Laboratory guidance for SARS-CoV-2 testing

CCDR

Volume 47-4: COVID-19: A Year Later

Diagnostic testing

Practical guidance for clinical laboratories for SARS-CoV-2 serology testing

Carmen Charlton1,2,3, Jamil Kanji1,2,4, Vanessa Tran5,6, Julianne Kus5,6, Jonathan Gubbay5,6, Carla Osiowy7, Jason Robinson8, Inna Sekirov9, Michael Drebot7, Todd Hatchette10, Derek Stein11,12, Nadia El-Gabalawy7, Amanda Lang13, Lei Jiao10, Paul Levett11, Heidi Wood7, Christian Therrien14, L Robbin Lindsay7, Muhammad Morshed9, Jessica Forbes6, Antonia Dibernardo7 on behalf of the Canadian Public Health Laboratory Network (CPHLN) Serology Working Group

Affiliations

1 Public Health Laboratory, Alberta Precision Laboratories, Edmonton, AB

2 Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB

3 Li Ka Shing Institute of Virology, Edmonton, AB

4 Division of Infectious Diseases, Department of Medicine, University of Alberta, Edmonton, AB

5 Public Health Ontario Laboratory, Toronto, ON

6 Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON

7 National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB

8 Provincial Laboratory Services, Division of Clinical Chemistry, Health PEI, Charlottetown, PE

9 British Columbia Centre for Disease Control Public Health Laboratory, Vancouver, BC

10 Newfoundland and Labrador Public Health Microbiology Laboratory, St. John’s, NL

11 Department of Medical Microbiology and Infectious Disease, University of Manitoba, Winnipeg, MB

12 Cadham Provincial Laboratory, Winnipeg, MB

13 Roy Romanow Provincial Laboratory, Regina, SK

14 Laboratoire de santé publique du Québec, Institut national de santé publique du Québec, Sainte-Anne-de-Bellevue, QC

Correspondence

carmen.charlton@albertaprecisionlabs.ca

Suggested citation

Charlton CL, Kanji JN, Tran V, Kus JV, Gubbay J, Osiowy C, Robinson JL, Sekirov I, Drebot M, Hatchette T, Stein DR, El-Gabalawy N, Lang A, Jiao L, Levett PN, Wood H, Therrien C, Lindsay LR, Morshed M, Forbes JD, Dibernardo A on behalf of the Canadian Public Health Laboratory Network (CPHLN) Serology Working Group. Practical guidance for clinical laboratories for SARS-CoV-2 serology testing. Can Commun Dis Rep 2021;47(4):171–83. https://doi.org/10.14745/ccdr.v47i04a01

Keywords: COVID-19, SARS-CoV-2, serological testing, serology algorithms

Abstract

The landscape of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) diagnostic testing is rapidly evolving. While serology testing has limited diagnostic capacity for acute infection, its role in providing population-based information on positivity rates and informing evidence-based decision making for public health recommendations is increasing. With the global availability of vaccines, there is increasing pressure on clinical laboratories to provide antibody screening and result interpretation for vaccinated and non-vaccinated individuals. Here we present the most up-to-date data on SARS-CoV-2 antibody timelines, including the longevity of antibodies, and the production and detection of neutralizing antibodies. Additionally, we provide practical guidance for clinical microbiology laboratories to both verify commercial serology assays and choose appropriate testing algorithms for their local populations.

Introduction

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in December 2019 resulted in a rapid global development of molecular, antigenic and serological assays for the diagnosis of acute infection and identification of past infection. While molecular testing is widely accepted as the gold standard for diagnosis of acute infection, the role of serology is limited to special clinical cases as an adjunct for diagnosisFootnote 1. The SARS-CoV-2 antibodies are not reliably detected fewer than 7–21 days post symptom onset, making their utility in diagnosis of acute infection of limited valueFootnote 2Footnote 3Footnote 4. However, in cases such as multiple inflammatory syndrome in adults (MIS-A) or in children (MIS-C), a positive result on a serological assay can help guide clinical management in the absence of a positive molecular test result or when molecular testing is unavailable, and positive serology results have been included as part of the clinical case definition for MIS-CFootnote 5. Outside these very specific clinical scenarios, the role of serology assays is to examine population-based prevalence rates of SARS-CoV-2, and to help inform public health decisions.

To improve the utility of serology testing, an international standard is required to allow direct comparison of assays between laboratories. Population-based studies linking quantitative serology results to clinical outcomes will be needed to help determine what level of antibody may correlate with immunity to infection. Such approaches would be similar to what has been done with other viruses (e.g. rubella), where an international standard is used to calibrate assays and a quantitative serological immunoglobulin G (IgG) is used to determine immunity (more than 10 IU/ml), or susceptibility (less than 10 IU/ml) to infection, based on correlation with clinical outcomesFootnote 6Footnote 7. Multiple quantitative serological assays have been submitted to accreditation agencies globally, including the Food and Drug Administration (FDA), Health Canada and European Council (Conseil européen); however, to date in North America, there remains a gap in approved quantitative assays that have a correlation with immunity from infection (i.e. neutralizing antibodies). While this will not impact clinical decision making on an individual level, this remains a critical gap in the interpretation and utility of SARS-CoV-2 serology testing for serosurveillance studies.

Here we examine the current knowledge of serological testing, discuss assay limitations, describe how clinical laboratories can both validate these assays and implement appropriate algorithms for local patient populations, discuss the role for differentiating antibodies derived from natural infection versus those that are vaccine-derived and consider options for detection of neutralizing antibodies.

Seroconversion timelines

The antibody response to SARS-CoV-2 is relatively well studied; however, there is substantial variability in seroconversion timelines given the heterogeneity amongst populations studied (i.e. disease severity, age, presence of comorbidities, etc.), serologic tests used and serologic markers analyzed. In general, the overall range of seroconversion regardless of the type of antibody, is estimated to be between four and 14 days post-onset of symptomsFootnote 2Footnote 8Footnote 9. An early study evaluated seroconversion rates in 173 patients and reported median seroconversion times for total antibody (Ab), immunoglobulin M (IgM), and IgG that were 11, 12 and 14 days, respectively. However, the authors reported that fewer than 40% of patients had detectable antibodies within one week of onset of illness and this rose to 100% (total Ab), 94% (IgM) and 80% (IgG) by day 15 in the same patientsFootnote 10. In contrast, another study reported the seroconversion rate of immunoglobulin A (IgA) was similar to IgM, with a median seroconversion of five days (IQR 3–6) after symptom onset compared with 14 days for IgGFootnote 11. Despite earlier detection of IgM in those studies, the time course for IgM and IgG seroconversion rates are similar for SARS-CoV-2 compared with other infectious diseases, where IgM preceded IgG by weeks. Indeed, both IgG and IgM detection signals were found to plateau six days after the first positive serology test among 285 coronavirus disease 2019 (COVID-19) patientsFootnote 9. Thus, while studies agree that seroconversion occurs within 4–14 days of symptom onset and that IgG and IgM seroconversion dynamics are similar, the considerable variability leads to poor sensitivity of antibody testing for diagnosis of acute COVID-19, which has been well documented in the literatureFootnote 1Footnote 2Footnote 12.

Antibody longevity

The length of time that antibody responses persists, and possibly confer protection from reinfection, is pivotal to understanding SARS-CoV-2 infection dynamics (detailed timeline for antibody detection can be seen inFootnote 9). Using sequential serum samples from 65 patients and 31 seropositive healthcare workers, Seow et al.Footnote 13 showed seroconversion of IgM, IgG and IgA occurred in more than 95% of cases when sampled equal to or greater than eight days post-symptom onset. Neutralizing antibody kinetics were consistent with other acute viral infectionsFootnote 13, with an initial peak at 3–4 weeks (magnitude of peak dependent on disease severity) followed by declining neutralizing antibody titres. Interestingly, patients with a high peak infectivity dose maintained neutralizing antibody titres longer compared with patients with a lower peak infectivity doseFootnote 13. Neutralizing antibody decline occurred simultaneously with declines in IgG titres against SARS-CoV-2 spike (S) glycoprotein and receptor-binding domain (RBD), as well as IgM and IgA binding to S glycoprotein and RBD. In patients with mild to moderate disease, IgG antibody titres were found to be stable for up to five months, with a significant correlation between anti-spike binding titres and neutralizationFootnote 14.

Additional studies using plaque reduction neutralization test (PRNT)/microneutralization assays in combination with anti-spike enzyme-linked immunosorbent assay (ELISA) procedures have further validated the findings of longitudinal neutralization antibody duration Footnote 15Footnote 16. Previously observed declines in antibody titres during the first few months after infection is expected as short-lived plasma cells are depleted; however, when these are replaced by long lived antibody secreting cells, neutralizing antibodies will persist for several months in most individualsFootnote 17.

Interestingly, longitudinal antibody longevity studies in serum and saliva showed that IgM and IgA levels rapidly declined following peak levels Footnote 19, while IgG remained relatively stable in both biofluids (up to 105 days post symptom onset). Similarly, others found that 92.3% of patients (N=427) remained IgG positive 3–6 months post symptom onsetFootnote 19. While most evidence suggests that IgM and IgA antibody levels drop significantly compared with IgG isotypes, in patients with a remote history of COVID-19, they appear to play a key role in the initial neutralizing antibody response. Serum IgA was shown to contribute to virus neutralization up to one month following symptom onsetFootnote 20, while IgM was shown to neutralize SARS-CoV-2 in both pseudoviral particle and wild type virus assaysFootnote 21.

Lessons on antibody longevity for SARS-CoV-2 may be learned by studying other human coronaviruses; a strategy that has been reviewed extensively elsewhereFootnote 22 A recent systematic review highlighted the kinetics, protection correlations and antibody association with disease severity among human coronaviruses. In general, antibody responses to other human coronaviruses, such as SARS-CoV, Middle Eastern respiratory syndrome coronavirus (MERS-CoV) and seasonal coronaviruses, are present for one year after infection; in some cases, antibodies may persist for longerFootnote 23. Moreover, other evidence suggests that serum antibody titres remained relatively high two years after SARS-CoV infection, and up to 55% of patients had detectable antibodies at three years post infectionFootnote 24. However, no detectable anti-SARS-CoV antibodies were observed in patients six years post infectionFootnote 25. Similarly, antibodies against MERS-CoV have been shown to persist for approximately three yearsFootnote 26, although the persistence of anti-MERS-CoV antibodies depended on disease severity, as patients with subclinical or mild disease had low or undetectable levels of antibodies two years after infectionFootnote 27. In contrast, seroprevalence studies of human coronavirus (HCoV)-229E and HCoV-OC43 suggested that antibody titres wane significantly one year after infectionFootnote 22. The persistence of anti-SARS-CoV-2 antibody response remains to be seen although given the association of the antibody signal and disease severityFootnote 13, it is likely that persistence of antibodies will correlate with disease severity.

