key: cord-0733369-5mf1r24m authors: Chau, Cindy H.; Strope, Jonathan D.; Figg, William D. title: COVID‐19 Clinical Diagnostics and Testing Technology date: 2020-07-08 journal: Pharmacotherapy DOI: 10.1002/phar.2439 sha: 6f0af73257e6b995fe875a235de60e7dd92d6c80 doc_id: 733369 cord_uid: 5mf1r24m Given the global nature of the COVID‐19 pandemic, the need for disease detection and expanding testing capacity remains a critical priority. This review discusses the technological advances in testing capability and methodology that are currently employed or in development for detecting the novel coronavirus. We describe the current clinical diagnostics and technology, including molecular and serological testing approaches, for SARS‐CoV‐2 testing as well as address their advantages and limitations. Nucleic acid amplification technology for molecular diagnostics remains the gold standard for virus detection. We highlight alternative molecular detection techniques used for developing novel COVID‐19 diagnostics on the horizon. Antibody response against SARS‐CoV‐2 remains poorly understood and proper validation of serology tests is necessary to demonstrate their accuracy and clinical utility. In order to bring the pandemic under control, we must speed up the development of rapid and widespread testing through improvements in clinical diagnostics and testing technology as well as access to these tools. In December of 2019, the World Health Organization (WHO) was notified by Chinese officials of a pneumonia of unknown etiology that had affected nearly 44 individuals. On January 7, 2020, Chinese researchers isolated a novel coronavirus now known as SARS-CoV-2 that causes the disease COVID- 19 . Proper mitigation of continued spread of the virus relies on rapid and accurate laboratory testing to confirm positive cases. One of the first published reports of an asymptomatic transmission of disease came from a family cluster in China, 1 opening the possibility that positive patients could unknowingly be infecting others and that the number of positive cases was being underreported. Analysis of the viral load in an asymptomatic patient in China found that the levels were similar to those of symptomatic patients. 2 These findings present a challenge to successful contact tracing and exposure control. Taken together, the data underlay the importance of not only physical distancing, but more importantly expansive testing and isolation of positive cases regardless of the presence of symptoms to contain and mitigate the SARS-CoV-2 pandemic. It remains critical that clinicians understand the relationships between test results from virologic and serologic testing and the performance of these diagnostic assays in order to prescribe appropriate drug therapy. Accurate interpretation and assessment of viral exposure and/or immunological response may impact pharmacotherapy decisions. As no "cure" currently exists for SARS-CoV-2 infection, clinical utility of validated serologic testing may have a role in clinical decision making as antibody detection may be used as an indicator of the stage of COVID-19 progression to aid in management of the disease. Thus, it is essential that patients who are seropositive be identified in order to properly design treatments. There are many different ways to test for SARS-CoV-2. In this review, we briefly describe the current and future approaches for clinical testing for COVID-19 including molecular and serological testing as well as their benefits and limitations. We reviewed original papers published in PubMed since the COVID-19 outbreak up to the time of writing using the following terms: COVID-19, SARS-CoV-2, diagnostics, testing, RT-PCR, serology, and antigen. This article is protected by copyright. All rights reserved Genetic information for the viral genome SARS-CoV-2 originally appeared in the community online resource (https://virological.org). Subsequently, updated sequences of the genome from around the world have been deposited in the viral sequence database curated by the Global Initiative on Sharing All Influenza Data (GISAID) and GenBank (accession number MN908947; NCBI Reference Sequence: NC_045512). Nucleic acid amplification technology (NAAT) based on polymerase chain reaction (PCR), which is regarded as the "gold standard" for virus detection. The diagnosis of COVID-19 relies on the detection of the SARS-CoV-2 RNA by real-time reverse transcriptase polymerase chain reaction (RT-PCR). 3, 4 The viral RNA is detectable in nasopharyngeal and oropharyngeal swab samples as well as respiratory secretions during the acute phase of viral infection. Genetic material is extracted from patient samples and reverse transcriptase is used to make a complementary DNA strand from the viral RNA. Selective amplification of the target nucleic acid from the sample is achieved by designing target-specific forward and reverse primers flanking regions of interest (ROIs) that are unique to the SARS-CoV-2 genome (Figure 1 ): replicase open reading frame (ORF1a/b), spike (S), envelop (E), membrane (M), and nucleocapsid (N). The genome of SARS-CoV-2 has been characterized and compared to other coronaviruses to identify homology. 