Detecting the Coronavirus (COVID-19) Pravin Pokhrel, Changpeng Hu, and Hanbin Mao* Department of Chemistry and Biochemistry, Kent State University, Kent, OH, USA (44240) Keywords: Diagnosis, Detection Kits, RT-PCR, Immunoassay, False Negative, False Positive, Sensitivity, Specificity, Pointof-care testing (POCT) ABSTRACT: The COVID-19 pandemic has created huge damage to society and brought panics around the world. Such panics can be ascribed to the seemingly deceptive features of the COVID-19: compared to other deadly viral outspreads, it has medium transmission and mortality rates. As a result, the severity of the causative coronavirus, SARS-CoV-2, was deeply underestimated by the society at the beginning of the COVID-19 outbreak. Based on this, in this review, we define the viruses with features similar to those of SARS-CoV-2 as the Panic Zone viruses. To contain those viruses, accurate and fast diagnosis followed by effective isolation and treatment of patients are pivotal at the early stage of virus breakouts. This is especially true when there is no cure or vaccine available for a transmissible disease, which is the case for current COVID-19 pandemic. As of June 2020, more than one hundred kits for the COVID-19 diagnosis on the market are surveyed in this review. It is of critical importance to rationally use these kits for the efficient management and control of the Panic Zone viruses. Therefore, we discuss guidelines to select diagnostic kits at different outbreak stages of the Panic Zone viruses, SARS-CoV-2 in particular. While it is of utmost importance to use nucleic-acid based detection kits with low false negativity (high sensitivity) at the early stage of an outbreak, the low false positivity (high specificity) gains its importance at later stages of the outbreak. When the society is set to reopen from the lock-down stage of the COVID-19 pandemic, it becomes critical to have immunoassay based kits with high specificity to identify people who can safely return to the society after their recovery of SARS-CoV-2 infections. Finally, since a massive attack from a viral pandemic requires a massive defense from the whole society, we urge both government and private sectors to research and develop affordable and reliable point-of-care testing (POCT) kits, which can be used massively by the general public (and therefore called as massive POCT) to contain Panic Zone viruses in future. 1. Background Since the beginning of the 21st century, our world has been facing unprecedented crises of deadly viruses like Zika, Ebola, SARS, MERS, and so forth. The epidemics of these viral diseases were sparked either by the evolution of pre-existing viruses or by the emergence of new viral species. Such diseases have already caused colossal damage to the society. Loss of lives struck the most, but the consequences aftermath was equally dreadful: the psychological wellbeing of survivors and socio-economic fallout were rather distressing. Now, in December 2019, the world was hit by another virus known as SARSCoV-2 (the disease associated with this virus is called COVID19). [Figure 1] 1.1. The Panic Zone viruses Compared to other viruses, SARS-CoV-2 has a medium reproduction rate (R0=2.25*) and a medium mortality rate of 5.7%*1 (as of June 7, 2020, *subject to change). Such mediocre characteristics give a rather deceiving impression of this virus. When the virus first started in China, it did not draw immediate attention to the public due to its seemingly “benign” appearance. Indeed, compared to the Death Zone viruses which include Ebola and smallpox (see definition in Figure 1), this disease was considered merely as another type of influenza even among health professionals. However, the virus soon revealed its damaging nature. Staying untreated, the disease spread out quickly to overwhelm the health systems in a society. This eventually caused panics in the general public. People rushed to see doctors even if they developed very mild or even unrelated symptoms, which overran hospitals. This is because in modern society, the production system of healthcare supplies is profit driven2. Decisions regarding the management of disease can no longer be made based solely on scientific grounds. Unless a disease poses a specific risk to a wide population, its mere presence in a localized area or population may not be significant from a business perspective. As a result, necessary resources such as PPE (Personal Protective Equipment) are in short supply to fight pandemic diseases promptly. Due to these reasons, the diseases in the Panic Zone (see Figure 1 for definition) often wreck huge collateral damages due to its paralyzing role for the whole society. In the Panic Zone, SARS was most recently contained by means of massive syndromic surveillance, prompt isolation of patients, and strict quarantine of all contacts. By interrupting all human-to-human transmissions, SARS was effectively eradicated in 20033. Although there are striking similarities between SARS and COVID-19, the differences in the virus characteristics will ultimately determine whether the same measures for SARS will also be successful for the current COVID-19 outbreak. COVID-19 differs from the SARS in terms of infectious period, transmissibility, clinical severity, and extent of the community spread3. Although COVID-19 has lower transmissibility than SARS4, many more COVID-19 patients have mild symptoms that contribute to the rapid spread of the virus as these patients are often missed and not isolated. 1.2. The early detections It is generally true that for a rapidly transmitting disease with no cure or vaccine available, the most effective way to curb its spread is the early detection to isolate patients5,6. The first step to achieve this is to identify those patients using detection kits. Never before is a virus detection system so critical to contain a viral outbreak as dangerous as COVID-19. As shown in Figure 2, for the five countries with similar age distribution and hospital resources, the more extensive the early tests on the COVID19, the lower the overall mortality rates in a country. Indeed, Korea and Germany conducted a substantial number of the tests right at the beginning of the COVID-19 outbreak. Correspondingly, their death rates are among the lowest so far (Figure 2, inset). This confirmed the importance of the early testing to curb the spreading of the COVID-19. [Figure 2] In this review, we first describe the COVID-19 outbreak briefly. Given the importance of the diagnosis for this deadly pandemic disease, we then survey the detection kits used for the COVID-19. After summarizing the challenges facing current commercial kits, we discuss emerging techniques to address these issues. Next, we propose and discuss guidelines to use various kits during different stages of the COVID-19 outbreak. Finally, we wrap up by proposing the research and development of affordable point-of-care testing (POCT) kits that can be used massively (massive POCT) to battle these viral pandemics in the future. 2. The COVID-19 outbreak 2.1. COVID-19 Timeline In December 2019, a cluster of pneumonia cases were reported in Wuhan, China7. The causative virus of that disease was determined as SARS-CoV-2 (later this disease was called as COVID-19, Corona Virus Disease 2019, by the WHO) since the virus shared ~80% genome from the SARS-CoronaVirus8. On January 11, 2020, the first death caused by this virus was reported in China. This disease was highly contagious and therefore was declared by the WHO as a Public Health Emergency of International Concern (PHEIC) within a month after the first case. On March 11, WHO declared COVID-19 a pandemic disease as it started to spread across the globe. 2.2. Clinical characteristics of COVID-19 Based on current epidemiological researches, the clinical characteristics for COVID-19 appeared in 1-14 days after the infection and most patients developed symptoms within 3-7 days9. The common symptoms include fever, coughing, and body weakness. A few patients developed nasal congestion, running nose, pharyngalgia, myodynia, and diarrhea. In severe cases, by the end of the first week, the disease can develop into dyspnea and/or hypoxia. In deadly cases, the disease can quickly progress to acute respiratory distress syndrome, septic shock, coagulation disorders, and multiple organ failure9. It is noteworthy that patients with high viral loads may have low or insignificant fever during the infection. Some children and neonates did not have typical symptoms, but they presented with gastrointestinal symptoms such as vomiting and diarrhea or presented with depression or shortness of breath10. The elderly and patients with chronic underlying diseases had poor prognosis11. 2.3. Epidemiology of COVID-19 People are generally susceptible to the SARS-CoV-2 infection at all ages. The infection is transmitted by droplet (direct inhalation of droplets from the sneeze, cough, or talking of an infected person) or contact (contacting the virus deposited on the object surface, which then enters the body via the mouth, nose, eyes, or other mucous membrane12). Study showed a higher viral load in the nasal cavity than the throat, suggesting the nasal sampling is a more effective approach to detect the virus. There was no difference in the viral load between symptomatic and asymptomatic patients13, the latter of which can also transmit the disease14. Guan et al. reported that some patients were tested positive for SARS-CoV-2 in stool and urine samples also9. 