Impact of disease severity and age

Given that many SARS-CoV-2 infections are subclinical or asymptomatic, it is critically import that the antibody response (including titres, seroconversion and time to seronegativity) in these patient populations be well understood. It is now widely recognized that titres of SARS-CoV-2 antibodies are positively associated with clinical severity of disease. Zhao et al.,Footnote 10 first reported that a significantly higher titre of total antibody was independently associated with a worse clinical classification (p=0.006) at 2-weeks post illness onset. A high total antibody titre was hypothesized to be a risk factor for critical illness and that it may even be used as a surrogate marker for worse clinical prognosis. A similar observation was made in 289 COVID-19 patients clinically categorized as having mild, moderate or severe infection. Patients with a severe infection had significantly higher levels of S1-specific IgA and IgG compared with those with a mild infectionFootnote 28. Moreover, S1-specific IgG was detectable after two weeks in only 20% of patients in the mild group compared with 100% of patients in the severe and moderate groups.

The impact of disease severity on seroconversion kinetics and their relationship to neutralizing properties in serum is not well understood. When compared to patients with mild symptoms, those with severe symptoms had a significantly faster time to IgG seroconversion (median 22 versus 11 days, respectively) with approximately 10% of patients with mild symptoms never seroconvertingFootnote 29. Detectable IgG levels were still observed more than 75 days post-symptom onset in patients who had seroconverted. Interestingly, even at 90 days post-symptom onset a small number (N=3) of patients developed total antibody levels below the limit of detection of commercial assays yet still had a detectable neutralizing response (titre range of 8–48). This observation is in direct contrast to that reported previously, where sera from 20% of discharged patients had no neutralizing properties despite sera from 100% of patients showing seroconversionFootnote 30. Furthermore, in a small study, the development of IgM in patients with severe disease was delayed (N=6; eight days) compared with mild disease (N=39; six days)Footnote 31. Jiang et al.Footnote 32 evaluated rates of seroconversion in non-severely ill patients with COVID-19, as well as asymptomatic patients, concluding that different IgM/IgG kinetics exist depending on the severity of the disease. Indeed, the authors reported that IgG seroconversion occurred among 94% of symptomatic and 85% of asymptomatic patients while IgM seroconversion occurred in 74% of symptomatic patients and only 31% of asymptomatic patients (p<0.001). Interestingly, the authors also reported that the median time to seroconversion (IgM or IgG) among the asymptomatic group was significantly shorter compared with the symptomatic group (median seven days from first positive polymerase chain reaction assay vs. 14 days; p<0.001).

While it is generally accepted that antibodies persist for longer periods in severe cases of COVID-19, there is considerable variation among studies even when normalized for the clinical severity. For example, IgG in mild to moderate disease appears to persist anywhere from 3–5 monthsFootnote 14Footnote 33. A study evaluating symptomatic and asymptomatic patients with COVID-19 associated pneumonia (apparent or subtle on radiography) found antibodies were more frequently detected among symptomatic patients (100% vs. 71%; measured at two and five months post symptom onset)Footnote 34. Furthermore, titres decreased significantly between the two time points. Another study comparing IgG and neutralizing antibody levels in asymptomatic versus symptomatic patients showed 40% of the asymptomatic patients became seronegative in the early convalescent phase (2–3 months post symptom onset) compared with only 12.9% of symptomatic patientsFootnote 35. Interestingly, Choe et al.Footnote 36 also reported that neutralizing antibody titre correlated with severity of disease, suggesting that patients with severe disease may be more protected from reinfection compared with patients with subclinical or asymptomatic infection. It should be noted that to date, there are only a few documented cases of reinfectionFootnote 37Footnote 38Footnote 39Footnote 40, which suggests that either other immune mechanisms (such as the T cell-mediated response) may contribute to protection against SARS-CoV-2 reinfection, or that, as a function of the short time that SARS-CoV-2 has been circulating and combined with implementation of public health restrictions, sufficient time has not yet passed for re-infection to be detected. Additionally, as most of these early studies examined the level of anti-nucleocapsid antibodies, association of high levels of antibody with poorer clinical outcome may be related to the production of anti-nucleocapsid rather than anti-spike antibodies.

Although there are less data on the relationship between age and the antibody response in COVID-19, recent evidence suggests there may be distinct antibody responses in children and adults. It has been shown that unlike adults, who produced robust levels of anti-S and anti-nucleocapsid (N) antibodies, children produced less anti-N and neutralizing antibodies Footnote 41. The clinical significance of this is unknown, although it is interesting considering disease severity is positively associated with age.

The large variability in how the antibody response was measured (i.e. different assays, platforms, methods, and antigenic targets) makes it difficult to make direct comparisons between studies. Despite this limitation, it is clear that not all infected individuals will mount an antibody response and that the level of antibodies may wane over time. This has significant implications for the interpretation of antibody testing for diagnostic purposes, especially for use as a surrogate marker of immunity to SARS-CoV-2. At best, anti-SARS-CoV-2 antibodies have good positive predictive agreement for neutralizing properties; however, the negative predictive agreement is poor.

How to implement testing in the clinical laboratory

Validation of antibody tests for infectious disease often depends on studies aimed to calculate the diagnostic sensitivity and specificity (i.e. correlating assay response to true positives and true negatives). In the case of SARS-CoV-2, this is inherently difficult for several reasons. Evidence suggests that humoral response and resultant seropositivity are affected by the severity of infection (presence/absence of detectable antibody), the time since symptom onset (antibody longevity) and the antigen target (antibody isotypes and viral protein recognition). Moreover, because the sensitivity of ribonucleic acid (RNA) testing is dependent on the time of sample collection in disease and the quality of sample, discordant serology results are often difficult to interpret for most clinical laboratories that do not have access to neutralization assays. Therefore, it is recommended that clinical laboratories offer assays that have undergone extensive review by a governing body such as Health Canada or the FDA, or by published peer-reviewed performance analyses when available. The requirement is on the clinical laboratory to ensure serology assays are used as intended and meet local accreditation standards.

The complexity and scope of internal verification studies required for laboratories to offer serology testing is highly dependent on the method of choice. Many clinical laboratories will choose to offer automated chemiluminescent immunoassay methods using existing instrumentation and infrastructure that are Health Canada or FDA emergency use authorization approved. When using these validated methodologies, with provided performance characteristics, a modified method verification may be acceptable with reduced rigour of testing to ensure the assay is fit for purpose. However, modifications to a validated method, such as using a different specimen type like a dried blood spot or altering the manufacturer’s cut-off, requires complete method validation prior to patient testing (considerations for verification and validation are shown in Table 1). In the case of a laboratory developed test, a complete method validation is required. A detailed flow diagram depicting additional possible scenarios for method validation and verification was published previouslyFootnote 42.

Table 1: Minimal requirements for validation and verification of qualitative assays
Item Verification Validation
Requirements Assay methodology and reagents must be unchanged from the manufacturer's instructions. Assay performance and methodology must be assessed and determined in the local testing population.
Purpose Laboratory verifies that the operators using their assay platform in their laboratory environment obtain the same performance characteristics with the assay method as described in the manufacturer's validation data. Laboratory validates all performance characteristics in their laboratory and all sample types to be used.
Sample number A statistically significant number of samples (generally 50 minimum) must be used in the evaluation process to cover the full range of expected results for the intended use A statistically significant number of samples (generally 50 positive and 50 negative run over 5–10 days) must be used, and cover the full range of expected results
Statistical analyses A statistical correlation with existing validated methods or comparisons with known outcomes ("gold standard") are required for qualitative methods. % CV, SD and 95% CI are recommended A statistical correlation with existing validated methods or comparisons with known outcomes ("gold standard") are required for qualitative methods. % CV, SD and 95% CI are recommended
Calculations Confirmation of Clinical Evaluation is the minimum to be tested
Sensitivity/specificity: Minimum of 20 samples (10 positive, 10 negative), or a recommended 100 samples: 50 samples valid for the method that are positive for SARS-CoV-2 RNA and 50 negative samples valid for the method that are negative for SARS-CoV-2 RNA OR that have been tested by a validated comparator immunoassay and were positive (N=50) or negative (N=50) for SARS-CoV-2 antibody. Apply the binary classification test ("Test outcome vs. condition") to determine both characteristics. Determine CI%.
Precision: is defined as the closeness of agreement between independent test results obtained under conditions of the assay run (includes repeatability and reproducibility). Minimum of 20 samples: 10 replicates each of one positive and one negative sample; 10 RNA positive and 10 RNA negative samples. Create aliquots and freeze all aliquots necessary for testing to avoid freeze-thaw variability between repeats. If comparing to another immunoassay, a range of S/CO values within the samples would be desirable.
Repeatability: Assay 10 replicates of the positive and negative samples in a single run. Determine the SD and % CV for the S/CO values.
Reproducibility: Assay the 20 specimen aliquots on three different days. Determine the SD and % CV for the S/CO values for each specimen.
All performance characteristics must be tested
Sensitivity/specificity: Recommended 100 samples: 50 samples valid for the method that are positive for SARS-CoV-2 RNA and 50 negative samples valid for the method that are negative for SARS-CoV-2 RNA OR that have been tested by a validated comparator immunoassay and were positive (N=50) or negative (N=50) for SARS-CoV-2 antibody. Apply the binary classification test ("Test outcome vs. condition") to determine both characteristics. Determine CI%.
Diagnostic (clinical) sensitivity is defined as the percentage of individuals with the target condition (as determined by the diagnostic accuracy criteria) whose test values are positive.
Diagnostic (clinical) specificity is defined by the percentage of individuals without the target condition (as determined by the diagnostic accuracy criteria) whose test values are negative.
Target specificity (cross-reactivity) ensures the test is specific only for the analyte of interest. This is determined by testing other pathogens within the same family or disease group.
Positive predictive value is defined as the percentage of individuals with a positive test result who have the target condition (as determined by the diagnostic accuracy criteria). Consider evaluating parameters as a function of the population prevalence.
Negative predictive value is defined as the percentage of subjects with a negative test result who do not have the target condition (as determined by the diagnostic accuracy criteria). Consider evaluating parameters as a function of the population prevalence.
Precision: Create aliquots and freeze all aliquots necessary for testing to avoid freeze-thaw variability between repeats. If comparing to another immunoassay, a range of S/CO values within the samples would be desirable.
Repeatability: Assay 10 replicates of the positive and negative samples in a single run. Determine the SD and % CV for the S/CO values.
Reproducibility: Assay the 20 specimen aliquots on 3 different days. Determine the SD and % CV for the S/CO values for each specimen.
Normal values: 120 specimens should be run to establish normal range of values for local testing population.
Other considerations When possible, laboratories should consider using multiple operators to perform verification, particularly when result interpretation is required.
If the assay documentation does not include a full validation report or incomplete performance characteristics as recognized by technical organizations, then a user laboratory validation is required.
When possible, laboratories should consider using multiple operators to perform validation, particularly when result interpretation is required.