5 The genome of SARS-CoV-2 shares 89% nucleotide identity with bat SARS-like-CoVZXC21 and 82% with that of human SARS-CoV. Preferred targets or ROIs include ORF1a/b, ORF1b-nsp14, nonstructural RNA dependent RNA polymerase (RdRp), S, E, or N gene of SARS-CoV-2. A detailed genetic analysis of SARS-CoV-2 with other coronaviruses, those with low and high case fatality rates, found that pathogenicity is predicted most by the regions of the spike and nucleocapsid proteins. 6 A recent study has identified selective mutations in the SARS-COV-2 genome; therefore, it is essential to avoid certain ROIs when designing primers and probes. 7 Although the mutation rate of SARS-CoV-2 remains to be determine, it is estimated to be moderate compared to influenza. Based on the natural viral mutation and recombination rates, targeting more than one ROI can mitigate the risk of loss of sensitivity should potential mutations arise. 8 Monitoring of genomic variations in SARS-CoV-2 is ongoing in case assay re-design is needed. This article is protected by copyright. All rights reserved Target selection can affect assay performance in terms of specificity and sensitivity as well as cross reactivity due to conserved regions. The validity of a test is measured by its analytical and clinical sensitivity and specificity. Analytical sensitivity is the assays ability to reliably detect the minimum concentration of a substance in a sample (also referred to as limit of detection) whereas analytical specificity refers to the ability to detect only the desired analyte in a specimen without cross reacting with other substances. Clinical sensitivity measures how accurately a test identifies positive patients who are infected. A test with 95% sensitivity will identify 95% of patients who have the disease and produces false negatives in 5% of patients (5 out of every 100 patients who are positive test negative); a lower sensitivity test means higher false negative results. Clinical specificity, the inverse of sensitivity, determines how accurately a test identifies negative patients who do not have COVID-19. A test with 95% specificity will identify 95% of patients who do not have the disease and incorrectly identify 5% of patients who test positive but actually are not infected; a lower specificity test means higher false positives. A test with good analytical sensitivity and specificity does not necessarily correlate with clinical sensitivity and specificity. (See section on Assay Limitations for a discussion on implications of false positive and negative tests.) The WHO has established protocols for various testing kits developed around the world with a listing of the targeted viral regions ( Table 1 ). One of the first RT-PCR assays published for SARS-CoV-2 was developed by Germany Charité and targeted the RdRp, E, and N genes of the virus. The assay, designed using synthetic nucleic acid technology and in the absence of available virus isolate or original patient specimens, demonstrated that targeting the E and RdRp gene was highly sensitive while the N gene was slightly less sensitive. 3 Numerous RT-PCR assays targeting various combinations of the ROIs have since been designed to improve and maximize detection sensitivity and specificity. 9 Initially in the United States, production and validation of RT-PCR based testing kits were completed by the Centers for Disease Control and Prevention (CDC) following federal rules and regulations. The first kits sent out to state health agencies were sidelined by a faulty reagent in the negative control and one primer set (CDC N3) that did not perform well which was later excluded from the kits. 10 Subsequent studies validated the specificity and sensitivity of the primer-probe sets This article is protected by copyright. All rights reserved from various assays. A study conducted by the University of Washington compared the performance of their primer sets against those developed by Germany Charité 3 and the US CDC primers (CDC N1 and CDC N2) in the current CDC testing kit. The study found high specificity among all primer sets and variable sensitivity, with the CDC N2 and the Germany E-gene sets being more sensitive than others. 11 Another study (in preprint) examined the sensitivity and efficiency of four common RT-PCR assays developed by the US CDC, China CDC, Germany Charité, and Hong Kong University (HKU). 12 This study also found that while all primer-probe sets can detect SARS-CoV-2, the most sensitive primer-probe sets are the Germany E gene, HKU-ORF1, HKU-N and US CDC N1. While the US CDC N2 primers had background cross-reactivity, it did not interfere with outcomes of the combined N1 and N2 assay when testing clinical samples from COVID-19 patients. 12 Other studies have also compared the US CDC N1 and N2 against commercial test kits, reporting similar specificity with variable sensitivity. 