3. Diagnosis of the COVID-19 As discussed in the Background, in the absence of effective therapeutic drugs or vaccines for COVID-19, it is essential to detect the disease at an early stage and immediately isolate infected patients. Currently, there are three methods in clinical practice to diagnose COVID-19, which are summarized below. 3.1. Chest CT Imaging Studies showed that chest CT images contained characteristic features for COVID-19 patients. The hallmarks of these CT images include ground glass opacities, crazy-paving pattern, consolidative opacities, septal thickening, and the reverse-halo sign15–18. These features demonstrate a highly organized pattern of pneumonia16. Unlike these features, nodules, cystic changes, bronchiectasis, pleural diffusion, and lymphadenopathy are less common18. Despite such features, the Centers for Disease Control (CDC) in the US does not currently recommend CT to diagnose COVID-19. Laboratory testing of the virus remains the reference standard, even if the CT findings are suggestive of SARSCoV-2 infections19. This is because features of the chest imaging from COVID-19 patients may overlap with other infections caused by influenza, H1N1, or SARS-CoV20,21. However, studies on the sensitivity of CT imaging over RTPCR (Reverse Transcription - Polymerase Chain Reaction, which is considered as the reference standard for laboratory testing of SARS-CoV-2, see section 3.2 below) showed that CT imaging is more sensitive and rather reliable in detecting SARSCoV-2 infections during certain stage of the COVID-19. Fang et al. studied 51 patients with COVID-19 symptoms based on their clinical manifestations and epidemiological histories22. They found that the chest CT scan was more sensitive (98%) than the RT-PCR method (71%). This study was limited by the number of subjects involved. However, another study involving more than 1000 patients reached similar conclusions23. Among 1014 patients, 59% were RT-PCR positive, from which 97% showed positive CT features. In addition, 75% of RT-PCR negative patients showed positive CT features. To further validate this, Ai et al. studied multiple RT-PCR testing and serial CT imaging in a selected group. They found 60 - 93% people who were RT-PCR negative showed initial positive CT images consistent with SARS-CoV-2 infections. From the patients in the recovery stage, 42% showed improvement in CT features before their RT-PCR results turned negative. According to these diagnostic studies, RT-PCR assays were not as sensitive and reliable as CT images in certain stages of the COVID-19. The false negative results from RT-PCR assays can be detrimental to the control of the COVID-19, especially at the beginning of the outbreak. The caveat for the CT scans is that at an early stage of infection, the lungs of a patient may not develop damaging features that can be picked up by CT scans, increasing its false negative rate. In addition, the COVID-19 CT features share similarities with other viral pneumonia, resulting in false positive detections. Nevertheless, given the rapid spreading of the COVID-19, the priority is to identify any suspicious case for patient isolation and proper treatment. In the context of emergency disease control, some false-positive cases (i.e. compromised specificity) may be acceptable. It is the false negative cases, due to the poor sensitivity of testing methods, that present a threat to public health at the beginning of an outbreak. In some cases, chest CT imaging showed positive SARSCoV-2 infection while RT-PCR testing was negative22. These findings suggest that a combination of clinical symptoms, epidemiologic history, and CT imaging of a patient may be instrumental to identify SARS-CoV-2 infections at the time when chemical detection kits are in short supply. 3.2. Nucleic acid based methods After identification of the SARS-CoV-2 as the causative virus for this pandemic, the SARS-CoV-2 genome was quickly sequenced24, from which unique sequences have been identified for COVID-19 diagnosis. Reverse transcriptase-polymerase chain reaction (RT-PCR) is a nucleic acid amplification assay that has long been used routinely for the detection of RNA viruses in clinical settings25. In RT-PCR, reverse transcriptase is first used to convert RNA to its complementary DNA, which is amplified by PCR (polymerase chain reaction). There are variants of RT-PCR methods that share the same mechanism while differing in the detection strategy. For example, real time RTPCR reads fluorescent signals during PCR amplification26 to quantify the target, whereas nested RT-PCR uses two sets of primers to avoid non-specific PCR amplifications27. The SARS-CoV-2 genes targeted for detection so far include the RdRP gene, Nucleocapsid (N) gene, E gene, Spike protein (S gene), and ORF1ab gene. Chu et al. used two different onestep real-time RT-PCR approaches to detect ORF1ab and N genes of the viral genome28. This assay showed a high dynamic range of 0.0002-20 TCID50 (50% tissue culture infective dose) per reaction and the detection limit below 10 RNA copies per reaction. Later, WHO developed a technical guidance including the protocols from different countries to aid COVID-19 diagnosis29. According to this compilation, in the US, CDC developed a real time RT-PCR diagnostic kit with detection limits as low as 4-10 RNA copies per µl. Scientists from Germany used the E gene for the first-line screening and the RdRP gene for confirmatory testing29. This method further increased sensitivity to detect as low as 5.6 RNA copies per reaction for the E gene and 3.8 RNA copies per reaction for the RdRP gene. In Hongkong, the N gene was used as the first-line screening while the ORF1b as the confirmatory testing29. In France, two RdRP genes were used for initial screening followed by the confirmatory E gene testing29. In Japan, nested RT-PCR was used,29 which significantly reduced non-specific target amplification, leading to decreased false-positive results (i.e. increased specificity). In general, the sensitivity of these assays ranges from 3.8 to 10 RNA copies per reaction, with high specificities. In the public health emergency, highly sensitive methods are desirable. Although studies have shown that RT-PCR may be less sensitive than CT imaging at certain stages of the COVID19, its specificity makes it superior to other methods to detect SARS-CoV-2. It is of critical importance to rationally choose specific diagnostic methods to battle viral outbreaks. Any negligence or compromise in the diagnosis may lead to devastating consequences. Wang et al. suggested combining RT-PCR with other methods as well as epidemiological history of patients to diagnose SARS-CoV-2 infection more credibly30. Indeed, the Chinese authority has adopted this approach to diagnose COVID-19 in Wuhan by combining RT-PCR with CT scans23. Studies also showed that the sensitivity of RT-PCR varies with the specimen types. To et al. revealed that the saliva samples were more promising to be used in RT-PCR31 while Yam et al. concluded that testing more than one specimen could significantly maximize the sensitivity of the RT-PCR testing32. These findings suggest it is rather important to apply nucleic acid based kits with optimized conditions to maximize their diagnosis potency. In particular, the finding of effective SARS-CoV-2 detection in the non-invasive saliva33 provides a convenient way to develop affordable point-of-care testing kits that can be massively used by the general public (see Sections 4 and 5 below). Table 1 lists the nucleic acid based kits used for the diagnosis of COVID-19. The sensitivity of those kits ranges from 1001000 copies/mL. 3.3. Immunoassays Immunoassay is another established diagnostic method. This method detects viral protein antigens or serum antibodies in patients who have been exposed to the SARS-CoV-2. These antibody tests are important in detecting prior infections. In the SARS-CoV-2 infection, studies have shown that the seroconversion in the patient-generally starts after a week of the first symptom34. In a study of post symptomatic patients, Amanat et al. detected high IgA and IgM immune responses35. Using recombinant viral proteins, this immunoassay could detect antibodies as early as 3 days after the development of the first symptom. Liu et al. reported that the accuracy of the ELISA for IgG and IgM antibodies was more than 80%36. The efficacy of the immunoassay also depends on the specificity of the antigens used to capture the antibodies from the patients. Between the spike (S) proteins and nucleocapsid (N) proteins, the sensitivity of the S proteins is higher for the antibody capture. Among various spike proteins, the S1 protein has shown more capabilities to bind to SARS-CoV-2 antibodies37. In a comparative study, both ELISA and colloidal gold immunochromatographic kits showed equal sensitivity with 100% specificity for the SARS-CoV-2 detection38. Several immunoassay kits are already on market for emergency detection of COVID-19 specific antibodies (see Table 2). However, the major problem of this method is that it only works for post-symptomatic patients who must have an immune response to the SARS-CoV-2. At this stage, some patients may already be critically ill. Other drawbacks of immunoassay include changes in viral load over the course of infection39, potential cross reactivity (less specific)40, and low sensitivity with respect to nucleic-acid based methods. Nevertheless, immunoassays are faster41 and cheaper than the RT-PCR methods. They can be used for rapid screening of previous SARS-CoV-2 infections. This is particularly useful in the reopening stage of the society at which people recovered from previous COVID-19 infections and therefore, immune to the virus, can safely re-engage to the society. The method also has a unique advantage of identifying individuals who have strong immune responses against the virus and therefore, can serve as potential donors for therapeutic and research purposes. 