Method verifications are likely to be the most common form of method evaluation performed by clinical laboratories at present. At minimum, clinical laboratories should verify manufacturer claims by assessing the diagnostic sensitivity and specificity, assay precision (reproducibility), cross-reactivity of non-SARS-CoV-2 antibodies and interfering substances (such as hemoglobin, lipids or biotin) commonly found in their patient population. A summary of the minimum suggested sample sizes for establishing diagnostic sensitivity and specificity are provided in Table 2. The clinical laboratory must verify the assay performance using statistical correlations and comparisons based on manufacturer claims. Careful scrutiny is required in the case of discrepant results, and arbitrator testing can be performed by external laboratories performing the same methodology. If verification specimens are sourced locally, patient history may be considered to reconcile test performance and to further characterize the testing population (outpatient, hospitalized or intensive care unit, and timing of sample collections). Furthermore, it may be prudent to interrogate the signal obtained from the assay in expected positives that may be in the equivocal range because some patient antibody titres may have waned over time, and assay cut-offs were assigned with only limited samples. A particular challenge during verification is resolving suspected false positive serologic results given the variable disease prevalenceFootnote 43. Potential approaches to resolve discrepancies include testing for another assay or target antigen, reviewing the clinical history of the patient case, including time since symptom onset and contact tracing, or reviewing prior SARS-CoV-2 RNA testing results.

Table 2: Theoretical number of samples required for establishing diagnostic sensitivity and specificity estimates by error margin and confidence interval
Estimated sensitivity or specificity Estimate with 2% error Estimate with 5% error
Confidence Confidence
90% 95% 99% 90% 95% 99%
90% 610 864 1,493 98 138 239
92% 466 707 1,221 75 113 195
94% 382 542 935 61 87 150
95% 372 456 788 60 73 126
96% 260 369 637 42 59 102
97% 197 279 483 32 45 77
98% 133 188 325 21 30 52
99% 67 95 164 11 15 26

Consideration of orthogonal testing algorithms for severe acute respiratory syndrome coronavirus 2 serology

In general, laboratories should strive to use serological assays with manufacturer-claimed sensitivity of greater than 95% and specificity of greater than 99.5% Footnote 1. Many of the SARS-CoV-2 specific antibody assays currently approved for use in North America meet these performance criteria when used aloneFootnote 44Footnote 45Footnote 46Footnote 47Footnote 48. However, when testing is performed in low prevalence populations or in patients with a low pre-test probability of disease the positive predictive value or post-test odds will be unacceptably low despite excellent specificity. Therefore, laboratory professionals may adopt an orthogonal testing strategy to improve the specificity and positive predictive values of serologic test interpretation for SARS-CoV-2Footnote 49Footnote 50Footnote 51. In orthogonal testing, samples that test positive or equivocal on an initial test are re-tested using a second test to confirm or refute the resultFootnote 1. Orthogonal testing strategies have been recommended by public health authorities in North America in low prevalence populationsFootnote 1Footnote 52, and detailed protocols are availableFootnote 53. Most of the orthogonal testing approaches for SARS-CoV-2 antibodies involve two independent tests, each with unique assay design characteristics such as antigen typeFootnote 54Footnote 55Footnote 56 or assay formatsFootnote 57. However, these approaches may have practical limitations in terms of implementation because the specific epitope targeted in each manufacturers assay is unknown or not providedFootnote 58. Improvements in sensitivity can also be accomplished in orthogonal testing approaches by reducing cut-off values for commercial high-throughput automated SARS-CoV-2 assaysFootnote 59; however, alteration of manufacturer recommendations requires full validation of the new cut-off values prior to clinical use.

When users are considering either a single serological testing algorithm or a two-step (orthogonal) testing algorithm, they must consider the reason for testing, the intended use of the data generated and the expected prevalence of SARS-CoV-2 in the population of interest. For example, use of serology for special clinical case testing (e.g. in MIS-C cases) or in seroepidemiological studies, requires high assay sensitivity and high negative predictive value in early infection (more than two weeks) and late convalescence, respectively. Indeed, in these scenarios reporting true cases should be the priority; although false positives are not desirable, they can be tolerated more so than false negatives. For example, in seroepidemiological studies, a sensitive assay with a good positive predictive value is critical to identify true cases to provide robust population level estimates of seropositivity. In contrast, when screening potential convalescent plasma donors, the presence of SARS-CoV-2 antibody is criticalFootnote 60, and a positive predictive value more than 99% should be required. Because high negative predictive value is less important in this scenario, orthogonal testing is recommended in both low and high population prevalence levelsFootnote 56. These examples illustrate the need for careful consideration of orthogonal testing strategies that are tailored to the intended use of the serological data. As a result of these different scenarios, SARS-CoV-2 antibody testing strategies will vary based on site-specific requirements.

Serological assays to determine severe acute respiratory syndrome coronavirus 2 antibody neutralization potency

To better understand and characterize SARS-CoV-2 immunity after natural infection or vaccination, functional assays such as virus neutralizing tests are required. The previous/current gold standard methodologies to detect and quantify SARS-CoV-2 neutralizing antibodies have used cell-culture-based infection assays, which block viral entry into cells in vitro. These live-virus assays use wild type SARS-CoV-2 virus incubated with dilutions of a patient’s sera or plasma. The mixture is then added to susceptible cells to determine if the sera inhibits or neutralizes the cytopathological effect or plaque reduction is observed. PRNT provide a means to quantitate neutralization titres associated with an individual’s clinical specimen. However, live virus assays require biosafety level-3 containment, are labour-intensive and due to the biologic variation associated with these assays they can be difficult to standardizeFootnote 61Footnote 62Footnote 63.

Neutralization assays that use pseudotyped viruses, such as the vesicular stomatitis virus or lentivirus-based systems that incorporate SARS-CoV-2 spike protein, can be used in biosafety level-2 laboratoriesFootnote 64. An example of a basic procedure for establishing a pseudotype assay using lentivirus particles involves transfecting a “packaging” cell line, such as HEK 293T cells, with a number of plasmids to produce safe, non-replicative viral particles expressing the spike protein. Transfected plasmids include a reporter-expressing plasmid, a plasmid encoding the SARS-CoV-2 spike and a number of plasmids encoding lentiviral proteins required for assembling viral particles. The transfected cells produce the pseudotype viruses, which can then be used to infect permissive cells expressing the SARS-CoV-2 receptor, angiotensin-converting enzyme 2 (ACE2), to measure a decrease in cytopathological effect, via the reporter signal, or by plaque reduction in the presence of patient serum.

Although pseudotype virus formats alleviate biosafety limitations, they have similar drawbacks to conventional PRNTs in that they are difficult to standardize across laboratories, and the assay characteristics may vary depending on culture conditions, virus strains and cell lines used. Furthermore, these cell-based assays require highly skilled personnel, are low throughput and have suboptimal turnaround times for clinical decision making. These drawbacks make implementation of pseudotype viral assays in the clinical laboratory impractical.

Recently, several ELISA-based surrogate neutralization assays that detect antibodies targeting the viral spike protein RBD have come to marketFootnote 65Footnote 66Footnote 67. These more rapid assays are based on antibody-associated blockage of the interaction between the spike RBD and the ACE2 receptor. The procedure by Abe et al.,Footnote 67 uses immobilized ACE2 and soluble biotinylated RBD (which exhibited increased sensitivity to other reagent configurations), and provides a direct comparison with conventional ELISAs (detecting antibodies that bind RBD) in a plate format.

A number of commercial assays using a similar ELISA-based platform have also become available (e.g. GenScript cPass, Cayman SARS-CoV-2 antibody ELISA) that indirectly and semi-quantitatively measure the neutralizing capability of SARS-CoV-2 antibodies. The GenScript surrogate virus neutralization test (sVNT) kit has been compared with conventional cell-based neutralization assaysFootnote 68Footnote 69Footnote 70 and displayed good specificity, and comparable sensitivity to virus culture-based assays, but demonstrated somewhat reduced efficacy in identifying samples with a lower level of virus neutralization potency. This observation may be explained by the specific detection of antibodies targeting the RBD in the assay, as non-RBD neutralizing antibodies are not targeted in the sVNT assays. It should also be noted that unlike functional PRNT or cell-based assays, surrogates may detect non-neutralizing antibodies in some samples. However, the advantage of both commercial and non-commercial ELISA-based sVNT assays is the ease of use and the potential for automation and standardization. Moreover, with appropriate validations, these may be incorporated into conventional virus neutralizing testing algorithms and offer an important tool to assess neutralization in clinical specimens.

Additional standardization is underway to compare cell-based neutralization assays with surrogate tests utilizing pseudotype viruses or ELISA-based competitive binding assaysFootnote 71. Good correlation was observed between a modified ELISA-based surrogate assay with a conventional PRNT and spike pseudotyped viral vector-based platformsFootnote 67. Antibody titres between TCID50 neutralization tests and lentiviral/vesicular stomatitis virus pseudotype assays correlated wellFootnote 71. It should be noted that although cell-based neutralization assays, such as PRNTs or micro-neutralization tests, are the reference standards for detection and quantification of neutralizing antibodies in clinical specimens, the biological nature of these test may lead to some variability in titres and sensitivity when comparing inter-laboratory resultsFootnote 72. The utilization of proficiency panels made up of well pedigreed control and patient sera/plasma samples facilitates standardization between laboratories.

