Skip to main content
Wiley - PMC COVID-19 Collection logoLink to Wiley - PMC COVID-19 Collection
. 2020 Sep 21;31(2):e2170. doi: 10.1002/rmv.2170

Viral infection neutralization tests: A focus on severe acute respiratory syndrome‐coronavirus‐2 with implications for convalescent plasma therapy

Daniele Focosi 1,, Fabrizio Maggi 2, Paola Mazzetti 2, Mauro Pistello 3
PMCID: PMC7536930  PMID: 33350017

Summary

Viral neutralization tests (VNTs) have long been considered old‐fashioned tricks in the armamentarium of fundamental virology, with laboratory implementation for a limited array of viruses only. Nevertheless, they represent the most reliable surrogate of potency for passive immunotherapies, such as monoclonal or polyclonal antibody therapy. The recent interest around therapy with convalescent plasma or monoclonal antibodies for the Covid‐19 pandemic has paralleled the revival of VNTs. We review here the available methods by dissecting variations for each fundamental component of the VNT (i.e., virus type and dose, replication‐competent cell line, serum, and detection system).

Keywords: convalescent plasma, high through put neutralizing antibodies, turnaround time, viral neutralization


Abbreviations

BSL

biosafety level

Covid‐19

coronavirus disease 2019

CP

convalescent plasma

CPE

cytopathic effect

CCID

cell culture infectious dose

CLIA

chemiluminescence immunoassay

CMIA

chemiluminescent microparticle immunoassay

CRNT

chemiluminescence reduction neutralization test

ECDC

European Centre for Disease Prevention and Control

ECLIA

electrochemilunescence immunoassay

ELISA

enzyme‐linked immunosorbent assay

FDA

Food and Drug Administration

FFA

focus‐forming assay

FFU

focus‐forming unit

FRNT

focus neutralization reduction test

MERS‐CoV

Middle East respiratory syndrome‐related coronavirus

MN

microneutralization

nAb

neutralizing antibodies

PFU

plaque‐forming units

PRNT

plaque reduction neutralization test

RBD

receptor‐binding domain

RCCL

replication‐competent cell line

RLU

relative light units

TCID

tissue culture infectious dose

SARS‐CoV‐2

severe acute respiratory syndrome coronavirus 2

TMPRSS2

transmembrane protease, serine 2

VNT

viral neutralization test

1. INTRODUCTION

The magnitude of neutralizing antibody (nAb) responses to SARS‐CoV‐2 is extremely variable, and a significant fraction of convalescent individuals have comparatively low to undetectable levels of plasma nAb. 1 , 2 Exact quantification of nAb has implications for studying duration of immunity (acquired either by natural infection or vaccination), and selection of convalescent plasma (CP) donors, 3 or relative potency 4 and durability 5 of monoclonal antibody therapies. In fact, neutralizing potency of sera was found to be greater in patients who went on to resolve infection, compared with those that died from Covid‐19, 6 and CP therapy is more efficacious in patients receiving units with highest titres of nAb. 3

SARS‐CoV‐2 Spike (S) protein is the main surface protein of SARS‐CoV‐2 and the target of neutralizing activity: it consists of an N‐terminal S1 subunit responsible for virus—receptor binding and a C‐terminal S2 subunit responsible for virus—cell membrane fusion. S1 itself consists of an N‐terminal domain and a receptor‐binding domain (RBD). Most coronavirus nAb target the RBD, while a few target regions in the S2 subunit or the S1/S2 proteolytic cleavage site. 7 By depleting sera of subunit‐specific antibodies to determine the contribution of these individual subunits to the antigen‐specific nAb response, Steffen et al. demonstrated that epitopes within RBD are the target of a majority of the nAb in the human polyclonal antibody response. 8 Barnes et al. classified nAbs into three categories 1 : VH3‐53 hNAbs with short CDRH3s that block angiotensin‐converting enzyme 2 (ACE2) and bind only to up RBDs, 2 ACE2‐blocking hNAbs that bind both up and down RBDs and can contact adjacent RBDs, 3 hNAbs that bind outside the ACE2 site and recognize up and down RBDs. 9

S variants that resist commonly elicited nAb are now present at low frequencies in circulating SARS‐CoV‐2 populations. 10 While vaccines are still under development and reinfection from next pandemic waves is still under observation, CP donor selection represents the most urgent challenge.

To date more than 90,000 patients across the world have already been treated with CP, mostly in non‐randomized studies (65,000 in the United States only thanks to expanded access programs approved by FDA 11 ). The efficacy of CP therapy is believed to rely on nAb content. 3 Convalescent patients should hence be screened for the presence of nAb levels, and donations collected only from convalescent individuals with high nAb titres. In the setting of Covid‐19, several regulatory authorities recommend threshold values, but none of them specifies nAb assay details that could alter test output: FDA says that ‘when a measurement of nAb titres is available, we recommend nAb titres of at least 1:160. A titre of 1:80 may be considered acceptable if an alternative matched unit is not available. When the measurement of nAb titres is not available, consider storing a retention sample from the CP donation for determining antibody titres at a later date. 12 The ECDC basically endorse FDA recommendations, suggesting that immunocompromised recipients are transfused with CP units having a titre ≥1:320. 13 As previously said, those thresholds have poor meaning unless details are disclosed, making trial results poorly comparable.

Neutralizing antibody assessment has historically been performed using time‐consuming and hazardous methods that required high technical skills. While high throughput platform surrogate tests having substantially shorter turnaround time and good correlations with nAb are heavily under study, old‐fashioned methods remain the gold standard for exact nAb quantification. The Covid‐19 pandemic has had the side benefit of expediting research on nAb testing upgrades. This manuscript reviews the principle behind classical nAb testing by dissecting each key component (as depicted in Figure 1), and the developments that have been released in the last years.

FIGURE 1.

