key: cord-1021586-5jmplsgi authors: Taylor, Sean C. title: A Practical Approach to SARS-CoV-2 Testing in a Pre and Post-Vaccination Era date: 2021-10-08 journal: Journal of clinical virology plus DOI: 10.1016/j.jcvp.2021.100044 sha: 6d9f3eb79c668e1c0b8b6811367bcf43ec827df8 doc_id: 1021586 cord_uid: 5jmplsgi As countries globally are in the process of planning, introducing or implementing mass vaccination strategies while continuing to deal with the ongoing SARS-CoV-2 pandemic, an evolution in testing strategies may be required to minimize spread in mixed vaccinated and non-vaccinated populations. This mini-review explores the key public health questions associated with the widely varying efficacy of commercially available vaccines and their persistence of protection in the context of a growing number of variant virus strains. A new strategy for SARS-CoV-2 testing that accommodates the current and evolving pandemic paradigm is proposed. Covid-19 has led to the rapid innovation and commercialization of a myriad of tests for early detection of SARS-CoV-2 [1] [2] [3] [4] . Unfortunately, there has been little vetting of their efficacy and accuracy with real world samples in rigorous clinical trials due to the urgent population testing requirements to minimize transmission and spread [5] [6] [7] [8] . Thus, the government and private clinical diagnostics sectors have been obliged to filter through the flood of available commercial assays and create in house strategies to vet, choose and adopt individual approaches to SARS-CoV-2 population testing without global standards [8] . In the rush to contain the spread of infection through testing, mistakes have been made with the adoption of rapid blood and/or saliva tests with such poor accuracy that they were discarded costing some governments millions of dollars [9] [10] [11] . There has also been concern around the cutoff cycle for many qPCR molecular tests that may have resulted in a large number of false positive results upending the lives of those individuals requiring quarantine [12, 13] . The mass production and distribution of vaccines from multiple commercial suppliers has raised further questions and concerns pertaining to testing: 1. Since different commercial vaccines have widely different SARS-CoV-2 strain-dependent efficacies ranging from 10% to 95% [14] [15] [16] , how is protection assured post-vaccination in the context of circulating wild type and variant strains? 2. How long does immunity persist after administration of vaccines? 3. What tests are relevant and provide the most actionable information in populations of mixed vaccinated and non-vaccinated individuals? The rate of data produced and published pertaining to SARS-CoV-2 has eclipsed most other fields of study since the onset of the pandemic requiring almost daily literature reviews to filter the solid articles that are advancing the field [17] [18] [19] . There is now strong evidence correlated from multiple peer-reviewed articles in solid journals to answer the above questions. All vaccines are designed to elicit an immune response to either the spike protein of SARS-CoV-2 or the attenuated virus [14, 15, 20] . The goal being to train the immune system to engage the virus with a robust immune response with front-line, neutralizing antibodies (nAb's) along with T and B cells. Neutralizing antibody (nAb) levels post-vaccination have been shown to be highly predictive of vaccine efficacy and immune protection from symptomatic SARS-CoV-2 infection [21] [22] [23] . Their early presence within 14 days of infection has been negatively correlated to COVID-19 morbidity [24] . However, two recent studies demonstrated that 30% of elderly people over 80 years of age receiving the Biontech/Pfizer BNT162b2 vaccine and 71.5% of immunocompromised patients receiving the mRNA-1273 (Moderna) vaccine did not elicit a measurable nAb response [25, 26] . Furthermore, the level of protection from infection by the native SARS-CoV-2 and the emerging variants has been shown to differ greatly between the vaccines [14, 15] . Fortunately, there is a growing body of evidence correlating a strong vaccine or infection induced nAb response to immunity from the SARS-CoV-2 wild type and variant forms of the virus [27] [28] [29] [30] [31] [32] [33] [34] [35] [36] [37] . Taken together, these data warrant the measurement of neutralizing antibodies post-vaccination. Although a global consensus relating neutralizing antibody titers to vaccine efficacy and protection has not yet been reached, some very recent articles support this premise. Of note, the Gilbert et al. pre-print paper demonstrates that vaccine efficacy drops precipitously below 90% when neutralizing antibody titers are below 100 WHO International Standard (IS) Units/mL [22] . The Feng pre-print manuscript shows that neutralizing antibody titers below about 200 WHO IS Units per mL equates to vaccine efficacy below 80% [23] . Finally, the Khoury Nature Medicine article supports only 50% protection from vaccines when neutralizing antibody titers are approximately 54 WHO IS Units per mL. These articles represent some of the first published evidence supporting a neutralizing antibody titer-based dependence on vaccine efficacy and protection summarized in Table 1 . Note: These cut-offs have been interpreted from the above-mentioned articles and have not been Authorized by the US Food and Drug Administration. Many commercial manufacturers of serology assays have therefore responded by supplying tests that measure non-functional, binding immunoglobulins (typically IgG's) to spike protein and/or its receptor binding domain (RBD) which are the primary target for nAbs [2, 3, 38] . However, it has recently been shown that only a fraction of these IgG's are in fact neutralizing [39] . Also, the total IgG immune response to spike protein or RBD from vaccinated individuals does not closely correlate to vaccine efficacy or presence of functional, nAbs [26, 40, 41] underlining the requirement to specifically measure nAb levels. The current, gold standard, neutralization tests require live cells and either live SARS-CoV-2 virus or a less dangerous pseudovirus [42] [43] [44] . In either case, these tests require days to complete and an elevated biosecurity level laboratory (BSL 3) (when working with live SARS-CoV-2). Therefore, these live virus/cell neutralization tests are expensive, dangerous and low throughput so not amenable to large population testing. A recent high-throughput, ELISA-based test termed cPass surrogate virus neutralization test (sVNT) (cPass sVNT) [2, 45] has been designed, commercialized and EUA authorized by FDA to specifically detect nAbs at much lower cost, and with highly correlating data to the live cell/virus assays ( Figure 1 ) [2, 34, 38, [45] [46] [47] [48] . Molecular (qPCR) testing has been used exclusively for early detection (within one to two A major question associated with the mass vaccination efforts globally is the duration of protection [33, 65] . Longitudinal human studies detecting both total IgG as well as neutralizing- 6 specific antibodies support a decline in their levels over time and animal studies have shown a direct correlation to neutralizing antibody levels and susceptibility to SARS-CoV-2 reinfection [33, 40, 41, 51-54, 57-59, 61, 64] . Although these data have not yet been fully verified in humans, some recent work strongly supports neutralizing antibody titers as a protective correlate to SARS-CoV-2 infection that can be used as potential measurable molecular determinants that trigger the requirement for a booster shot [22, 23, 27] . The recently initiated human challenge studies should substantiate the current body of data [66, 67] . nAbs will likely play a role and can be measured semi-quantitatively to investigate the correlation of their levels post-infection/recovery and/or post-vaccination with protection. Therefore, to mitigate continued pandemic waves of infection globally, the periodic measurement of nAb titer levels may reveal a minimal titer required for protection and the potential requirement for booster vaccinations. SARS-CoV-2 pandemic control continues to be a challenge globally and is further complicated by the distribution of different vaccines with varying efficacies towards the variant strains [14] [15] [16] . The question around the longevity of immunity post-vaccination remains unanswered but the periodic measurement of nAb titers may give a good indication. To minimize the potential for future outbreaks, a revised testing strategy to assure initial and continued protection postvaccination will likely be required. This could include a combination of qPCR (for non-vaccinated and susceptible people) and nAb (for previously infected and vaccinated people) assays. cPass sVNT could be a useful tool for substantiating individual immunity in a SARS-CoV-2 postvaccination era. The cPass SARS-CoV-2 Neutralization Antibody Test is CE Marked for diagnostic use in European (2021)). The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. Performance of four automated SARS-CoV-2 serology assay platforms in a large cohort including susceptible COVID-19 negative and COVID-19 positive patients Dual Purpose Serology Test: Highly Accurate Infection Tracing and Neutralizing Antibody Response Detection Evaluation of Six Commercial Mid-to High-Volume Antibody and Six Point-of-Care Lateral Flow Assays for Detection of SARS-CoV-2 Antibodies Diagnostics for SARS-CoV-2 detection: A comprehensive review of the FDA-EUA COVID-19 testing landscape Effectiveness of tests to detect the presence of SARS-CoV-2 virus, and antibodies to SARS-CoV-2, to inform COVID-19 diagnosis: a rapid systematic review The estimation of diagnostic accuracy of tests for COVID-19: A scoping review Evaluating 10 Commercially Available SARS-CoV-2 Rapid Serological Tests by Use of the STARD (Standards for Reporting of Diagnostic Accuracy Studies) Method Guidance for the design and reporting of studies evaluating the clinical performance of tests for present or past SARS-CoV-2 infection Paid $20 million for new coronavirus tests. They didn't