Commercially available, high-throughput serological assays that measure the binding of antibodies to various viral antigens have been directly compared to neutralization antibody titres in patient samples using PRNT or sVNT assaysFootnote 73Footnote 74Footnote 75. The results from these comparisons show that commercial serology assays are sensitive for the detection of total antibodies but are less robust at predicting the neutralization titre relative to conventional (e.g. PRNT) or sVNT assays. Furthermore, the reliability of commercial high-throughput platforms that are specific for spike/RBD protein antigens to infer neutralization titre equivalents may depend upon time frames for specimen collection and the specific antibody induction responses of an individual. However, more recent studies have described a strong correlation between anti-RBD antibody concentrations and spike/RBD-ACE2 inhibiting antibody titres (r>0.86, p<0.001)Footnote 76Footnote 77, with the possibility of anti-nucleocapsid antibodies (signal to cut-off greater than or equal to 5.0) serving as a surrogate for screening of high neutralizing antibody titre plasma (greater than or equal to 160)Footnote 78.

Detecting antibodies derived from vaccination versus natural infection

With the roll-out of SARS-CoV-2 vaccines, the detection and quantification of vaccine-induced antibody by current commercial assays needs to be verified in relation to the detection of natural immunity following infection. A difference in the ability to detect antibodies from both natural infection and vaccination has been seen with other viral infections, including varicella zoster virus (VZV), where commercial assays can readily detect IgG antibodies in people who have had a natural infection but many commercial assays are less robust in detecting vaccine-induced antibodiesFootnote 79. Multiple studies are currently ongoing to determine what SARS-CoV-2 antibodies are detected with commercial assays, and if there are variances in detection based on the type of vaccine received by an individual. The current lack of serum from vaccinated individuals makes it challenging for a clinical laboratory to incorporate appropriate serology algorithms, particularly as both vaccine and laboratory assay targets are proprietary. Furthermore, with new vaccine candidates emerging, the variability in vaccine type and antigen may limit the ability of current commercial anti-SARS-CoV-2 serology tests to determine whether a patient has been vaccinated. Moreover, there is no evidence that the presence and titre of anti-SARS-CoV-2 antibodies can be extrapolated to immune status of vaccinated patients.

The role for differentiation of antibodies derived from natural infection versus vaccine-induced antibodies does not have importance for clinical management, and there are currently no Health Canada approved anti-SARS-CoV-2 assays specific for vaccinated populations. In the first four months following vaccination, it is likely that natural versus vaccine-induced antibodies can be differentiated using a combination of anti-nucleocapsid and anti-spike assays. In Canada, for example, where only spike-based vaccines are currently being used, a positive anti-nucleocapsid could indicate natural infection, and a negative anti-nucleocapsid but positive anti-spike could indicate vaccine-induced antibodies. However, this process is not straightforward as negative anti-nucleocapsid but positive anti-spike antibody profiles have been identified in those following natural infection (anti-spike antibodies persist longer than anti-nucleocapsid Footnote 50Footnote 80). The landscape becomes more complicated after four months or longer because anti-nucleocapsid antibodies are expected to decline among patients who have recovered following natural infection. Therefore, current assays cannot determine whether a patient had positive anti-spike antibodies and negative anti-nucleocapsid antibodies due to a history of infection or a vaccination. From a public health perspective, it is important to understand the overall immunity of a population, and whether immunity is derived from vaccine or natural infection is academic. In jurisdictions using only spike-based vaccines, the use of spike or receptor binding domain assays should be considered as the primary targets for screening to ensure both scenarios are detected in surveillance studies. Conversely, the presence of vaccination-induced antibodies may be problematic for clinical testing; for example, in the diagnosis of MIS-C. As such, clinical serology testing is not recommended for individuals who have received the COVID-19 vaccine, especially if an assay that detects anti-N antibody is not available. Better markers of immune status are needed that are simple, reproducible and robust.

The use of serology in determination of immunity is challenging. There are currently no commercially available assays to determine immune status, which can only be evaluated using PRNT assays. In the absence of a universal standard, or a surrogate marker for immunity, the detection of antibodies in serological assays, regardless of the signal strength, cannot determine with confidence whether an individual would be susceptible or immune to a subsequent challenge with SARS-CoV-2. Therefore, the value of an “immunity passport”, where individuals with detectable antibody have fewer public health restrictions, is limited and may harm public health efforts aiming to decrease the spread of infection.

Towards the end of 2020, a series of SARS-CoV-2 variants of concern have emerged within different geographic regions of the world, such as the United Kingdom, South Africa and BrazilFootnote 81. These variants include specific mutations within the spike protein (K417N, E484K, N501Y) that are shared among all independent variant lineagesFootnote 82. The most pressing concern with these variants is the increased transmissibilityFootnote 83 associated with them; but also troubling is the aspect of immune escape, with the potential to evade detection and thus diagnosis. These mutations have been shown to affect neutralization properties depending on whether monoclonal or polyclonal antibodies were testedFootnote 84Footnote 85Footnote 86. Furthermore, the P.1 variant first found circulating in Manaus, Brazil, was associated with re-infection in immunocompetent individualsFootnote 87 which presumably would make it more difficult to interpret results from serology and PRNT assays targeting wild-type virus or viral antigens.

Currently there is no recommendation for pre or post-vaccine immunity screeningFootnote 88, as there is a lack of correlation between antibody detection and immunity to infection, and little work has been done to understand antibody production in immunocompromised populations. To understand what level of antibody could correlate with immunity from infection, more in-depth studies are needed, including creation of an international standard to allow comparison of antibody levels between different vaccines, assays and laboratories.

Conclusion

Implementation of SARS-CoV-2 serology in the clinical laboratory is challenging, and laboratory professionals must be aware of the limitations of these assays. There are a number of unknown factors that affect these assays, and guidelines and recommendations for their use in clinical laboratories are ever evolving. Here we present the most up-to-date testing recommendations in Canada, and provide practical guidance for laboratories to choose appropriate serological assays and employ the best testing algorithms for their local populations.

Authors’ statement

  • CLC — Conceptualized project, original draft, review of manuscript, final editing of manuscript
  • JNK, VT, JVK, JG, CO, JLR, MD, TH, DRS, AL, LJ, PNL, HW, CT, LRL, MM, JDF, AD — Conceptualized project, original draft, review of manuscript
  • NE — Review of manuscript, coordination of drafts

All authors have approved the final version sent for publication and are accountable for all aspects of the work.

Competing interests

None.

Acknowledgements

Dr. J Kanji is now with the Division of Infectious Diseases, Department of Medicine, University of Calgary, Calgary, Alberta.

Funding

None.

References

Footnote 1

Van Caeseele P, Bailey D, Forgie SE, Dingle TC, Krajden M; Canadian Public Health Laboratory Network; Canadian Society of Clinical Chemists; Association of Medical Microbiology and Infectious Disease Canada; Canadian Association for Clinical Microbiology and Infectious Diseases; COVID-19 Immunity Task Force. SARS-CoV-2 (COVID-19) serology: implications for clinical practice, laboratory medicine and public health. CMAJ 2020;192(34):E973-9. https://doi.org/10.1503/cmaj.201588

Return to footnote 1 referrer

Footnote 2

Charlton CL, Kanji JN, Johal K, Bailey A, Plitt SS, MacDonald C, Kunst A, Buss E, Burnes LE, Fonseca K, Berenger BM, Schnabl K, Hu J, Stokes W, Zelyas N, Tipples G. Evaluation of Six Commercial Mid- to High-Volume Antibody and Six Point-of-Care Lateral Flow Assays for Detection of SARS-CoV-2 Antibodies. J Clin Microbiol 2020;58(10):e01361-20. https://doi.org/10.1128/JCM.01361-20

Return to footnote 2 referrer

Footnote 3

Grzelak L, Temmam S, Planchais C, Demeret C, Tondeur L, Huon C, Guivel-Benhassine F, Staropoli I, Chazal M, Dufloo J, Planas D, Buchrieser J, Rajah MM, Robinot R, Porrot F, Albert M, Chen KY, Crescenzo-Chaigne B, Donati F, Anna F, Souque P, Gransagne M, Bellalou J, Nowakowski M, Backovic M, Bouadma L, Le Fevre L, Le Hingrat Q, Descamps D, Pourbaix A, Laouénan C, Ghosn J, Yazdanpanah Y, Besombes C, Jolly N, Pellerin-Fernandes S, Cheny O, Ungeheuer MN, Mellon G, Morel P, Rolland S, Rey FA, Behillil S, Enouf V, Lemaitre A, Créach MA, Petres S, Escriou N, Charneau P, Fontanet A, Hoen B, Bruel T, Eloit M, Mouquet H, Schwartz O, van der Werf S. A comparison of four serological assays for detecting anti-SARS-CoV-2 antibodies in human serum samples from different populations. Sci Transl Med 2020;12(559):eabc3103. https://doi.org/10.1126/scitranslmed.abc3103

Return to footnote 3 referrer

Footnote 4

Lee CY, Lin RT, Renia L, Ng LF. Serological Approaches for COVID-19: Epidemiologic Perspective on Surveillance and Control. Front Immunol 2020;11:879. https://doi.org/10.3389/fimmu.2020.00879

Return to footnote 4 referrer

Footnote 5

Centers for Disease Control and Prevention. Information for Healthcare Providers about Multisystem Inflammatory Syndrome in Children (MIS-C). Atlanta (GA): CDC; 2021. https://www.cdc.gov/mis-c/hcp/index.html

Return to footnote 5 referrer

Footnote 6

World Health Organization. Manual for the Laboratory-based Surveillance of Measles, Rubella, and Congenital Rubella Syndrome. Third Edition Edition. WHO: June 2018. https://www.who.int/immunization/monitoring_surveillance/burden/laboratory/manual/en/

Return to footnote 6 referrer

Footnote 7

Public Health Agency of Canada. Elimination of Measles, Rubella and Congenital Rubella Syndrome in Canada: Documentation and Verification Report. PHAC; 2011. https://www.canada.ca/en/public-health/services/immunization/vaccine-preventable-diseases/elimination-measles-rubella-congenital-rubella-syndrome-canada-documentation-verification-report.html