FIGURE 1

Summary of the key components of a viral neutralization assay

1.1. The replication‐competent cell line

The virus—serum mixture is generally added onto confluent cell monolayer. In the case of SARS‐CoV‐2, several cell lines naturally express high levels of ACE2: the—by far—mostly frequently used cells are African green monkey (Chlorocebus sabaeus) Vero E6 (a.k.a. Vero 1008, ATCC® CRL‐1586™) or Vero CC‐81 (a.k.a. Vero CCL‐81 or Vero WHO, ATCC® CCL‐81™) kidney epithelial cells. At a dose of 3 × 103, 1.5 × 104 or 3 × 104 cells/well monolayers that are 70%, 80%, and 90% confluent, respectively, are generated in 24 h with both cell lines. Vero CCL‐81 result in about 2 folds higher foci formation per well relative to Vero E6, despite releasing less viral genome copies as detected by RT‐quantitative polymerase chain reaction (qPCR). 14 Alternatively, human lung epithelial cells CALU‐3, human hepatoma (Huh7.5 and Huh7), human gastric adenocarcinoma AGS and MKN have proven as effective as Vero cells. 14

Mammalian cell lines not expressing human ACE2 can be transduced or plasmid‐transfected with ACE2 (e.g., human lung epithelial cells A549, 15 human embryo kidney [HEK] 293T 16 , 17 or 293FT, or BHK21 18 , 19 or human connective tissue HT1080 20 ). Rodent 3T3 and SHHC17 cell lines are instead not permissive. 14

Stable introduction of the S activating Transmembrane protease, serine 2 (TMPRSS2) further enhanced susceptibility to infection by 5–10 folds. 19 , 21

Schmidt et al. have reported that HT1080/ACE2cl.14 and Huh7.5 cell lines are significantly more adherent than 293T‐derived cell lines and are hence recommended (for HIV‐1 and vesicular stomatitis virus [VSV] pseudotype assays, respectively; see paragraph below) in high throughput situations, as great care is necessary when using 293T‐derived cells whose adhesive properties during washing steps are suboptimal. 20

1.2. The viral challenge and virus quantification

Intact virions or several different surrogates can be used to represent the viral challenge, as detailed below.

1.2.1. Intact virions

The challenging dose has variable amounts of virus. Virus quantity can be determined with protein assays (such as haemagglutination assays for influenza viruses, the colorimetric bicinchonic acid assay, or single radial immunodiffusion assay), plaque assays (reported as plaque‐forming units [PFU] per ml), endpoint dilution assay (reported as median tissue culture infectious dose [TCID50] or cell culture infectious dose [CCID50], that is, the amount of virus required to kill 50% of infected host cells or to produce a cytopathic effect [CPE] in 50% of inoculated tissue culture cells).

Plaque assays have both an immunostaining variant (focus‐forming assay, with virus quantity reported as focus‐forming unit per ml) and, for firefly luciferase‐tagged recombinant viruses, 22 a luminometric variant (with quantity reported in relative light units). 17

More modern methods of virus quantification include transmission electron microscopy, tuneable resistive pulse sensing, flow cytometry, quantitative PCR, or ELISA: they have shorter turnaround times but do not provide information about virus viability. Hence, it is easily inferred that TCID is generally preferred to define the challenging dose, 23 and the challenging value is generally 100 TCID50 of input virus per well, 24 presenting a difficulty for viruses which replicate to low titre in cell culture (such as the majority of recent A [H3N2] isolates). Assuming that the same cell system is used, that the virus forms plaques on those cells, and that no procedures are added which would inhibit plaque formation, according to Poisson distribution, 1 TCID50 is approximatively 0.69 PFU.

1.2.2. Pseudotyped viruses

SARS‐CoV‐2 has been classified as a category B pathogen, but biosafety caution applicable to the category A pathogens are encouraged when handling it. Manipulation of SARS‐CoV‐2 should only be carried out in a biosafety level 3 (BSL3) facility with negative air pressure. Cell culture procedures are carried out in BSL2 and moved to BSL3 when ready for viral infection. 25

Pseudotyped viruses provide a safe viral entry model because of their inability to produce infectious progeny virus. In the pseudoparticle neutralization test (ppNT; a.k.a. pseudovirion neutralization assay [PsVNA]), a single‐cycle, replication‐defective virus (e.g., retroviruses such as replication‐defective HIV‐1, third‐generation lentiviral [pLV], or G‐protein‐deficient VSV 18 , 20 ) is pseudotyped with the surface protein from the virus against which NAb should be measured. This activity can be run in BSL2 facilities. For SARS‐CoV‐2, the expression of full‐length S protein was enhanced over 10‐fold by deleting the last C‐terminal 18 18 or 19 19 , 26 amino acids of the cytoplasmic tail, or by codon optimization. 17 Such methodology has previously been used to produce pseudotyped viruses for SARS‐CoV‐1 27 and MERS‐CoV. 28 Modification of a single amino acid in the Furin cleavage site of S (R682Q) enhanced infectious particle production another 10‐fold. With all enhancing elements combined, the titre of pseudotyped particles reached almost 106 infectious particles/ml. 19 Nevertheless, lower Spike densities in pseudotypes viruses could affect the avidity of bivalent antibodies, particularly those that are unable to engage two S‐protein monomers within a single trimer and whose potency is dependent on engaging 2 adjacent trimers.

1.2.3. VSV pseudotypes

Assembly of the VSV occurs at the plasma membrane and involves budding of virions from the cell surface. During budding, VSV acquires an envelope consisting of a lipid bilayer derived from the plasma membrane and spike proteins consisting of trimers of the VSV‐glycoprotein (VSV‐G). When the VSV‐G is absent and the glycoprotein from a heterologous virus is complacently expressed in cells infected with recombinant vesicular stomatitis virus with protein G deletion (rVSV‐dG), the glycoprotein of the heterologous virus could be assembled into the VSV membrane (30). Recently, VSVdG‐luc bearing S chimeras has been used to study the cell entry and receptor usage for SARS‐CoV‐2 and other lineage B betacoronaviruses. 29 A PsVNA assay for SARS‐CoV‐2, which consists of pseudotyped VSV bearing the full‐length Sprotein of SARS‐CoV‐2 and Huh7 cell, has been successfully tested. 30 Interestingly, the absence of proof‐reading activity in VSV‐L polymerase has been exploited to generate virus stocks with greater diversity (especially in S protein) than authentic SARS‐CoV‐2. 10 The assay provides results in 12–16 h.

Because single‐cycle, replication‐defective pseudotypes viruses do not allow for any viral spread and this could impact the sensitivity of the VNT, replication‐competent VSV/SARS‐CoV‐2 chimeric viruses have been generated for usage in multicycle replication‐based assays. 20

1.2.4. Lentiviral pseudotypes

Lentiviral pseudotype bearing the truncated spike protein of SARS‐CoV‐2 was also constructed and used to study the virus entry and its immune cross‐reactivity with SARS‐CoV‐1. 20 , 31 , 32 In one study, Moloney murine leukemia virus (MMLV) was able to pseudotype the VSV‐G glycoprotein efficiently but was unable to pseudotype the SARS‐CoV‐2 Spike protein. 16 However, in a different study, MMLV was able to pseudotype the SARS‐CoV‐2 Spike protein. 24 These differences could be due to the constructs used to express the Spike protein with the former study expressing a full‐length Spike protein, whilst the latter study expressed a Spike protein with a deletion in the C‐terminal 19 amino acid (aa) that could aid better expression as alluded to earlier. 33 In contrast, pLV pseudotyped both glycoproteins efficiently; however, much higher titres of pLV‐G particles were produced. Among all the tested mammalian cells, HEK 293Ts expressing human ACE2 (hACE2) were most efficiently transduced using the pLV‐S system. 16 The neutralizing activity of both CP and human monoclonal antibodies measured using each virus correlated quantitatively with nAb measured using an authentic SARS‐CoV‐2 neutralization assay. The assay provides results in approximatively 48 h.