13, 14 To accelerate testing capacity, the US Food and Drug Administration (FDA) released updated guidance on February 29, 2020 opening up the production of diagnostic kits to state labs, laboratories certified under Clinical Laboratory Improvement Amendments (CLIA), and commercial diagnostic developers. 15 Clinical laboratories have the option of either using the CDC primers or other established primers/protocols listed on the WHO website, to produce their own "laboratory-developed tests" for in-house use. Since release of the guidance, the FDA has and continues to issue Emergency Use Authorizations (EUAs) and a comprehensive listing of all commercial diagnostic tests are updated on the FDA's website (https://www.fda.gov/medical-devices/coronavirus-disease-2019covid-19-emergency-use-authorizations-medical-devices/vitro-diagnostics-euas). EUAs issued to the singular developing laboratory such as ones based at research/academic institutions and hospitals can also be found at the same FDA website. At the time of writing, the US FDA has issued 149 individuals EUAs (commercial and laboratory-developed tests), which includes 125 molecular tests, 23 antibody tests and 1 antigen test. The FDA lists all individual EUAs for molecular, antibody, and antigen tests and provides links to these tests the include information on the assay platform, gene targets of the test as well as performance characteristics (sensitivity and specificity). The WHO partners with the Foundation for Innovative New Diagnostics (FIND) for developing urgently needed This article is protected by copyright. All rights reserved diagnostic tests. FIND's SARS-CoV-2 diagnostic pipeline tracks all tests commercially available or in development worldwide (https://www.finddx.org/covid-19/pipeline/). FIND is conducting independent evaluations of SARS-Cov-2 testing platforms to verify the limit of detection of each test (as reported by the manufacturers) and the clinical performance of the test (https://www.finddx.org/covid-19/sarscov2-eval-molecular/). Diagnostic testing errors can result in false positives and/or false negatives that stem from improper sample collection, testing procedural errors, and variability in assay performance (sensitivity/specificity). Diagnostic tests need to be evaluated to determine their sensitivity and specificity, ideally by comparison with a "gold standard." The lack of a clear-cut gold-standard diagnostic test for SARS-CoV-2 (because the virus is new) makes evaluation of test accuracy challenging; therefore, assay performance needs to be compared to a reference standard. The FDA recommends selecting a comparator assay that has established high sensitivity with an internationally recognized standard or FDA SARS-CoV-2 Reference Panel and set the criteria at a minimum of 95% positive and negative agreement as acceptable clinical performance of the assay. With the strategy to "test, isolate, contact trace and quarantine", a false positive result mistakenly identifies a patient as having COVID-19 resulting in unnecessary contact tracing and quarantine. False negatives prevent proper contact tracing and quarantine efforts and importantly miss isolating the individual who can now infect others. Timing is critical when testing for SARS-CoV-2 to prevent inaccurate results. Depending on the test rendered, testing early or late in the infection cycle could lead to false negative results. For molecular based assays, the optimal testing window occurs near the end of the first week and into the beginning of the second week after symptom onset ( Figure 2 ). However, positive tests are possible near the first day of symptom onset. 16 Persistent positive tests or positive tests in discharged patients after symptom resolution have been documented. In a report from South Korea CDC, 285 of a reported 447 re-positive tests were followed; they concluded that there is no evidence that those testing positive long after symptom onset and resolution are re-infected or still infectious. 17 The duration of viral RNA shedding is variable and depend on disease severity; This article is protected by copyright. All rights reserved prolonged viral RNA shedding of SARS-CoV-2 is detectable by RT-PCR in patients recovering from COVID-19 for up to 37 days. 18, 19 Thus, detection of viral RNA does not necessarily reflect the presence of infectious virus and prolonged viral RNA detection following recovery does not necessarily indicate infectiousness. The implications of false negatives become increasingly significant as governments move forward with reopening and loosening stay at home restrictions. Recent studies have shed light and caution on the interpretation of negative test results (particularly early in the course of infection). One study demonstrated a highly variable false-negative rate for SARS-CoV-2 RT-PCR testing, suggesting one out of five people suspected of COVID-19 may test negative based on pooled analysis of data from 7 studies. 