Return to footnote 7 referrer

Footnote 8

Grzelak L, Temmans S, Planchais S, Demeret C, Huon C, Guivel-Benhassine F, Staropoli I, Chazal M, Dufloo J, Planas D, Buchrieser J, Rajah MM, Robinot R, Porrot F, Albert M, Chen K-Y, Crescenzo B, Donati F, Anna F, Souque F, Gransagne M, Bellalou J, Nowakowski M, Backovic M, Bouadma L, Le Fevre L, Le Hingrat Q, Descamps D, Pourbaix A, Yazdanpanah Y, Tondeur L, Besombes C, Ungeheuer M-N, Mellon G, Morel P, Rolland S, Rey F, Behillil S, Enouf V, Lemaitre A, Créach M-A, Petres S, Escriou N, Charneau P, Fontanet A, Hoen B, Bruel T, Eloit M, Mouquet H, Schwartz O, van der Werf S. SARS-CoV-2 serological analysis of COVID-19 hospitalized patients, pauci-symptomatic individuals and blood donors. medRxiv. 2020.04.21.20068858. https://doi.org/10.1101/2020.04.21.20068858

Return to footnote 8 referrer

Footnote 9

Long QX, Liu BZ, Deng HJ, Wu GC, Deng K, Chen YK, Liao P, Qiu JF, Lin Y, Cai XF, Wang DQ, Hu Y, Ren JH, Tang N, Xu YY, Yu LH, Mo Z, Gong F, Zhang XL, Tian WG, Hu L, Zhang XX, Xiang JL, Du HX, Liu HW, Lang CH, Luo XH, Wu SB, Cui XP, Zhou Z, Zhu MM, Wang J, Xue CJ, Li XF, Wang L, Li ZJ, Wang K, Niu CC, Yang QJ, Tang XJ, Zhang Y, Liu XM, Li JJ, Zhang DC, Zhang F, Liu P, Yuan J, Li Q, Hu JL, Chen J, Huang AL. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat Med 2020;26(6):845-8. https://doi.org/10.1038/s41591-020-0897-1

Return to footnote 9 referrer

Footnote 10

Zhao J, Yuan Q, Wang H, Liu W, Liao X, Su Y, Wang X, Yuan J, Li T, Li J, Qian S, Hong C, Wang F, Liu Y, Wang Z, He Q, Li Z, He B, Zhang T, Fu Y, Ge S, Liu L, Zhang J, Xia N, Zhang Z. Antibody Responses to SARS-CoV-2 in Patients With Novel Coronavirus Disease 2019. Clin Infect Dis 2020;71(16):2027-34. https://doi.org/10.1093/cid/ciaa344

Return to footnote 10 referrer

Footnote 11

Guo L, Ren L, Yang S, Xiao M, Chang D, Yang F, Dela Cruz CS, Wang Y, Wu C, Xiao Y, Zhang L, Han L, Dang S, Xu Y, Yang QW, Xu SY, Zhu HD, Xu YC, Jin Q, Sharma L, Wang L, Wang J. Profiling Early Humoral Response to Diagnose Novel Coronavirus Disease (COVID-19). Clin Infect Dis 2020;71(15):778-85. https://doi.org/10.1093/cid/ciaa310

Return to footnote 11 referrer

Footnote 12

Suhandynata RT, Hoffman MA, Kelner MJ, McLawhon RW, Reed SL, Fitzgerald RL. Longitudinal Monitoring of SARS-CoV-2 IgM and IgG Seropositivity to Detect COVID-19. J Appl Lab Med 2020;5(5):908-20. https://doi.org/10.1093/jalm/jfaa079

Return to footnote 12 referrer

Footnote 13

Seow J, Graham C, Merrick B, Acors S, Pickering S, Steel KJ, Hemmings O, O'Byrne A, Kouphou N, Galao RP, Betancor G, Wilson HD, Signell AW, Winstone H, Kerridge C, Huettner I, Jimenez-Guardeño JM, Lista MJ, Temperton N, Snell LB, Bisnauthsing K, Moore A, Green A, Martinez L, Stokes B, Honey J, Izquierdo-Barras A, Arbane G, Patel A, Tan MK, O'Connell L, O'Hara G, MacMahon E, Douthwaite S, Nebbia G, Batra R, Martinez-Nunez R, Shankar-Hari M, Edgeworth JD, Neil SJ, Malim MH, Doores KJ. Longitudinal observation and decline of neutralizing antibody responses in the three months following SARS-CoV-2 infection in humans. Nat Microbiol 2020;5(12):1598-607. https://doi.org/10.1038/s41564-020-00813-8

Return to footnote 13 referrer

Footnote 14

Wajnberg A, Amanat F, Firpo A, Altman DR, Bailey MJ, Mansour M, McMahon M, Meade P, Mendu DR, Muellers K, Stadlbauer D, Stone K, Strohmeier S, Simon V, Aberg J, Reich DL, Krammer F, Cordon-Cardo C. Robust neutralizing antibodies to SARS-CoV-2 infection persist for months. Science 2020;370(6521):1227-30. https://doi.org/10.1126/science.abd7728

Return to footnote 14 referrer

Footnote 15

Ripperger TJ, Uhrlaub JL, Watanabe M, Wong R, Castaneda Y, Pizzato HA, Thompson MR, Bradshaw C, Weinkauf CC, Bime C, Erickson HL, Knox K, Bixby B, Parthasarathy S, Chaudhary S, Natt B, Cristan E, El Aini T, Rischard F, Campion J, Chopra M, Insel M, Sam A, Knepler JL, Capaldi AP, Spier CM, Dake MD, Edwards T, Kaplan ME, Scott SJ, Hypes C, Mosier J, Harris DT, LaFleur BJ, Sprissler R, Nikolich-Žugich J, Bhattacharya D. Orthogonal SARS-CoV-2 Serological Assays Enable Surveillance of Low-Prevalence Communities and Reveal Durable Humoral Immunity. Immunity 2020;53(5):925-933.e4. https://doi.org/10.1016/j.immuni.2020.10.004

Return to footnote 15 referrer

Footnote 16

Lau EH, Tsang OT, Hui DS, Kwan MY, Chan WH, Chiu SS, Ko RL, Chan KH, Cheng SM, Perera RA, Cowling BJ, Poon LL, Peiris M. Neutralizing antibody titres in SARS-CoV-2 infections. Nat Commun 2021;12(1):63. https://doi.org/10.1038/s41467-020-20247-4

Return to footnote 16 referrer

Footnote 17

Hartley GE, Edwards ES, Aui PM, Varese N, Stojanovic S, McMahon J, Peleg AY, Boo I, Drummer HE, Hogarth PM, O'Hehir RE, van Zelm MC. Rapid generation of durable B cell memory to SARS-CoV-2 spike and nucleocapsid proteins in COVID-19 and convalescence. Sci Immunol 2020;5(54):eabf8891. https://doi.org/10.1126/sciimmunol.abf8891

Return to footnote 17 referrer

Footnote 18

Isho B, Abe KT, Zuo M, Jamal AJ, Rathod B, Wang JH, Li Z, Chao G, Rojas OL, Bang YM, Pu A, Christie-Holmes N, Gervais C, Ceccarelli D, Samavarchi-Tehrani P, Guvenc F, Budylowski P, Li A, Paterson A, Yue FY, Marin LM, Caldwell L, Wrana JL, Colwill K, Sicheri F, Mubareka S, Gray-Owen SD, Drews SJ, Siqueira WL, Barrios-Rodiles M, Ostrowski M, Rini JM, Durocher Y, McGeer AJ, Gommerman JL, Gingras AC. Persistence of serum and saliva antibody responses to SARS-CoV-2 spike antigens in COVID-19 patients. Sci Immunol 2020;5(52):eabe5511.

Return to footnote 18 referrer

Footnote 19

Maine GN, Lao KM, Krishnan SM, Afolayan-Oloye O, Fatemi S, Kumar S, VanHorn L, Hurand A, Sykes E, Sun Q. Longitudinal characterization of the IgM and IgG humoral response in symptomatic COVID-19 patients using the Abbott Architect. J Clin Virol 2020;133:104663. https://doi.org/10.1016/j.jcv.2020.104663

Return to footnote 19 referrer

Footnote 20

Sterlin D, Mathian A, Miyara M, Mohr A, Anna F, Claër L, Quentric P, Fadlallah J, Devilliers H, Ghillani P, Gunn C, Hockett R, Mudumba S, Guihot A, Luyt CE, Mayaux J, Beurton A, Fourati S, Bruel T, Schwartz O, Lacorte JM, Yssel H, Parizot C, Dorgham K, Charneau P, Amoura Z, Gorochov G. IgA dominates the early neutralizing antibody response to SARS-CoV-2. Sci Transl Med 2021;13(577):eabd2223. https://doi.org/10.1126/scitranslmed.abd2223

Return to footnote 20 referrer

Footnote 21

Gasser R, Cloutier M, Prévost J, Fink C, Ducas É, Ding S, Dussault N, Landry P, Tremblay T, Laforce-Lavoie A, Lewin A, Beaudoin-Bussières G, Laumaea A, Medjahed H, Larochelle C, Richard J, Dekaban GA, Dikeakos JD, Bazin R, Finzi A. Major role of IgM in the neutralizing activity of convalescent plasma against SARS-CoV-2. Cell Rep 2021;34(9):108790. https://doi.org/10.1016/j.celrep.2021.108790

Return to footnote 21 referrer

Footnote 22

Sariol A, Perlman S. Lessons for COVID-19 Immunity from Other Coronavirus Infections. Immunity 2020;53(2):248-63. https://doi.org/10.1016/j.immuni.2020.07.005

Return to footnote 22 referrer

Footnote 23

Huang AT, Garcia-Carreras B, Hitchings MD, Yang B, Katzelnick LC, Rattigan SM, Borgert BA, Moreno CA, Solomon BD, Trimmer-Smith L, Etienne V, Rodriguez-Barraquer I, Lessler J, Salje H, Burke DS, Wesolowski A, Cummings DA. A systematic review of antibody mediated immunity to coronaviruses: kinetics, correlates of protection, and association with severity. Nat Commun 2020;11(1):4704. https://doi.org/10.1038/s41467-020-18450-4

Return to footnote 23 referrer

Footnote 24

Wu LP, Wang NC, Chang YH, Tian XY, Na DY, Zhang LY, Zheng L, Lan T, Wang LF, Liang GD. Duration of antibody responses after severe acute respiratory syndrome. Emerg Infect Dis 2007;13(10):1562-4. https://doi.org/10.3201/eid1310.070576