1.2.5. Engineered viruses

Genetically engineered viruses harbouring a reporter gene that is exploited for the detection system (see dedicated chapter below) have been developed by several research groups: examples include luciferases (firefly, Renillareniformis or nanoluciferase 15 ), green fluorescent protein 34 or mNeonGreen integration into ORF7. 35 Among luciferases, Gaussia princeps luciferase, the smallest known to date, is naturally secreted into mammalian cell culture media, thus avoiding the cell lysis step. 36 The results can be obtained by automatically counting positive cell number at 5–12 h after infection, making the assay convenient and high‐throughput. In the case of mNeonGreen, VNT in Vero CC‐81 cells have shown high correlation with VNT using intact virions. 37 When a chemiluminescent reporter is used, the assay is called chemiluminescence reduction neutralization test (CRNT). 38

1.2.6. Nonviral alternatives

SARS‐CoV‐2 Spike trimer fused to a constitutively fluorescent protein (Gamillus, isolated from Olindias formosus) has also been developed, providing a versatile tool for high‐throughput screening and phenotypic characterization of SARS‐CoV‐2 entry inhibitors. 39

The Promega HiBiT/LgBiT® system can be exploited for HiBiT‐tagged VNT (hiVNT): genome‐free virus‐like particles are incorporated with a small luciferase peptide (HiBiT) and their entry into LgBiT‐expressing target cells reconstitutes NanoLuc luciferase readily detected by a luminometer within 3 h. 40

1.3. The patient’ serum

It is important to heat‐inactivate serum or plasma samples from Covid‐19 patients at 56°C for 30 min to 1 h before performing the assay to destroy residual viral particles: this step is less crucial for SARS‐CoV‐2 because of rare and low‐titre viremia. It has been reported that complement deposition on virus envelope may lead to infection‐enhancement which may mask the neutralizing effects of Abs contained in serum or plasma samples. 41 Although many laboratories start with a 1:10 dilution, for serum or plasma samples it is recommended starting with a 1:100 dilution to avoid potential impurities that may affect the sensitivity of the assay. Dulbecco's minimal essential medium (DMEM) supplemented with NaHCO3, hydroxyethylpiperazine ethane sulfonic acid (HEPES) buffer, penicillin, streptomycin, and 1% foetal bovine serum, also used as cell culture medium, is typically used for the dilutions. In the case of mAbs, 10 µg is a commonly used starting dose: however, this will depend on the neutralizing capability of the mAb.

Since EDTA chelates calcium and blocks the complement cascade, EDTA‐anticoagulated plasma samples cannot be used in VNT that detect CPE induced by complement‐dependent cytotoxicity (e.g., PRNT). On the contrary, CRNT can instead use whole blood. 38

1.4. The detection system

The classical plaque reduction neutralization test (PRNT) is performed in a 24‐well format in duplicate for each serum dilution. The virus‐serum‐cell mixture is left for 1 h at 37°C in a 5% CO incubator. 42 Then, the supernatant is removed and the cells overlaid with 1% agarose in cell culture medium (generally Minimum Essential Medium with 2% foetal bovine serum). After 3–5 days incubation at 36.5°C in a 5% CO2, the plates are fixed and stained. Positive and negative controls and a virus back‐titration are included in each assay. A microscope is used to detect plaques due to CPE on the replication‐competent cell line (RCCL) monolayer. 43 Antibody titres are defined as reciprocal of the highest serum dilution that resulted in >90% (PRNT90) or > 50% (PRNT50) reduction in the number of plaques. When PRNT is run in 96‐well plates it is called microneutralization (MN) assay (or when, pseudotyped viruses are used, pseudotype MN [pMN] assay). 44

In the alternative focus neutralization reduction test (FRNT) cells, the development of visible spots is dependent on the time it takes for viral protein production to occur (rather than to cause lysis as in PRNT) and for infectious virus to spread to neighbouring susceptible cells. Cells (as explained above, Vero WHO cells are preferred) are hence stained after 24–48 h incubation using an antiviral serum as the primary antibody and a secondary horseradish peroxidase‐labelled IgG targeting the Fc of the primary antibody. 14 The signal can be developed using a precipitate forming 3,3′,5,5′‐tetramethylbenzidine substrate, and the number of infected cells per well are counted using an ELISpot analyser (e.g., ImmunoSpot® 5 Image analyser, CTL Europe GmbH 45 ). Again, antibody titres are defined as reciprocal of the highest serum dilution that resulted in >90% (FRNT90) or > 50% (FRNT50) reduction in the number of foci. The FRNT is also amenable to a 96‐well plate format.

Alternatively, the infrared staining technique (relying on secondary Ab IRDye 800CW to stain virus‐infected cells, and DRAQ5TM Fluorescent Probe Solution stain the nucleus) holds the advantage of its capability to measure cell viability (in addition to measuring antiviral activity) using readings at 700 nm (800CW, measuring viral infection) and 800 nm (DRAQ5, measuring cell viability) on the Odyssey Sa Infrared Imaging System. 45

As previously stated, when the firefly luciferase reporter gene is inserted in the viral construct, cells are lysed and assayed for luciferase expression. 17 , 22

The evaluability of the PRNT technique may be further improved by overlaying the cells with cellulose or by using specific antibodies to detect remaining viral antigens in the cells. 46

1.5. Correlation with high‐throughput serological platforms

Many studies have investigated correlations between PRNT and other serological assays. While several studies only correlated PRNT with in‐house ELISAs, 21 , 47 , 48 , 49 , 50 Table 1 summarizes studies comparing PRNT with other marketed serological platforms based on enzymes (ELISA), chemiluminesce (CLIA), eletrochemiluminescence (ECLIA) or chemiluminescent microparticle immunoassays, and targeting different viral antigens in the setting of interventional 65 , 66 or observational 26 , 42 , 46 , 51 , 52 , 53 , 54 , 55 trials.

TABLE 1.

Summary of published correlation studies of PRNT with marketed serological assays (r‐values indicate Spearman correlation coefficient; x: r not available).