20 Therefore, waiting 1 to 3 days after symptom onset can lessen the chances of a false negative result. If there is high clinical suspicion for infection by SARS-CoV-2, then serial testing is recommended to reduce the false negative rate. Recent studies have begun to investigate clinical performance of SARS-CoV-2 molecular testing to determine optimal time frames for appropriate repeat testing with one study suggesting 15 to 20 days after a positive test and the same or next 2 days after a negative test in patients with a high suspicion for infection. 21 Understanding the predictive value of NAAT with regards to time from exposure and symptom onset is important as the assay may not have been appropriately validated against a clinically meaningful reference standard for detecting SARS-CoV-2 in the absence of symptoms (during earlier stages of the disease) or in asymptomatic individuals. 22 The sensitivity of a test must be high for adequate containment measures and assessment of clinical sensitivity in asymptomatic patients is warranted. Other limitations of RT-PCR testing that can hinder use, include contamination issues, timeconsuming sample handling 23 and variable detection of SARS-CoV-2 depending on the type of clinical specimen. 24 Commercial developers have been challenged to improve the real-time PCR method and produce assays with ease of sample collection and/or processing and shorter run times to decrease turnaround times. This will enable tests to be conducted in the field at local hospitals, during doctor visits, and at mobile testing centers. In addition, the unprecedented need for testing resulted in a severe shortage of reagents and supplies including collection swabs, transport media, extraction kits, This article is protected by copyright. All rights reserved and RT-PCR enzyme mixtures. To circumvent shortages, suitable alternative solutions were investigated such as comparing the use of various swabs and transport media for SARS-CoV-2 testing. 25 Saliva as a non-invasive specimen for detection of SARS-CoV-2 represents a suitable reliable alternative to help ease the global shortage of swabs for sampling and personal protective equipment used during collection. 26, 27 The first saliva-based test, developed and validated by Rutgers University, was issued an EAU to allow testing of saliva specimens as an alternative to samples collected through swabbing. Many of the limitations seen in available testing modalities is the necessity for expensive bulky machines, trained technical staff, and reagents. 28 These tests are most often performed in large labs at commercial diagnostic facilities or academic centers. Samples must therefore be sent to these labs, increasing both time and coordination. Improvements to the performance of the assay are available with the use of high throughput platforms, such as the cobas 6800/8800 systems 4 , which involve an automated workflow to enable high throughput testing with minimal hands-on time, and still offering fast, reliable results (e.g., 96 results in about three hours or 384 results for the cobas 6800 System and 960 results for the cobas 8800 System in 8 hours). Other diagnostics that use high throughput platforms for COVID-19 testing that have received an EUA are listed on the FDA website. The FDA granted an EUA for the first point-of-care (POC) diagnostic, the Xpert Xpress SARS-CoV-2 test. 29 The test comes as a self-contained kit where a patient sample is loaded into a cartridge system and gives a readout in 45 minutes without the need of additional reagents or trained personnel. This technology allows for medical personnel in the hospital to make rapid assessments and clinical decisions. Another POC diagnostic that received the FDA's EUA is the Accula SARS-Cov-2 test, a handheld device that also uses PCR technology to detect SARS-CoV-2 in throat and nasal swab specimens. Specimens are added to a buffer to solubilize the samples and then an aliquot is dispensed in a cassette that contains the proper controls, enzymes, and reagents as well as a detection strip for the assay that is automated from sample extraction to nucleic acid amplification. Test results are displayed by the visualization of blue test lines on the detection strip after 30 minutes. Perhaps the This article is protected by copyright. All rights reserved most rapid POC diagnostic kit with EUA approval from the FDA is the ID NOW COVID-19 test which delivers positive results within 5 minutes and negative results in 13 minutes. The platform eliminates thermocycling and performs isothermal nucleic acid amplification technology with a sequence-specific fluorescent probe readout. All of the above POC tests enable the convenience of on-site testing and rapid detection of SARS-CoV-2. Limitations of these POC testing system are that they are not built for high throughput assessments, can often be more expensive than other options, and have variable sensitivity due to the more rapid nature of the test. Studies have been conducted to examine the performance of these POC tests compared to standard RT-PCR or in detection of low levels of viral RNA. 30, 31 One study (comparing Xpert Xpress, ID NOW, and the ePlex systems) found that while all three had 100% specificity (did not exhibit false positive results), Xpert Xpress performed well and had the lowest limit of detection and highest sensitivity, while the ePlex and ID NOW had lower sensitivities. 