Return to footnote 24 referrer

Footnote 25

Tang F, Quan Y, Xin ZT, Wrammert J, Ma MJ, Lv H, Wang TB, Yang H, Richardus JH, Liu W, Cao WC. Lack of peripheral memory B cell responses in recovered patients with severe acute respiratory syndrome: a six-year follow-up study. J Immunol 2011;186(12):7264-8. https://doi.org/10.4049/jimmunol.0903490

Return to footnote 25 referrer

Footnote 26

Payne DC, Iblan I, Rha B, Alqasrawi S, Haddadin A, Al Nsour M, Alsanouri T, Ali SS, Harcourt J, Miao C, Tamin A, Gerber SI, Haynes LM, Al Abdallat MM. Persistence of Antibodies against Middle East Respiratory Syndrome Coronavirus. Emerg Infect Dis 2016;22(10):1824-6. https://doi.org/10.3201/eid2210.160706

Return to footnote 26 referrer

Footnote 27

Zhao J, Alshukairi AN, Baharoon SA, Ahmed WA, Bokhari AA, Nehdi AM, Layqah LA, Alghamdi MG, Al Gethamy MM, Dada AM, Khalid I, Boujelal M, Al Johani SM, Vogel L, Subbarao K, Mangalam A, Wu C, Ten Eyck P, Perlman S, Zhao J. Recovery from the Middle East respiratory syndrome is associated with antibody and T-cell responses. Sci Immunol 2017;2(14):eaan5393. https://doi.org/10.1126/sciimmunol.aan5393

Return to footnote 27 referrer

Footnote 28

Kowitdamrong E, Puthanakit T, Jantarabenjakul W, Prompetchara E, Suchartlikitwong P, Putcharoen O, Hirankarn N, Putcharoen O, Hirankarn N. Antibody responses to SARS-CoV-2 in patients with differing severities of coronavirus disease 2019. PLoS One 2020;15(10):e0240502. https://doi.org/10.1371/journal.pone.0240502

Return to footnote 28 referrer

Footnote 29

Marklund E, Leach S, Axelsson H, Nyström K, Norder H, Bemark M, Angeletti D, Lundgren A, Nilsson S, Andersson LM, Yilmaz A, Lindh M, Liljeqvist JÅ, Gisslén M. Serum-IgG responses to SARS-CoV-2 after mild and severe COVID-19 infection and analysis of IgG non-responders. PLoS One 2020;15(10):e0241104. https://doi.org/10.1371/journal.pone.0241104

Return to footnote 29 referrer

Footnote 30

Kalkan Yazıcı M, Koç MM, Çetin NS, Karaaslan E, Okay G, Durdu B, Sümbül B, Doymaz MZ. Discordance between Serum Neutralizing Antibody Titers and the Recovery from COVID-19. J Immunol 2020;205(10):2719-25. https://doi.org/10.4049/jimmunol.2000840

Return to footnote 30 referrer

Footnote 31

Shen L, Wang C, Zhao J, Tang X, Shen Y, Lu M, Ding Z, Huang C, Zhang J, Li S, Lan J, Wong G, Zhu Y. Delayed specific IgM antibody responses observed among COVID-19 patients with severe progression. Emerg Microbes Infect 2020;9(1):1096-101. https://doi.org/10.1080/22221751.2020.1766382

Return to footnote 31 referrer

Footnote 32

Jiang C, Wang Y, Hu M, Wen L, Wen C, Wang Y, Zhu W, Tai S, Jiang Z, Xiao K, Faria NR, De Clercq E, Xu J, Li G. Antibody seroconversion in asymptomatic and symptomatic patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Clin Transl Immunology 2020;9(9):e1182. https://doi.org/10.1002/cti2.1182

Return to footnote 32 referrer

Footnote 33

Ibarrondo FJ, Fulcher JA, Goodman-Meza D, Elliott J, Hofmann C, Hausner MA, Ferbas KG, Tobin NH, Aldrovandi GM, Yang OO. Rapid Decay of Anti-SARS-CoV-2 Antibodies in Persons with Mild Covid-19. N Engl J Med 2020;383(11):1085-7. https://doi.org/10.1056/NEJMc2025179

Return to footnote 33 referrer

Footnote 34

Choe PG, Kang CK, Suh HJ, Jung J, Song KH, Bang JH, Kim ES, Kim HB, Park SW, Kim NJ, Park WB, Oh MD. Waning Antibody Responses in Asymptomatic and Symptomatic SARS-CoV-2 Infection. Emerg Infect Dis 2021;27(1):327-9. https://doi.org/10.3201/eid2701.203515

Return to footnote 34 referrer

Footnote 35

Long QX, Tang XJ, Shi QL, Li Q, Deng HJ, Yuan J, Hu JL, Xu W, Zhang Y, Lv FJ, Su K, Zhang F, Gong J, Wu B, Liu XM, Li JJ, Qiu JF, Chen J, Huang AL. Clinical and immunological assessment of asymptomatic SARS-CoV-2 infections. Nat Med 2020;26(8):1200-4. https://doi.org/10.1038/s41591-020-0965-6

Return to footnote 35 referrer

Footnote 36

Choe PG, Kang CK, Suh HJ, Jung J, Kang E, Lee SY, Song KH, Kim HB, Kim NJ, Park WB, Kim ES, Oh MD. Antibody Responses to SARS-CoV-2 at 8 Weeks Postinfection in Asymptomatic Patients. Emerg Infect Dis 2020;26(10):2484-7. https://doi.org/10.3201/eid2610.202211

Return to footnote 36 referrer

Footnote 37

Tillett RL, Sevinsky JR, Hartley PD, Kerwin H, Crawford N, Gorzalski A, Laverdure C, Verma SC, Rossetto CC, Jackson D, Farrell MJ, Van Hooser S, Pandori M. Genomic evidence for reinfection with SARS-CoV-2: a case study. Lancet Infect Dis 2021;21(1):52-8. https://doi.org/10.1016/S1473-3099(20)30764-7

Return to footnote 37 referrer

Footnote 38

To KK, Hung IF, Ip JD, Chu AW, Chan WM, Tam AR, Fong CH, Yuan S, Tsoi HW, Ng AC, Lee LL, Wan P, Tso E, To WK, Tsang D, Chan KH, Huang JD, Kok KH, Cheng VC, Yuen KY. COVID-19 re-infection by a phylogenetically distinct SARS-coronavirus-2 strain confirmed by whole genome sequencing. Clin Infect Dis 2020 Aug;ciaa1275. (Online head of print). https://doi.org/10.1093/cid/ciaa1275

Return to footnote 38 referrer

Footnote 39

Van Elslande J, Vermeersch P, Vandervoort K, Wawina-Bokalanga T, Vanmechelen B, Wollants E, Laenen L, André E, Van Ranst M, Lagrou K, Maes P. Symptomatic SARS-CoV-2 reinfection by a phylogenetically distinct strain. Clin Infect Dis 2020;ciaa1330. (Online head of print). https://doi.org/10.1093/cid/ciaa1330

Return to footnote 39 referrer

Footnote 40

Prado-Vivar B, Becerra-Wong M, Guadalupe JJ, Márquez S, Gutierrez B, Rojas-Silva P, Grunauer M, Treuba G, Barragan V, Cardenas P. A case of SARS-CoV-2 reinfection in Ecuador. Lancet Infect Dis. 2020. (Online head of print). https://doi.org/10.1016/s1473-3099(20)30910-5

Return to footnote 40 referrer

Footnote 41

Weisberg SP, Connors TJ, Zhu Y, Baldwin MR, Lin WH, Wontakal S, Szabo PA, Wells SB, Dogra P, Gray J, Idzikowski E, Stelitano D, Bovier FT, Davis-Porada J, Matsumoto R, Poon MM, Chait M, Mathieu C, Horvat B, Decimo D, Hudson KE, Zotti FD, Bitan ZC, La Carpia F, Ferrara SA, Mace E, Milner J, Moscona A, Hod E, Porotto M, Farber DL. Distinct antibody responses to SARS-CoV-2 in children and adults across the COVID-19 clinical spectrum. Nat Immunol 2021;22(1):25-31. https://doi.org/10.1038/s41590-020-00826-9

Return to footnote 41 referrer

Footnote 42

National Association of Testing Authorities. Australia (NATA). General Accreditation Guidance - Validation and verification of quantitative and qualitative test methods. Australia: NATA; 2018. https://www.nata.com.au/phocadownload/gen-accreditation-guidance/Validation-and-Verification-of-Quantitative-and-Qualitative-Test-Methods.pdf

Return to footnote 42 referrer

Footnote 43

Farnsworth CW, Anderson NW. SARS-CoV-2 Serology: Much Hype, Little Data. Clin Chem 2020;66(7):875-7. https://doi.org/10.1093/clinchem/hvaa107

Return to footnote 43 referrer

Footnote 44

Kohmer N, Westhaus S, Rühl C, Ciesek S, Rabenau HF. Brief clinical evaluation of six high-throughput SARS-CoV-2 IgG antibody assays. J Clin Virol 2020;129:104480. https://doi.org/10.1016/j.jcv.2020.104480

Return to footnote 44 referrer

Footnote 45

Manthei DM, Whalen JF, Schroeder LF, Sinay AM, Li SH, Valdez R, Giacherio DA, Gherasim C. Differences in Performance Characteristics Among Four High-Throughput Assays for the Detection of Antibodies Against SARS-CoV-2 Using a Common Set of Patient Samples. Am J Clin Pathol 2021;155(2):267-79. https://doi.org/10.1093/ajcp/aqaa200

Return to footnote 45 referrer

Footnote 46

Merrill AE, Jackson JB, Ehlers A, Voss D, Krasowski MD. Head-to-Head Comparison of Two SARS-CoV-2 Serology Assays. J Appl Lab Med 2020;5(6):1351-7. https://doi.org/10.1093/jalm/jfaa125

Return to footnote 46 referrer

Footnote 47

Perkmann T, Perkmann-Nagele N, Breyer MK, Breyer-Kohansal R, Burghuber OC, Hartl S, Aletaha D, Sieghart D, Quehenberger P, Marculescu R, Mucher P, Strassl R, Wagner OF, Binder CJ, Haslacher H. Side-by-Side Comparison of Three Fully Automated SARS-CoV-2 Antibody Assays with a Focus on Specificity. Clin Chem 2020;66(11):1405-13. https://doi.org/10.1093/clinchem/hvaa198