4. Ideal characteristics of diagnostic methods Diagnostic testing has become indispensable for diagnosis, prognoses, and monitoring the progress of different diseases. Efficient diagnostic testing is an important intervention for pandemic management and control. WHO has developed the ASSURED criteria as a benchmark to decide if a test efficiently addresses the needs for disease control: Affordable, Sensitive, Specific, User-friendly, Rapid and robust, Equipment-free and Deliverable to end-users42. It is ideal to have all the criteria fulfilled in a single test. In practice, however, testing methods can rarely fit all the ASSURED criteria. In pandemics for example, rapid and sensitive methods are dearly needed at the beginning of an outbreak. But many kits available require qualified laboratories and personnel for testing. In such a case, accommodation of the ASSURED principles must be taken to facilitate the testing. In a pandemic, it is always important to understand the nature of the pathogen before developing efficient diagnostic tests. Translating the tests into the point-of-care (POC)43 mode can help decision-making and improve the efficiency of the treatment. POC provides rapid and actionable information for patient management and care at the time when it is most needed. Many affordable POC testing kits such as lateral flow immunoassays44 are also appropriate for resource-limited settings in middle- or low-income countries where laboratory infrastructure is weak. One example for affordable POC testing is the Rapid HIV test45 in which HIV infection can be quickly determined at home using paper strips on specimens such as blood samples. Due to the requirements of easy usage and cheap price, they often use colloidal gold based immunoassay mechanisms. Such POC testing kits perhaps represent the best solution to fight fast transmitting pandemics. 5. Emerging techniques to detect SARS-CoV-2 Given a variety of problems associated with current clinical diagnosis for the SARS-Cov-2 (Section 3), here, we discuss some promising techniques that may address these issues. 5.1 Isothermal amplification for nucleic acid targets Although RT-PCR is a widely used method in the confirmatory screening of COVID-19 infections (see Section 3.2), it is time consuming and requires sophisticated laboratory facility and trained personal to operate46. To simplify the testing procedures, isothermal nucleic acid amplifications have been developed. These methods do not require any thermal cycler to perform the amplification and therefore, can be carried out in a simple water bath at a constant temperature of 40-65 ᵒC47. One promising isothermal nucleic acid amplification approach is Reverse Transcription Loop Mediated Isothermal Amplification (RT-LAMP). In this method, the RNA genome of SARSCoV-2 is first reverse transcribed to cDNA, which is then amplified using four to six target-specific primers. Prior to the LAMP amplification, a dumbbell shaped single-stranded DNA (ssDNA) is formed through the annealing and the strand displacing cycle on both ends of the target sequence with the help of the primers and a strand-displacing polymerase. The looped ssDNA on each end then serves as a seed for the LAMP amplification cycle48–51. As a result, the target sequence is amplified exponentially, which is detected by turbidimetry52 or fluorescence53/colorimetry48. As an example, the RNA extraction and LAMP amplification have been performed in the same tube51,54. This method has the LOD ranging from 80 to 500 SARS-Cov-2 RNA copies per milliliter, which is comparable to the RT-PCR assay. To improve the LOD, El-Tholoth et al. developed a two-stage closed tube test (named Penn-RAMP) by combining LAMP with Recombinase Polymerase Amplification55. In the Penn-RAMP, each amplification was performed at a separate compartment in a single tube followed by mixing. The method demonstrated 10 times higher sensitivity than LAMP or RT-PCR alone. In other developments, Zhang group56 and Chiu group57 integrated the LAMP with the CRISPR-based SHERLOCK (see Table 1)58 and CRISPR-Cas12 based methods, respectively, to detect the SARS-Cov-2 RNA with a detection limit as low as 10 copies/µl on a point-of-care testing (POCT) format. Some commercial COVID-19 diagnostic kits based on isothermal RT-LAMP assays are already on the market (see Table 1). Abbott ID NowTM COVID-19 is such an example. This method only requires 5 minutes to give positive results. Recently however, issues on the false negativity have been raised for the Abbott ID NowTM because of its relatively high LOD 59. This may be attributed to the compromised performance of the RdRP target60,61 used in this assay, which is found to be mutating and evolving62. Rolling circle amplification (RCA)63 is another isothermal amplification method that gives sensitive detection of nucleic acids. In this method, a segment of the target genome is circularized and amplified by a highly processive strand-displacing DNA polymerase. Wang et al. used this method to develop a highly sensitive and efficient assay for SARS-CoV64. Compared to the LAMP assay, the RCA method is simpler since it requires fewer steps and it can be performed at room temperature. The method offers high sensitivity comparable to RTPCR65 since it amplifies the target sequence by ~10,000 folds. In addition, it presents high specificity as the RCA is initiated only after the formation of a circular template upon which a specific primer is hybridized65. Therefore, RCA reduces falsepositive results often encountered in PCR-based assays. A major difficulty in this method is that it requires a circular template whose preparation is dependent on the length of a linear template and the ligation efficiency of the circularization. Inappropriate design of complementary sequences therefore results in the failure of amplifications. 5.2 Lateral flow based detection on nucleic acids and proteins The nucleic acid-based isothermal amplifications discussed above partially overcome the limitations of conventional RTPCR assays as they do not require sophisticated laboratory facilities while their turnaround time is short. However, these methods still require trained staff to operate various sample collection and processing steps. To address these problems, paperbased lateral flow assays (LFAs) have gained interest because of their low cost, easy manufacturing, and full compatibility with POCT, which allows them to be conveniently performed by anyone at home. In LFAs, both nucleic acid detection methods and immunoassays can be utilized. The device is often made of papers with immobilized capture probes. Upon binding with nucleic acid targets, the probes give a visible signal66–68. Such methods still require initial nucleic acid extraction and amplification steps, the latter of which can be accomplished by the PCR or isothermal amplifications as discussed above. On the POCT platform, all those steps are integrated in a single device. Reboud et. al.68 developed a paper-plastic lateral flow method to detect nucleic acids of malaria. They used a foldable paper in which extraction of malaria genome and LAMP amplification of target sequences were performed at separate locations. The LAMP amplified DNA was carried by capillary flow to the detection zone, giving a visible color change68. Similarly, Byers et al developed a 2D paper network to perform immunoassay for the detection of nucleic acids of SARS-CoV-2 with the POCT format69. Although nucleic acid-based lateral flow assays are sensitive, lateral flow immunoassays have gained interest in the massive surveillance of COVID-19 pandemic because of their simplicity and cheap cost. Currently, IgM/IgG rapid test kits are available for qualitative antibody test of COVID-19. Many such commercial devices have already been developed (See Table 2). One problem associated with the immunoassay based lateral flow assay is the weak signal, which results in reduced sensitivity66. Various signal enhancement strategies therefore have been proposed. A promising signal amplification strategy in lateral flow assays is the use of colloidal gold nanoparticles conjugated with the probes. Upon binding with the target, the gold nanoparticles linked to the capture probe aggregate to change the color, enhancing the signal70. Other signal amplification strategies include solvent evaporation for analyte preconcentrations, nanoparticle catalyzed nanoparticle labeled assays, and ion concentration polarization methods71. Due to the low-cost requirement of the POCT, detection in the LFA is usually achieved by visual inspection. To improve detection sensitivity, cameras in smartphones have been used72. These cameras are sensitive to subtle color changes and hence provide more effective color detection than traditional RGB sensors or the naked eye73. For improved read-out of the results and data processing, machine learning algorithm could also be used74. Smartphones can also be coupled with external adapters to integrate external biosensor platforms for more versatile POC testing75. 5.3 Other emerging methods As discussed in section 4, diagnostic tests developed so far rarely meet all the ASSURED criteria. The most important features for the SARS-CoV-2 detection are sensitivity, specificity, and efficiency (throughput and cost-effectiveness). In addition to the approaches discussed in the sections 5.1 and 5.2, other emerging methods have been developed to improve these features. To improve the sensitivity, methods with single-molecule detection capability can be used76–78. As an example, the single molecule enzyme linked immunosorbent assay (ELISA) has been developed to offer detection limit of subfemtomolar protein concentrations79. In this method, each microscopic bead decorated with specific antibodies is loaded into individual femtoliter wells. Sensing was accomplished by the ELISA on each bead, whereas the excellent concentration detection limit was achieved by a large array of such beads. To be applied in clinical setting, however, this method requires special equipment, increasing its cost. To increase the specificity, Proximity Ligation Assay (PLA) has been developed. the method utilizes two or more DNAtagged aptamers or antibodies for bindings of multiple targets80. The DNA tags on the probes are amplified only when the two different targets are in close proximity. The multiple targets ensure the specificity of the target detection. However, this method requires intact SARS-CoV-2 virus particles from which two different targets are present for positive detections. This demands stringent sample processing steps. To increase the throughput, fast sequencing such as next generation sequencing81 and DNA microarray82 can be used. In the case of COVID-19, evidences have suggested that the SARSCoV-2 is rapidly evolving while infecting people. Therefore, it is critical to rapidly identify the genome of the causative agent83. The DNA microarray has been used in high-throughput identification of mutations in SARS-CoV-284. However, for these methods, the time limiting step becomes the sample collection, which must be performed one-at-a-time. In addition, these methods involve rather advanced equipment with high cost, therefore, they may not be appropriate for the economic and rapid screening in the COVID-19 pandemic. 6. Rationales in choosing diagnostic methods in the COVID19 outbreak As stated in the introduction, diagnosis becomes one of the most important approaches to curb a viral outbreak such as COVID-19, which does not have a cure or vaccine. As shown in Figure 3, intervention such as identification of patients for isolation at the early stage before the inflection point of the viral spreading will significantly slow down the transmission of the virus. It will not only delay the time at which the peak occurs, but also reduces the magnitude of the peak population. While decreased peak magnitude directly reduces the burden on hospitals, the delay of the peak occurrence gives more time for the public to prepare well for the peak-time challenge. Both are expected to decrease the mortality rate. Such an early intervention heavily relies on the quality and quantity of the detection kits for specific viruses. Since the start of the COVID-19 outbreak, many diagnostic kits have been developed in different countries (see Tables 1-3). With the increase in the number of diagnostic tests, it is difficult for policymakers, laboratories, and other endusers to make rational decisions on the selection and use of these tests. As a result, tests have been used unnecessarily and incorrectly, with results misinterpreted. Here, based on the epidemiology of the COVID-19 and the available diagnostic kits on the market, we suggest some guidelines to rationally select kits for efficient disease control and suppression. In particular, we will discuss the relative importance of sensitivity and specificity85,86 of different assays in the fight against the COVID-19 pandemic. [Figure 3] Among all current methods, nucleic acid based kits are considered the most reliable because of their excellent sensitivity and specificity. This is not surprising since these methods target unique sequences in the viral genome for identification. Due to these advantages, it becomes a detection of choice at the beginning of a viral outbreak (Figure 4). At this stage, it is critical to identify and isolate all possible patients before the virus enters an exponential growth stage (around the inflection point, see Figure 3). Therefore, it is important to reduce the false negative results of the diagnosis. To achieve this, high sensitivity is a necessity. The PCR amplification used in various RT-PCR kits can detect as low as 100 copies/mL reaction (see Table 1), which is equivalent to 0.167 attoMolar (for a reaction volume of 100 microliters). It is noteworthy that high sensitivity is often accompanied with increased false positive results87,88. But at the beginning of a viral outbreak, some false positive level may be tolerated. Since there are not so many infected patients at the initial stage of the outbreak, the chance of cross contamination from COVID-19 patients to these false positive cases is small, even if they are isolated together (but well protected by PPE) in spacious locations such as convention centers. When the viral outbreak becomes stronger, false positive cases should be reduced (i.e. specificity increased) as much as possible due to the increasing cross contamination concerns. [Figure 4] Due to the extensive amplifications, isothermal amplification-based methods89 (see Table 1) usually have superior sensitivities albeit with increased false positive levels87,88 (see section 5.1). Therefore, at the beginning of an outbreak, isothermal amplification may be used first. However, this method usually involves many testing steps, therefore, it is more complex to run. Due to the same reason, its development and approval also take time, which makes the technique slow to be adopted at the beginning of an outbreak. With easy performance and fast approval, PCR-based kits still remain the gold standard at the beginning of a viral outbreak. Another means to reduce the false negativity in nucleic acid based testing is to perform CT scans. As discussed in Section 3.1, it can be more sensitive to diagnose COVID-19 using CT scans at certain stages of the disease. The caveat for the CT scan is its relatively low specificity (i.e. high false positive results), which may be tolerated at the initial stage of an outbreak. However, positive CT scans only diagnose patients at the later stage of their SARS-CoV-2 infections, which limits its use for early stage screening. The method is still valuable to quickly screen serious cases from mild ones. Due to limited testing kits and over-burdened clinical resources, many patients with mild symptoms have been self-isolated first. When their conditions deteriorate, it becomes important to streamline life-threatening cases as soon as these patients are sent to the hospital. Due to the fast performance and interpretation of CT scans within tens of minutes as demonstrated in China hospitals for example, these patients can be quickly identified, followed by appropriate treatment to save lives. Immunoassays work well only after the human body develops antibodies against the viruses. Therefore, these kits are not appropriate to detect infection cases at the early stage of an infection at which patients may be asymptomatic. Given that asymptomatic patients also transmit COVID-1914, it is not recommended to use immunoassays at the beginning of the pandemic. In the current COVID-19 breakout, we have often seen that during the exponential increase stage of the disease (around the inflection point, see Figure 3), there have been insufficient number of nucleic acid-based kits to test all suspicious cases. Current strategy to solve this issue is rather passive. These precious testing kits are reserved only for more serious cases. For the patients with light symptoms, they were sent home for selfisolation. The immunoassay can be used to test those patients after their symptoms lasted about one week. Since these immunoassays are cheaper, faster, and easier to perform44 with respect to nucleic acid based methods, they can be quickly and massively conducted by staff at drive-thru stations for example. This is particularly important during the society reopening stage of the COVID-19 pandemic in which the recurrence of the disease must be avoided while the lifestyle is set to be normal again (Figure 4). In this stage where the society is set to reopen, it is important to ensure that there is no recurrence of the COVID-19 breakout. To this end, one of the most important approaches is to identify people who have been previously infected with the COVID-19, and therefore immune to the SARS-Cov-2 virus. Since these people are clear of viral load, only immunoassay based detection can be used for this purpose. It is a fatal mistake for the whole society if false positive cases are high in such screening. In such cases, people who have not been exposed to the virus and therefore, vulnerable to the COVID-19, are wrongly identified as immune to the disease. This misidentification will expose them to the SARS-Cov-2 infection, which increases the chance for the recurrence of the COVID-19 in a recovering society. In the future, affordable POC testing (POCT) kits as discussed in section 4 may present a viable direction to address the bottleneck diagnosis problem caused by shortage of testing kits. These kits can be performed at home for self-isolated people with mild symptoms. If they are tested positively by the POCT kits, their conditions will be closely monitored for further medical treatments or other interventions. The inherent properties of these POCT kits (cheap, fast, and easy-to-use) afford their massive usage by the general public to fight with pandemic. We therefore name such an approach a massive POCT strategy. Given there is no such massive POCT product on the market for the COVID-19 diagnosis yet, research and development on the affordable POCT kits are dearly needed at this stage for virus detections. 7. Conclusions and Perspectives In summary, like other viruses in the Panic Zone, the SARSCoV-2 has caused unexpected damage to society. During the outbreak of the COVID-19, most studies have focused on the potential causes and epidemiology of the virus while the information on the epidemic prevention is obscure. From the data we have collected so far, it is imperative to carry out the diagnosis to isolate and treat patients at the early breakout stage of the viruses in the Panic Zone. This is especially important for the virus without a cure or vaccine. The burden of accurate and rapid diagnosis falls on the detection kits used for the SARSCoV-2 detection, which include nucleic acid based methods and immunoassays. Given the epidemiology of the COVID-19 and the features of available detection kits, it is crucial to reduce false negative results (i.e. increased sensitivity) at the expense of some false positive level (i.e. reduced specificity) during the early stage of the outbreak. It becomes important to reduce the false positivity in later stages of the outbreak, especially when the society is poised to reopen from the lock-down stage. Although nucleic acid based detection kits, RT-PCR in particular, offer best solutions so far to these requirements because of their high sensitivity and specificity, immunoassays can well supplement the detection armory due to their cheaper price, simpler operation, and faster detection time. The use of immunoassays is especially useful at the later stages of the virus outbreak when people who have been recovered from the COVID-19 are identified for their reengagement to the society. We believe a massive attack from a Panic Zone viral outbreak requires a massive defense from the whole society. The best approach to deal with this massive attack is the development of cheap, fast, and easyto-use point-of-care testing (POCT) kits that can be used in a massive fashion by the general public. In the future, intensive research and development on the so-called massive POCT kits for Panic Zone viruses therefore should be encouraged both by the government and private sectors. AUTHOR INFORMATION Corresponding Author *Hanbin Mao, PhD Department of Chemistry and Biochemistry, Kent State University, OH, USA hmao@kent.edu; (+1) 330-672 9380 850 University Esplanade, Kent, OH, 44242-0001, P.O. Box 5190 ORCID: 0000-0002-6720-9429 15. Author Contributions 16. The manuscript was written through contributions of all authors. All authors have given approval to the final version of the manuscript. 17. Funding Sources HM thanks NIH (R01 CA236350) and NSF (CBET-1904921) for financial support. 18. REFERENCES 1. Baud, D.; Qi, X.; Nielsen-Saines, K.; Favre, G. Real estimates of mortality following COVID-19 infection. Lancet Infect. Dis. (2020). DOI: 10.1016/ S1473-3099(20)30195-X 2. Cohen, D.; Carter, P. WHO and the pandemic flu “conspiracies”. BMJ 340, (2010). DOI: 10.1136/bmj.c2912 3. Wilder-Smith, A.; Chiew, C. J.; Lee, V. J. Can we contain the COVID-19 outbreak with the same measures as for SARS? Lancet Infect. Dis. (2020). DOI: 10.1016/S1473-3099(20)30129-8 4. Du, Z.; Xu, X.; Wu, Y.; Wang, L.; Cowling, B. J.; Meyers, L. A. Serial Interval of COVID-19 among Publicly Reported Confirmed Cases - Volume 26, Number 6—June 2020 - Emerging Infectious Diseases journal CDC. DOI:10.3201/eid2606.200357. 5. WHO. Managing epidemics: Key facts about major deadly diseases. (2018) https://www.who.int/publications/i/item/managing-epidemics-key-facts-ab… 6. Knobler, S.; Mahmoud, A.; Lemon, S.; Mack, A.; Sivitz, L.; Oberholtzer, K. Learning from SARS: Preparing for the Next Disease Outbreak: Workshop Summary. National Academies Press (US), (2004). DOI: 10.17226/10915 7. WHO. Novel Coronavirus (2019-nCoV) situation report-1. (January 21, 2020). https://www.who.int/emergencies/diseases/novel-coronavirus-2019/situati… 8. Guo, Y.; Cao, Q.; Hong, Z.; Tan, Y.; Chen, S.; Jin, H.; Tan, K.; Wang, D.; Yan, Y. The origin, transmission and clinical therapies on coronavirus disease 2019 (COVID-19) outbreak–an update on the status. Mil. Med. Res. 7, 1–10 (2020). DOI: 10.1186/s40779020-00240-0 9. Guan, W.; Ni, Z.; Hu, Y.; Liang, W.; Ou, C.; He, J.; Liu, L.; Shan, H.; Lei, C.; Hui, D.S.C. et al. Clinical characteristics of coronavirus disease 2019 in China. N. Engl. J. Med. (2020). DOI: 10.1056/NEJMoa2002032 10. Zeng, L. K.; Tao, X. W.; Yuan, W. H.; Wang, J.; Liu, X.; Liu, Z. S. First case of neonate infected with novel coronavirus pneumonia in China. Zhonghua Er Ke Za Zhi Chin. J. Pediatr. 58, E009 (2020). DOI: 10.3760/cma.j.issn.0578-1310.2020.0009 11. Chen, N.; Zhou, M.; Dong, X.; Qu, J.; Gong, F.; Han, Y.; Qui, Y.; Wang, J.; Liu, Y.; Wei, Y. et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. The Lancet 395, 507–513 (2020). DOI: 10.1016/S0140-6736(20)30211-7 12. Lu, C.; Liu, X.; Jia, Z. 2019-nCoV transmission through the ocular surface must not be ignored. The Lancet 395, e39 (2020). DOI: 10.1016/S0140-6736(20)30313-5 13. Zou, L.; Ruan, F.; Huang, M.; Liang, L.; Huang, H.; Hong, Z.; Yu, J.; Kang, M.; Song, Y.; Xia, J. et al. SARS-CoV-2 viral load in upper respiratory specimens of infected patients. N. Engl. J. Med. 382, 1177–1179 (2020). DOI: 10.1056/NEJMc2001737 14. Rothe, C.; Schunk, M.; Sothmann, P.; Bretzel, G.; Froeschl, G.; Wallrauch, C.; Zimmer, T.; Thiel, V.; Janke, C.; Guggemos, W. et al. Transmission of 2019-nCoV infection from an 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. asymptomatic contact in Germany. N. Engl. J. Med. 382, 970– 971 (2020). DOI: 10.1056/NEJMc2001468 Bernheim, A.; Mei, X.; Huang, M.; Yang, Y.; Fayad, Z.A.; Zhang, N.; Diao, K.; Lin, B.; Zhu, X.; Li, K. et al. Chest CT findings in coronavirus disease-19 (COVID-19): relationship to duration of infection. Radiology 200463 (2020). DOI: 10.1148/radiol.2020200463 Kong, W.; Agarwal, P. P. Chest imaging appearance of COVID19 infection. Radiol. Cardiothorac. Imaging 2, e200028 (2020). DOI: 10.1148/ryct.2020200028 Pan, F.; Ye, T.; Sun, P.; Gui, S.; Liang, B.; Li, L.; Zheng, D.; Wang, J.; Hesketh, R. L.; Yang, L. et al. Time course of lung changes on chest CT during recovery from 2019 novel coronavirus (COVID-19) pneumonia. Radiology 200370 (2020). DOI: 10.1148/radiol.2020200370 Shi, H.; Han, X.; Jiang, N.; Cao, Y.; Alwalid, O.; Gu, J.; Fan, Y.; Zheng, C. et al. Radiological findings from 81 patients with COVID-19 pneumonia in Wuhan, China: a descriptive study. Lancet Infect. Dis. (2020). DOI: 10.1016/S1473-3099(20)300864 Zu, Z. Y.; Jiang, M. D.; Xu, P. P.; Chen, W.; Ni, Q. Q.; Lu, M. G.; Zhang, L. J. et al. Coronavirus disease 2019 (COVID-19): a perspective from China. Radiology 200490 (2020). DOI: 10.1148/radiol.2020200490 Hosseiny, M.; Kooraki, S.; Gholamrezanezhad, A.; Reddy, S.; Myers, L. Radiology perspective of coronavirus disease 2019 (COVID-19): lessons from severe acute respiratory syndrome and Middle East respiratory syndrome. Am. J. Roentgenol. 1–5 (2020). DOI: 10.2214/AJR.20.22969 American Roentgen Ray Society. Novel coronavirus (COVID19) imaging features overlap with SARS and MERS: COVID19’s imaging features are variable and nonspecific, but the imaging findings reported thus far do show. ScienceDaily https://www.sciencedaily.com/releases/2020/02/200228142018.htm. Fang, Y.; Zhang, H.; Xie, J.; Lin, M.; Ying L.; Pang, P.; Ji, W. Sensitivity of chest CT for COVID-19: comparison to RT-PCR. Radiology 200432 (2020). DOI: 10.1148/radiol.2020200432 Ai, T.; Yang, Z.; Hou, H.; Zhan, C.; Chen, C.; Lv, W.; Tao, Q.; Sun, Z.; Xia, L. Correlation of chest CT and RT-PCR testing in coronavirus disease 2019 (COVID-19) in China: a report of 1014 cases. Radiology 200642 (2020). DOI: 10.1148/radiol.2020200642 Heymann, D. L.; Shindo, N. COVID-19: what is next for public health? The Lancet 395, 542–545 (2020). DOI: 10.1016/S01406736(20)30374-3 Bustin, S. A.; Mueller, R. Real-time reverse transcription PCR (qRT-PCR) and its potential use in clinical diagnosis. Clin. Sci. 109, 365–379 (2005). DOI: 10.1042/CS20050086 Bustin, S. A. Quantification of mRNA using real-time reverse transcription PCR (RT-PCR): trends and problems. J. Mol. Endocrinol. 29, 23–39 (2002). DOI: 10.1677/jme.0.0290023 Chen, H.; Guo, J.; Wang, C.; Luo, F.; Yu, X.; Zhang, W.; Li, J.; Zhao, D.; Xu, D.; Gong, Q. et al. Clinical characteristics and intrauterine vertical transmission potential of COVID-19 infection in nine pregnant women: a retrospective review of medical records. The Lancet 395, 809–815 (2020). DOI: 10.1016/S01406736(20)30360-3 Chu, D. K. W.; Pan, Y.; Cheng, S. M. S.; Hui, K. P. Y.; Krishnan, P.; Liu, Y.; Ng, D. Y. M; Wan, C. K. C.; Yang, P.; Wang, Q. et al. Molecular diagnosis of a novel coronavirus (2019-nCoV) causing an outbreak of pneumonia. Clin. Chem. 66, 549–555 (2020). DOI: 10.1093/clinchem/hvaa029 WHO. Summary of available protocols to detect SARS-CoV-2. (2020) https://www.who.int/docs/default-source/coronaviruse/whoinhouseassays.p… Wang, C.; Horby, P. W.; Hayden, F. G.; Gao, G. F. A novel coronavirus outbreak of global health concern. The Lancet 395, 470– 473 (2020). DOI: 10.1016/S0140-6736(20)30185-9 To, K. K.; Tsang, O. T.; Yip, C. C.; Chan, K.; Wu, T.; Chan, J. M. C.; Leung, W.; Chik, T. S.; Choi, C. Y.; Kandamby, D. H. et al. Consistent detection of 2019 novel coronavirus in saliva. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. (2020). DOI: 10.1093/cid/ciaa149 32. Yam, W. C.; Chan, K. H.; Poon, L. L. M.; Yuen, K. Y.; Seto, W. H.; Peiris, J. S. M. Evaluation of reverse transcription-PCR assays for rapid diagnosis of severe acute respiratory syndrome associated with a novel coronavirus. J. Clin. Microbiol. 41, 4521–4524 (2003). DOI: 10.1128/jcm.41.10.4521-4524.2003 33. Rutgers Today. New Rutgers Saliva Test for Coronavirus Gets FDA Approval. (April 13, 2020) https://www.rutgers.edu/news/new-rutgers-saliva-test-coronavirus-gets-f… 34. Gao, Y.; Yuan, Y.; Li, T. T.; Wang, W. X.; Li, Y. X.; Li, A.; Han, F. M. Evaluation the auxiliary diagnosis value of antibodies assays for detection of novel coronavirus (SARS-Cov-2) causing an outbreak of pneumonia (COVID-19). medRxiv (2020). DOI: 10.1101/2020.03.26.20042044 35. Amanat, F.; Stadbauer, D.; Strohmeier, S.; Nguyen, T. H. O.; Chromikova, V.; McMahon, M.; Jiang, K.; Arunkumar, G. A.; Jurczyszak, D.; Polanco, J. et al. A serological assay to detect SARS-CoV-2 seroconversion in humans. Nat Med (2020). DOI: 10.1038/s41591-020-0913-5 36. Liu, W.; Liu, L.; Kou, G.; Zheng, Y.; Ding, Y.; Ni, W.; Wang, Q.; Tan, Li,; Wu, W.; Tang, S.; et al. Evaluation of Nucleocapsid and Spike Protein-based ELISAs for detecting antibodies against SARS-CoV-2. J. Clin. Microbiol. (2020). DOI: 10.1128/JCM.00461-20 37. Okba, N. M.; Muller, M. A.; Li, W.; Wang, C.; GeurtsvanKessel, C. H.; Corman, V. M.; Lamers, M. M.; Sikkema, R. S.; Bruin, E.; Chandler, F. D. et al. SARS-CoV-2 specific antibody responses in COVID-19 patients. Emerg Infect Dis (2020). DOI: 10.3201/eid2607.200841 38. Xiang, J.; Yan, M.; Li, H.; Liu, T.; Lin, C.; Huang, S; Shen, C. Evaluation of Enzyme-Linked Immunoassay and Colloidal GoldImmunochromatographic Assay Kit for Detection of Novel Coronavirus (SARS-Cov-2) Causing an Outbreak of Pneumonia (COVID-19). medRxiv (2020). DOI: 10.1101/2020.02.27.20028787 39. Kassanjee, R.; Pilcher, C. D.; Busch, M. P.; Murphy, G.; Facente, S. N.; Keating, S. M.; Mckinney, E.; Marson, K.; Price, M. A.; Martin, J. N. et al. Viral load criteria and threshold optimization to improve HIV incidence assay characteristics-a CEPHIA analysis. AIDS Lond. Engl. 30, 2361 (2016). DOI: 10.1097/QAD.0000000000001209 40. Tate, J.; Ward, G. Interferences in immunoassay. Clin. Biochem. Rev. 25, 105 (2004). 41. Sheridan, C. Fast, portable tests come online to curb coronavirus pandemic. Nat. Biotechnol. (2020) DOI:10.1038/d41587-02000010-2. 42. Peeling, R. W., Holmes, K. K., Mabey, D.; Ronald, A. Rapid tests for sexually transmitted infections (STIs): the way forward. Sex. Transm. Infect. 82, v1–v6 (2006). DOI: 10.1136/sti.2006.024265 43. US FDA. Coronavirus (COVID-19) Update: FDA Issues New Policy to Help Expedite Availability of Diagnostics. FDA (2020) https://www.fda.gov/news-events/press-announcements/coronavirus-covid-1… 44. Hesterberg, L. K.; Crosby, M. A. An overview of rapid immunoassays. Lab. Med. 27, 41–46 (1996). DOI: 10.1093/labmed/27.1.41 45. Branson, B. M. Point-of-Care Rapid Tests for HIV Antibodies. Lab. Med. 27, 288–295 (2003). DOI: 10.1046/j.1439-0477.2003. 03049.x 46. Bustin, S. A.; Nolan, T. Pitfalls of quantitative real-time reversetranscription polymerase chain reaction. J. Biomol. Tech. JBT 15, 155 (2004). 47. Silva, S. J. R.; Pardee, K.; Pena, L. Loop-Mediated Isothermal Amplification (LAMP) for the Diagnosis of Zika Virus: A Review. Viruses 12, 19 (2020). DOI: 10.3390/v12010019 48. Zhang, Y.; Odiwuor, N.; Xiong, J.; Sun, L.; Nyaruaba, R. O.; Wei, H.; Tanner, N. A. Rapid Molecular Detection of SARS- 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. CoV-2 (COVID-19) Virus RNA Using Colorimetric LAMP. medRxiv (2020). DOI: 10.1101/2020.02.26.20028373 Yu, L.; Wu, S.; Hao, X.; Dong, X.; Mao, L.; Pelechano, V.; Chen, W.; Yin, X. Rapid detection of COVID-19 coronavirus using a reverse transcriptional loop-mediated isothermal amplification (RT-LAMP) diagnostic platform. Clin. Chem. (2020). DOI: 10.1093/clinchem/hvaa102 Park, G.; Ku, K.; Baek, S.; Kim, S.; Kim, S.; Kim, B.; Maeng, J. Development of Reverse Transcription Loop-mediated Isothermal Amplification (RT-LAMP) Assays Targeting SARS-CoV-2. J. Mol. Diagn. (2020). DOI: 10.1016/j.jmoldx.2020.03.006 Huang, W. E.; Lim, B.; Hsu, C.; Xiong, D.; Wu, W.; Yu, Y.; Jia, H.; Wang, Y.; Zeng, Y.; Ji, M. et al. RT-LAMP for rapid diagnosis of coronavirus SARS-CoV-2. Microb. Biotechnol. (2020). DOI: 10.1111/1751-7915.13586 Yuan, X.; Wang, Y.; Meng, K.; Zhang, Y.; Xu, H.; Ai, W. LAMP real-time turbidity detection for fowl adenovirus. BMC Vet. Res. 15, 256 (2019). DOI: 10.1186/s12917-019-2015-5 Zheney, M.; Kaziyev, Z.; Kassenova, G.; Zhao, L.; Liu, W.; Liang, L.; Li, G. Real-time fluorescence loop-mediated isothermal amplification assay for direct detection of egg drop syndrome virus. BMC Vet. Res. 14, 49 (2018). DOI: 10.1186/s12917-0181364-9 Jiang, M.; Pan, W.; Aratehfar, A.; Fang, W.; Ling, L.; Fang, H.; Daneshnia, F.; Yu, J.; Liao, W.; Pei, H. et al. Development and validation of a rapid single-step reverse transcriptase loop-mediated isothermal amplification (RT-LAMP) system potentially to be used for reliable and high-throughput screening of COVID-19. medRxiv (2020). DOI: 10.1101/2020.03.15.20036376 El-Tholoth, M.; Bau, H. H.; Song, J. A single and two-stage, closed-tube, molecular test for the 2019 Novel Coronavirus (COVID-19) at home, clinic, and points of entry. (2020). DOI: 10.26434/chemrxiv.11860137.v1 Zhang, F.; Abudayyeh, O. O.; Jonathan, S. G. A protocol for detection of COVID-19 using CRISPR diagnostics. (2020). https://broad.io/sherlockprotocol. Broughton, J. P.; Deng, X.; Yu, G.; Fasching, C. L.; Singh, J.; Streithorst, J.; Granados, A.; Sotomayor-Gonzalex, A.; Zorn, K.; Gopez, A. et al. Rapid detection of 2019 novel coronavirus SARS-CoV-2 using a CRISPR-based DETECTR lateral flow assay. MedRxiv (2020). DOI: 10.1101/2020.03.06.20032334 Kellner, M. J.; Koob, J. G.; Gootenberg, J. S.; Abudayyeh, O. O.; Zhang, F. SHERLOCK: nucleic acid detection with CRISPR nucleases. Nat. Protoc. 14, 2986–3012 (2019). DOI: 10.1038/s41596-019-0210-2 Basu, A.; Zinger, T.; Inglima, K.; Woo, K.; Atie, O.; Yurasits, L.; See, B.; Rosenfeld, M. E. A. Performance of the rapid Nucleic Acid Amplification by Abbott ID NOW COVID-19 in nasopharyngeal swabs transported in viral media and dry nasal swabs, in a New York City academic institution. bioRxiv (2020). DOI: 10.1101/2020.05.11.089896 Loeffelholz, M. J.; Alland, D.; Butler-Wu, S. M.; Pandey, U.; Perno, C. F.; Nava, A.; Carroll, K. C.; Mostafa, H.; Davies, E.; McEwan, A.; Rakeman, J. L. et al. Multicenter Evaluation of the Cepheid Xpert Xpress SARS-CoV-2 Test. J. Clin. Microbiol. (2020). DOI: 10.1128/JCM.00926-20 Visseaux, B.; Hingrat, Q. L.; Collin, G.; Bouzid, D.; Lebourgeois, S.; Pluart, D. L.; Deconinck, L.; Lescure, F-X.; Leucet, J-C.; Bouadma, D.et al. Evaluation of the QIAstat-Dx Respiratory SARS-CoV-2 Panel, the first rapid multiplex PCR commercial assay for SARS-CoV-2 detection. J. Clin. Microbiol. (2020). DOI: 10.1128/JCM.00630-20 Pachetti, M.; Marini, B.; Benedetti, F.; Giudici, F.; Mauro, E.; Storici, P.; Masciovecchio, C.; Angeletti, S.; Ciccozzi, M.; Gallo, R. C. et al. Emerging SARS-CoV-2 mutation hot spots include a novel RNA-dependent-RNA polymerase variant. J. Transl. Med. 18, 1–9 (2020). DOI: 10.1186/s12967-020-02344-6 Liu, D.; Daubendiek, S. L.; Zillman, M. A.; Ryan, K; Kool, E. T. Rolling circle DNA synthesis: small circular oligonucleotides as efficient templates for DNA polymerases. J. Am. Chem. Soc. 118, 1587–1594 (1996). DOI: 10.1021/ja952786k 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. Wang, B.; Potter, S. J.; Lin, Y.; Cunningham, A. L.; Dwyer, D. E.; Su, Y.; Ma, X.; Hou, Y.; Saksena, N. K. Rapid and sensitive detection of severe acute respiratory syndrome coronavirus by rolling circle amplification. J. Clin. Microbiol. 43, 2339–2344 (2005). DOI: 10.1128/JCM.43.5.2339-2344.2005 Zhao, W.; Ali, M. M.; Brook, M. A.; Li, Y. Rolling circle amplification: applications in nanotechnology and biodetection with functional nucleic acids. Angew. Chem. Int. Ed. 47, 6330–6337 (2008). DOI: 10.1002/anie.200705982. Koczula, K. M.; Gallotta, A. Lateral flow assays. Essays Biochem. 60, 111–120 (2016). DOI: 10.1042/EBC20150012 Zhang, D.; Broyles D.; Hunt, E. A.; Dikici, E.; Daunert, S.; Deo, S. K. A paper-based platform for detection of viral RNA. Analyst 142, 815–823 (2017). DOI: 10.1039/c6an02452a Reboud, J.; Xu, G.; Garrett, A.; Adriko, M.; Yang, Z.; Tukahebwa, E. M.; Rowell, C.; Cooper, J. M. Paper-based microfluidics for DNA diagnostics of malaria in low resource underserved rural communities. Proc. Natl. Acad. Sci. 116, 4834–4842 (2019). DOI: 10.1073/pnas.1812296116 Byers, K. M.; Bird, A. R.; Cho, H. D.; Linnes, J. C. Fully Dried Two-Dimensional Paper Network for Enzymatically Enhanced Detection of Nucleic Acid Amplicons. ACS Omega 5, 4673–4681 (2020). DOI: 10.1021/acsomega.0c00115 Tanaka, R.; Yuhi, T.; Nagatani, N.; Endo, T.; Kerman, K.; Takamura, Y.; Tamiya, E. A novel enhancement assay for immunochromatographic test strips using gold nanoparticles. Anal. Bioanal. Chem. 385, 1414–1420 (2006). DOI: 10.1007/s00216-0060549-4 Giri, B. A perspective on the sensitivity of paper-analytical devices for bioanalysis. J. Appl. Bionalysis 2, 6 (2016). DOI: 10.17145/jab.16.002 Martinez, A. W.; Phillips, S. T.; Carrilho, E.; Thomas 3rd, S. W.; Sindi, H.; Whitesides, G. M. Simple telemedicine for developing regions: camera phones and paper-based microfluidic devices for real-time, off-site diagnosis. Anal. Chem. 80, 3699–3707 (2008). DOI: DOI: 10.1021/ac800112r Priye, A.; Bird, S. W.; Light, Y. K.; Ball, C. S.; Negrete, O. A.; Meagher, R. J. A smartphone-based diagnostic platform for rapid detection of Zika, chikungunya, and dengue viruses. Sci. Rep. 7, 1–11 (2017). DOI: 10.1038/srep44778 Berg, B.; Cortazar, B.; Tseng, D.; Ozkan, H.; Feng, S.; Wei, Q.; Chan, R. Y.; Burbano, J.; Farooqui, Q.; Lewinski, M. et al. Cellphone-based hand-held microplate reader for point-of-care testing of enzyme-linked immunosorbent assays. ACS Nano 9, 7857– 7866 (2015). DOI: 10.1021/acsnano.5b03203 Laksanasopin, T.; Guo, T. W.; Nayak, S.; Sridhara, A. A.; Xie, S.; Olowookere, O. O.; Cadinu, P.; Meng, F.; Chee, N. H.; Kim, J. et al. A smartphone dongle for diagnosis of infectious diseases at the point of care. Sci. Transl. Med. 7 (2015). DOI: 10.1126/scitranslmed.aaa0056 Koirala, D.; Yu, Z.; Dhakal, S.; Mao, H. Detection of Single Nucleotide Polymorphism Using Tension-Dependent Stochastic Behavior of a Single-Molecule Template. J. Am. Chem. Soc. 133, 9988–9991 (2011). DOI: 10.1021/ja201976r Koirala, D.; Shrestha, P.; Emura, T.; Hidaka, K.; Mandal, S.; Endo, M.; Sugiyama, H.; Mao, H. Single-Molecule Mechanochemical Sensing Using DNA Origami Nanostructures. Angew. Chem. Int. Ed. 53, 8137–8141 (2014). DOI: 10.1002/anie.201404043 Kaur, A.; Sapkota, K.; Dhakal, S. Multiplexed Nucleic Acid Sensing with Single-Molecule FRET. ACS Sens. 4, 623–633 (2019). DOI: 10.1021/acssensors.8b01373 Rissin, D. M.; Kan, C. W.; Campbell, T. G.; Howes, S. C.; Fournier, D. R.; Song, L.; Piech, T.; Patel, P. P.; Chang, L.; Rivnak, A. J. et al. Single-molecule enzyme-linked immunosorbent assay detects serum proteins at subfemtomolar concentrations. Nat. Biotechnol. 28, 595 (2010). DOI: 10.1038/nbt.1641 Greenwood, C.; Ruff, D.; Kirvell, S.; Johnson, G.; Dhillon, H. S.; Bustin, S. A. Proximity assays for sensitive quantification of proteins. Biomol. Detect. Quantif. 4, 10–16 (2015). DOI: 10.1016/j.bdq.2015.04.002 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. 91. 92. 93. 94. 95. 96. 97. 98. Metzker, M. L. Sequencing technologies—the next generation. Nat. Rev. Genet. 11, 31–46 (2010). DOI: 10.1038/nrg2626 Khan, M. J.; Trabuco, A. C.; Alfonso, H. L.; Figueiredo, M. L.; Batista, W. C.; Badra, S. J.; Figueiredo, L. T.; Lavrador, M. A.; Aquino, V. J. DNA microarray platform for detection and surveillance of viruses transmitted by small mammals and arthropods. PLoS Negl. Trop. Dis. 10, (2016). DOI: 10.1371/journal.pntd.0005017 Shen, Z.; Xiao, Y.; kang, L.; Ma, W.; Shi, L.; Zhang, L.; Zhou, Z.; Yang, J.; Zhong, J.; Yang, D. et al. Genomic Diversity of Severe Acute Respiratory Syndrome–Coronavirus 2 in Patients With Coronavirus Disease 2019. Clin Infect Dis (2020). DOI: 10.1093/cid/ciaa203 Long, W.; Xiao, H.; Gu, X.; Zhang, Q.; Yang, H.; Zhao, G.; Liu, J. A universal microarray for detection of SARS coronavirus. J. Virol. Methods 121, 57–63 (2004). DOI: 10.1016/j.jviromet.2004.06.016 Nouvellet, P.; Garske, T.; Mills, H. L.; Nedjati-Gilani, G.; Hinsley, W.; Blake, I. M.; Kerkhove, M. D. V.; Cori, A.; Dorigatti, I.; Jombart, T. et al. The role of rapid diagnostics in managing Ebola epidemics. Nature 528, S109 (2015). DOI: 10.1038/nature16041 Fegan, D. F. Evaluation of diagnostic tests: the epidemiological approach. (Retrieved on June 3, 2020) http://www.fao.org/3/X4946E/x4946e0b.htm. Lalkhen, A. G.; McCluskey, A. Clinical tests: sensitivity and specificity. Contin. Educ. Anaesth. Crit. Care Pain 8, 221–223 (2008). DOI: 10.1093/bjaceaccp/mkn041 McNamara, L. A. & Martin, S. W. 1 - Principles of Epidemiology and Public Health. in Principles and Practice of Pediatric Infectious Diseases (Fifth Edition). Elsevier. (2018). DOI:10.1016/B978-0-323-40181-4.00001-3. Udugama, B.; Kadhiresan, P.; Kozlowski, H. N.; Malekjahani, A.; Osborne, M.; Li, V. Y. C.; Chen, H.; Mubareka, S.; Gubbay, J. B.; Chan, W. C. W. Diagnosing COVID-19: The Disease and Tools for Detection. ACS Nano (2020). DOI: 10.1021/acsnano.0c02624 Dietz, K. The estimation of the basic reproduction number for infectious diseases. Stat. Methods Med. Res. 2, 23–41 (1993). DOI: 10.1177/096228029300200103 Lipsitch, M.; Moxon, E. R. Virulence and transmissibility of pathogens: what is the relationship? Trends Microbiol. 5, 31–37 (1997). DOI: 10.1016/S0966-842X(97)81772-6 Guerra, F. M.; Bolotin, S.; Lim, G.; Heffernan, J.; Deeks, S. L.; Li, Y.; Crowcroft, N. S. The basic reproduction number (R0) of measles: a systematic review. Lancet Infect. Dis. 17, e420–e428 (2017). DOI: 10.1016/S1473-3099(17)30307-9 Kretzschmar, M.; Teunis, P. F.; Pebody, R. G. Incidence and reproduction numbers of pertussis: estimates from serological and social contact data in five European countries. PLoS Med. 7, (2010). DOI: 10.1371/journal.pmed.1000291 Australian Government Department of Health. Mumps Laboratory Case Definition (LCD). (2015) https://www1.health.gov.au/internet/main/publishing.nsf/Content/cda-phl… Ferguson, N. M.; Cummings, D. A. T.; Fraser, C.; Cajka, J. C.; Cooley, P. C.; Burke, D. S. Strategies for mitigating an influenza pandemic. Nature 442, 448–452 (2006). DOI: 10.1038/nature04795 Fraser, C.; Donnelly, C. A.; Cauchemez, S.; Hanage, W. P.; Van Kerkhove, M. D.; Hollingsworth, T. D.; Griffin, J.; Baggaley, R. F.; Jenkins, H. E.; Lyons, E. J. et al. Pandemic potential of a strain of influenza A (H1N1): early findings. science 324, 1557–1561 (2009). DOI: 10.1126/science.1176062 Kucharski, A. J.; Althaus, C. The role of superspreading in Middle East respiratory syndrome coronavirus (MERS-CoV) transmission. Euro Surveill. 20 (2015). DOI: 10.2807/15607917.es2015.20.25.21167 Khan, A.; Naveed, M.; Dur-e-Ahmad, M.; Imran, M. Estimating the basic reproductive ratio for the Ebola outbreak in Liberia and Sierra Leone. Infect. Dis. Poverty 4, 13 (2015). DOI: 10.1186/s40249-015-0043-3 99. Gani, R.; Leach, S. Transmission potential of smallpox in contemporary populations. Nature 414, 748–751 (2001). DOI: 10.1038/414748a 100. Chen, J. Pathogenicity and transmissibility of 2019-nCoV—a quick overview and comparison with other emerging viruses. Microbes Infect. (2020). DOI: 10.1016/j.micinf.2020.01.004 101. Roser, M.; Ritchie, H.; Ortiz-Ospina, E. Coronavirus Disease (COVID-19) – Statistics and Research. Our World Data (2020). https://ourworldindata.org/coronavirus FIGURE 1. Viruses with high transmission rates (R0) are less fatal. R0 is the reproduction rate of a virus, which measures its transmissibility77. Solid curve represents an inverse fitting between the mortality rate and the R0, which has been proposed as the trade-off principle between the virulence and transmissibility of virus91. The inverse function fits well except for the two viruses in the Death Zone (blue), which is defined to have a rather high mortality rate. The Panic Zone contains viruses with medium levels of transmission and mortality rates. The data used here are taken from references92–100. 11 FIGURE 2. Critical importance of the early detection in the COVID-19 outbreak. COVID-19 daily tests are shown for 5 countries with similar medical resources and age distributions. Inset shows the mortality rates (percentage of the death cases among all confirmed COVID19 cases) as of 06/07/2020 vs the number of the early detections per thousand population performed during 03/04/2020 - 03/26/2020. The early detection data for each country101 are taken from different periods (marked by stretches) to reflect the timing of the outbreak in Asia, Europe, and North America (~2 weeks apart). The inset data are linearly fit (r=-0.94), which indicates a negative correlation between the early detection and the mortality rate. 12 FIGURE 3. Intervention of the COVID-19 outbreak. The intervention at an early stage (before the inflection point, which is the point where the half width of a Gaussian peak is equivalent to the sigma of the Gaussian) of a viral breakout is the key to slow down the transmission of the virus. It not only decreases the peak value of newly confirmed daily cases, but also saves the time to increase the hospital capacity, each of which reduces the overall mortality rate. 13 FIGURE 4. Schematic diagram of the relative usage of nucleic acid vs antibody detection methods (left y axis) and the relative importance of sensitivity vs specificity (right y axis) in the detection of SARS-CoV-2 virus during the COVID-19 pandemic. At the initial breakout stage, nucleic acid-based detection methods are important because of their high sensitivity with low false negativity. It allows quick isolation of infected individuals for timely treatment and disease containment. At the healing stage when a society is set to reopen, antibody detection methods are important to identify individuals immune to the disease due to previous COVID-19 infections. Highly specific immunoassays with low false positivity are desirable to correctly identify these individuals who are safe to return to the society. Interestingly, the same pattern can be used to describe individual cases of COVID-19 infections. 14 Table 1: Kits based on nucleic acid detection Authorization Manufacturer Mechanism Target LOD Time - 1drop Inc RT-PCR RdRP, E 8 cp/rxn ~2 h 05/11/2020* 1drop Inc. RT-PCR E, RdRp 200 cp/mL - 3B BlackBio Biotech India Ltd RT-PCR RdRP, E, N - - 3D Medicines A*Star Tan Tock Seng Hospital of Singapore RT-PCR - - - RT-PCR - - - 3/27/2020* Abbott Diagnostics Scarborough, Inc. RT-LAMP RdRP 5-13 min 3/18/2020* Abbott Molecular RT-PCR RdRP, N Abbott Molecular Inc. RT-PCR RdRp, N 125 GE/mL 100-200 cp/mL 100 cp/mL ƛ 03/2020 # € 05/11/2020* 04/22/2020* 02/2020 # 05/13/2020* ** - Altona Diagnostics GmbH RT-PCR N, S 0.1 PFU/mL Anatolia Geneworks RT-PCR - - - Applied DNA Sciences, Inc. RT-PCR RNaseP 5 cp/rxn - ARUP Laboratories RT-PCR - - Assurance Scientific Laboratories RT-PCR 37 cp/rxn - - Atila BioSystems, Inc RT-PCR N1, N2, RNaseP N, ORF1ab 4 cp/µL - AUSDiagnostics RT-PCR - - - Avellino Lab USA, Inc. RT-PCR RNase P (RP) 55 cp/µL 4/8/2020* Becton, Dickinson & Company RT-PCR N, RP 40 GE/mL 1-2 days - 4/2/2020* Becton, Dickinson and Company RT-PCR N (N1, N2) 40 GE/mL 90 min 05/15/2020* 4/10/2020* 03/2020# 3/25/2020* 3/26/2020*, 3/2/2020#, 1/2020$ 05/21/2020* 3/23/2020* 05/01/2020* 05/06/2020* 05/01/2020* ** 2/4/2020* 3/20/2020* 03/2020 4/3/2020* 03/2020# 05/22/2020* 4/8/2020* **** 3/19/2020* - Beijing Applied RT-PCR ORF1ab, N, E 1000 cp/mL BGI Genomics Co. Ltd. RT-PCR ORF1ab 100 cp/mL - BGI Wuhan Biotech Co., Ltd RT-PCR ORF1ab 100 cp/mL 90 min BioCore Co., Ltd. RT-PCR N, RdRp 500 cp/mL - BioFire Defense, LLC RT-PCR ORF1ab, ORF8 330 cp/mL 50 min BioFire Diagnostics, LLC Multiplex RT-PCR - - - N, RdRp, E 300 GE/mL - BioMérieux SA RT-PCR Bioneer RT-PCR - - - Bio-Rad Laboratories, Inc Endpoint RT-PCR N1, N2 625 cp/mL - BioReference Laboratories RT-PCR - - - Centers for Disease Control and Prevention's (CDC) RT-PCR N1, N2. RP 4-10 cp/µL - Cepheid RT-PCR N2, E 250 cp/mL 45 min CerTest BioTec RT-PCR - - - Co-Diagnostics, Inc RT-PCR RdRP 600 cp/spl - Credo Diagnostics Biomedical RT-PCR - - - Daan Gene Co., Ltd., Sun Yat-sen University RT-PCR ORF1ab, N 500 cp/mL 110 min Dba SpectronRx RT-PCR N, E 5 cp/rxn - DiaCarta, Inc RT-PCR E, N, ORF1ab 100 cp/mL - Diagnostic Solutions Laboratory RT-PCR - - - DiaSorin Molecular LLC RT-PCR ORF1ab, S 500 cp/mL 1-1.5 h Diatherix Eurofins RT-PCR - - - 15 N1, N2, RNaseP 5 cp/mL - RT-PCR ORF1ab, N, E 300 cp/mL - Sequencing - - - GeneMatric, Inc. RT-PCR RdRp, N 50 cp/rxn - Genetic Signatures RT-PCR - - - GenMark Diagnostics, Inc. RT-PCR, electrowetting and sensing - 10^5 cp/mL 2h Genomica/PharmMar Group RT-PCR - - - 05/15/2020* Flugent Therapeutics, LLC RT-PCR 04/17/2020* Fosun Pharma USA Inc. Fulgent Genetics/MedScan laboratory ** 05/14/2020* 03/2020 # 3/19/2020* 03/2020 # 4/16/2020* GenoSensor, LLC RT-PCR E, N, ORF1ab 1 cp/µL - 4/6/2020* Gnomegen LLC RT-PCR N (N1, N2) 8 GE/rxn - 05/08/2020* Gnomegen LLC RT-PCR N1, N2 10 cp/rxn - N1, N2, RNaseP 4.8 cp/µL - 06/01/2020* Gravity Diagnostics, LLC RT-PCR 05/14/2020* Holigic, Inc. Transcription Mediated Amplification 3/16/2020* Hologic, Inc. RT-PCR - InBios International, Inc RT-PCR E, N, ORF1ab Integrated DNA technologies/Danaher RT-PCR - 0.01 TCID50/mL 10-2 TCID50/m L 12.5 GE/rxn - 4/7/2020* * 4/1/2020* #, ORF1ab - Ipsum Diagnostics, LLC RT-PCR N, RP 8.5 cp/µL - € JN Medsys RT-PCR - - - ψ Kogene Biotech RT-PCR - - - KorvaLabs, Inc RT-PCR N1, N2, RP 200 cp/rxn - LabGenomics Co., Ltd. 4/16/2020* 04/29/2020* RT-PCR RdRp, E 20 GE/mL - Laboratory Corporation of America (LabCorp) RT-PCR Rnase P (RP), N 6.25 cp/µL - LGC, Biosearch Technologies RT-PCR - - - Luminex Corporation RT-PCR 75 GE/uL - 3/27/2020* Luminex Molecular Diagnostics, Inc. RT-PCR ORF1ab, N ORF1ab, N gene, E 1.5 cp/µL 4h 4/15/2020* Maccura Biotechnology (USA) LLC RT-PCR E, N, ORF1ab 1 cp/µL 2h 3/23/2020* Mesa Biotech Inc. RT-PCR and colorimetry N 100 cp/rxn 30 min 3/30/2020* NeuMoDx Molecular RT-PCR - - - 3/30/2020* 3/20/2020*, 2/2020#, ƛ 3/26/2020 NeuMoDx Molecular, Inc RT-PCR Nsp2, N 150 cp/mL Novacyt/Primerdesign RT-PCR - - - OPTI Medical Systems, Inc. RT-PCR N1, N2 0.7 cp/µL - 3/16/2020* * 4/3/2020* 05/06/2020* 04/18/2020* 02/2020# 3/24/2020* 06/04/2020* 3/20/2020* *** OSANG Healthcare RT-PCR RdRp, N, E 0.5 cp/µL - OsangHealthCare RT-PCR - - - PerkinElmer, Inc. RT-PCR ORF1ab, N 20 cp/mL - Phosphorus Diagnostics LLC RT-qPCR N1, N2, RNaseP 5 cp/µL - Primerdesign Ltd. RT-PCR - 0.33 cp/µL - - - - - 500 cp/mL - Promedical Lateral Flow Immunoassay RT-PCR 3/30/2020* QIAGEN GmbH 3/17/2020* Quest Diagnostics Infectious Disease, Inc. RT-PCR N1 and N3 136 cp/mL - Quidel Corporation RT-PCR pp1ab 0.8 cp/µL 75 min 3/23/2020*, 3/2020# 16 05/18/2020* 03/2020$ 04/29/2020* 3/12/2020* 05/04/2020* 4/3/3030* Quidel Corporation RT-PCR Pp1ab Rendu Biotechnology RT-LAMP - Endpoint RT-PCR N1 Roche Molecular Systems, Inc. RT-PCR E - 3 hrs SANSURE Bio-tech Co., Ltd RT-PCR ORF1ab, N 200 cp/mL 90 min Sansure BioTech Inc. RT-PCR ORF1ab, N 200 cp/mL - ScienCell Research Laboratories RT-PCR N (N1, N2) 500 cp/µL - ORF1ab, RdRp, E 0.5 cp/µL - 1 cp/µL - - - - - RT-PCR 04/27/2020* SEASUN BIOMATERIALS RT-PCR 05/21/2020* Seasun Biomaterials, Inc. RT-LAMP 02/2020 , See Gene RT-PCR ORF1ab, N ORF1ab, RNaseP - Seegene, Inc. RT-PCR E, RdRp, N - Shanghai Bio Germ RT-PCR ORF1ab, N - Shanghai GeneoDx Biotech Co., Ltd RT-PCR ORF1ab, N 05/06/2020* 2/2020 , , # $$ $$$ 05/21/2020* 03/2020 # 3/13/2020* 03/2020# 04/20/2020* 03/2020 # 2/29/2020* - - Rheonix, Inc. SD Biosensor, Inc 04/21/2020* - 625 GE/mL 04/23/2020* # $$ 1.28x104 Genome eq/mL - Shanghai ZJ Bio-tech Co., Ltd. RT-PCR ORF1ab, N, E 4167 cp/mL 1000 cp/mL 500 cp/mL 1000 cp/mL 1-4.5 cp/µL - - 90 min 90 min 90 min Sherlock BioSciences, Inc. CRISPR SolGent RT-PCR ORF1ab, N, RNaseP - SolGent Co., Ltd. RT-PCR N, ORF1 200 cp/mL - Systaaq Diagnostic Products RT-PCR - - - - Thermo Fisher Scientific, Inc. RT-PCR S, N 10 GE/rxn 4h TIB MolBiol Synthesalabor RT-PCR E - - Trax Management Services Inc. RT-PCR RNaseP 50 cp/mL - Ustar RT-PCR ORF1ab, N - 90 min Vision Medicals PCR (Clinical Sequencing Assay) - - - RT-PCR RP 25 cp/rxn - RT-PCR ORF1ab, N - 75 min Wadsworth Center, New York State Department of Public Health's (CDC) Wuhan Easydiagnosis 17 Table 2: Kits based on antibodies detection Authorization 04/26/2020* Manufacturer Abbott Laboratories, Inc. Mechanism Chemiluminescent microparticle immunoassay Immunoassay Target LOD Time IgG - - IgG IgM - - *** Assure Tech *** Autobio Diagnostics Immunoassay - - - Autobio Diagnostics Co. Ltd. Lateral Flow Immunoassay IgG IgM - 50 min *** Beijing Decombio Biotechnology Immunoassay IgG IgM - - *** Beijing Diagreat Biotechnologies Beijing Kewei Clinical Diagnostic Reagent Immunoassay IgG IgM - - Immunoassay IgG IgM - - 04/24/2020* *** *** Beijing O&D Biotech Colloidal gold - - - *** *** Beroni Group BioMedomics Immunoassay Immunoassay IgG IgM IgG IgM - - - BiOSCiENCE Immunoassay IgM IgG - 30 min *** BTNX Immunoassay IgG IgM - - 4/1/2020* Cellex Immunoassay IgG IgM - - ChemBio Diagnostic System Immunoassay IgG IgM - - Chembio Diagnostic System, Inc Immunoassay IgG IgM - - Core Technology Immunoassay IgG IgM - - DiaSorin Inc. Chemiluminescent Immunoassay IgG - - *** Diazyme Laboratories Immunoassay IgG IgM - - *** Eachy Biopharmaceuticals Immunoassay IgG IgM - - Eagle Bioscience Immunoassay IgG, IgM - - EUROIMMUN US Inc. ELISA IgG - 2.5 h Guangdong Hecin Immunoassay IgM - - *** Guangzhou Wondfo Immunoassay - - 15 min *** Hangzhou AllTest Biotech Immunoassay IgG IgM - - *** Hangzhou Biotest Biotech Immunoassay IgG IgM - - Hangzhou Biotest Biotech Co., Ltd. Lateral Flow Immunoassay IgG IgM - 30 min *** 4/14/2020* *** 04/24/2020* 05/04/2020* - 06/04/2020* *** Hangzhou Clongene Biotech Immunoassay IgG IgM - - *** *** Hangzhou Testsealabs Biotechnology Healgen Scientific Immunoassay Immunoassay IgG IgM IgG IgM - - 05/29/2020* Healgen Scientific LLC Lateral Flow Immunoassay IgG IgM - 10 min - INNOVITA (Tangshan) Immunoassay IgG IgM - 15 min *** Jiangsu Macro & Micro-Test Med-Tech Colloidal gold IgG IgM - - *** Lifeassay Diagnostics Immunoassay IgG IgM - - *** Medical Systems Biotechnology Immunoassay IgG IgM - - Mount Sinai Laboratory Immunoassay IgG NA - Nanjing Liming Bio-Products Immunoassay IgG IgM - - Nanjing Vazyme Immunoassay IgM, IgG - 10 min *** NanoResearch Immunoassay IgG IgM - - *** Nantong Diagnos Biotechnology Colloidal gold - - - *** Nirmidas Biotech Immunoassay IgG IgM - - Ortho Clinical Diagnostics, Inc Lateral Flow Immunoassay IgG IgM - 10-15 min PCL Immunoassay IgG IgM - - 4/15/2020* *** - 4/14/2020* *** 18 *** PharmaTech Immunoassay IgG IgM - - 05/08/2020* Quidel Corporation Lateral Flow Immunoassay Nucleocapsid protein - 15 min 06/02/2020* Roche Diagnostics Immunoassay IL-6 - 18 min 05/02/2020* Roche Diagnostics Immunoassay - - 18 min *** SD Biosensor Immunoassay IgG IgM - - *** Shenzhen Landwind Medical Immunoassay IgG IgM - - Siemens Healthcare Diagnostics Inc. Chemiluminescent Immunoassay Total Antibodies - 10 min 05/29/2020* Snibe Diagnostics Immunoassay IgG IgM - - *** Telepoint Medical Services Immunoassay IgG IgM - - *** Tianjin Beroni Biotechnology Immunoassay IgG IgM - - IgG IgM - - - - - - - 15 min 02/2020 # 06/04/2020* 04/30/2020* *** Wadsworth Center, New York, State Department of health Xiamen innoDx Bio-tech Chemiluminescence immunoassay Microsphere Immunoassay Immunoassay Zuhai Livzon Diagnostics Colloidal gold Vibrant America Clinical Labs Total Antibodies IgG IgM IgG IgM 19 Table 3: Kits based on ‘not identified’ mechanism Authorization Manufacturer Mechanism Target LOD Time - Biological Technologies Co., LTD - - - - - (Chongqing) Bio-tech, Co., Ltd - - Health Technology Co., Ltd - Bio-tech Co., Ltd - Medical Technology Co., Ltd - Biotechnologies (Hangzhou) Ltd - - - - - Biomedicine Co., Ltd. - - - - *US EUA Authorized, **US EUA Planned, ***US Notified FDA under section IV.D, ****US EUA Submitted, #European Union Conformity Marked, $The National Medical Product Administration Authorized China, $$Korea Ministry of Food and Drug Safety, $$$Philippines Food and Drug Administration, €Singapore Health Sciences Authority, personal authorization for clinical use, ƛEUA India, ψKorea Centers for Disease Control and the Korea Food and Drug Administration -Data Not Available References for kits in table 1-3: https://www.fda.gov/medical-devices/emergency-situations-medical-device…. http://ph.china-embassy.org/eng/sgdt/t1760281.htm. https://www.modernhealthcare.com/safety/coronavirus-test-tracker-commer…. https://www.finddx.org/covid-19/pipeline/. 20
Photograph
Photograph