PRNT Ref. n Anti‐S1 RBD Anti‐S1 Anti‐S1/S2 trimers Anti‐N Anti‐S + Anti‐N
ELISA (OD ratio) CLIA (AU/ml) CMIA (S/CO) ECLIA (COI) ELISA (OD ratio) CLIA (AU/ml)
Fortress Wantai Beckman Coulter Access® Euroimmun Ortho Vitros DiaSorin Abbott Roche Elecsys® Epitope EDI™ Mikrogen recom Well Diazyme DZ‐Lite SARS‐CoV‐2 IgG CLIA test YHLO
Total IgG IgG IgA IgG IgG IgG Total IgG IgG IgG IgG
BEP2000® LIAISON® Architect® Cobas® BEP2000® DZ‐Lite 3000 Plus® iFlash 1800
VSV‐pseudotyped Vero E6 PRNT50PRNT90 26 181 x x
England/02/2020 and lentivirus pseudotyped (r = 0.83) Vero E6 PRNT50 42 51 0.58 0.88
German isolate Vero E6 PRNT50 46 10 + 31 0.51/0.92 0.63/0.98
PRNT90 0.85/0.88 0.79/0.93
(HIV/NanoLuc)‐SARS‐CoV‐2 pseudotype 51 30 0.75 0.72
VSV/NG/NanoLuc‐SARS‐CoV‐2 pseudotype 0.70 0.69
M16502 isolate Vero PRNT50 52 47 x x x x x x
n.a. n.a n.a 53 26 0.69 0.76 0.56 0.45 0.35
Isolate Vero E6 PRNT50 PRNT90 54 38 x x
SARS‐CoV‐2/USA‐WA1/2020 Vero‐E6‐TMPRSS2 cells PRNT50 55 126 x x
Germany/BavPat1/2020 Vero n.a. 56 100 0.72 0.76 x x
hCoV‐19/Italy/UniSR1/2020 Vero E6 n.a. 57 46 x x
MLV‐based pseudotype ACE2‐transduced HeLa PRNT50 58 164 x x x
n.a. Vero n.a. 59 44 x x
Lombardy isolate Vero E6 PRNT50 60 304 x
SARS‐CoV‐2//Finland/1/2020 Vero E6 Titre >40 61 70 x x x x
Italian isolate Vero E6 n.a. 62 18 x
mNeonGreen SARS‐CoV‐2 Vero E6 PRNT50 63 47 0.48 0.52 0.37
2019‐nCov/Italy‐INMI1 Vero E6 PRNT50 64 181 x x

Reference 60 also reports correlation with Acro Biotech 2019‐nCoV IgG and IgM assays and with Xiamen Biotime SARS‐CoV‐2 IgG and IgM assays, not reported in this table.

Abbreviations: PRNT, plaque reduction neutralization test; VSV, vesicular stomatitis virus.

Although several assays correlated better than others, even the best performing serological assays had poor correlation results, implying that anti‐SARS‐CoV‐2 nAb should be titrated using a VNT to optimize CP therapy. One major cause could be that, despite IgM, IgG, and IgA are capable of mediating neutralization, VNT titres correlated better with binding levels of IgM and IgA1 than IgG. 67 In addition, the quaternary structure of S protein available on infected RCCL is hardly replicated by recombinant antigens bound on solid substrates: for this reasons, alternative high‐throughput methods of antibody quantification based on Spike expression in cell lines are being developed, 68 but no correlation studies with VNT have.

In the largest study to date, ROC analysis showed Euroimmune anti‐S1 IgG ELISA AUC outperformed 6 different in‐house ELISAs and pseudotyped PRNT at predicting PRNT titres >1:100 against the native isolate. A cut‐off value of 9.1 S/CO in the Euroimmune ELISA identified 65% of donations above the 1:100 nAb threshold with no false identification of donations below this nAb threshold. 42

1.6. PseudoNAb ACE2‐competing assays

New‐generation, cell‐free, protein‐based pseudo‐nAb assays (a.k.a. surrogate virus neutralization test [sVNT]) have been developed, where cells are replaced by receptors, and the virus is replaced by surface proteins. Among them, a competitive serological assay can simultaneously determine an individual's seropositivity against the SARS‐CoV‐2 S protein RBD and estimate the neutralizing capacity of anti‐S antibodies to block interaction with the human ACE2 required for viral entry. 69 In an ELISA‐based assay, Zheng et al. presented natively‐folded S protein RBD‐containing antigens via avidin‐biotin interactions. Sera are then supplemented with soluble ACE2‐Fc to compete for RBD‐binding serum antibodies, and antibody binding was quantified. A comparison of signals from untreated serum and ACE2‐Fc‐treated serum reveals the presence of antibodies that compete with ACE2 for RBD binding. 70 This test is performed on the same platform and in parallel with an ELISA for the detection of antibodies against the RBD. 71 , 72

An entirely different approach is based on the antibody detection by agglutination PCR (ADAP) methodology. A cell‐free neutralization polymerase chain reaction (PCR) assay using SARS‐CoV‐2 S protein and human ACE2 receptor‐DNA conjugates has been developed to quantify nAbs. Briefly, the neutralizing antibodies in the specimen will engage with S1‐DNA conjugate in step 1 to decrease S1‐DNA binding with ACE2‐DNA in step 2. Even this assay can be run in BSL2 and provide results in 2−3 h. 73

1.7. Clinical correlates for nAb titres

Sixty percent of 24 hospital personnel with mild Covid‐19 developed nAb titres <1:20. 54 Patients with mild Covid‐19 disease produced stronger nAb responses than asymptomatic individuals. 67 , 74 Significantly higher nAb titres were accordingly observed in patients with severe forms versus asymptomatic carriers. 21 An infection without fever had a negative predictive value of 92% for nAb titres >1:200. 56 Aziz et al. reported that a history of reduced taste or smell, fever, chills/hot flashes, pain while breathing, pain in arms/legs, as well as muscle pain and weakness were significantly associated with the presence of nAb in those with mild to moderate infection. 75 Infection with the recently described S protein variant 614G produced higher levels of nAb when compared to viruses possessing the 614D variant (6).