Return to footnote 47 referrer

Footnote 48

Prince HE, Givens TS, Lapé-Nixon M, Clarke NJ, Schwab DA, Batterman HJ, Jones RS, Meyer WA 3rd, Kapoor H, Rowland CM, Haji-Sheikhi F, Marlowe EM. Detection of SARS-CoV-2 IgG Targeting Nucleocapsid or Spike Protein by Four High-Throughput Immunoassays Authorized for Emergency Use. J Clin Microbiol 2020;58(11):e01742-20. https://doi.org/10.1128/JCM.01742-20

Return to footnote 48 referrer

Footnote 49

Pflüger LS, Bannasch JH, Brehm TT, Pfefferle S, Hoffmann A, Nörz D, van der Meirschen M, Kluge S, Haddad M, Pischke S, Hiller J, Addo MM, Lohse AW, Schulze Zur Wiesch J, Peine S, Aepfelbacher M, Lütgehetmann M. Clinical evaluation of five different automated SARS-CoV-2 serology assays in a cohort of hospitalized COVID-19 patients. J Clin Virol 2020;130:104549. https://doi.org/10.1016/j.jcv.2020.104549

Return to footnote 49 referrer

Footnote 50

Ripperger TJ, Uhrlaub JL, Watanabe M, Wong R, Castaneda Y, Pizzato HA, Thompson MR, Bradshaw C, Weinkauf CC, Bime C, Erickson HL, Knox K, Bixby B, Parthasarathy S, Chaudhary S, Natt B, Cristan E, T El Aini, Rischard F, Campion J, Chopra M, Insel M, Sam A, Knepler JL, Capaldi AP, Spier CM, Dake MD, Edwards T, Kaplan ME, Scott SJ, Hypes C, Mosier J, Harris DT, LaFleur BJ, Sprissler R, Nikolich-Žugic J, Bhattacharya D. Detection, prevalence, and duration of humoral responses to SARS-CoV-2 under conditions of limited population exposure. medRxiv. 2020. (Online ahead of print). https://doi.org/10.1101/2020.08.14.20174490

Return to footnote 50 referrer

Footnote 51

Xu G, Emanuel AJ, Nadig S, Mehrotra S, Caddell BA, Curry SR, Nolte FS, Babic N. Evaluation of Orthogonal Testing Algorithm for Detection of SARS-CoV-2 IgG Antibodies. Clin Chem. 2020;66(12)1531–7. https://doi.org/10.1093/clinchem/hvaa210

Return to footnote 51 referrer

Footnote 52

Centers for Disease Control and Prevention. Interim Guidelines for COVID-19 Antibody Testing. Atlanta (GA); CDC; updated March 2021. https://www.cdc.gov/coronavirus/2019-ncov/lab/resources/antibody-tests-guidelines.html

Return to footnote 52 referrer

Footnote 53

Stadlbauer D, Amanat F, Chromikova V, Jiang K, Strohmeier S, Arunkumar GA, Tan J, Bhavsar D, Capuano C, Kirkpatrick E, Meade P, Brito RN, Teo C, McMahon M, Simon V, Krammer F. SARS-CoV-2 Seroconversion in Humans: A Detailed Protocol for a Serological Assay, Antigen Production, and Test Setup. Curr Protoc Microbiol 2020;57(1):e100. https://doi.org/10.1002/cpmc.100

Return to footnote 53 referrer

Footnote 54

Coste AT, Jaton K, Papadimitriou-Olivgeris M, Greub G, Croxatto A. Comparison of SARS-CoV-2 serological tests with different antigen targets. J Clin Virol 2021;134:104690. https://doi.org/10.1016/j.jcv.2020.104690

Return to footnote 54 referrer

Footnote 55

DomBourian MG. Annen K, Huey L, Andersen G, Merkel PA, Jung S, Dominguez SR, Knight V. Analysis of COVID-19 convalescent plasma for SARS-CoV-2 IgG using two commercial immunoassays. J Immunol Methods 2020;486:112837. https://doi.org/10.1016/j.jim.2020.112837

Return to footnote 55 referrer

Footnote 56

Turbett SE, Anahtar M, Dighe AS, Garcia Beltran W, Miller T, Scott H, Durbin SM, Bharadwaj M, Thomas J, Gogakos TS, Astudillo M, Lennerz J, Rosenberg ES, Branda JA. Evaluation of Three Commercial SARS-CoV-2 Serologic Assays and Their Performance in Two-Test Algorithms. J Clin Microbiol 2020;59(1):e01892–20. https://doi.org/10.1128/JCM.01892-20

Return to footnote 56 referrer

Footnote 57

Hoste AC, Venteo A, Fresco-Taboada A, Tapia I, Monedero A, López L, Jebbink MF, Pérez-Ramírez E, Jimenez-Clavero MA, Almonacid M, Muñoz P, Guinea J, Vela C, van der Hoek L, Rueda P, Sastre P. Two serological approaches for detection of antibodies to SARS-CoV-2 in different scenarios: a screening tool and a point-of-care test. Diagn Microbiol Infect Dis 2020;98(4):115167. https://doi.org/10.1016/j.diagmicrobio.2020.115167

Return to footnote 57 referrer

Footnote 58

Pallett SJ, Jones R, Pallett MA, Rayment M, Mughal N, Davies GW, Moore LS. Characterising differential antibody response is integral to future SARS-CoV-2 serostudies. J Infect 2020;81(6):e28–30. https://doi.org/10.1016/j.jinf.2020.07.029

Return to footnote 58 referrer

Footnote 59

Perkmann T, Perkmann-Nagele N, Oszvar-Kozma M, Koller T, Breyer M-K, Breyer-Kohansal R, Burghuber OC, Harti S, Aletaha D, Sieghart D, Quehenberger P, Marculescu R, Mucher P, Radakovics A, Strassl R, Leitner G, Wagner OF, Binder CJ, Haslacher H. Increasing both specificity and sensitivity of SARS-CoV-2 antibody tests by using an adaptive orthogonal testing approach. medRxiv. 2020.11.05.20226449. (Online ahead of print). https://doi.org/10.1101/2020.11.05.20226449

Return to footnote 59 referrer

Footnote 60

Ng DL, Goldgof GM, Shy BR, Levine AG, Balcerek J, Bapat SP, Prostko J, Rodgers M, Coller K, Pearce S, Franz S, Du L, Stone M, Pillai SK, Sotomayor-Gonzalez A, Servellita V, Martin CS, Granados A, Glasner DR, Han LM, Truong K, Akagi N, Nguyen DN, Neumann NM, Qazi D, Hsu E, Gu W, Santos YA, Custer B, Green V, Williamson P, Hills NK, Lu CM, Whitman JD, Stramer SL, Wang C, Reyes K, Hakim JM, Sujishi K, Alazzeh F, Pham L, Thornborrow E, Oon CY, Miller S, Kurtz T, Simmons G, Hackett J Jr, Busch MP, Chiu CY. SARS-CoV-2 seroprevalence and neutralizing activity in donor and patient blood. Nat Commun 2020;11(1):4698. https://doi.org/10.1038/s41467-020-18468-8

Return to footnote 60 referrer

Footnote 61

Kaufer AM, Theis T, Lau KA, Gray JL, Rawlinson WD. Laboratory biosafety measures involving SARS-CoV-2 and the classification as a Risk Group 3 biological agent. Pathology 2020;52(7):790–5. https://doi.org/10.1016/j.pathol.2020.09.006

Return to footnote 61 referrer

Footnote 62

Lee WT, Girardin RC, Dupuis AP, Kulas KE, Payne AF, Wong SJ, Arinsburg S, Nguyen FT, Mendu DR, Firpo-Betancourt A, Jhang J, Wajnberg A, Krammer F, Cordon-Cardo C, Amler S, Montecalvo M, Hutton B, Taylor J, McDonough KA. Neutralizing Antibody Responses in COVID-19 Convalescent Sera. J Infect Dis 2021;223(1):47–55. https://doi.org/10.1093/infdis/jiaa673

Return to footnote 62 referrer

Footnote 63

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. https://doi.org/10.1002/cpmc.105

Return to footnote 63 referrer

Footnote 64

Crawford KH, Eguia R, Dingens AS, Loes AN, Malone KD, Wolf CR, Chu HY, Tortorici MA, Veesler D, Murphy M, Pettie D, King NP, Balazs AB, Bloom JD. Protocol and Reagents for Pseudotyping Lentiviral Particles with SARS-CoV-2 Spike Protein for Neutralization Assays. Viruses 2020;12(5):513. https://doi.org/10.3390/v12050513

Return to footnote 64 referrer

Footnote 65

Tan CW, Chia WN, Qin X, Liu P, Chen MI, Tiu C, Hu Z, Chen VC, Young BE, Sia WR, Tan YJ, Foo R, Yi Y, Lye DC, Anderson DE, Wang LF. A SARS-CoV-2 surrogate virus neutralization test based on antibody-mediated blockage of ACE2-spike protein-protein interaction. Nat Biotechnol 2020;38(9):1073–8. https://doi.org/10.1038/s41587-020-0631-z

Return to footnote 65 referrer

Footnote 66

Muruato AE, Fontes-Garfias CR, Ren P, Garcia-Blanco MA, Menachery VD, Xie X, Shi PY. A high-throughput neutralizing antibody assay for COVID-19 diagnosis and vaccine evaluation. Nat Commun 2020;11(1):4059. https://doi.org/10.1038/s41467-020-17892-0

Return to footnote 66 referrer

Footnote 67

Abe KT, Li Z, Samson R, Samavarchi-Tehrani P, Valcourt EJ, Wood H, Budylowski P, Dupuis AP 2nd, Girardin RC, Rathod B, Wang JH, Barrios-Rodiles M, Colwill K, McGeer AJ, Mubareka S, Gommerman JL, Durocher Y, Ostrowski M, McDonough KA, Drebot MA, Drews SJ, Rini JM, Gingras AC. A simple protein-based surrogate neutralization assay for SARS-CoV-2. JCI Insight 2020;5(19):e142362. https://doi.org/10.1172/jci.insight.142362

Return to footnote 67 referrer

Footnote 68

Meyer B, Reimerink J, Torriani G, Brouwer F, Godeke GJ, Yerly S, Hoogerwerf M, Vuilleumier N, Kaiser L, Eckerle I, Reusken C. Validation and clinical evaluation of a SARS-CoV-2 surrogate virus neutralisation test (sVNT). Emerg Microbes Infect 2020;9(1):2394–403. https://doi.org/10.1080/22221751.2020.1835448