32 Another study determined that the POC test (ID NOW) was less sensitive compared to the traditional RT-PCR assay. 33 There is urgent need to develop a more rapid, accurate, and point-of-care assay to control and minimize the spread of infection in the community. Alternative molecular detection techniques have employed both LAMP-and CRISPR-based methods. Reverse transcription loop-mediated isothermal amplification (RT-LAMP) method is another nucleic acid amplification technique that amplifies small numbers of DNA and RNA templates with high specificity and sensitivity under isothermal conditions. 34 LAMP is a one-step amplification reaction that requires no thermocycling steps, resulting in shorter "sample-to-answer" times than conventional PCR. Thus, the assay is ideally suited for field-based nucleic acid diagnostics due to rapid results (usually 30 to 60 minutes) and minimal Accepted Article requirements for expensive instrumentation or reagents. Previous RT-LAMP assays with improved specificity, visualization techniques or readouts have been developed for detecting MERS-CoV and SARS-CoV. Several LAMP-based diagnostics are in the pipeline for detecting SARS-CoV-2 that have been developed by laboratories worldwide including South Korea, 35, 36 China, [37] [38] [39] and the United Kingdom 40 as well as in the United States with LAMP and CRISPR technology developed in combination (discussed in the next section). A rapid colorimetric assay using RT-LAMP is being developed for the qualitative detection of virus directly from saliva. 41 The powerful genome editing technology known as CRISPR (clustered regularly interspaced short palindromic repeats) has also been recently exploited to develop rapid, inexpensive, POC diagnostics for COVID-19 testing. The RNA-targeting CRISPR associated enzyme Cas13 can be programmed to target and destroy single stranded RNA viruses. 42 A platform termed SHERLOCK (specific highsensitivity enzymatic reporter unlocking) was developed to combine isothermal preamplification with Cas13 to detect single molecules of RNA or DNA. 43, 44 The CRISPR-Cas13-based SHERLOCK COVID-19 detection protocol searches for unique nucleic acid targets (SARS-CoV-2 Orf1ab and S genes) and uses a dipstick as the visual readout in less than an hour. 45 This article is protected by copyright. All rights reserved The antigen test for SARS-CoV-2 is a diagnostic test designed for rapid detection of viral proteins (antigens). While the main advantage of these antigen tests is the speed of the test, they are often plagued with inaccurate results and have lower sensitivity and specificity than nucleic acid assays. Previous rapid antigen tests for influenza (H1N1) and respiratory syncytial virus were found to be less sensitive compared with RT-PCR. 49, 50 Development of antibodies that are specific to viral antigens is often difficult and time consuming and antigen tests must undergo rigorous quality control measures to ensure specificity and sensitivity. Although antigen tests are very specific for the virus, they cannot detect all active infections and thus have a higher chance of false negatives than RT-PCR. FDA issued an EUA to the first antigen assay, the Sofia 2 SARS Antigen FIA test, which employs immunofluorescence-based lateral flow technology in a sandwich design that is used to detect the viral nucleocapsid protein in nasopharyngeal and nasal swab specimens with test results in 15 minutes. Future research on the clinical performance of antigen testing compared to NAAT is highly encouraged to determine their diagnostic utility. Outside of nucleic acid amplification-based diagnostics, serological or antibody tests for COVID-19 is another promising testing modality. Both molecular and serological tests are intended for patients at different stages of the disease process with RT-PCR for detection of current active infection and serology tests for later stages since these assays detect the presence of SARS-CoV-2 antibodies produced by the bodies humoral immune system. Serologic testing enables the understanding of how patients produce antibodies to SARS-CoV-2, and assays can detect IgA, IgM, IgG, or total antibody. IgM antibodies can be detected as early as 5 days after new infection, with higher levels detected in the second and third week 51, 52 . Other studies detected optimal IgM antibodies towards the end of the first week that peaked during the third week after symptom onset, and may correspond to the emergence of negative results by RT-PCR tests as the viral load is cleared (Figure 2) . 