1.8. nAb decline

While the overall antibody responses for other beta coronaviruses typically declines after 6–12 months, 76 SARS‐CoV‐specific nAb usually persist for 2 years. 77 In most of Covid‐19 inpatients, nAb reached a plateau 2 weeks post‐symptom onset and then declined, reaching a low or undetectable level ≥40 days post‐symptom onset. 21 In less severe cases, nAb in serum reached a peak about 4 weeks after disease onset but dropped to a lower level about 6 weeks later. 78 An earlier IgG antibody response against the S2 domain of the S protein could better mediate virus neutralization, as previously suggested for SARS‐CoV‐1 nAb targeting the S2 domain. 26 , 79 , 80 , 81 Analyses at a 1‐month interval on 31 convalescent individuals showed that RBD‐specific IgG slightly decreased between 6 and 10 weeks after symptoms onset but RBD‐specific IgM decreased much more abruptly. Similarly, a significant decrease in the capacity of CP to neutralize pseudo particles bearing SARS‐CoV‐2 S wild‐type or its D614G variant has been reported. 82

The magnitude of the nAb response is correlated with disease severity, but this does not affect the kinetics of the nAb response. Whilst some individuals with high peak ID50 (>10,000) maintained titres >1000 at >60 days after onset of symptoms, some with lower peak ID50 had titres approaching baseline within a 94 days follow‐up. 83 Neutralizing activity increased with time after the onset of symptoms, reaching a peak at 31–35 days. At this point, the number of sera having nAb titres ≥160 was about 93% (PRNT50) and 54% (PRNT90). Sera with high SARS‐CoV‐2 antibody levels (≥960 in‐house ELISA RBD titres) showed maximal activity, but not all high titre sera contained nAb. 48

Sterling et al. reported that while the specific antibody response to SARS‐CoV‐2 included IgG, IgM, and IgA, the latter contributed to a much larger extent to VNT titre, as compared to IgG, but declined after just 1 month. 84 In 27 patients Ma et al. estimated that convalescent patients' RBD‐specific IgG reach an undetectable level approximatively 273 days after hospital discharge, while the predicted decay times are 150 and 108 days for IgM and IgA, respectively. 85

Wajnberg et al. reported that more than 90% of infected individuals with mild‐to‐moderate Covid‐19 experience robust IgG antibody responses against S protein, based on a dataset of 19,860 individuals screened at Mount Sinai Health System in New York City, which were stable for at least a period approximating 3 months, and correlated with neutralization of authentic SARS‐CoV‐2. 86 In another series of 30 patients, Wang et al. reported that SARS‐CoV‐2‐specific nAb titres were low for the first 7–10 days after symptom onset and increased after 2–3 weeks. The median peak time for nAbs was 33 days after symptom onset. nAb titres in 93.3% (28/30) of the patients declined gradually over the 3‐months study period, with a median decrease of 34.8% (IQR 19.6%–42.4%). NAb titres increased over time in parallel with the rise in IgG antibody levels, correlating well at week 3 (r = 0.41, p < 0.05). 87 Similarly, Crawford et al. reported in a series of 34 patients that nAb titres declined an average of about four‐fold from one to four months post‐symptom onset. 88 The decline in anti‐RBD antibodies was not related to the number of donations but strongly correlated with the number of days after symptoms onset (r = 0.821). 89

The rapid decline in nAb may be attributed to the rapid decay of IgM in the acute phase. However, the relative contribution of IgG to nAb increased and that of IgM further decreased after 6 weeks after symptom onset. 12 Accordingly, Lei et al. reported that the titres of neutralizing antibodies in asymptomatic individuals gradually vanished in 2 months. 74

Gontu et al. reported that robust IgM, IgG, and VNT responses to SARS‐CoV‐2 persist, in the aggregate, for at least 100 days post‐symptom onset. However, a notable acceleration in decline in virus neutralization titres ≥160, a value suitable for CP therapy, was observed starting 60 days after first symptom onset. 90

2. CONCLUSIONS

The assays described above are adaptable to high‐throughput and are useful tools in the evaluation of serologic immunity conferred by vaccination or prior SARS‐CoV‐2 infection, as well as the potency of CP or human monoclonal antibodies.

Endorsing specific protocols and disclosing them in guidelines and recommendations will largely facilitate a comparison between clinical trials.

AUTHOR CONTRIBUTIONS

Focosi Daniele designed the paper, searched relevant literature and wrote the first draft. Maggi Fabrizio analysed the literature and created figure and table. Maggi Fabrizio, Mazzetti Paola, and Pistello Mauro critically revised the manuscript.

3.

DATA AVAILABILITY STATEMENT

This manuscript contains no original data.

References

REFERENCES

  • 1. Robbiani DF, Gaebler C, Muecksch F, et al. Convergent antibody responses to SARS‐CoV‐2 in convalescent individuals. Nature. 2020;7821(584):437–442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Suthar MS, Zimmerman MG, Kauffman RC, et al. Rapid generation of neutralizing antibody responses in COVID‐19 patients. Cell Rep Med. 2020;1(3):100040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Focosi D, Tang J, Anderson A, Tuccori M. Convalescent plasma therapy for Covid‐19: state of the art. Clin Microbiol Rev. 2020;33(4):e00072‐20. 10.1101/2020040097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Pinto D, Park YJ, Beltramello M, et al. Cross‐neutralization of SARS‐CoV‐2 by a human monoclonal SARS‐CoV antibody. Nature. 2020;583(7815):290‐295. [DOI] [PubMed] [Google Scholar]
  • 5. Robbie GJ, Criste R, Dall'acqua WF, et al. A novel investigational Fc‐modified humanized monoclonal antibody, motavizumab‐YTE, has an extended half‐life in healthy adults. Antimicrob Agents Chemother. 2013;57(12):6147‐6153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Tighe PJ, Urbanowicz RA, Fairclough L, et al. Potent anti‐SARS‐CoV‐2 antibody responses are associated with better prognosis in hospital inpatient COVID‐19 disease. 2020. 10.1101/2020.08.22.20176834. [DOI] [Google Scholar]
  • 7. Jiang S, Hillyer C, Du L. Neutralizing antibodies against SARS‐CoV‐2 and other human coronaviruses. Trends Immunol. 2020;41(5):355‐359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Steffen TL, Stone ET, Hassert M, et al. The receptor binding domain of sars‐cov‐2 spike is the key target of neutralizing antibody in human polyclonal sera. 2020. 10.1101/2020.08.21.261727. [DOI] [Google Scholar]
  • 9. Barnes CO, Jette CA, Abernathy ME, et al. Structural classification of neutralizing antibodies against the sars‐cov‐2 spike receptor‐binding domain suggests vaccine and therapeutic strategies. 2020. 10.1101/2020.08.30.273920. [DOI] [Google Scholar]
  • 10. Weisblum Y, Schmidt F, Zhang F, et al. Escape from neutralizing antibodies by SARS‐CoV‐2 spike protein variants. 2020. 10.1101/2020.07.21.214759 [DOI] [PMC free article] [PubMed]
  • 11. Martinez‐Resendez MF, Castilleja‐Leal F, Torres‐Quintanilla A, et al. Initial experience in mexico with convalescent plasma in COVID‐19 patients with severe respiratory failure, a retrospective case series. 2020. 10.1101/2020.07.14.20144469. [DOI] [Google Scholar]
  • 12. Yao X‐Y, Liu W, Li Z‐Y, et al. Neutralizing and binding antibody kinetics of COVID‐19 patients during hospital and convalescent phases. 2020. 10.1101/2020.07.18.20156810. [DOI] [Google Scholar]
  • 13. Commission E . An EU Programme of COVID‐19 Convalescent Plasma Collection and Transfusion. Guidance on Collection, Testing, Processing, Storage, Distribution and Monitored Use 2020. Retrieved from https://ec.europa.eu/health/sites/health/files/blood_tissues_organs/docs/guidance_plasma_covid19_en.pdf.
  • 14. Stone ET, Geerling E, Steffen TL, et al. Characterization of cells susceptible to SARS‐COV‐2 and methods for detection of neutralizing antibody by focus forming assay. 2020. 10.1101/08.20.259838. [DOI]
  • 15. Xie X, Muruato AE, Zhang X, et al. A nanoluciferase SARS‐CoV‐2 for rapid neutralization testing and screening of anti‐infective drugs for COVID‐19. bioRxiv. 2020. 10.1101/2020.06.22.165712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Tandon R, Mitra D, Sharma P, Stray SJ, Bates JT, Marshall GD. Effective screening of SARS‐CoV‐2 neutralizing antibodies in patient serum using lentivirus particles pseudotyped with SARS‐CoV‐2 spike glycoprotein. 2020. 10.1101/2020.05.21.20108951. [DOI] [PMC free article] [PubMed]
  • 17. Thompson C, Grayson N, Paton R, et al. Neutralising antibodies to SARS Coronavirus 2 in Scottish blood donors ‐ a pilot study of the value of serology to determine population exposure. 2020. 10.1101/2020.04.13.20060467. [DOI] [PMC free article] [PubMed]
  • 18. Xiong H‐L, Wu Y‐T, Cao J‐L, et al. Robust neutralization assay based on SARS‐CoV‐2 S‐bearing vesicular stomatitis virus (VSV) pseudovirus and ACE2‐overexpressed BHK21 cells. bioRxiv. 2020. 10.1101/2020.04.08.026948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Johnson MC, Lyddon TD, Suarez R, et al. Optimized pseudotyping conditions for the SARS‐CoV2 spike glycoprotein. 2020. 10.1101/2020.05.28.122671. [DOI] [PMC free article] [PubMed]
  • 20. Schmidt F, Weisblum Y, Muecksch F, et al. Measuring SARS‐CoV‐2 neutralizing antibody activity using pseudotyped and chimeric viruses. J Exp Med. 2020;217(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Brochot E, Demey B, Touze A, et al. Anti‐spike, anti‐nucleocapsid and neutralizing antibodies in SARS‐CoV‐2 inpatients and asymptomatic carriers. medRxiv. 2020. 10.1101/2020.05.12.20098236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Garcia V, Krishnan R, Davis C, et al. High‐throughput titration of luciferase‐expressing recombinant viruses. JoVE. 2014;91:51890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Mendoza EJ, Manguiat K, Wood H, Drebot M. Two detailed plaque assay protocols for the quantification of infectious SARS‐CoV‐2. Curr Protoc Microbiol. 2020;57(1):ecpmc105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Rowe T, Abernathy RA, Hu‐Primmer J, et al. Detection of antibody to avian influenza A (H5N1) virus in human serum by using a combination of serologic assays. J Clin Microbiol. 1999;37(4):937‐943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Ferrara F, Temperton N. Pseudotype neutralization assays: from laboratory bench to data analysis. Methods Protocols. 2018;1(1).8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Meyer B, Torriani G, Yerly S, et al. Validation of a commercially available SARS‐CoV‐2 serological immunoassay. medRxiv. 2020. 10.1101/2020.05.02.20080879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Temperton NJ, Chan PK, Simmons G, et al. Longitudinally profiling neutralizing antibody response to SARS coronavirus with pseudotypes. Emerg Infect Dis. 2005;11(3):411‐416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Grehan K, Ferrara F, Temperton N. An optimised method for the production of MERS‐CoV spike expressing viral pseudotypes. Methods (Orlando). 2015;2:379‐384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Letko M, Marzi A, Munster V. Functional assessment of cell entry and receptor usage for SARS‐CoV‐2 and other lineage B beta coronaviruses. Nature Microbiol. 2020;5(4):562‐569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Nie J, Li Q, Wu J, et al. Establishment and validation of a pseudovirus neutralization assay for SARS‐CoV‐2. Emerg Microbes Infect. 2020;9(1):680‐686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Ou X, Liu Y, Lei X, et al. Characterization of spike glycoprotein of SARS‐CoV‐2 on virus entry and its immune cross‐reactivity with SARS‐CoV. Nat Commun. 2020;11(1):1620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Crawford KHD, Eguia R, Dingens AS, et al. Protocol and reagents for pseudotyping lentiviral particles with SARS‐CoV‐2 spike protein for neutralization assays. Viruses. 2020;12(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Zheng Y, Larragoite ET, Lama J, et al. Neutralization assay with SARS‐CoV‐1 and SARS‐CoV‐2 spike pseudotyped murine leukemia virions. 2020. 10.1101/2020.07.17.207563. [DOI] [PMC free article] [PubMed]
  • 34. Noval MG, Kaczmarek ME, Koide A, et al. High titers of multiple antibody isotypes against the SARS‐CoV‐2 Spike receptor‐binding domain and nucleoprotein associate with better neutralization. 2020. 10.1101/2020.08.15.252353. [DOI]
  • 35. Xie X, Muruato A, Lokugamage KG, et al. An infectious cDNA clone of SARS‐CoV‐2. Cell Host Microbe. 2020;27(5):841‐848. e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Zeng C, Evans JP, Pearson R, et al. Neutralizing antibody against SARS‐CoV‐2 spike in COVID‐19 patients, health care workers and convalescent plasma donors: a cohort study using a rapid and sensitive high‐throughput neutralization assay. 2020. 10.1101/2020.08.02.20166819. [DOI] [PMC free article] [PubMed]
  • 37. Muruato AE, Fontes‐Garfias CR, Ren P, et al. A high‐throughput neutralizing antibody assay for COVID‐19 diagnosis and vaccine evaluation. Nat Commun. 2020;11(1):4059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Tani H, Tan L, Kimura M, et al. Evaluation of SARS‐CoV‐2 neutralizing antibodies using a vesicular stomatitis virus possessing SARS‐CoV‐2 spike protein. 2020. 10.1101/2020.08.21.262295. [DOI] [PMC free article] [PubMed]
  • 39. Zhang Y, Wang S, Wu Y, et al. Virus‐free and live‐cell visualizing SARS‐CoV‐2 cell entry for studies of neutralizing antibodies and compound inhibitors. 2020. 10.1101/2020.07.22.215236. [DOI] [PMC free article] [PubMed]
  • 40. Miyakawa K, Jeremiah SS, Ohtake N, et al. Rapid quantitative screening assay for SARS-CoV-2 neutralizing antibodies using HiBiT‐tagged virus‐like particles. 2020. 10.1101/2020.07.20.20158410. [DOI] [PMC free article] [PubMed]
  • 41. Montefiori DC. Role of complement and Fc receptors in the pathogenesis of HIV‐1 infection. Springer Seminars Immunopathol. 1997;18(3):371‐390. [DOI] [PubMed] [Google Scholar]
  • 42. Harvala H, Robb M, Watkins N, et al. Convalescent plasma therapy for the treatment of patients with COVID‐19: assessment of methods available for antibody detection and their correlation with neutralising antibody levels. medRxiv. 2020. 10.1101/2020.05.20.20091694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Wang X, Guo X, Xin Q, et al. Neutralizing antibodies responses to SARS‐CoV‐2 in COVID‐19 inpatients and convalescent patients. 2020. 10.1101/2020.04.15.20065623. [DOI]
  • 44. Amanat F, White KM, Miorin L, et al. An in vitro microneutralization assay for SARS‐CoV‐2 serology and drug screening. Curr Protoc Microbiol. 2020;58(1):e108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Oark J‐G, Oladduni FS, Chiem K, et al. Rapid in vitro assays for screening neutralizing antibodies and antivirals against SARS‐CoV‐2. 2020. 10.1101/2020.07.22.216648. [DOI] [PMC free article] [PubMed]
  • 46. Okba NMA, Müller MA, Li W, et al. Severe acute respiratory syndrome coronavirus 2‐specific antibody responses in coronavirus disease patients. Emerg Infect Dis. 2020;26(7):1478‐1488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Oguntuyo KY, Stevens CS, Hung C‐T, et al. Quantifying absolute neutralization titers against SARS‐CoV‐2 by a standardized virus neutralization assay allows for cross‐cohort comparisons of COVID‐19 sera. 2020. 10.1101/2020.08.13.20157222. [DOI] [PMC free article] [PubMed]
  • 48. Salazar E, Kuchipudi SV, Christensen PA, et al. Relationship between anti‐spike protein antibody titers and SARS‐CoV‐2 in vitro virus neutralization in convalescent plasma. 2020. 10.1101/2020.06.08.138990. [DOI]
  • 49. Lee WT, Girardin RC, Dupuis AP, et al. Neutralizing antibody responses in COVID‐19 convalescent sera. medRxiv. 2020. 10.1101/2020.07.10.20150557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Rathe JA, Hemann EA, Eggenberger J, et al. SARS‐CoV‐2 serologic assays in control and unknown populations demonstrate the necessity of virus neutralization testing. 2020. 10.1101/2020.08.18.20177196. [DOI] [PMC free article] [PubMed]
  • 51. Luchsinger LL, Ransegnola B, Jin D, et al. Serological analysis of New York city COVID19 convalescent plasma donors. 2020. 10.1101/2020.06.08.20124792.medRxiv. [DOI] [Google Scholar]
  • 52. Stroemer A, Grobe O, Rose R, Fickenscher H, Lorentz T, Krumbholz A. Diagnostic accuracy of six commercial SARS‐CoV‐2 IgG/total antibody assays and identification of SARS‐CoV‐2 neutralizing antibodies in convalescent sera. medRxiv. 2020. 10.1101/2020.06.15.20131672. [DOI] [Google Scholar]
  • 53. Mueller L, Ostermann PN, Walker A, et al. Sensitivity of commercial anti‐SARS‐CoV‐2 sSerological assays in a high‐prevalence setting. medRxiv. 2020. 10.1101/2020.06.11.20128686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Rijkers G, Murk J-L, Wintermans B, et al. Differences in antibody kinetics and functionality between severe and mild SARS-CoV-2 infections. medRxiv. 2020. 10.1101/2020.06.11.20128686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Klein S, Pekosz A, Park H‐S, et al. Sex, age, and hospitalization drive antibody responses in a COVID‐19 convalescent plasma donor population. medRxiv. 2020. 10.1101/2020.06.26.20139063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Weidner L, Gänsdorfer S, Unterweger S, et al. Quantification of SARS‐CoV‐2 antibodies with eight commercially available immunoassays. J Clin virology official Publ Pan Am Soc Clin Virology. 2020;129:104540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Criscuolo E, Diotti RA, Strollo M, et al. Poor correlation between antibody titers and neutralizing activity in sera from SARS‐CoV‐2 infected subjects. 2020. 10.1101/2020.07.10.20150375. [DOI] [PMC free article] [PubMed]
  • 58. Suhandynata RT, Hoffman MA, Huang D, et al. Commercial serology assays predict neutralization activity against SARS‐CoV‐2. medRxiv; 2020. 10.1101/2020.07.10.20150946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Marklund E, Leach S, Axelsson H, et al. Serum‐IgG responses to SARS‐CoV‐2 after mild and severe COVID‐19 infection and analysis of IgG non‐responders. medRxiv. 2020. 10.1101/2020.07.11.20151324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Bonelli F, Sarasini A, Zierold C, et al. Clinical and analytical performance of an automated serological test that identifies S1/S2 neutralizing IgG in COVID‐19 patients semiquantitatively. J Clin Microbiol. 2020.58(9):e01224‐20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Jääskeläinen AJ, Kuivanen S, Kekäläinen E, et al. Performance of six SARS‐CoV‐2 immunoassays in comparison with microneutralisation. J Clin Virology Official Publ Pan Am Soc Clin Virol. 2020;129:104512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Meschi S, Colavita F, Bordi L, et al. Performance evaluation of Abbott ARCHITECT SARS‐CoV‐2 IgG immunoassay in comparison with indirect immunofluorescence and virus microneutralization test. J Clin Virology Official Publ Pan Am Soc Clin Virol. 2020;129:104539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Gniadek TJ, Thiede JM, Matchett WE, et al. SARS‐CoV‐2 neutralization and serology testing of COVID‐19 convalescent plasma from donors with non‐severe disease. 2020. 10.1101/2020.08.07.242271. [DOI] [PMC free article] [PubMed]
  • 64. Mazzini L, Martinuzzi D, Hyseni I, et al. Comparative analyses of SARS‐CoV‐2 binding (IgG, IgM, IgA) and neutralizing antibodies from human serum samples. 2020. 10.1101/2020.08.10.243717. [DOI] [PMC free article] [PubMed]
  • 65. Duan K, Liu B, Li C, et al. Effectiveness of convalescent plasma therapy in severe COVID‐19 patients. Proc Natl Acad Sci USA. 2020;117(17):9490‐9496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Shen C, Wang Z, Zhao F, et al. Treatment of 5 critically ill patients with COVID‐19 with convalescent plasma. J Am Med Assoc. 2020;323(16):1582‐1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Klingler J, Weiss S, Itri V, et al. Role of IgM and IgA antibodies to the neutralization of SARS‐CoV‐2. 2020. 10.1101/2020.08.18.20177303. [DOI]
  • 68. Horndler L, Delgado P, Balabanov I, et al. Flow cytometry multiplexed method for the detection of neutralizing human antibodies to the native SARS‐CoV‐2 spike protein 2020. 10.1101/2020.08.24.20180661. [DOI] [PMC free article] [PubMed]
  • 69. Johnson M, Wagstaffe H, Gilmour KC, et al. Evaluation of a novel multiplexed assay for determining IgG levels and functional activity to SARS‐CoV‐2. 2020. 10.1101/2020.07.20.213249. [DOI] [PMC free article] [PubMed]
  • 70. Byrnes JR, Zhou XX, Lui I, et al. A SARS‐CoV‐2 serological assay to determine the presence of blocking antibodies that compete for human ACE2 binding. 2020. 10.1101/05.27.20114652. [DOI]
  • 71. Bošnjak B, Stein SC, Willenzon S, et al. Novel surrogate virus neutralization test reveals low serum neutralizing anti‐SARS‐CoV‐2‐S antibodies levels in mildly affected COVID‐19 convalescents. 2020. 10.1101/2020.07.12.20151407. [DOI]
  • 72. Abe KT, Li Z, Samson R, et al. A simple protein‐based SARS‐CoV‐2 surrogate neutralization assay. 2020. 10.1101/2020.07.10.197913. [DOI] [PMC free article] [PubMed]
  • 73. Danh K, Karp DG, Robinson PV, et al. Detection of SARS-CoV-2 neutralizing antibodies with a cell-free PCR assay. 2020. 10.1101/2020.05.28.20105692. [DOI]
  • 74. Lei Q, Li Y, Hou H, et al. Antibody dynamics to SARS-CoV-2 in asymptomatic COVID-19 infections. 2020. 10.1101/2020.07.09.20149633. [DOI] [PMC free article] [PubMed]
  • 75. Aziz NA, Corman VM, Echterhoff AKC, et al. Seroprevalence and correlates of SARS‐CoV‐2 neutralizing antibodies: results from a population‐based study in Bonn Germany; 2020. 10.1101/2020.08.24.20181206. [DOI] [PMC free article] [PubMed]
  • 76. Chan KH, Cheng VC, Woo PC, et al. Serological responses in patients with severe acute respiratory syndrome coronavirus infection and cross‐reactivity with human coronaviruses 229E, OC43, and NL63. Clin Diagnostic Laboratory Immunol. 2005;12(11):1317‐1321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Liu W, Fontanet A, Zhang PH, et al. Two‐year prospective study of the humoral immune response of patients with severe acute respiratory syndrome. J Infect Dis. 2006;193(6):792‐795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Flehmig B, Schindler M, Ruetalo N, et al. Longitudinal analysis of virus load, serum antibody levels and virus neutralizing activity in vitro in cases with less severe COVID‐19. 2020. 10.1101/2020.08.20.20174912. [DOI]
  • 79. Duan J, Yan X, Guo X, et al. A human SARS‐CoV neutralizing antibody against epitope on S2 protein. Biochem Biophys Res Commun. 2005;333(1):186‐193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Zhong X, Yang H, Guo ZF, et al. B‐cell responses in patients who have recovered from severe acute respiratory syndrome target a dominant site in the S2 domain of the surface spike glycoprotein. J Virology. 2005;79(6):3401‐3408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Keng C‐T, Zhang A, Shen S, et al. Amino acids 1055 to 1192 in the S2 region of severe acute respiratory syndrome coronavirus S protein induce neutralizing antibodies: implications for the development of vaccines and antiviral agents. J Virology. 2005;79(6):3289‐3296. 10.1128/jvi.79.6.3289-3296.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Beaudoin‐Bussières G, Laumaea A, Anand SP, et al. Decline of humoral responses against SARS‐CoV‐2 spike in convalescent individuals. 2020. 10.1101/2020.07.09.194639. [DOI] [PMC free article] [PubMed]
  • 83. Seow J, Graham C, Merrick B, et al. Longitudinal evaluation and decline of antibody responses in SARS‐CoV‐2 infection medRxiv. 2020. 10.1101/2020.07.09.20148429. [DOI] [Google Scholar]
  • 84. Sterlin D, Mathian A, Miyara M, et al. IgA Dominates the Early Neutralizing Antibody Response to SARS‐CoV‐2.; 2020. 10.1101/2020.06.10.20126532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Ma H, Zhao D, Zeng W, et al. Decline of SARS‐CoV‐2‐specific IgG, IgM and IgA in convalescent COVID‐19 patients within 100 days after hospital discharge. Sci China Life Sci. 2020. 10.1007/s11427-020-1805-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Wajnberg A, Amanat F, Firpo A, et al. SARS‐CoV‐2 infection induces robust, neutralizing antibody responses that are stable for at least three months. 2020. 10.1101/2020.07.14.20151126. [DOI]
  • 87. Wang K, Long Q‐X, Deng H‐J, et al. Longitudinal dynamics of the neutralizing antibody response to SARS‐CoV‐2 infection. 2020. 10.1101/2020.07.14.20151159. [DOI] [PMC free article] [PubMed]
  • 88. Crawford KH, Dingens AS, Eguia R, et al. Dynamics of neutralizing antibody titers in the months after SARS‐CoV‐2 infection. 2020. 10.1101/2020.08.06.20169367. [DOI] [PMC free article] [PubMed]
  • 89. Perreault J, Tremblay T, Fournier M‐J, et al. Longitudinal analysis of the humoral response to SARS‐CoV‐2 spike RBD in convalescent plasma donors. 2020. 10.1101/2020.07.16.206847. [DOI]
  • 90. Gontu A, Srinivasan S, Salazar E, et al. Limited window for donation of convalescent plasma with high live‐virus neutralizing antibodies for COVID‐19 immunotherapy. 2020. 10.1101/2020.08.21.261909. [DOI] [PMC free article] [PubMed]

Associated Data

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

Data Availability Statement

This manuscript contains no original data.


Articles from Reviews in Medical Virology are provided here courtesy of Wiley

RESOURCES