work Diagnostic accuracy of serological tests for covid-19: systematic review and meta-analysis Flawed Tests: China's Quality Control Prob lem in Leading Global COVID-19 Fight. The Los Angeles Times To Interpret the SARS-CoV-2 Test, Consider the Cycle Threshold Value Preanalytical issues and cycle threshold values in SARS-CoV-2 real-time RT-PCR testing: should test results include these? COVID-19 Vaccine Decisions: Considering the Choices and Opportunities SARS-CoV-2 vaccines Covid-19: Where are we on vaccines and variants? Scientists are drowning in COVID-19 papers. Can new tools keep them afloat Methodological quality of COVID-19 clinical research Pandemic publishing: Medical journals strongly speed up their publication process for COVID-19 Developing a SARS-CoV-2 Vaccine at Warp Speed Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection Immune Correlates Analysis of the mRNA-1273 COVID-19 Vaccine Efficacy Trial Correlates of protection against symptomatic and asymptomatic SARS-CoV-2 infection Delayed production of neutralizing antibodies correlates with fatal COVID-19 Age-dependent immune response to the Biontech/Pfizer BNT162b2 COVID-19 vaccination Humoral and cellular immune response and safety of two-dose SARS-CoV-2 mRNA-1273 vaccine in solid organ transplant recipients What level of neutralising antibody protects from COVID-19? COVID-19-neutralizing antibodies predict disease severity and survival Neutralizing antibodies correlate with protection from SARS-CoV-2 in humans during a fishery vessel outbreak with high attack rate. medRxiv Antibodies elicited by SARS-CoV-2 infection and boosted by vaccination neutralize an emerging variant and SARS-CoV-1 Neutralization of SARS-CoV-2 spike 69/70 deletion, E484K and N501Y variants by BNT162b2 vaccine-elicited sera mRNA-1273 vaccine induces neutralizing antibodies against spike mutants from global SARS-CoV-2 variants Humoral immune responses and neutralizing antibodies against SARS-CoV-2; implications in pathogenesis and protective immunity Validation of a commercially available indirect assay for SARS-CoV-2 neutralising antibodies using a pseudotyped virus assay Reduced neutralization of SARS-CoV-2 B. 1.1. 7 variant by convalescent and vaccine sera Effectiveness of the BNT162b2 Covid-19 Vaccine against the B.1.1.7 and B.1.351 Variants CoV2373 Covid-19 Vaccine against the B.1.351 Variant Head-to-head evaluation on diagnostic accuracies of six SARS-CoV-2 serological assays The plasmablast response to SARS-CoV-2 mRNA vaccination is dominated by non-neutralizing antibodies that target both the NTD and the RBD Antibody Responses after a Single Dose of SARS-CoV-2 mRNA Vaccine Detection of SARS-CoV-2 antibodies formed in response to the BNT162b2 and mRNA-1237 mRNA vaccine by commercial antibody tests Challenges in Laboratory Diagnosis of the Novel Coronavirus SARS-CoV-2. Viruses Laboratory testing of SARS-CoV, MERS-CoV, and SARS-CoV-2 (2019-nCoV): Current status, challenges, and countermeasures Protocol and reagents for pseudotyping lentiviral particles with SARS-CoV-2 Spike protein for neutralization assays A SARS-CoV-2 surrogate virus neutralization test based on antibody-mediated blockage of ACE2-spike protein-protein interaction Evaluation of a SARS-CoV-2 surrogate virus neutralization test for detection of antibody in human, canine, cat and hamster sera Early detection of neutralizing antibodies against SARS-CoV-2 in COVID-19 patients in Thailand A comparison of SARS-CoV-2 antibody assays Optimal COVID-19 quarantine and testing strategies COVID-19 testing: One size does not fit all Rapid decline of neutralizing antibodies against SARS-CoV-2 among infected healthcare workers Rapid decay of anti-SARS-CoV-2 antibodies in persons with mild Covid-19 Longitudinal observation and decline of neutralizing antibody responses in the three months following SARS-CoV-2 infection in humans Declining prevalence of antibody positivity to SARS-CoV-2: a community study of 365,000 adults Robust neutralizing antibodies to SARS-CoV-2 infection persist for months Semiquantitative, high throughput analysis of SARS-CoV-2 neutralizing antibodies: Measuring the level and duration of immune response antibodies post infection/vaccination Critical role of neutralizing antibody for SARS-CoV-2 reinfection and transmission Evaluation of the mRNA-1273 vaccine against SARS-CoV-2 in nonhuman primates DNA vaccine protection against SARS-CoV-2 in rhesus macaques BNT162b vaccines protect rhesus macaques from SARS-CoV-2 A SARS-CoV-2 infection model in mice demonstrates protection by neutralizing antibodies Correlates of protection induced by vaccination Isolation of potent SARS-CoV-2 neutralizing antibodies and protection from disease in a small animal model Development and structural basis of a two-MAb cocktail for treating SARS-CoV-2 infections SARS-COV-2, can you be over it? Arguments for the Immune passport COVID-19 human challenge studies in the UK. The Lancet Respiratory Medicine Evaluating use cases for human challenge trials in accelerating SARS-CoV-2 vaccine development