16, 19, 51, 53 Another report determined earlier IgA seroconversion than IgM 54 or that IgA is detected at higher This article is protected by copyright. All rights reserved levels than IgM. 55 IgG antibodies become detectable later in the infection course around 7-14 days after symptom onset and levels remained relatively high until 6 weeks. 51, 52 While the data remains premature, a small study of 285 patients with COVID-19 reported that 100% of patients tested positive for IgG within 20 days after symptom onset. Seroconversion for IgG and IgM occurred simultaneously or sequentially between the third and fourth weeks, and both levels plateaued within 6 days after seroconversion. 56 Asymptomatic patients may seroconvert later in the course of infection or may not at all. 57 Another study found that asymptomatic patients have significantly longer duration of viral shedding, lower IgG and neutralizing serum antibody levels compared to the symptomatic group, suggesting that asymptomatic patients may mount a weaker antibody response and immunity may diminish within months of infection. 58 This article is protected by copyright. All rights reserved been granted EUAs using various platforms, including ELISAs and CIAs, and all authorized tests are qualitative rather than quantitative. A comprehensive listing of all commercially marketed antibody tests granted EUAs are updated on the FDA and FIND websites on a rolling basis. Serology tests are currently being developed for the detection of antibodies (in blood plasma or serum) to two main antigenic targets, the N protein and S protein, including subunits S1 and S2 as well as the receptor-binding domain (RBD) of SARS-CoV-2. The target selection determines crossreactivity and specificity with N being more conserved across coronaviruses than S and within S, the RBD is more conserved than either subunit. Proper serology testing requires understanding the performance characteristics and limitations of these tests. Serological tests have varying levels of specificity and sensitivity and can detect past infections in those who are RT-PCR negative. False positives can result from cross-reactivity with pre-existing antibodies from previous infections such as other coronaviruses (229E, NL63, OC43, and HKU1 that cause the common cold; SARS-CoV or MERS-CoV; an antibody test with low specificity); whereas negative results may be due to antibodies that have not yet formed during the early stages of infections. Initial studies have shown that N and S1 protein ELISAs could detect SARS-CoV-2 with high specificity; RBD and N protein ELISAs are more sensitive than S1 assays in detecting antibodies in patients with mild infections. 61 Researchers also found that antibodies to N protein are more sensitive than to S protein during early infection using a liquid phase immunoassay called luciferase immunoprecipitation systems. 62 A meta-analysis was recently conducted to investigate the performance of all available antibody tests and found that tests using the S antigen are more sensitive than N antigen-based tests. A combined IgG/IgM test displayed better sensitivity than measuring either antibody type alone with ELISA tests being more sensitive than lateral flow immunoassays. 63 A recent study found high sensitivity and specificity of the RBD for antibody detection and its strong association with SARS-CoV-2 neutralizing antibodies. 59 Studies are ongoing to determine the clinical performance characteristics of various antibody tests across different platforms. 64, 65 Accepted Article This article is protected by copyright. All rights reserved The prevalence of SARS-CoV-2 antibody positive individuals in the US population remains to be determined and is based on the different rates of infection across various locations and populations. The positive and negative predictive values (PPV and NPV) of antibody tests will affect the overall outcome of testing and are determined by the prevalence of the disease and the sensitivity/specificity of the test. PPV is the probability that a patient who tests positive truly has the disease and NPV is the chance that one who tests negative has not been infected. In the setting of low prevalence of SARS-CoV-2 infection, even a highly specific test can give a large number of false positives; thus, the PPV will be low. The FDA has summarized the expected performance of the tests it has authorized based on the information FDA reviewed when deciding whether or not to grant these tests an EUA and assuming a prevalence of 5% for PPV and NPV calculations. We refer the reader to the FDA's website for the performance characteristics of the individual tests which have received EUA status (https://www.fda.gov/medical-devices/emergency-situations-medical-devices/eua-authorizedserology-test-performance). Antibody response against SARS-CoV-2 remains poorly understood and the clinical utility of tests is unclear until the assays are properly validated to demonstrate their accuracy. Massive efforts are currently underway to