Return to footnote 68 referrer

Footnote 69

Valcourt EJ, Manguiat K, Robinson A, Chen JC, Dimitrova K, Philipson C, Lamoureux L, McLachlan E, Schiffman Z, Drebot MA, Wood H. Evaluation of a commercially-available surrogate virus neutralization test for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Diagn Microbiol Infect Dis 2021;99(4):115294. https://doi.org/10.1016/j.diagmicrobio.2020.115294

Return to footnote 69 referrer

Footnote 70

Taylor SC, Hurst B, Charlton CL, Bailey A, Kanji JN, McCarthy MK, Morrison TE, Huey L, Annen K, DomBourian MG, Knight V. A New SARS CoV-2 Dual Purpose Serology Test: Highly Accurate Infection Tracing and Neutralizing Antibody Response Detection. J Clin Microbiol. 2021;JCM.02438-20. https://doi.org/10.1128/JCM.02438-20

Return to footnote 70 referrer

Footnote 71

Riepler L, Rössler A, Falch A, Volland A, Borena W, von Laer D, Kimpel J. Comparison of Four SARS-CoV-2 Neutralization Assays. Vaccines (Basel) 2020;9(1):13. https://doi.org/10.3390/vaccines9010013

Return to footnote 71 referrer

Footnote 72

Perera RA, Mok CK, Tsang OT, Lv H, Ko RL, Wu NC, Yuan M, Leung WS, Chan JM, Chik TS, Choi CY, Leung K, Chan KH, Chan KC, Li KC, Wu JT, Wilson IA, Monto AS, Poon LL, Peiris M. Serological assays for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), March 2020. Euro Surveill 2020;25(16):2000421. https://doi.org/10.2807/1560-7917.ES.2020.25.16.2000421

Return to footnote 72 referrer

Footnote 73

Bonelli F, Sarasini A, Zierold C, Calleri M, Bonetti A, Vismara C, Blocki FA, Pallavicini L, Chinali A, Campisi D, Percivalle E, DiNapoli AP, Perno CF, Baldanti F. 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. https://doi.org/10.1128/JCM.01224-20

Return to footnote 73 referrer

Footnote 74

Valdivia A, Torres I, Latorre V, Francés-Gómez C, Albert E, Gozalbo-Rovira R, Alcaraz MJ, Buesa J, Rodríguez-Díaz J, Geller R, Navarro D. Inference of SARS-CoV-2 spike-binding neutralizing antibody titers in sera from hospitalized COVID-19 patients by using commercial enzyme and chemiluminescent immunoassays. Eur J Clin Microbiol Infect Dis 2021;40(3):485–94. https://doi.org/10.1007/s10096-020-04128-8

Return to footnote 74 referrer

Footnote 75

Patel EU, Bloch EM, Clarke W, Hsieh YH, Boon D, Eby Y, Fernandez RE, Baker OR, Keruly M, Kirby CS, Klock E, Littlefield K, Miller J, Schmidt HA, Sullivan P, Piwowar-Manning E, Shrestha R, Redd AD, Rothman RE, Sullivan D, Shoham S, Casadevall A, Quinn TC, Pekosz A, Tobian AA, Laeyendecker O. Comparative Performance of Five Commercially Available Serologic Assays To Detect Antibodies to SARS-CoV-2 and Identify Individuals with High Neutralizing Titers. J Clin Microbiol 2021;59(2):e02257–20. https://doi.org/10.1128/JCM.02257-20

Return to footnote 75 referrer

Footnote 76

L’Huillier AG, Meyer B, Andrey DO, Arm-Vernez I, Baggio S, Didierlaurent A, Eberhardt CS, Eckerle I, Grasset-Salomon C, Huttner A, Posfay-Barbe KM, Royo IS, Pralong JA, Vuilleumier N, Yerly S, Siegrist CA, Kaiser L. Geneva Centre for Emerging Viral Diseases. Antibody persistence in the first 6 months following SARS-CoV-2 infection among hospital workers: a prospective longitudinal study. Clin Microbiol Infect. 2021;S1198-743X(21)00031-8. https://doi.org/10.1016/j.cmi.2021.01.005

Return to footnote 76 referrer

Footnote 77

Iyer AS, Jones FK, Nodoushani A, Kelly M, Becker M, Slater D, Mills R, Teng E, Kamruzzaman M, Garcia-Beltran WF, Astudillo M, Yang D, Miller TE, Oliver E, Fischinger S, Atyeo C, Iafrate AJ, Calderwood SB, Lauer SA, Yu J, Li Z, Feldman J, Hauser BM, Caradonna TM, Branda JA, Turbett SE, LaRocque RC, Mellon G, Barouch DH, Schmidt AG, Azman AS, Alter G, Ryan ET, Harris JB, Charles RC. Persistence and decay of human antibody responses to the receptor binding domain of SARS-CoV-2 spike protein in COVID-19 patients. Sci Immunol 2020;5(52):eabe0367. https://doi.org/10.1126/sciimmunol.abe0367

Return to footnote 77 referrer

Footnote 78

Wendel S, Fontão-Wendel R, Fachini R, Candelaria G, Scuracchio P, Achkar R, Brito M, Reis LF, Camargo A, Amano M, Machado R, Araujo D, Soares C, Durigon E. A longitudinal study of convalescent plasma (CCP) donors and correlation of ABO group, initial neutralizing antibodies (nAb), and body mass index (BMI) with nAb and anti-nucleocapsid (NP) SARS-CoV-2 antibody kinetics: proposals for better quality of CCP collections. Transfusion 2021. (Online head of print). https://doi.org/10.1111/trf.16323

Return to footnote 78 referrer

Footnote 79

National Advisory Committee on Immunization (NACI). Varicella (chickenpox) vaccine: Canadian Immunization Guide. Ottawa (ON): NACI; 2018. https://www.canada.ca/en/public-health/services/publications/healthy-living/canadian-immunization-guide-part-4-active-vaccines/page-24-varicella-chickenpox-vaccine.html

Return to footnote 79 referrer

Footnote 80

Fenwick C, Croxatto A, Coste AT, Pojer F, André C, Pellaton C, Farina A, Campos J, Hacker D, Lau K, Bosch BJ, Gonseth Nussle S, Bochud M, D’Acremont V, Trono D, Greub G, Pantaleo G. Changes in SARS-CoV-2 Spike versus Nucleoprotein Antibody Responses Impact the Estimates of Infections in Population-Based Seroprevalence Studies. J Virol 2021;95(3):e01828–20. https://doi.org/10.1128/JVI.01828-20

Return to footnote 80 referrer

Footnote 81

Kupferschmidt K. New coronavirus variants could cause more reinfections, require updated vaccines. Science 2021. https://www.sciencemag.org/news/2021/01/new-coronavirus-variants-could-cause-more-reinfections-require-updated-vaccines

Return to footnote 81 referrer

Footnote 82

European Centre for Disease Prevention and Control. Risk Assessment: Risk related to spread of new SARS-CoV-2 variants of concern in the EU/EEA. ECDC; 2020. https://www.ecdc.europa.eu/en/publications-data/covid-19-risk-assessment-spread-new-sars-cov-2-variants-eueea

Return to footnote 82 referrer

Footnote 83

World Health Organization. SARS-CoV-2 Variants. Geneva (Switzerland): WHO; 2020. https://www.who.int/csr/don/31-december-2020-sars-cov2-variants/en/

Return to footnote 83 referrer

Footnote 84

Greaney AJ, Loes AN, Crawford KH, Starr TN, Malone KD, Chu HY, Bloom JD. Comprehensive mapping of mutations in the SARS-CoV-2 receptor-binding domain that affect recognition by polyclonal human plasma antibodies. Cell Host Microbe 2021;29(3):463–476.e6. https://doi.org/10.1016/j.chom.2021.02.003

Return to footnote 84 referrer

Footnote 85

Wibmer CK, Ayres F, Hermanus T, Madzivhandila M, Kgagudi P, Lambson BE, de Oliveira T, Vermeulen M, van der Berg K, Rossouw T, Boswell M, Ueckermann V, Meiring S, von Gottberg A, Cohen C, Morris L, Bhiman JN, Moore PL. SARS-CoV-2 501Y.V2 escapes neutralization by South African COVID-19 donor plasma. bioRxiv. 2021.01.18.427166. (Online head of print). https://doi.org/10.1101/2021.01.18.427166

Return to footnote 85 referrer

Footnote 86

Wang Z, Schmidt F, Weisblum Y, Muecksch F, Barnes CO, Finkin S, Schaefer-Babajew D, Cipolla M, Gaebler C, Lieberman JA, Oliveira TY, Yang Z, Abernathy ME, Huey-Tubman KE, Hurley A, Turroja M, West KA, Gordon K, Millard KG, Ramos V, Da Silva J, Xu J, Colbert RA, Patel R, Dizon J, Unson-O'Brien C, Shimeliovich I, Gazumyan A, Caskey M, Bjorkman PJ, Casellas R, Hatziioannou T, Bieniasz PD, Nussenzweig MC. mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants. Nature. 2021;592:616–22. (Online ahead of print). https://doi.org/10.1038/s41586-021-03324-6

Return to footnote 86 referrer

Footnote 87

Naveca F, da Costa C, Nascimento V, Souza V, Corado A, Nascimento F, Costa A, Duarte D, Silva G, Mejia M, Pessoa K, Goncalves L, Brandao MJ, Jesus M, Pinto R, Silva M, Mattros T, Abdalla L, Santos JH, Costa-Filho R, Wallau GL, Siqueira MM, Delatorre E, Graf T, Bello G, Resende PC. SARS-CoV-2 reinfection by the new Variant of Concern (VOC) P.1 in Amazonas, Brazil. Virological. Jan 17, 2021. https://virological.org/t/sars-cov-2-reinfection-by-the-new-variant-of-concern-voc-p-1-in-amazonas-brazil/596

Return to footnote 87 referrer

Footnote 88

National Advisory Committee on Immunization (NACI). Recommendations on the use of COVID-19 vaccines. Ottawa (ON): NACI; 2021. https://www.canada.ca/en/public-health/services/immunization/national-advisory-committee-on-immunization-naci/recommendations-use-covid-19-vaccines.html

Return to footnote 88 referrer

Page details

Date modified: