (72). Phage display is a method that involves cloning the entire repertoire of genes from the variable regions of human antibodies and inserting them into phages harboring the coat protein gene. This results in the display of exogenous genes on the phage surface as fusion proteins, creating an antibody library. Screening this phage library with target proteins facilitates the rapid isolation of antibodies with high affinity for the desired target (73,74). Phage display technology outperforms the conventional hybridoma method in terms of speed, efficiency, and simplicity. Furthermore, it can be coupled with Nb technology to generate novel Nb variants characterized by reduced molecular weight, enhanced stability and increased neutralizing capacity (75).

RBD NAbs

RBD NAbs are considered to be the most abundant and potent of the SARS-CoV-2 Nabs (76,77). A prior investigation into the humoral immune response to SARS-CoV-2 infection showed that antibodies blocking the interaction between RBD and ACE2 led to a decrease in viral RNA expression to levels below detection, suggesting a crucial role for RBD-specific neutralizing antibodies in the response to SARS-CoV-2 infection (78). A number of RBD antibody categorization systems have been proposed, with the one devised by Barnes et al (76) being the most widely accepted. The approach employs a classification system comprising four categories, which are delineated by the structural characteristics and binding sites of the NAbs (Fig. 3). Class 1 RBD NAbs are characterized by their immunoglobulin heavy chain variable region (IGHV) of 3–53 or 3–66 genetic origin, short complementarity-determining region (CDR)3 of antibody heavy chains (CDRH3) of <15 residues in length, RBD ‘up’-state binding and ACE2 blocking capability. C105 (79), LY-CoV016 (27), B38 (80), CB6 (81) and CT-P59 (31), 1–20, 4–20 (49), 910–30 (82), S2E12 (83) and S2K146 (84) are representative examples of Class 1 RBD NAbs. They predominantly target or overlap with the ACE2 binding site at the RBM, competing with ACE2 for binding at this site. However, these antibodies do not bind adjacent RBDs (85,86).

Class 2 RBD antibodies are also able to bind to the ACE2 binding site by recognizing both the ‘up’- and ‘down’-state RBDs, with added specificity to neighboring RBDs. Unlike the Class 1 IGHV3-53 antibodies, Class 2 antibodies have CDRH3 loops that are >15 residues. The prominence of Class 2 antibodies in the RBD-targeting fraction of plasma may be partially attributed to their binding capacity to both the ‘up’ and ‘down’ states of RBD. They are typically produced by germline genes, including variable heavy-chain (VH) 1–2, VH1-69 and VH3-53 (87). C135, C110 (87), C144, C002, C104, C119, C121 (76), DH1041, DH1042 and DH1043 (88) belong to this class of RBD NAbs. These antibodies not only directly obstruct ACE2 engagement but can also bridge adjacent ‘down’ state RBDs, locking S proteins in a ‘closed’ pre-fusion state to inhibit S-ACE2 engagement. Class 1 and Class 2 NAbs do not cross-neutralize viruses due to the limited conservation of RBM structures in different β-coronavirus species. By contrast, a number of SARS-CoV-2 NAbs that can identify conserved RBD epitopes further away from the ACE2 engaging site have been reported to exhibit considerable cross-neutralizing ability. Such non-RBM-targeting RBD antibodies are categorized into structural classes 3 or 4. Class 3 RBD NAbs can identify both the ‘up’ state and ‘down’ state RBDs but do not inhibit ACE2 binding, whilst Class 4 RBD NAbs do not recognize the ‘down’ RBD conformation. For example, CR3022 is a weak Class 4 cross-reactive neutralizing antibody and is one of the most broad-spectrum coronavirus mAbs identified to date (89,90). The cryptic site targeted by CR3022 shares 86% similarity among SARS and SARS-CoV-2 (91). Furthermore, the Class 3 antibody S309, which was first identified in a blood sample from a patient with SARS in 2003, can inhibit a range of associated coronaviruses, including SARS-CoV-2. S309 recognizes a proteoglycan epitope accessible in both the ‘up’ and ‘down’ conformations of RBD but differs in the mode of binding to RBM on SARS-CoV-2 (25). In addition, the IgG1κ mAb sotrovimab was developed from the S309 antibody and was previously approved under emergency use authorization for mild-to-moderate COVID-19 cases in patients aged ≥12 years and weighing >40 kg because of its ability to reduce the risk of disease progression (92). However, since sotrovimab only targets a single viral antigenic epitope, resistance can readily develop in patients (93).

Other antibodies, such as DH1047 (94), ADI-56046, ADI-55689 (95) and ADG-2 (96), can block the interaction between ACE2 and S to inhibit SARS-CoV, SARS-CoV-2 and Bat SARS-like coronavirus infection. Specifically, DH1047 binds the SARS-CoV-2 RBD at an epitope outside of the N-terminal end of the RBM, unlike other known non-cross-neutralizing antibodies. Furthermore, the epitopes targeted by the ADG-2 antibody overlap but differ from DH1047. They are linked by rotation around the longitudinal axis of fragment antigen-binding (Fab), with the ADI-56046, ADI-55689 and ADG-2 antibodies preferring the ACE2 engaging region of RBD. These cross-neutralizing antibodies contact S proteins by an angle of proximity to the viral surface in a relatively horizontal manner (96), which indicates the importance of a highly conserved recognition pattern and horizontal angle of proximity for cross-neutralizing activity (95,97).

Using high-throughput surface plasmon resonance analysis and Cryo-EM structure determination, RBD antibodies can be classified into seven different ‘communities’ ranging from RBD-1 to RBD-7 according to their binding epitopes, giving rise to another categorization system (98). This categorization system offers a more detailed landscape of RBD NAbs, which can be used to complement the four aforementioned classes. Possessing non-overlapping epitopes and considerable potency, the neutralizing effects of RBD-1 to −4 clusters are highly susceptible to deletions and mutations in emerging SARS-CoV-2 variants. By contrast, RBD-5 to −7 antibodies are generally less potent, but the epitopes they target are highly conserved and therefore more resistant to mutations. Therefore, combining and/or engineering these antibodies into multivalent formulas can produce mutagenesis-resistant NAb therapeutic cocktails (98).

NTD NAbs

The NTD is an essential locus for the development of vaccines (49). NTD-targeting NAbs account for 5–20% of all S protein-targeting mAbs derived from memory B cells of patients with SARS-CoV-2 infections (99). The first NTD-targeting NAb, 4A8, was reported by Chi et al (100). Instead of blocking the S-ACE2 binding, the majority of NTD NAbs impede the fusion state change of the S protein (101). A number of NTD NAbs, such as S2L28, S2M28 and S2X333, have been reported to obstruct the TMPRSS2-independent infection pathway, a noteworthy pathway in S-protein-mediated infection of human lung cells (99,102). In general, NTD NAbs possess a common structure and genetic origin, use a similar set of VH genes and bind to the S protein in a similar manner (103). Notable examples of NTD-targeting NAbs include 1–68, 1–87, 2–51 (49), DH1049, DH1050.1, DH1050.2 (88) and 4A8 (48), which are all generated from the VH1-24 gene segment. In particular, the IGHV1-24 expression in B cells is ~10 times higher in patients with COVID-19 (5–8%) (85,104) compared with that in healthy individuals (0.4–0.8%) (105), underscoring the importance of the N-terminal domain (NTD)-targeting NAbs in the immune response to SARS-CoV-2. Consistent with the genetic and structural features of NTD NAbs are their binding sites. The NTD NAbs previously isolated by McCallum et al (99) were all reported to target a specific site (site 1). Other previous studies have also identified the same binding mechanism and subsequently named it the ‘antigenic supersite’ (99,103). The NTD is highly glycosylated, but site 1 has the largest non-glycosyl-modified surface on the NTD domain. A list of antibodies targeting the NTD antigenic supersite and their genetic origins are summarized in Table I.

Table I.

NTD antibodies and their neutralizing capacity.

Table I.

NTD antibodies and their neutralizing capacity.

NTH antibody (coding VH gene)Neutralizing mechanismBinding site(Refs.)
COV2-2676 (VH1-69), COV2-2489 (VH4-39)Inhibits post-attachment stepsSupersite(168)
4A8 (VH1-24)Limits the structural shifts of the S proteinSupersite(100)
S2X333 (VH3-33), S2M28 (VH3-33), S2L28 (VH3-21), S2X28 (VH3-30)Blocks membrane fusion, Fc-mediated effector functions promote Ab-dependent cytophagy and Ab-dependent cytotoxicity in vivoSupersite(99)
FC05 (VH1-24)Restricts conformational transition of RBD and blocks membrane fusionSupersite(106)
CM25 (VH1-24)Not reportedSupersite(169)
DH1050.1 (VH1-24)Not reportedSupersite(88)
4-8 (VH1-69), 2–17 (VH1-69), 5–24 (VH3-33)Not reportedSupersite(49)
ADI-56479 (not mentioned)Binds an epitope on adjacent S trimers which decreases their densitySupersite(107)
5-7 (VH1-46)Limits the structural shifts of the S proteinNon-supersite(170)
P008_056 (VH3-21)Blocks access to the ACE2 engaging siteNon-supersite(171)
C1520 (VH3-48), C1565 (VH3-48)Prevents the cleavage of the S2′ site or destabilizes S1Non-supersite(172)

[i] VH, variable heavy-chain; Ab, antibody; Fc, fragment crystallizable; S protein, spike protein; ACE2, angiotensin converting enzyme 2; NTD, N-terminal domain; RBD, receptor binding domain.

Although NAbs generally achieved effective neutralization against the original 2019 strain of SARS-CoV-2, with the emergence of variants of concerns (VOCs), the NTD region has been reported to be highly variable. Antibodies targeting the supersite of NTD appeared to be especially susceptible to potency loss, with a large number losing their affinity to α, β and γ VOCs. However, numerous NTD NAbs, such as 5–7, C1520 and C1565, remain capable of binding other sites, thereby remaining effective even for the ο variant BA.1. However, NTD NAbs combined with RBD antibodies have been proposed to be a more effective therapeutic cocktail for treating the variants. The combination of FC05 with H014, HB27 and P17 (106), as well as the combination of ADI-56479 with ADI-56443 (107), are both examples of combining NTD-targeting mAbs with RBD-targeting mAbs, and have been reported to result in lower virus escape compared with either class of NAbs when used alone. This combination was also proposed to reduce S protein mutations that lead to neutralization escape.

S2 NAbs

In a study investigating SARS-CoV-2-related IgG antibodies, the majority of healthy individuals who had not been exposed to SARS-CoV-2 exhibited the presence of IgG antibodies targeting the S2 subunit in their sera (108). Numerous studies have previously suggested that S2-targeting antibody responses against SARS-CoV-2 are associated with superior outcomes for patients and broader neutralization, suggesting the crucial protective effect of this type of antibodies (109111). S2 is more conserved compared with S1 and shows 63–98% sequence similarity among the same protein from seven different human coronaviruses (112). S2 contains numerous conserved antigenic sites, including the stem helix, the FP and the hinge region. S2 NAbs tend to inhibit the formation of the six-helix bundle structures by HR1 or HR2, thereby blocking membrane fusion and viral entry. Certain S2 antibodies are summarized in Table II. Nevertheless, S2 NAbs appear to rely on the Fc mechanism for protection in vivo, although combination with the Fc has been reported to be a promising strategy in the development of antibody drugs (113).

Table II.

S2 antibodies and their neutralizing capacity.

Table II.

S2 antibodies and their neutralizing capacity.

S2 antibodyNeutralizing mechanismBinding site(Refs.)
S2P6Inhibits membrane fusionStem helix(173)
CC40.8Inhibits membrane fusionStem helix(174)
WS6Inhibits membrane fusion and post-viral attachmentStem helix(175)
CV3-25Inhibits membrane fusionStem helix(176)
COV44-79, COV44-62Inhibits membrane fusionFusion peptide(177)
C77G12, VN01H1Inhibits membrane fusionFusion peptide(178)
76E1Inhibits membrane fusionFusion peptide(179)
3A3, RAY53Inhibits membrane fusionHinge region(112)

[i] S, spike protein.

Nbs

Heavy-chain-only antibodies (HCAbs) in camelids include two constant structural domains CH2 and CH3, a hinge region and a variable domain heavy-chain (VHH), and retain complete antigen binding capability (Fig. 4A) (114). In cartilaginous fish, their immunoglobulin new antigen receptors (NARs) consist of a homodimer of five constant domains and a variable domain (V-NAR) (Fig. 4A) (115). Recombinantly expressed VHH and V-NAR domains exhibit remarkable structural stability under extreme temperature and pH conditions, and their antigen-binding capabilities are comparable to those of HCAbs (116). These fragments represent the minimal functional units necessary for antigen targeting. Due to their low molecular weight (<15 kDa), VHH and V-NAR are also classed as Nbs. The modularity and small size of Nbs allow them to be readily linked to other molecules, rendering them optimal for generating bispecific or multispecific antibodies with ideal affinity or effectiveness (117). Furthermore, VHH-72 is an Nb that has been previously generated by immunizing camelids with SARS-CoV and middle East respiratory syndrome coronavirus RBDs. After the attachment of human IgG Fc to induce bivalency, it was reported to be able to neutralize the SARS-CoV-2 pseudovirus in vitro and can be expressed in transiently transfected ExpiCHO cells (101).

VHHs consist of four conserved sequence regions that surround three highly variable CDRs, whilst V-NARs possess two CDRs (CDR1 and CDR3) (118,119) (Fig. 4B). CDR3 is the primary binding domain responsible for 60–80% of antigen interactions (120). The SARS-CoV-2 spike protein exhibits an average spacing of 25 nm, which is not optimal for the 5–10 nm range required for efficient B-cell response activation. Consequently, this larger spacing results in inadequate stimulation of B cells and complement recruitment, leading to a less efficient and transient neutralizing antibody response (121). Compared with the poor diversity in CDR loop lengths in conventional antibodies, VHH and V-NAR possess long protruding CDR3 loops that allow them to access more occluded antigenic epitopes. Due to their low molecular weight and weak cohesive interactions between monomers, Nbs can be readily concatenated through genetic engineering to form multimers. When designed as therapeutic agents, this property allows for the creation of multivalent and bispecific antibodies, enhancing their binding capabilities to antigens (122).

Immunization of animals with specific antigens is the first step in Nb production (Fig. 4C). Multiple immunizations are typically administered to stimulate the immune system in the animal into producing antibodies against specific antigens. After isolating the B lymphocytes from the blood of immunized animals, molecular biology techniques, such as reverse transcription PCR, are used to amplify the antibody genes from the B cells. Specific screening methods, such as phage display or yeast surface display technology, are then used to select Nb genes with high affinity from the amplified antibody gene library, which are subsequently cloned into the expression vectors. These vectors are transformed into suitable host cells, such as Escherichia coli or mammalian cells, for recombinant protein expression. Nbs are then purified from cell culture supernatants or cell lysates using a range of purification methods, such as affinity chromatography, ion exchange chromatography and gel filtration. The purified Nbs undergo numerous bioactivity assays, cell-based assays and animal experiments, to verify their affinity and specificity. Depending on the requirements, further modification and optimization of the Nbs may be performed to enhance their stability and affinity (123).

Although VHHs are not of human origin, they exhibit low immunogenicity because of the substantial sequence identity with the human VH gene family III (124). However, due to the evolutionary distance between sharks and humans, V-NARs from sharks exhibit minimal sequence identity with the human VH and variable region light chain structural domains (~30%). Therefore, humanization of V-NARs would be desirable prior to clinical application, which at present is the routine practice. However, in fully humanized VHH, conserved hydrophilic amino acids in framework region (FR)2 are changed, leading to unsatisfactory solubility and stability. Therefore, partial humanization is typically performed. Traditionally, there are two main approaches for humanization, CDR grafting and resurfacing. CDR grafting is a process in which the CDR region is directly grafted from a heterologous antibody to human FRs (125). Resurfacing is the replacement of exposed FR residues on the surface of non-human antibodies with corresponding residues of human antibody FRs, minimizing the immunogenicity of the antibody in humans (126). Humanized antibodies can also be produced directly through the immunization of humanized mice. Moreover, it is theoretically possible to directly extract antibody gene clones from humans and combine them with Nb. A full Nb library was constructed by grafting the CDR sequences from a natural antibody repertoire derived from healthy donor blood onto human IGHV gene framework regions. A total of 18 unique single-domain antibodies were isolated from the library, which demonstrated efficient and specific binding to the SARS-CoV-2 RBD and were categorized into three competitive groups (127). Among the isolated antibodies, n3088 and n3130 were found to bind to a concealed epitope of the SARS-CoV-2 RBD and exhibited significant neutralizing activity.

Due to their stability during long-term storage, VHHs serve as a viable therapeutic option for future epidemic responses (128). Their favorable biophysical attributes facilitate large-scale production via prokaryotic expression systems in a matter of weeks, which allows for their swift deployment in an emergency situation (129). A Nb termed Nb6 has been previously reported to stabilize two adjacent RBDs in the ‘down’ state after binding to S proteins, which may then reorganize the second and third binding sites of Nb6 to maintain the closed conformation, causing the RBD to detach from ACE2. Moreover, the affinity and neutralizing capacity of Nb6 was reported to be enhanced further after dimerization and trimerization, where the trimerized version of Nb6 showed picomolar-range neutralization and femtomolar-range affinity for the SARS-CoV-2 RBD. Efficient neutralization was even maintained after nebulization, lyophilization and thermal processing (130).

The majority of mAbs must be generated in mammalian cells and require intravenous injection. In comparison, Nbs are produced in bacteria or yeast and can be delivered to the lungs by inhalation, which can confer significant advantages for SARS-CoV-2 treatment. Possible benefits include low systemic exposure, rapid onset of action and high concentrations at the lesion site (131). After immunization of alpacas with SARS-CoV-2 S protein, Xiang et al (132) identified a large number of Nbs with high affinity to S protein RBD by using a custom-designed Nb platform technology. Through further screening, purification and testing, a number of neutralizing Nbs with exceptional antiviral ability were identified. Among them, Nb21 was reported to bind to RBD with picomolar-range affinity and displayed neutralizing ability against viruses. Upon formation into multivalent antibodies, their antiviral capabilities are further enhanced. In particular, Nb21 was reported to be highly thermally stable and retained the same antiviral capacity after lyophilization and nebulization. An inhalable nebulizer based on this antibody, Pittsburgh inhalable Nb-21, was reported to be effective against severe SARS-CoV-2 infection in hamsters in an in vitro viral infection assay (133). Nbs, such as NIH-CoVnb-112, Nb11-59, bn03, 2–3-Fc, Nb22, RBD-1-2G, pan-Sarbecovirus Nbs, TP17, TP86, R14 and S43 have all also been demonstrated to exert positive neutralizing effects against SARS-CoV-2 following respiratory administration (134141).

Challenges and current countermeasures

ADE following infection

Not all antibodies against viruses are protective (142). Antibodies against the dengue virus protein expressed by the dengue fever virus can provoke severe reactions mediated by the Fc receptor, namely the ADE of infection (143). ADE is frequently detected in monocytes, macrophages and B cells and is typically mediated by IgG Fc receptors (FcγRs) or complement receptors (144,145). Typically, the ADE of viral infections uses the FcγR. The Fab domains of antibodies with the ADE phenotype bind to viral particles and the Fc domains bind to cells containing FcγR, allowing the virus to enter the host cell through the FcγR, circumventing the involvement of specific receptors (146). For SARS-CoV-2, two forms of ADE mechanisms have been identified. The first form acts through RBD antibodies inducing ADE through FcγRs (147). MW05, the first infection-enhancing SARS-CoV-2 antibody reported, was demonstrated to contribute to ADE by crosslinking its Fc region with FcγRIIB (148). The second ADE pathway is induced by NTD antibodies changing the conformation of the S protein and inducing the open state of the RBD, thereby promoting its binding to ACE2 (149). The ACE2 receptor has also been reported to be partially responsible for ADE (150).

ADE has been reported to impact the degranulation of mast cells, contributing to heart damage or multisystem inflammatory syndrome (151,152). Due to the complex nature of the immune system in vivo, whether complement-dependent cytotoxicity and/or antibody-dependent cell-mediated cytotoxicity occurs during ADE will likely depend on the balance of virus removal and infection augmentation. ADE risk has been previously associated with the concentration of antibodies (153). The balance between neutralizing and deleterious antibodies in the body favors the neutralization of SARS-CoV-2 (154). In vivo experiments prior to clinical trials are necessary for avoiding ADE. Furthermore, non-neutralizing antibodies have been reported to be mostly responsible for ADE. Focusing on the RBM or other highly neutralizing sites may mitigate these drawbacks. Likewise, engineering antibodies to avoid contact with FcRs or using Nbs without a Fc region may also reduce these side effects. Additionally, studies have identified associations between ADE and specific epitopes on the RBD and NTD of SARS-CoV-2, providing crucial insight for the development of safe and effective Nabs (88,155).

Viral variants escaping neutralizing antibodies

Neutralizing antibody development is challenged by the continuous emergence of mutant variants (156). As an RNA virus, SARS-CoV-2 undergoes constant mutation during replication and under the pressure of antibody selection. The first prevalent S protein mutation that received worldwide attention was D614G, a site that does not come into direct contact with any RBD antibodies, and therefore had no significant impact on antibody neutralization activity. By contrast, the ο variant, which was first detected at the end of 2021, rapidly replaced the δ variant as the major epidemic strain worldwide due to its high transmissibility. The ο variants with enhanced resistance to NAbs also challenged vaccination and infection-induced immunity, rendering therapeutic mAbs ineffective (157). Continued mutational evolution of the virus has led to the emergence of a wide range of variants with greater growth advantage, which evaded almost all current neutralizing antibody drugs and vaccinated or convalescent plasma, as represented by the XBB strain, BQ 1.1 strain and CH 1.1 strain, subvariants of the ο variant of the SARS-CoV-2 (158). Breakthrough infections with the BA.2 and BA.5 subvariants of SARS-CoV-2 have reduced the diversity of NAb binding sites and increased the proportion of non-NAb clones. This, in turn, has increased the selective pressure on the humoral immune response, fostering the convergent evolution of RBD (158,159). Subsequent analysis uncovered that the ο variant harbors 15 mutation sites within the RBD, which confer the ability to circumvent antibody neutralization. Among them, the K417, E484, G446 and S371 mutations mediated the binding inhibition of Class 1–4 antibodies, respectively. Specifically, Class 4 antibodies were rendered ineffective against the variants, leading to a marked reduction in the plasma neutralization capacity among recovered and vaccinated individuals (84,160).

Antibodies against conserved epitopes are promising in dealing with the ever-emerging variants. Starr et al (83) previously identified a broad-spectrum neutralizing antibody referred to as S2H97, which binds to a previously unidentified hidden epitope on RBD, causing a conformational change in the RBD, thereby preventing the binding of ACE2. This antibody was reported to retain neutralizing activity against a wide range of strains of Sarbecovirus, including SARS-CoV and SARS-CoV-2. Antibodies which interact with the same binding site as S2H97 could not be found in the blood of convalescent individuals, which is most likely due to the fact that these epitopes are not easily accessible and therefore cannot sufficiently trigger an immune response (83).

Through biological computing technologies, antibodies with superior neutralizing potency can be designed and produced. An antibody named AI-1028, which is a modified S2H97 with an improved neutralization capacity compared with S2H97, was previously reported to be capable of broadly neutralizing Sarbecovirus, including the emerging ο subvariants XBB, BQ.1.1 and BA.2.3.20 (161).

It is of strategic importance to predict the direction of virus evolution to anticipate possible mutant strains in advance. Cao et al (162) developed a computational model for predicting the trend of mutational evolution in SARS-CoV-2 using a high-throughput deep mutation scanning method. Specifically, mutations in BA.5 and BA.2 that may escape existing herd immunity were analyzed, identified and validated.

The simultaneous use of multiple antibodies that target distinct epitopes are known as cocktail therapeutic regimens, which presents potential for disease treatment and advancement of novel antibodies. In cases where existing antibody products exhibit diminished ability to neutralize viral variants, it would appear logical to modify them using engineering techniques, such as site mutation, creating bispecific or multispecific antibodies, bivalent or multivalent constructs (163,164). Furthermore, the combination of soluble cytokine receptors or exosomes with antiviral Nbs has increased effectiveness. c19s130Fc is a bispecific therapeutic that hinders both the IL-6 signaling pathway and the SARS-CoV-2 RBD. It is created by fusing a soluble cytokine receptor with an antiviral Nb, enabling it to block viral entry and dampen the inflammatory response induced by the virus simultaneously (165). In addition, researchers have engineered exosomes with a S-protein-targeting Nb and human IFN-β bound to MFG-E8, creating a dual-action system. The Nbs on the exosome surface bind to the SARS-CoV-2 S protein, blocking its entry into host cells, while the encapsulated IFN-β is delivered to infected cells to trigger antiviral responses and boost the expression of interferon-stimulated genes (166). In summary, further exploration of antibody engineering is justified to improve the neutralization capacity of available antibodies.

Future perspectives

Novel NAbs that can target different parts of SARS-CoV-2 proteins, including the S protein, are constantly being discovered. It is likely that highly effective novel antibodies will be screened in the future for treating patients with COVID-19, especially during the early stages of the disease. Combination therapy using multiple different NAbs may improve treatment efficacy and reduce the likelihood of resistance. Furthermore, the development of long-acting NAbs may reduce the required frequency of administration and improve patient compliance. This may involve modifications in the engineering process of the antibody to extend its half-life in the body. NAbs can be used not only for treatment but also for pre- or post-exposure prophylaxis, particularly in high-risk groups, such as healthcare workers and the elderly. Broad-spectrum neutralizing antibodies, such as the SA55 and SA58 antibody combination, can be administered as a nasal spray to establish rapid short-acting prophylaxis in the respiratory tract. These antibodies can also be injected during the initial stages of infection to provide medium-to-long-term prophylaxis, which is particularly suitable for the protection of high-risk healthcare workers, patients with compromised immune systems who cannot be vaccinated and the elderly (162). Therefore, investigations into enhanced delivery methods will likely contribute to the advancement and use of NAbs. Nebulized inhalation and nasal dripping have also demonstrated promising outcomes in animal models (138140). Further research into the mechanism of NAbs to understand how they interact with the different regions of the virus may be helpful in designing more effective antibodies and vaccines. Reducing the cost of producing NAbs by optimizing the production process would make them more widely available and economically viable. To aid end, research institutions and companies worldwide should strengthen their collaboration and share resources and data to accelerate the R&D of neutralizing antibodies. Moreover, large-scale clinical trials are warranted to assess the safety and efficacy of neutralizing antibodies in different populations. Governments and regulatory agencies should also provide policy support to accelerate the R&D and approval processes of NAbs, whilst ensuring product quality and safety.

Conclusion

The present review summarized the current research progress on antibodies against SARS-CoV-2 by considering the target sites of antibodies, the SARS-CoV-2 invasion mechanism, as well as the preparation methods, structural properties, mechanisms of action and clinical applications of different NAbs. Following the analysis of a broad body of research, a number of conclusions can be drawn. Firstly, NAbs serve a role in the prevention and treatment of SARS-CoV-2. These antibodies are capable of recognizing and binding to key parts of SARS-CoV-2, preventing viral invasion into host cells and viral replication. Screening and evaluation of SARS-CoV-2 NAbs is another required step for optimization. Through the screening of a large number of candidate antibodies, antibodies with high neutralizing activity have been identified, providing a foundation for subsequent research and applications. Additionally, evaluating these antibodies for numerous parameters, such as affinity, stability and safety may ensure their effectiveness and safety in clinical applications. Furthermore, a series of notable advancements have been made in the practical clinical application of SARS-CoV-2 NAbs, especially in the elderly, immunosuppressed or critically ill patients. Moreover, NAbs can be used as an emergency treatment to reduce viral replication and disease severity. For individuals at high risk of exposure to SARS-CoV-2, such as healthcare workers, NAbs can be used as a preventive treatment. NAbs can also be combined with vaccines as a ‘passive immunization’ strategy to provide immediate protection whilst awaiting the establishment of the ‘active immunization’ response induced by the vaccine. For patients who continue to experience long-term symptoms after recovery, NAbs may also help alleviate symptoms and improve quality of life. In summary, the research and application of SARS-CoV-2 NAbs not only provide a notable therapeutic tool for understanding the COVID-19 pandemic, but also hold potential preventive and therapeutic value for future outbreaks. With increased understanding and the advancement of technology, the clinical application of NAbs will likely become more widespread, making a greater contribution to global public health security.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant nos. 81973995 and 82170131) and the Program for HUST Academic Frontier Youth Team (grant no. 2018QYTD14).

Availability of data and materials

Not applicable.

Authors' contributions

The present manuscript was written by TZ, DY, LT and YH. Images were prepared by TZ and DY. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

ADE

antibody-dependent enhancement

ORF

open reading frame

ACE2

angiotensin converting enzyme 2

CDR

complementarity-determining region

CDRH3

CDR 3 of antibody heavy chains

CTD

C-terminal domain

COVID-19

coronavirus disease 2019

FcγR

IgG Fc receptor

FP

fusion peptide

HR

heptagonal repeat

mAb

monoclonal antibody

Nb

nanobody

NAb

neutralizing antibody

NTD

N-terminal domain

R&D

research and development

RBD

receptor binding domain

RBM

receptor-binding motif

SARS-CoV-2

severe acute respiratory syndrome coronavirus 2

sgRNA

subgenomic RNA

TMPRSS2

transmembrane protease serine 2

VHH

variable domain heavy-chain

References

1 

Aktas G: A comprehensive review on rational and effective treatment strategies against an invisible enemy; SARS Cov-2 infection. Exp Biomed Res. 3:293–311. 2020. View Article : Google Scholar

2 

Wu Z and McGoogan JM: Characteristics of and Important Lessons From the Coronavirus Disease 2019 (COVID-19) Outbreak in China: Summary of a Report of 72 314 Cases From the Chinese Center for Disease Control and Prevention. JAMA. 323:1239–1242. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Aktas G, Balci B, Yilmaz S, Bardak H and Duman TT: Characteristics of Covid-19 infection with the original SARS-Cov-2 virus and other variants: A comparative review. J Bionic Mem. 2:96–112. 2022.

4 

Ceasovschih A, Sorodoc V, Shor A, Haliga RE, Roth L, Lionte C, Onofrei Aursulesei V, Sirbu O, Culis N, Shapieva A, et al: Distinct features of vascular diseases in COVID-19. J Inflamm Res. 16:2783–2800. 2023. View Article : Google Scholar : PubMed/NCBI

5 

Khalid A, Ali Jaffar M, Khan T, Abbas Lail R, Ali S, Aktas G, Waris A, Javaid A, Ijaz N and Muhammad N: Hematological and biochemical parameters as diagnostic and prognostic markers in SARS-COV-2 infected patients of Pakistan: A retrospective comparative analysis. Hematology. 26:529–542. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Aktas G: Hematological predictors of novel Coronavirus infection. Rev Assoc Med Bras (1992). 67 (Suppl 1):S1–S2. 2021. View Article : Google Scholar

7 

Fiolet T, Kherabi Y, MacDonald CJ, Ghosn J and Peiffer-Smadja N: Comparing COVID-19 vaccines for their characteristics, efficacy and effectiveness against SARS-CoV-2 and variants of concern: A narrative review. Clin Microbiol Infect. 28:202–221. 2022. View Article : Google Scholar : PubMed/NCBI

8 

Zheng B, Zhao Q, Yang W, Feng P, Xin C, Ying Y, Yang B, Han B, Zhu J, Zhang M and Li G: Small-molecule antiviral treatments for COVID-19: A systematic review and network meta-analysis. Int J Antimicrob Agents. 63:1070962024. View Article : Google Scholar : PubMed/NCBI

9 

Saul S and Einav S: Old drugs for a new virus: Repurposed approaches for combating COVID-19. ACS Infect Dis. 6:2304–2318. 2020. View Article : Google Scholar : PubMed/NCBI

10 

Crawford KHD, Dingens AS, Eguia R, Wolf CR, Wilcox N, Logue JK, Shuey K, Casto AM, Fiala B, Wrenn S, et al: Dynamics of neutralizing antibody titers in the months after severe acute respiratory syndrome coronavirus 2 infection. J Infect Dis. 223:197–205. 2021. View Article : Google Scholar : PubMed/NCBI

11 

Prévost J, Gasser R, Beaudoin-Bussières G, Richard J, Duerr R, Laumaea A, Anand SP, Goyette G, Benlarbi M, Ding S, et al: Cross-Sectional Evaluation of Humoral Responses against SARS-CoV-2 Spike. Cell Rep Med. 1:1001262020. View Article : Google Scholar : PubMed/NCBI

12 

Li M, Wang H, Tian L, Pang Z, Yang Q, Huang T, Fan J, Song L, Tong Y and Fan H: COVID-19 vaccine development: milestones, lessons and prospects. Signal Transduct Target Ther. 7:1462022. View Article : Google Scholar : PubMed/NCBI

13 

Wakefield TW, Strieter RM, Wilke CA, Kadell AM, Wrobleski SK, Burdick MD, Schmidt R, Kunkel SL and Greenfield LJ: Venous thrombosis-associated inflammation and attenuation with neutralizing antibodies to cytokines and adhesion molecules. Arterioscler Thromb Vasc Biol. 15:258–268. 1995. View Article : Google Scholar : PubMed/NCBI

14 

Cagdas D: Convalescent plasma and hyperimmune globulin therapy in COVID-19. Expert Rev Clin Immunol. 17:309–316. 2021. View Article : Google Scholar : PubMed/NCBI

15 

Li L, Zhang W, Hu Y, Tong X, Zheng S, Yang J, Kong Y, Ren L, Wei Q, Mei H, et al: Effect of convalescent plasma therapy on time to clinical improvement in patients with severe and life-threatening COVID-19: A Randomized clinical trial. JAMA. 324:460–470. 2020. View Article : Google Scholar : PubMed/NCBI

16 

Tang J, Grubbs G, Lee Y, Golding H and Khurana S: Impact of convalescent plasma therapy on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibody profile in coronavirus disease 2019 (COVID-19) Patients. Clin Infect Dis. 74:327–334. 2022. View Article : Google Scholar : PubMed/NCBI

17 

Wang Y, Ma Y, Xu Y, Liu J, Li X, Chen Y, Chen Y, Xie J, Xiao L, Xiang Z, et al: Resistance of SARS-CoV-2 Omicron variant to convalescent and CoronaVac vaccine plasma. Emerg Microbes Infect. 11:424–427. 2022.PubMed/NCBI

18 

Cao W, Liu X, Hong K, Ma Z, Zhang Y, Lin L, Han Y, Xiong Y, Liu Z, Ruan L and Li T: High-Dose intravenous immunoglobulin in severe coronavirus disease 2019: A multicenter retrospective study in China. Front Immunol. 12:6278442021. View Article : Google Scholar : PubMed/NCBI

19 

Cao W, Liu X, Bai T, Fan H, Hong K, Song H, Han Y, Lin L, Ruan L and Li T: High-Dose intravenous immunoglobulin as a therapeutic option for deteriorating patients with coronavirus disease 2019. Open Forum Infect Dis. 7:ofaa1022020. View Article : Google Scholar : PubMed/NCBI

20 

Xiang HR, Cheng X, Li Y, Luo WW, Zhang QZ and Peng WX: Efficacy of IVIG (intravenous immunoglobulin) for corona virus disease 2019 (COVID-19): A meta-analysis. Int Immunopharmacol. 96:1077322021. View Article : Google Scholar : PubMed/NCBI

21 

Kindgen-Milles D, Feldt T, Jensen BEO, Dimski T and Brandenburger T: Why the application of IVIG might be beneficial in patients with COVID-19. Lancet Respir Med. 10:e152022. View Article : Google Scholar : PubMed/NCBI

22 

Breedveld FC: Therapeutic monoclonal antibodies. Lancet. 355:735–740. 2000. View Article : Google Scholar : PubMed/NCBI

23 

Buss NA, Henderson SJ, McFarlane M, Shenton JM and de Haan L: Monoclonal antibody therapeutics: History and future. Curr Opin Pharmacol. 12:615–622. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Ren Z, Shen C and Peng J: Status and developing strategies for neutralizing monoclonal antibody therapy in the omicron Era of COVID-19. Viruses. 15:12972023. View Article : Google Scholar : PubMed/NCBI

25 

Pinto D, Park YJ, Beltramello M, Walls AC, Tortorici MA, Bianchi S, Jaconi S, Culap K, Zatta F, De Marco A, et al: Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody. Nature. 583:290–295. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Hillenbrand M, Esslinger C, Seidenberg J, Weber M, Zingg A, Townsend C, Eicher B, Rutkauskaite J, Riese P, Guzman CA, et al: Fast-Track Discovery of SARS-CoV-2-neutralizing antibodies from human B Cells by direct functional screening. Viruses. 16:3392024. View Article : Google Scholar : PubMed/NCBI

27 

Gottlieb RL, Nirula A, Chen P, Boscia J, Heller B, Morris J, Huhn G, Cardona J, Mocherla B, Stosor V, et al: Effect of bamlanivimab as monotherapy or in combination with etesevimab on viral load in patients with mild to moderate COVID-19: A Randomized clinical trial. JAMA. 325:632–644. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Weinreich DM, Sivapalasingam S, Norton T, Ali S, Gao H, Bhore R, Musser BJ, Soo Y, Rofail D, Im J, et al: REGN-COV2, a neutralizing antibody cocktail, in outpatients with Covid-19. N Engl J Med. 384:238–251. 2021. View Article : Google Scholar : PubMed/NCBI

29 

Ji Y, Zhang Q, Cheng L, Ge J, Wang R, Fang M, Mucker EM, Chen P, Ma J, Zhang R, et al: Preclinical characterization of amubarvimab and romlusevimab, a pair of non-competing neutralizing monoclonal antibody cocktail, against SARS-CoV-2. Front Immunol. 13:9804352022. View Article : Google Scholar : PubMed/NCBI

30 

Evering TH, Chew KW, Giganti MJ, Moser C, Pinilla M, Wohl DA, Currier JS, Eron JJ, Javan AC, Bender Ignacio R, et al: Safety and efficacy of combination SARS-CoV-2 neutralizing monoclonal antibodies amubarvimab plus romlusevimab in nonhospitalized patients with COVID-19. Ann Intern Med. 176:658–666. 2023. View Article : Google Scholar : PubMed/NCBI

31 

Kim C, Ryu DK, Lee J, Kim YI, Seo JM, Kim YG, Jeong JH, Kim M, Kim JI, Kim P, et al: A therapeutic neutralizing antibody targeting receptor binding domain of SARS-CoV-2 spike protein. Nat Commun. 12:2882021. View Article : Google Scholar : PubMed/NCBI

32 

Wang YT, Allen RD, Kim K, Shafee N, Gonzalez AJ, Nguyen MN, Valentine KM, Cao X, Lu L, Pai CI, et al: SARS-CoV-2 monoclonal antibodies with therapeutic potential: Broad neutralizing activity and No evidence of antibody-dependent enhancement. Antiviral Res. 195:1051852021. View Article : Google Scholar : PubMed/NCBI

33 

Tian D, Sun Y, Xu H and Ye Q: The emergence and epidemic characteristics of the highly mutated SARS-CoV-2 Omicron variant. J Med Virol. 94:2376–2383. 2022. View Article : Google Scholar : PubMed/NCBI

34 

Guo H, Gao Y, Li T, Li T, Lu Y, Zheng L, Liu Y, Yang T, Luo F, Song S, et al: Structures of Omicron spike complexes and implications for neutralizing antibody development. Cell Rep. 39:1107702022. View Article : Google Scholar : PubMed/NCBI

35 

Muyldermans S: Applications of Nanobodies. Annu Rev Anim Biosci. 9:401–421. 2021. View Article : Google Scholar : PubMed/NCBI

36 

Xu J, Xu K, Jung S, Conte A, Lieberman J, Muecksch F, Lorenzi JCC, Park S, Schmidt F, Wang Z, et al: Nanobodies from camelid mice and llamas neutralize SARS-CoV-2 variants. Nature. 595:278–282. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Weiss SR and Navas-Martin S: Coronavirus pathogenesis and the emerging pathogen severe acute respiratory syndrome coronavirus. Microbiol Mol Biol Rev. 69:635–664. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Wu F, Zhao S, Yu B, Chen YM, Wang W, Song ZG, Hu Y, Tao ZW, Tian JH, Pei YY, et al: A new coronavirus associated with human respiratory disease in China. Nature. 579:265–269. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Yang H and Rao Z: Structural biology of SARS-CoV-2 and implications for therapeutic development. Nat Rev Microbiol. 19:685–700. 2021. View Article : Google Scholar : PubMed/NCBI

40 

Chen Y, Liu Q and Guo D: Emerging coronaviruses: Genome structure, replication, and pathogenesis. J Med Virol. 92:418–423. 2020. View Article : Google Scholar : PubMed/NCBI

41 

Kim D, Lee JY, Yang JS, Kim JW, Kim VN and Chang H: The Architecture of SARS-CoV-2 Transcriptome. Cell. 181:914–921.e10. 2020. View Article : Google Scholar : PubMed/NCBI

42 

Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, et al: SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 181:271–280.e8. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Huang Y, Yang C, Xu XF, Xu W and Liu SW: Structural and functional properties of SARS-CoV-2 spike protein: potential antivirus drug development for COVID-19. Acta Pharmacol Sin. 41:1141–1149. 2020. View Article : Google Scholar : PubMed/NCBI

44 

Walls AC, Park YJ, Tortorici MA, Wall A, McGuire AT and Veesler D: Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell. 181:281–292.e6. 2020. View Article : Google Scholar : PubMed/NCBI

45 

Wang Q, Zhang Y, Wu L, Niu S, Song C, Zhang Z, Lu G, Qiao C, Hu Y, Yuen KY, et al: Structural and Functional Basis of SARS-CoV-2 Entry by Using Human ACE2. Cell. 181:894–904.e9. 2020. View Article : Google Scholar : PubMed/NCBI

46 

Cai Y, Zhang J, Xiao T, Peng H, Sterling SM, Walsh RM Jr, Rawson S, Rits-Volloch S and Chen B: Distinct conformational states of SARS-CoV-2 spike protein. Science. 369:1586–1592. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Song W, Gui M, Wang X and Xiang Y: Cryo-EM structure of the SARS coronavirus spike glycoprotein in complex with its host cell receptor ACE2. PLoS Pathog. 14:e10072362018. View Article : Google Scholar : PubMed/NCBI

48 

Chi X, Yan R, Zhang J, Zhang G, Zhang Y, Hao M, Zhang Z, Fan P, Dong Y, Yang Y, et al: A neutralizing human antibody binds to the N-terminal domain of the Spike protein of SARS-CoV-2. Science. 369:650–655. 2020. View Article : Google Scholar : PubMed/NCBI

49 

Liu L, Wang P, Nair MS, Yu J, Rapp M, Wang Q, Luo Y, Chan JF, Sahi V, Figueroa A, et al: Potent neutralizing antibodies against multiple epitopes on SARS-CoV-2 spike. Nature. 584:450–456. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Yao H, Song Y, Chen Y, Wu N, Xu J, Sun C, Zhang J, Weng T, Zhang Z, Wu Z, et al: Molecular Architecture of the SARS-CoV-2 Virus. Cell. 183:730–738.e13. 2020. View Article : Google Scholar : PubMed/NCBI

51 

Fantini J, Di Scala C, Chahinian H and Yahi N: Structural and molecular modelling studies reveal a new mechanism of action of chloroquine and hydroxychloroquine against SARS-CoV-2 infection. Int J Antimicrob Agents. 55:1059602020. View Article : Google Scholar : PubMed/NCBI

52 

Fantini J, Chahinian H and Yahi N: Synergistic antiviral effect of hydroxychloroquine and azithromycin in combination against SARS-CoV-2: What molecular dynamics studies of virus-host interactions reveal. Int J Antimicrob Agents. 56:1060202020. View Article : Google Scholar

53 

Seyran M, Takayama K, Uversky VN, Adadi P, Mohamed Abd El-Aziz T, Soares AG, Kandimalla R, Tambuwala M, Hassan SS, Azad GK, et al: The structural basis of accelerated host cell entry by SARS-CoV-2†. FEBS J. 288:5010–5020. 2021. View Article : Google Scholar : PubMed/NCBI

54 

Lan J, Ge J, Yu J, Shan S, Zhou H, Fan S, Zhang Q, Shi X, Wang Q, Zhang L and Wang X: Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature. 581:215–220. 2020. View Article : Google Scholar : PubMed/NCBI

55 

Li F, Li W, Farzan M and Harrison SC: Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science. 309:1864–1868. 2005. View Article : Google Scholar : PubMed/NCBI

56 

Zhang J, Cai Y, Xiao T, Lu J, Peng H, Sterling SM, Walsh RM Jr, Rits-Volloch S, Zhu H, Woosley AN, et al: Structural impact on SARS-CoV-2 spike protein by D614G substitution. Science. 372:525–530. 2021. View Article : Google Scholar : PubMed/NCBI

57 

Jackson CB, Farzan M, Chen B and Choe H: Mechanisms of SARS-CoV-2 entry into cells. Nat Rev Mol Cell Biol. 23:3–20. 2022. View Article : Google Scholar : PubMed/NCBI

58 

Benton DJ, Wrobel AG, Xu P, Roustan C, Martin SR, Rosenthal PB, Skehel JJ and Gamblin SJ: Receptor binding and priming of the spike protein of SARS-CoV-2 for membrane fusion. Nature. 588:327–330. 2020. View Article : Google Scholar : PubMed/NCBI

59 

Bayati A, Kumar R, Francis V and McPherson PS: SARS-CoV-2 infects cells after viral entry via clathrin-mediated endocytosis. J Biol Chem. 296:1003062021. View Article : Google Scholar : PubMed/NCBI

60 

Jaimes JA, Millet JK and Whittaker GR: Proteolytic Cleavage of the SARS-CoV-2 Spike protein and the role of the novel S1/S2 Site. iScienc. 23:1012122020. View Article : Google Scholar

61 

Newcombe C and Newcombe AR: Antibody production: Polyclonal-derived biotherapeutics. J Chromatogr B Analyt Technol Biomed Life Sci. 848:2–7. 2007. View Article : Google Scholar : PubMed/NCBI

62 

Ascoli CA and Aggeler B: Overlooked benefits of using polyclonal antibodies. Biotechniques. 65:127–136. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Leenaars M and Hendriksen CF: Critical steps in the production of polyclonal and monoclonal antibodies: Evaluation and recommendations. ILAR J. 46:269–279. 2005. View Article : Google Scholar : PubMed/NCBI

64 

Zylberman V, Sanguineti S, Pontoriero AV, Higa SV, Cerutti ML, Morrone Seijo SM, Pardo R, Muñoz L, Acuña Intrieri ME, Alzogaray VA, et al: Development of a hyperimmune equine serum therapy for COVID-19 in Argentina. Medicina (B Aires). 80 (Suppl 3):S1–S6. 2020.

65 

Lopardo G, Belloso WH, Nannini E, Colonna M, Sanguineti S, Zylberman V, Muñoz L, Dobarro M, Lebersztein G, Farina J, et al: RBD-specific polyclonal F(ab´)2 fragments of equine antibodies in patients with moderate to severe COVID-19 disease: A randomized, multicenter, double-blind, placebo-controlled, adaptive phase 2/3 clinical trial. EClinicalMedicine. 34:1008432021. View Article : Google Scholar : PubMed/NCBI

66 

Vanhove B, Duvaux O, Rousse J, Royer PJ, Evanno G, Ciron C, Lheriteau E, Vacher L, Gervois N, Oger R, et al: High neutralizing potency of swine glyco-humanized polyclonal antibodies against SARS-CoV-2. Eur J Immunol. 51:1412–1422. 2021. View Article : Google Scholar : PubMed/NCBI

67 

Gaborit B, Dailly E, Vanhove B, Josien R, Lacombe K, Dubee V, Ferre V, Brouard S, Ader F, Vibet MA, et al: Pharmacokinetics and Safety of XAV-19, a Swine Glyco-humanized Polyclonal Anti-SARS-CoV-2 Antibody, for COVID-19-Related Moderate Pneumonia: A Randomized, Double-Blind, Placebo-Controlled, Phase IIa Study. Antimicrob Agents Chemother. 65:e01237212021. View Article : Google Scholar : PubMed/NCBI

68 

Vanhove B, Marot S, So RT, Gaborit B, Evanno G, Malet I, Lafrogne G, Mevel E, Ciron C, Royer PJ, et al: XAV-19, a swine glyco-humanized polyclonal antibody against SARS-CoV-2 spike receptor-binding domain, targets multiple epitopes and broadly neutralizes variants. Front Immunol. 12:7612502021. View Article : Google Scholar : PubMed/NCBI

69 

Singh R, Chandley P and Rohatgi S: Recent advances in the development of monoclonal antibodies and next-generation antibodies. Immunohorizons. 7:886–897. 2023. View Article : Google Scholar : PubMed/NCBI

70 

Safdari Y, Farajnia S, Asgharzadeh M and Khalili M: Antibody humanization methods-a review and update. Biotechnol Genet Eng Rev. 29:175–186. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Yu H, Borsotti C, Schickel JN, Zhu S, Strowig T, Eynon EE, Frleta D, Gurer C, Murphy AJ, Yancopoulos GD, et al: A novel humanized mouse model with significant improvement of class-switched, antigen-specific antibody production. Blood. 129:959–969. 2017. View Article : Google Scholar : PubMed/NCBI

72 

Pedrioli A and Oxenius A: Single B cell technologies for monoclonal antibody discovery. Trends Immunol. 42:1143–1158. 2021. View Article : Google Scholar : PubMed/NCBI

73 

Winter G and Milstein C: Man-made antibodies. Nature. 349:293–299. 1991. View Article : Google Scholar : PubMed/NCBI

74 

McCafferty J, Griffiths AD, Winter G and Chiswell DJ: Phage antibodies: Filamentous phage displaying antibody variable domains. Nature. 348:552–554. 1990. View Article : Google Scholar : PubMed/NCBI

75 

Chen F, Liu Z, Kang W, Jiang F, Yang X, Yin F, Zhou Z and Li Z: Single-domain antibodies against SARS-CoV-2 RBD from a two-stage phage screening of universal and focused synthetic libraries. BMC Infect Dis. 24:1992024. View Article : Google Scholar : PubMed/NCBI

76 

Barnes CO, Jette CA, Abernathy ME, Dam KA, Esswein SR, Gristick HB, Malyutin AG, Sharaf NG, Huey-Tubman KE, Lee YE, et al: SARS-CoV-2 neutralizing antibody structures inform therapeutic strategies. Nature. 588:682–687. 2020. View Article : Google Scholar : PubMed/NCBI

77 

Piccoli L, Park YJ, Tortorici MA, Czudnochowski N, Walls AC, Beltramello M, Silacci-Fregni C, Pinto D, Rosen LE, Bowen JE, et al: Mapping neutralizing and immunodominant sites on the SARS-CoV-2 spike receptor-binding domain by structure-guided high-resolution serology. Cell. 183:1024–1042.e21. 2020. View Article : Google Scholar : PubMed/NCBI

78 

Röltgen K, Powell AE, Wirz OF, Stevens BA, Hogan CA, Najeeb J, Hunter M, Wang H, Sahoo MK, Huang C, et al: Defining the features and duration of antibody responses to SARS-CoV-2 infection associated with disease severity and outcome. Sci Immunol. 5:eabe02402020. View Article : Google Scholar : PubMed/NCBI

79 

Barnes CO, West AP Jr, Huey-Tubman KE, Hoffmann MAG, Sharaf NG, Hoffman PR, Koranda N, Gristick HB, Gaebler C, Muecksch F, et al: Structures of Human Antibodies Bound to SARS-CoV-2 spike reveal common epitopes and recurrent features of antibodies. Cell. 182:828–842.e16. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Wu Y, Wang F, Shen C, Peng W, Li D, Zhao C, Li Z, Li S, Bi Y, Yang Y, et al: A noncompeting pair of human neutralizing antibodies block COVID-19 virus binding to its receptor ACE2. Science. 368:1274–1278. 2020. View Article : Google Scholar : PubMed/NCBI

81 

Shi R, Shan C, Duan X, Chen Z, Liu P, Song J, Song T, Bi X, Han C, Wu L, et al: A human neutralizing antibody targets the receptor-binding site of SARS-CoV-2. Nature. 584:120–124. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Banach BB, Cerutti G, Fahad AS, Shen CH, Oliveira De Souza M, Katsamba PS, Tsybovsky Y, Wang P, Nair MS, Huang Y, et al: Paired heavy- and light-chain signatures contribute to potent SARS-CoV-2 neutralization in public antibody responses. Cell Rep. 37:1097712021. View Article : Google Scholar : PubMed/NCBI

83 

Starr TN, Czudnochowski N, Liu Z, Zatta F, Park YJ, Addetia A, Pinto D, Beltramello M, Hernandez P, Greaney AJ, et al: SARS-CoV-2 RBD antibodies that maximize breadth and resistance to escape. Nature. 597:97–102. 2021. View Article : Google Scholar : PubMed/NCBI

84 

Cameroni E, Bowen JE, Rosen LE, Saliba C, Zepeda SK, Culap K, Pinto D, VanBlargan LA, De Marco A, di Iulio J, et al: Broadly neutralizing antibodies overcome SARS-CoV-2 Omicron antigenic shift. Nature. 602:664–670. 2022. View Article : Google Scholar : PubMed/NCBI

85 

Brouwer PJM, Caniels TG, van der Straten K, Snitselaar JL, Aldon Y, Bangaru S, Torres JL, Okba NMA, Claireaux M, Kerster G, et al: Potent neutralizing antibodies from COVID-19 patients define multiple targets of vulnerability. Science. 369:643–650. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Kim SI, Noh J, Kim S, Choi Y, Yoo DK, Lee Y, Lee H, Jung J, Kang CK, Song KH, et al: Stereotypic neutralizing VH antibodies against SARS-CoV-2 spike protein receptor binding domain in patients with COVID-19 and healthy individuals. Sci Transl Med. 13:eabd69902021. View Article : Google Scholar : PubMed/NCBI

87 

Greaney AJ, Starr TN, Barnes CO, Weisblum Y, Schmidt F, Caskey M, Gaebler C, Cho A, Agudelo M, Finkin S, et al: Mapping mutations to the SARS-CoV-2 RBD that escape binding by different classes of antibodies. Nat Commun. 12:41962021. View Article : Google Scholar : PubMed/NCBI

88 

Li D, Edwards RJ, Manne K, Martinez DR, Schäfer A, Alam SM, Wiehe K, Lu X, Parks R, Sutherland LL, et al: In vitro and in vivo functions of SARS-CoV-2 infection-enhancing and neutralizing antibodies. Cell. 184:4203–4219.e32. 2021. View Article : Google Scholar : PubMed/NCBI

89 

Tian X, Li C, Huang A, Xia S, Lu S, Shi Z, Lu L, Jiang S, Yang Z, Wu Y and Ying T: Potent binding of 2019 novel coronavirus spike protein by a SARS coronavirus-specific human monoclonal antibody. Emerg Microbes Infect. 9:382–385. 2020. View Article : Google Scholar : PubMed/NCBI

90 

ter Meulen J, van den Brink EN, Poon LL, Marissen WE, Leung CS, Cox F, Cheung CY, Bakker AQ, Bogaards JA, van Deventer E, et al: Human monoclonal antibody combination against SARS coronavirus: synergy and coverage of escape mutants. PLoS Med. 3:e2372006. View Article : Google Scholar : PubMed/NCBI

91 

Yuan M, Wu NC, Zhu X, Lee CD, So RTY, Lv H, Mok CKP and Wilson IA: A highly conserved cryptic epitope in the receptor binding domains of SARS-CoV-2 and SARS-CoV. Science. 368:630–633. 2020. View Article : Google Scholar : PubMed/NCBI

92 

Gupta A, Gonzalez-Rojas Y, Juarez E, Crespo Casal M, Moya J, Falci DR, Sarkis E, Solis J, Zheng H, Scott N, et al: Early Treatment for Covid-19 with SARS-CoV-2 Neutralizing Antibody Sotrovimab. N Engl J Med. 385:1941–1950. 2021. View Article : Google Scholar : PubMed/NCBI

93 

Rockett R, Basile K, Maddocks S, Fong W, Agius JE, Johnson-Mackinnon J, Arnott A, Chandra S, Gall M, Draper J, et al: Resistance Mutations in SARS-CoV-2 delta variant after sotrovimab use. N Engl J Med. 386:1477–1479. 2022. View Article : Google Scholar : PubMed/NCBI

94 

Martinez DR, Schaefer A, Gobeil S, Li D, De la Cruz G, Parks R, Lu X, Barr M, Manne K, Mansouri K, et al: A broadly neutralizing antibody protects against SARS-CoV, pre-emergent bat CoVs, and SARS-CoV-2 variants in mice. bioRxiv (Preprint). doi: 10.1101/2021.04.27.441655.

95 

Wec AZ, Wrapp D, Herbert AS, Maurer DP, Haslwanter D, Sakharkar M, Jangra RK, Dieterle ME, Lilov A, Huang D, et al: Broad neutralization of SARS-related viruses by human monoclonal antibodies. Science. 369:731–736. 2020. View Article : Google Scholar : PubMed/NCBI

96 

Rappazzo CG, Tse LV, Kaku CI, Wrapp D, Sakharkar M, Huang D, Deveau LM, Yockachonis TJ, Herbert AS, Battles MB, et al: Broad and potent activity against SARS-like viruses by an engineered human monoclonal antibody. Science. 371:823–829. 2021. View Article : Google Scholar : PubMed/NCBI

97 

Li D, Sempowski GD, Saunders KO, Acharya P and Haynes BF: SARS-CoV-2 Neutralizing Antibodies for COVID-19 Prevention and Treatment. Annu Rev Med. 73:1–16. 2022. View Article : Google Scholar : PubMed/NCBI

98 

Hastie KM, Li H, Bedinger D, Schendel SL, Dennison SM, Li K, Rayaprolu V, Yu X, Mann C, Zandonatti M, et al: Defining variant-resistant epitopes targeted by SARS-CoV-2 antibodies: A global consortium study. Science. 374:472–478. 2021. View Article : Google Scholar : PubMed/NCBI

99 

McCallum M, De Marco A, Lempp FA, Tortorici MA, Pinto D, Walls AC, Beltramello M, Chen A, Liu Z, Zatta F, et al: N-terminal domain antigenic mapping reveals a site of vulnerability for SARS-CoV-2. Cell. 184:2332–2347.e16. 2021. View Article : Google Scholar : PubMed/NCBI

100 

Chi XY, Yan RH, Zhang J, Zhang G, Zhang Y, Hao M, Zhang Z, Fan P, Dong Y, Yang Y, et al: A neutralizing human antibody binds to the N-terminal domain of the Spike protein of SARS-CoV-2. Science. 369:650–655. 2020. View Article : Google Scholar : PubMed/NCBI

101 

Wrapp D, De Vlieger D, Corbett KS, Torres GM, Wang N, Van Breedam W, Roose K, van Schie L; VIB-CMB COVID-19 Response Team; Hoffmann M, ; et al: Structural basis for potent neutralization of betacoronaviruses by single-domain camelid antibodies. Cell. 181:1436–1441. 2020. View Article : Google Scholar : PubMed/NCBI

102 

Hoffmann M, Kleine-Weber H and Pöhlmann S: A Multibasic Cleavage Site in the Spike Protein of SARS-CoV-2 is essential for infection of human lung cells. Mol Cell. 78:779–784.e5. 2020. View Article : Google Scholar : PubMed/NCBI

103 

Cerutti G, Guo Y, Zhou T, Gorman J, Lee M, Rapp M, Reddem ER, Yu J, Bahna F, Bimela J, et al: Potent SARS-CoV-2 neutralizing antibodies directed against spike N-terminal domain target a single supersite. Cell Host Microbe. 29:819–833.e7. 2021. View Article : Google Scholar : PubMed/NCBI

104 

Nielsen SCA, Yang F, Jackson KJL, Hoh RA, Röltgen K, Jean GH, Stevens BA, Lee JY, Rustagi A, Rogers AJ, et al: Human B Cell Clonal Expansion and Convergent Antibody Responses to SARS-CoV-2. Cell Host Microbe. 28:516–525.e5. 2020. View Article : Google Scholar : PubMed/NCBI

105 

Boyd SD, Gaëta BA, Jackson KJ, Fire AZ, Marshall EL, Merker JD, Maniar JM, Zhang LN, Sahaf B, Jones CD, et al: Individual variation in the germline Ig gene repertoire inferred from variable region gene rearrangements. J Immunol. 184:6986–6992. 2010. View Article : Google Scholar : PubMed/NCBI

106 

Wang N, Sun Y, Feng R, Wang Y, Guo Y, Zhang L, Deng YQ, Wang L, Cui Z, Cao L, et al: Structure-based development of human antibody cocktails against SARS-CoV-2. Cell Res. 31:101–103. 2021. View Article : Google Scholar : PubMed/NCBI

107 

Haslwanter D, Dieterle ME, Wec AZ, O'Brien CM, Sakharkar M, Florez C, Tong K, Rappazzo CG, Lasso G, Vergnolle O, et al: A Combination of Receptor-Binding Domain and N-Terminal Domain Neutralizing Antibodies Limits the Generation of SARS-CoV-2 Spike Neutralization-Escape Mutants. mBio. 12:e02473212021. View Article : Google Scholar : PubMed/NCBI

108 

Nguyen-Contant P, Embong AK, Kanagaiah P, Chaves FA, Yang H, Branche AR, Topham DJ and Sangster MY: S Protein-Reactive IgG and Memory B Cell Production after Human SARS-CoV-2 Infection Includes Broad Reactivity to the S2 Subunit. mBio. 11:e01991–20. 2020. View Article : Google Scholar : PubMed/NCBI

109 

Guo L, Wang Y, Kang L, Hu Y, Wang L, Zhong J, Chen H, Ren L, Gu X, Wang G, et al: Cross-reactive antibody against human coronavirus OC43 spike protein correlates with disease severity in COVID-19 patients: A retrospective study. Emerg Microbes Infect. 10:664–676. 2021. View Article : Google Scholar : PubMed/NCBI

110 

Zohar T, Loos C, Fischinger S, Atyeo C, Wang C, Slein MD, Burke J, Yu J, Feldman J, Hauser BM, et al: Compromised humoral functional evolution tracks with SARS-CoV-2 Mortality. Cell. 183:1508–1519.e12. 2020. View Article : Google Scholar : PubMed/NCBI

111 

Ma X, Zou F, Yu F, Li R, Yuan Y, Zhang Y, Zhang X, Deng J, Chen T, Song Z, et al: Nanoparticle vaccines based on the receptor binding Domain (RBD) and Heptad Repeat (HR) of SARS-CoV-2 elicit robust protective immune responses. Immunity. 53:1315–1330.e9. 2020. View Article : Google Scholar : PubMed/NCBI

112 

Silva RP, Huang Y, Nguyen AW, Hsieh CL, Olaluwoye OS, Kaoud TS, Wilen RE, Qerqez AN, Park JG, Khalil AM, et al: Identification of a conserved S2 epitope present on spike proteins from all highly pathogenic coronaviruses. Elife. 12:e837102023. View Article : Google Scholar : PubMed/NCBI

113 

Hsieh CL, Werner AP, Leist SR, Stevens LJ, Falconer E, Goldsmith JA, Chou CW, Abiona OM, West A, Westendorf K, et al: Stabilized coronavirus spike stem elicits a broadly protective antibody. Cell Rep. 37:1099292021. View Article : Google Scholar : PubMed/NCBI

114 

Hamers-Casterman C, Atarhouch T, Muyldermans S, Robinson G, Hamers C, Songa EB, Bendahman N and Hamers R: Naturally occurring antibodies devoid of light chains. Nature. 363:446–448. 1993. View Article : Google Scholar : PubMed/NCBI

115 

Tanaka Y, Nishikawa M, Kamisaki K, Hachiya S, Nakamura M, Kuwazuru T, Tanimura S, Soyano K and Takeda K: Marine-derived microbes and molecules for drug discovery. Inflamm Regen. 42:182022. View Article : Google Scholar : PubMed/NCBI

116 

Muyldermans S: Nanobodies: Natural single-domain antibodies. Annu Rev Biochem. 82:775–797. 2013. View Article : Google Scholar : PubMed/NCBI

117 

Jovčevska I and Muyldermans S: The therapeutic potential of nanobodies. BioDrugs. 34:11–26. 2020. View Article : Google Scholar : PubMed/NCBI

118 

Muyldermans S, Baral TN, Retamozzo VC, De Baetselier P, De Genst E, Kinne J, Leonhardt H, Magez S, Nguyen VK, Revets H, et al: Camelid immunoglobulins and nanobody technology. Vet Immunol Immunopathol. 128:178–183. 2009. View Article : Google Scholar : PubMed/NCBI

119 

Zielonka S, Empting M, Grzeschik J, Könning D, Barelle CJ and Kolmar H: Structural insights and biomedical potential of IgNAR scaffolds from sharks. MAbs. 7:15–25. 2015. View Article : Google Scholar : PubMed/NCBI

120 

Transue TR, De Genst E, Ghahroudi MA, Wyns L and Muyldermans S: Camel single-domain antibody inhibits enzyme by mimicking carbohydrate substrate. Proteins. 32:515–522. 1998. View Article : Google Scholar : PubMed/NCBI

121 

Bachmann MF, Mohsen MO, Zha L, Vogel M and Speiser DE: SARS-CoV-2 structural features may explain limited neutralizing-antibody responses. NPJ Vaccines. 6:22021. View Article : Google Scholar : PubMed/NCBI

122 

Steeland S, Vandenbroucke RE and Libert C: Nanobodies as therapeutics: Big opportunities for small antibodies. Drug Discov Today. 21:1076–1113. 2016. View Article : Google Scholar : PubMed/NCBI

123 

Holliger P and Hudson PJ: Engineered antibody fragments and the rise of single domains. Nat Biotechnol. 23:1126–1136. 2005. View Article : Google Scholar : PubMed/NCBI

124 

Vu KB, Ghahroudi MA, Wyns L and Muyldermans S: Comparison of llama VH sequences from conventional and heavy chain antibodies. Mol Immunol. 34:1121–1131. 1997. View Article : Google Scholar : PubMed/NCBI

125 

Zhang YF, Sun Y, Hong J and Ho M: Humanization of the Shark VNAR Single Domain Antibody Using CDR Grafting. Curr Protoc. 3:e6302023. View Article : Google Scholar : PubMed/NCBI

126 

Almagro JC and Fransson J: Humanization of antibodies. Front Biosci. 13:1619–1633. 2008.PubMed/NCBI

127 

Wu Y, Li C, Xia S, Tian X, Kong Y, Wang Z, Gu C, Zhang R, Tu C, Xie Y, et al: Identification of Human Single-Domain Antibodies against SARS-CoV-2. Cell Host Microbe. 27:891–898.e5. 2020. View Article : Google Scholar : PubMed/NCBI

128 

Hassanzadeh-Ghassabeh G, Devoogdt N, De Pauw P, Vincke C and Muyldermans S: Nanobodies and their potential applications. Nanomedicine (Lond). 8:1013–1026. 2013. View Article : Google Scholar : PubMed/NCBI

129 

Harmsen MM and De Haard HJ: Properties, production, and applications of camelid single-domain antibody fragments. Appl Microbiol Biotechnol. 77:13–22. 2007. View Article : Google Scholar : PubMed/NCBI

130 

Schoof M, Faust B, Saunders RA, Sangwan S, Rezelj V, Hoppe N, Boone M, Billesbølle CB, Puchades C, Azumaya CM, et al: An ultrapotent synthetic nanobody neutralizes SARS-CoV-2 by stabilizing inactive Spike. Science. 370:1473–1479. 2020. View Article : Google Scholar : PubMed/NCBI

131 

Van Heeke G, Allosery K, De Brabandere V, De Smedt T, Detalle L and de Fougerolles A: Nanobodies® as inhaled biotherapeutics for lung diseases. Pharmacol Ther. 169:47–56. 2017. View Article : Google Scholar : PubMed/NCBI

132 

Xiang Y, Nambulli S, Xiao Z, Liu H, Sang Z, Duprex WP, Schneidman-Duhovny D, Zhang C and Shi Y: Versatile and multivalent nanobodies efficiently neutralize SARS-CoV-2. Science. 370:1479–1484. 2020. View Article : Google Scholar : PubMed/NCBI

133 

Nambulli S, Xiang Y, Tilston-Lunel NL, Rennick LJ, Sang Z, Klimstra WB, Reed DS, Crossland NA, Shi Y and Duprex WP: Inhalable Nanobody (PiN-21) prevents and treats SARS-CoV-2 infections in Syrian hamsters at ultra-low doses. Sci Adv. 7:eabh03192021. View Article : Google Scholar : PubMed/NCBI

134 

Gai J, Ma L, Li G, Zhu M, Qiao P, Li X, Zhang H, Zhang Y, Chen Y, Ji W, et al: A potent neutralizing nanobody against SARS-CoV-2 with inhaled delivery potential. MedComm (2020). 2:101–113. 2021.PubMed/NCBI

135 

Li C, Zhan W, Yang Z, Tu C, Hu G, Zhang X, Song W, Du S, Zhu Y, Huang K, et al: Broad neutralization of SARS-CoV-2 variants by an inhalable bispecific single-domain antibody. Cell. 185:1389–1401.e18. 2022. View Article : Google Scholar : PubMed/NCBI

136 

Ma H, Zhang X, Zeng W, Zhou J, Chi X, Chen S, Zheng P, Wang M, Wu Y, Zhao D, et al: A bispecific nanobody dimer broadly neutralizes SARS-CoV-1 & 2 variants of concern and offers substantial protection against Omicron via low-dose intranasal administration. Cell Discov. 8:1322022. View Article : Google Scholar : PubMed/NCBI

137 

Wu X, Wang Y, Cheng L, Ni F, Zhu L, Ma S, Huang B, Ji M, Hu H, Li Y, et al: Short-Term Instantaneous Prophylaxis and Efficient Treatment Against SARS-CoV-2 in hACE2 Mice Conferred by an Intranasal Nanobody (Nb22). Front Immunol. 13:8654012022. View Article : Google Scholar : PubMed/NCBI

138 

Xiang Y, Huang W, Liu H, Sang Z, Nambulli S, Tubiana J, Williams KL Jr, Duprex WP, Schneidman-Duhovny D, Wilson IA, et al: Superimmunity by pan-sarbecovirus nanobodies. Cell Rep. 39:1110042022. View Article : Google Scholar : PubMed/NCBI

139 

Nagata K, Utsumi D, Asaka MN, Maeda R, Shirakawa K, Kazuma Y, Nomura R, Horisawa Y, Yanagida Y, Kawai Y, et al: Intratracheal trimerized nanobody cocktail administration suppresses weight loss and prolongs survival of SARS-CoV-2 infected mice. Commun Med (Lond). 2:1522022. View Article : Google Scholar : PubMed/NCBI

140 

Maeda R, Fujita J, Konishi Y, Kazuma Y, Yamazaki H, Anzai I, Watanabe T, Yamaguchi K, Kasai K, Nagata K, et al: A panel of nanobodies recognizing conserved hidden clefts of all SARS-CoV-2 spike variants including Omicron. Commun Biol. 5:6692022. View Article : Google Scholar : PubMed/NCBI

141 

Liu H, Wu L, Liu B, Xu K, Lei W, Deng J, Rong X, Du P, Wang L, Wang D, et al: Two pan-SARS-CoV-2 nanobodies and their multivalent derivatives effectively prevent Omicron infections in mice. Cell Rep Med. 4:1009182023. View Article : Google Scholar : PubMed/NCBI

142 

Bournazos S, Gupta A and Ravetch JV: The role of IgG Fc receptors in antibody-dependent enhancement. Nat Rev Immunol. 20:633–643. 2020. View Article : Google Scholar : PubMed/NCBI

143 

Wang TT, Sewatanon J, Memoli MJ, Wrammert J, Bournazos S, Bhaumik SK, Pinsky BA, Chokephaibulkit K, Onlamoon N, Pattanapanyasat K, et al: IgG antibodies to dengue enhanced for FcγRIIIA binding determine disease severity. Science. 355:395–398. 2017. View Article : Google Scholar : PubMed/NCBI

144 

Iwasaki A and Yang Y: The potential danger of suboptimal antibody responses in COVID-19. Nat Rev Immunol. 20:339–341. 2020. View Article : Google Scholar : PubMed/NCBI

145 

Ubol S and Halstead SB: How innate immune mechanisms contribute to antibody-enhanced viral infections. Clin Vaccine Immunol. 17:1829–1835. 2010. View Article : Google Scholar : PubMed/NCBI

146 

Haynes BF, Corey L, Fernandes P, Gilbert PB, Hotez PJ, Rao S, Santos MR, Schuitemaker H, Watson M and Arvin A: Prospects for a safe COVID-19 vaccine. Sci Transl Med. 12:eabe09482020. View Article : Google Scholar : PubMed/NCBI

147 

Yang Y and Xu F: Evolving understanding of antibody-dependent enhancement (ADE) of SARS-CoV-2. Front Immunol. 13:10082852022. View Article : Google Scholar : PubMed/NCBI

148 

Wang S, Peng Y, Wang R, Jiao S, Wang M, Huang W, Shan C, Jiang W, Li Z, Gu C, et al: Characterization of neutralizing antibody with prophylactic and therapeutic efficacy against SARS-CoV-2 in rhesus monkeys. Nat Commun. 11:57522020. View Article : Google Scholar : PubMed/NCBI

149 

Liu Y, Soh WT, Kishikawa JI, Hirose M, Nakayama EE, Li S, Sasai M, Suzuki T, Tada A, Arakawa A, et al: An infectivity-enhancing site on the SARS-CoV-2 spike protein targeted by antibodies. Cell. 184:3452–3466.e18. 2021. View Article : Google Scholar : PubMed/NCBI

150 

Wang Z, Deng T, Zhang Y, Niu W, Nie Q, Yang S, Liu P, Pei P, Chen L, Li H and Cao B: ACE2 can act as the secondary receptor in the FcγR-dependent ADE of SARS-CoV-2 infection. iScience. 25:1037202022. View Article : Google Scholar : PubMed/NCBI

151 

Tkaczyk C, Okayama Y, Woolhiser MR, Hagaman DD, Gilfillan AM and Metcalfe DD: Activation of human mast cells through the high affinity IgG receptor. Mol Immunol. 38:1289–1293. 2002. View Article : Google Scholar : PubMed/NCBI

152 

Darrell DO, Gherlone N, Fremont-Smith P, Tisdall P and Fremont-Smith M: Kawasaki Disease, Multisystem Inflammatory Syndrome in Children: Antibody-Induced Mast Cell Activation Hypothesis. J Pediatrics & Pediatr Med. 4:1–7. 2020. View Article : Google Scholar

153 

Ricke DO: Two Different Antibody-Dependent Enhancement (ADE) Risks for SARS-CoV-2 Antibodies. Front Immunol. 12:6400932021. View Article : Google Scholar : PubMed/NCBI

154 

Yahi N, Chahinian H and Fantini J: Infection-enhancing anti-SARS-CoV-2 antibodies recognize both the original Wuhan/D614G strain and Delta variants. A potential risk for mass vaccination? J Infect. 83:607–635. 2021.PubMed/NCBI

155 

Zhou Y, Liu Z, Li S, Xu W, Zhang Q, Silva IT, Li C, Wu Y, Jiang Q, Liu Z, et al: Enhancement versus neutralization by SARS-CoV-2 antibodies from a convalescent donor associates with distinct epitopes on the RBD. Cell Rep. 34:1086992021. View Article : Google Scholar : PubMed/NCBI

156 

Hachmann NP, Miller J, Collier AY, Ventura JD, Yu J, Rowe M, Bondzie EA, Powers O, Surve N, Hall K and Barouch DH: Neutralization Escape by SARS-CoV-2 Omicron Subvariants BA.2.12.1, BA.4, and BA.5. N Engl J Med. 387:86–88. 2022. View Article : Google Scholar : PubMed/NCBI

157 

Jiang XL, Zhu KL, Wang XJ, Wang GL, Li YK, He XJ, Sun WK, Huang PX, Zhang JZ, Gao HX, et al: Omicron BQ.1 and BQ.1.1 escape neutralisation by omicron subvariant breakthrough infection. Lancet Infect Dis. 23:28–30. 2023. View Article : Google Scholar : PubMed/NCBI

158 

Cao Y, Jian F, Wang J, Yu Y, Song W, Yisimayi A, Wang J, An R, Chen X, Zhang N, et al: Imprinted SARS-CoV-2 humoral immunity induces convergent Omicron RBD evolution. Nature. 614:521–529. 2023.PubMed/NCBI

159 

Röltgen K, Nielsen SCA, Silva O, Younes SF, Zaslavsky M, Costales C, Yang F, Wirz OF, Solis D, Hoh RA, et al: Immune imprinting, breadth of variant recognition, and germinal center response in human SARS-CoV-2 infection and vaccination. Cell. 185:1025–1040.e14. 2022. View Article : Google Scholar : PubMed/NCBI

160 

Liu L, Iketani S, Guo Y, Chan JF, Wang M, Liu L, Luo Y, Chu H, Huang Y, Nair MS, et al: Striking antibody evasion manifested by the Omicron variant of SARS-CoV-2. Nature. 602:676–681. 2022. View Article : Google Scholar : PubMed/NCBI

161 

Yang X, Duan H, Liu X, Zhang X, Pan S, Zhang F, Gao P, Liu B, Yang J, Chi X and Yang W: Broad sarbecovirus neutralizing antibodies obtained by computational design and synthetic library screening. J Virol. 97:e00610232023. View Article : Google Scholar : PubMed/NCBI

162 

Cao Y, Jian F, Zhang Z, Yisimayi A, Hao X, Bao L, Yuan F, Yu Y, Du S, Wang J, et al: Rational identification of potent and broad sarbecovirus-neutralizing antibody cocktails from SARS convalescents. Cell Rep. 41:1118452022. View Article : Google Scholar : PubMed/NCBI

163 

Ma H, Zhang X, Zheng P, Dube PH, Zeng W, Chen S, Cheng Q, Yang Y, Wu Y, Zhou J, et al: Hetero-bivalent nanobodies provide broad-spectrum protection against SARS-CoV-2 variants of concern including Omicron. Cell Res. 32:831–842. 2022. View Article : Google Scholar : PubMed/NCBI

164 

Mendon N, Ganie RA, Kesarwani S, Dileep D, Sasi S, Lama P, Chandra A and Sirajuddin M: Nanobody derived using a peptide epitope from the spike protein receptor-binding motif inhibits entry of SARS-CoV-2 variants. J Biol Chem. 299:1027322023. View Article : Google Scholar : PubMed/NCBI

165 

Ettich J, Werner J, Weitz HT, Mueller E, Schwarzer R, Lang PA, Scheller J and Moll JM: A Hybrid Soluble gp130/Spike-Nanobody Fusion Protein Simultaneously Blocks Interleukin-6 trans-Signaling and Cellular Infection with SARS-CoV-2. J Virol. 96:e01622212022. View Article : Google Scholar : PubMed/NCBI

166 

Lyu X, Imai S, Yamano T and Hanayama R: Preventing SARS-CoV-2 Infection Using Anti-spike Nanobody-IFN-β Conjugated Exosomes. Pharm Res. 40:927–935. 2023. View Article : Google Scholar : PubMed/NCBI

167 

Gruell H, Vanshylla K, Weber T, Barnes CO, Kreer C and Klein F: Antibody-mediated neutralization of SARS-CoV-2. Immunity. 55:925–944. 2022. View Article : Google Scholar : PubMed/NCBI

168 

Suryadevara N, Shrihari S, Gilchuk P, VanBlargan LA, Binshtein E, Zost SJ, Nargi RS, Sutton RE, Winkler ES, Chen EC, et al: Neutralizing and protective human monoclonal antibodies recognizing the N-terminal domain of the SARS-CoV-2 spike protein. Cell. 184:2316–2331.e15. 2021. View Article : Google Scholar : PubMed/NCBI

169 

Voss WN, Hou YJ, Johnson NV, Delidakis G, Kim JE, Javanmardi K, Horton AP, Bartzoka F, Paresi CJ, Tanno Y, et al: Prevalent, protective, and convergent IgG recognition of SARS-CoV-2 non-RBD spike epitopes. Science. 372:1108–1112. 2021. View Article : Google Scholar : PubMed/NCBI

170 

Cerutti G, Guo Y, Wang P, Nair MS, Huang Y, Yu J, Liu L, Katsamba PS, Bahna F, Reddem ER, et al: Neutralizing antibody 5–7 defines a distinct site of vulnerability in SARS-CoV-2 spike N-terminal domain. Cell Rep. 37:1099282021. View Article : Google Scholar : PubMed/NCBI

171 

Graham C, Seow J, Huettner I, Khan H, Kouphou N, Acors S, Winstone H, Pickering S, Galao RP, Dupont L, et al: Neutralization potency of monoclonal antibodies recognizing dominant and subdominant epitopes on SARS-CoV-2 Spike is impacted by the B.1.1.7 variant. Immunity. 54:1276–1289.e6. 2021. View Article : Google Scholar : PubMed/NCBI

172 

Wang Z, Muecksch F, Cho A, Gaebler C, Hoffmann HH, Ramos V, Zong S, Cipolla M, Johnson B, Schmidt F, et al: Analysis of memory B cells identifies conserved neutralizing epitopes on the N-terminal domain of variant SARS-Cov-2 spike proteins. Immunity. 55:998–1012.e8. 2022. View Article : Google Scholar : PubMed/NCBI

173 

Pinto D, Sauer MM, Czudnochowski N, Low JS, Tortorici MA, Housley MP, Noack J, Walls AC, Bowen JE, Guarino B, et al: Broad betacoronavirus neutralization by a stem helix-specific human antibody. Science. 373:1109–1116. 2021. View Article : Google Scholar : PubMed/NCBI

174 

Zhou P, Yuan M, Song G, Beutler N, Shaabani N, Huang D, He WT, Zhu X, Callaghan S, Yong P, et al: A human antibody reveals a conserved site on beta-coronavirus spike proteins and confers protection against SARS-CoV-2 infection. Sci Transl Med. 14:eabi92152022. View Article : Google Scholar : PubMed/NCBI

175 

Shi W, Wang L, Zhou T, Sastry M, Yang ES, Zhang Y, Chen M, Chen X, Choe M, Creanga A, et al: Vaccine-elicited murine antibody WS6 neutralizes diverse beta-coronaviruses by recognizing a helical stem supersite of vulnerability. Structure. 30:1233–1244.e7. 2022. View Article : Google Scholar : PubMed/NCBI

176 

Li W, Chen Y, Prévost J, Ullah I, Lu M, Gong SY, Tauzin A, Gasser R, Vézina D, Anand SP and Goyette G: Structural basis and mode of action for two broadly neutralizing antibodies against SARS-CoV-2 emerging variants of concern. Cell Rep. 38:1102102022. View Article : Google Scholar : PubMed/NCBI

177 

Dacon C, Tucker C, Peng L, Lee CD, Lin TH, Yuan M, Cong Y, Wang L, Purser L, Williams JK, et al: Broadly neutralizing antibodies target the coronavirus fusion peptide. Science. 377:728–735. 2022. View Article : Google Scholar : PubMed/NCBI

178 

Low JS, Jerak J, Tortorici MA, McCallum M, Pinto D, Cassotta A, Foglierini M, Mele F, Abdelnabi R, Weynand B, et al: ACE2-binding exposes the SARS-CoV-2 fusion peptide to broadly neutralizing coronavirus antibodies. Science. 377:735–742. 2022. View Article : Google Scholar : PubMed/NCBI

179 

Sun X, Yi C, Zhu Y, Ding L, Xia S, Chen X, Liu M, Gu C, Lu X, Fu Y, et al: Neutralization mechanism of a human antibody with pan-coronavirus reactivity including SARS-CoV-2. Nat Microbiol. 7:1063–1074. 2022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2024
Volume 30 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang T, Yang D, Tang L and Hu Y: Current development of severe acute respiratory syndrome coronavirus 2 neutralizing antibodies (Review). Mol Med Rep 30: 148, 2024
APA
Zhang, T., Yang, D., Tang, L., & Hu, Y. (2024). Current development of severe acute respiratory syndrome coronavirus 2 neutralizing antibodies (Review). Molecular Medicine Reports, 30, 148. https://doi.org/10.3892/mmr.2024.13272
MLA
Zhang, T., Yang, D., Tang, L., Hu, Y."Current development of severe acute respiratory syndrome coronavirus 2 neutralizing antibodies (Review)". Molecular Medicine Reports 30.2 (2024): 148.
Chicago
Zhang, T., Yang, D., Tang, L., Hu, Y."Current development of severe acute respiratory syndrome coronavirus 2 neutralizing antibodies (Review)". Molecular Medicine Reports 30, no. 2 (2024): 148. https://doi.org/10.3892/mmr.2024.13272