conduct large-scale validation studies on the performance of these assays, which is critical before they can be used in seroprevalence studies for disease surveillance. A collaborative effort by the FDA, NIH, CDC and Biomedical Advanced Research and Development Authority (BARDA) is currently underway to conduct the performance assessments and establish the validity of serological tests against a well-characterized set of clinical samples collected before and during the pandemic and correlate them with neutralization assays. The validation dataset is published on the openFDA website (https://open.fda.gov/apis/device/covid19serology/). Properly validated accurate test can then potentially be used in the future as a public health surveillance tool to monitor the epidemiological spread COVID-19, as a risk assessment tool to screen for immunity, or as a screening tool to identify donor patients who have antibodies for convalescent plasma treatment. This article is protected by copyright. All rights reserved Some companies have been developing at home sample packs. These test kits would include the necessary components and instructions to properly obtain a nasal swab sample while at home. The kits would then be mailed to a central laboratory for testing. On April 21, 2020 the FDA authorized the first molecular diagnostic test with a home collection option for LabCorp's RT-PCR assay to permit testing of samples self-collected by patients at home using the company's home collection kit. Subsequently, Rutgers University also received EUA designation for its at-home saliva self-collection assay. Everlywell's COVID-19 Test Home Collection Kit is the first standalone sample collection kit (for self-collecting a nasal sample at home) to be granted an EUA and can be paired with any authorized NAAT at any lab. At-home collection provides an additional solution that limits exposure and keeps patients isolated. However, there are still some concerns that may need to be addressed. The availability of nasopharyngeal swab supply may pose an issue if test needs continue to expand and swabs used for in-person testing are in short supply in some regions. Companies would need to develop alternative methods of sample collection, such as the use of oral swabs or fingerstick whole blood serology tests to provide some relief for the swab-based tests. Improper self-collection may affect specimen quality and subsequent test results. At-home sample collection may encounter issues with poor sampling and ongoing studies will need to examine whether samples (saliva, dried blood spot, swabs) self-collected by patients at home and monitored by a provider through telehealth are sufficient specimens for molecular and serology SARS-CoV-2 testing. 66 Additional limitations to these tests are the time incurred in shipping the test kits as well as the time for the laboratory assessment. In the face of a pandemic, testing, contact tracing, and isolation are all important parameters to contain the spread of COVID-19. While much is unknown about the new coronavirus, it is with certainty that SARS-CoV-2 infection with be a recurring disease. Mounting evidence from recent studies demonstrates the existence of viral transmission from presymptomatic and asymptomatic patients. 67 In the latter scenario, expansive testing remains a priority on the global level. Widespread testing must increase in order to effectively enact isolation procedures and contact tracing measures to This article is protected by copyright. All rights reserved detect asymptomatic infections in order to break transmission chains and attempt to flatten the curve. Asymptomatic cases in a family cluster with SARS-CoV-2 infection SARS-CoV-2 Viral Load in Upper Respiratory Specimens of Infected Patients Detection of 2019 novel coronavirus (2019-nCoV) by real-time RT-PCR Evaluation of a quantitative RT-PCR assay for the detection of the emerging coronavirus SARS-CoV-2 using a high throughput system A new coronavirus associated with human respiratory disease in China Genomic determinants of pathogenicity in SARS-CoV-2 and other human coronaviruses The establishment of reference sequence for SARS-CoV-2 and variation analysis Multiple assays in a real-time RT-PCR SARS-CoV-2 panel can mitigate the risk of loss of sensitivity by new genomic variants during the COVID-19 outbreak Improved molecular diagnosis of COVID-19 by the novel, highly sensitive and specific COVID-19-RdRp/Hel real-time reverse transcription-polymerase chain reaction assay validated in vitro and with clinical specimens The United States badly bungled coronavirus testing-but things may soon improve Comparative Performance of SARS-CoV-2 Detection Assays using Seven Different Primer/Probe Sets and One Assay Kit Analytical sensitivity and efficiency comparisons of SARS-COV-2 qRT-PCR primer-probe sets Comparison of Four Molecular In Vitro Diagnostic Assays for the Detection of SARS-CoV-2 in Nasopharyngeal Specimens Policy for Diagnostic Tests for Coronavirus Disease-2019 during the Public Health Emergency. Immediately in Effect Guidance for Clinical Laboratories, Commercial Manufacturers, and Food and Drug Administration Staff: US Food and Drug Administration Interpreting Diagnostic Tests for SARS-CoV-2. JAMA 2020. 17. Findings from investigation and analysis of re-positive cases Viral dynamics in mild and severe cases of COVID-19 Virological assessment of hospitalized patients with COVID-2019 Variation in False-Negative Rate of Reverse Transcriptase Polymerase Chain Reaction-Based SARS-CoV-2 Tests by Time Since Exposure Clinical Performance of SARS-CoV-2 Molecular Testing False Negative Tests for SARS-CoV-2 Infection -Challenges and Implications Potential preanalytical and analytical vulnerabilities in the laboratory diagnosis of coronavirus disease 2019 (COVID-19) Detection of SARS-CoV-2 in Different Types of Clinical Specimens Comparison of Copan Eswab and FLOQswab for COVID-19 PCR diagnosis: working around a supply shortage Saliva is a reliable tool to detect SARS-CoV-2 Novel Coronavirus Disease (COVID-19): Paving the Road for Rapid Detection and Point-of-Care Diagnostics Multicenter Evaluation of the Cepheid Xpert Xpress SARS-CoV-2 Test Detection of low levels of SARS-CoV-2 RNA from nasopharyngeal swabs using three commercial molecular assays The Detection of SARS-CoV-2 using the Cepheid Xpert Xpress SARS-CoV-2 and Roche cobas SARS-CoV-2 Assays Clinical Evaluation of Three Sample-To-Answer Platforms for the Detection of SARS-CoV-2 Comparison of Abbott ID Now and Abbott m2000 methods for the detection of SARS-CoV-2 from nasopharyngeal and nasal swabs from symptomatic patients Loop-mediated isothermal amplification (LAMP): principle, features, and future prospects Development of a reverse transcription-loop-mediated isothermal amplification as a rapid early-detection method for novel SARS-CoV-2 Development of Reverse Transcription Loop-Mediated Isothermal Amplification Assays Targeting SARS-CoV-2 A Novel Reverse Transcription Loop-Mediated Isothermal Amplification Method for Rapid Detection of SARS-CoV-2 Rapid detection of COVID-19 coronavirus using a reverse transcriptional loop-mediated isothermal amplification (RT-LAMP) diagnostic platform Accepted Article This article is protected by copyright. All rights reserved 39 Rapid and visual detection of CoV-2) by a reverse transcription loop-mediated isothermal amplification assay LAMP for rapid diagnosis of coronavirus SARS-CoV Rapid and extraction-free detection of SARS-CoV-2 from saliva with colorimetric LAMP Nucleic acid detection with CRISPR Multiplexed and portable nucleic acid detection platform with Cas13, Cas12a, and Csm6 SHERLOCK: nucleic acid detection with CRISPR nucleases A protocol for detection of COVID-19 using CRISPR diagnostics2020 Massively multiplexed nucleic acid detection using Cas13 CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity CRISPR-Cas12-based detection of SARS-CoV-2 Validity of two rapid point of care influenza tests and direct fluorecence assay in comparison of real time PCR for swine of origin Influenza virus Diagnostic Accuracy of Rapid Antigen Detection Tests for Respiratory Syncytial Virus Infection: Systematic Review and Meta-analysis Profiling Early Humoral Response to Diagnose Novel Coronavirus Disease (COVID-19) Accepted Article This article is protected by copyright. All rights reserved 52 Serum IgA, IgM, and IgG responses in COVID-19 Antibody Detection and Dynamic Characteristics in Patients with COVID-19 Distinct features of SARS-CoV-2-specific IgA response in COVID-19 patients IgA-Ab response to spike glycoprotein of SARS CoV-2 in patients with COVID-19: A longitudinal study Antibody responses to SARS-CoV-2 in patients with COVID-19 Different longitudinal patterns of nucleic acid and serology testing results based on disease severity of COVID-19 patients Clinical and immunological assessment of asymptomatic SARS-CoV-2 infections The RBD Of The Spike Protein Of SARS Group Coronaviruses Is A Highly Specific Target Of SARS-CoV-2 Antibodies But Not Other Pathogenic Human and Animal Coronavirus Antibodies Antibody responses to SARS-CoV-2 in patients of novel coronavirus disease Severe Acute Respiratory Syndrome Coronavirus 2 Specific Antibody Responses in Coronavirus Disease Detection of Nucleocapsid Antibody to SARS CoV-2 is More Sensitive than Antibody to Spike Protein in COVID-19 Patients Antibody Tests in Detecting SARS-CoV-2 Infection: A Meta-Analysis Performance Characteristics of Four High Throughput Immunoassays for Detection of IgG Antibodies against SARS-CoV-2 Accepted Article This article is protected by copyright. All rights reserved 65 Clinical performance of different SARS-CoV-2 IgG antibody tests Detection of SARS-CoV-2 RNA and Antibodies in Diverse Samples: Protocol to Validate the Sufficiency of Provider-Observed, Home-Collected Blood, Saliva, and Oropharyngeal Samples Evidence Supporting Transmission of Severe Acute Respiratory Syndrome Coronavirus 2 While Presymptomatic or Asymptomatic