Next Article in Journal
Mosquito Control Priorities in Florida—Survey Results from Florida Mosquito Control Districts
Next Article in Special Issue
Methodological Development of a Multi-Readout Assay for the Assessment of Antiviral Drugs against SARS-CoV-2
Previous Article in Journal
Antigen-Presenting Cells in the Airways: Moderating Asymptomatic Bacterial Carriage
Previous Article in Special Issue
Presence of SARS-CoV-2 in a Cornea Transplant
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Sewage Systems Surveillance for SARS-CoV-2: Identification of Knowledge Gaps, Emerging Threats, and Future Research Needs

1
Environmental and Occupational Hazards Control Research Center, Shahid Beheshti University of Medical Sciences, Tehran 35511, Iran
2
Department of Environmental Health Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran 35511, Iran
3
Department of Environmental Science and Technology, University of Maryland, College Park, MD 20740, USA
4
Environmental Service, Suez Water North America, Paramus, NJ 07652, USA
5
Department of Medical Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 35511, Iran
6
Water Purification Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran 19168, Iran
*
Authors to whom correspondence should be addressed.
Pathogens 2021, 10(8), 946; https://doi.org/10.3390/pathogens10080946
Submission received: 24 May 2021 / Revised: 4 July 2021 / Accepted: 22 July 2021 / Published: 28 July 2021
(This article belongs to the Collection SARS-CoV Infections)

Abstract

:
The etiological agent for novel coronavirus (COVID-19, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), not only affects the human respiratory system, but also the gastrointestinal tract resulting in gastrointestinal manifestations. The high rate of asymptomatic infected individuals has challenged the estimation of infection spread based on patients’ surveillance, and thus alternative approaches such as wastewater-based epidemiology (WBE) have been proposed. Accordingly, the number of publications on this topic has increased substantially. The present systematic review thus aimed at providing state-of-the-knowledge on the occurrence and existing methods for sampling procedures, detection/quantification of SARS-CoV-2 in sewage samples, as well as anticipating challenges and providing future research direction to improve the current scientific knowledge. Articles were collected from three scientific databases. Only studies reporting measurements of virus in stool, urine, and wastewater samples were included. Results showed that improving the scientific community’s understanding in these avenues is essential if we are to develop appropriate policy and management tools to address this pandemic pointing particularly towards WBE as a new paradigm in public health. It was also evident that standardized protocols are needed to ensure reproducibility and comparability of outcomes. Areas that require the most improvements are sampling procedures, concentration/enrichment, detection, and quantification of virus in wastewater, as well as positive controls. Results also showed that selecting the most accurate population estimation method for WBE studies is still a challenge. While the number of people infected in an area could be approximately estimated based on quantities of virus found in wastewater, these estimates should be cross-checked by other sources of information to draw a more comprehensive conclusion. Finally, wastewater surveillance can be useful as an early warning tool, a management tool, and/or a way for investigating vaccination efficacy and spread of new variants.

1. Introduction

During the years 2003–2004, the outbreak of severe acute respiratory syndrome coronavirus (SARS-CoV) afflicted people throughout the world and was poised to be the next pandemic. The virus, however, was responsible for just over 8400 cases with fewer than 1000 deaths in 29 countries and studies concerning infection-control measures were instrumental in severely curtailing the potential for a pandemic [1,2]. However, an outbreak of pneumonia of unknown etiology first reported in Wuhan (Hubei province, China) in the late 2019, has startled the world and caused the globe to undergo unprecedented change in a short space of time. Metagenomics sequencing of broncho alveolar lavage samples shed light on the association of this outbreak with a novel coronavirus (nCoV) [2]. The “novel coronavirus-infected pneumonia” was officially designated as SARS-CoV-2 after being provisionally named as 2019-nCoV [3,4]. On 31 January 2020, the World Health Organization (WHO) declared the outbreak to constitute a public-health emergency of international concern when the disease was reported in 114 countries and thence named this disease as Corona Virus Disease 2019 (COVID-19) on 11 February 2020 [5,6]. The status of the outbreak was upgraded from epidemic to pandemic on 11 March. By 2 July 2021, 182,653,642 confirmed cases, including 3,955,835 deaths, was officially announced all over the world, with distressing consequences on human health and economy, particularly in the United States, India, Brazil, and Russia, among others [7].
Current evidence for COVID-19 is primarily confined to flu-like symptoms with most affected patients suffering from fever, dry cough, and difficulty in breathing but some become seriously ill and even die [8,9]. The available epidemiological evidence strongly suggests that COVID-19 is primarily transmitted through respiratory droplets and contact routes [10]. Recent tracing of SARS-CoV-2 genetic material—viral RNA—in stool and urine of hospitalized COVID-19 patients [11,12,13], however, shed light on the rapidly evolving picture of this new disease. This pattern of spread is highly suggestive of virus dissemination by aqueous matrices, and the circulation of virus was speculated to be occurred from malfunctioning sewage works (sewer networks and wastewater treatment plants (WWTP)) in the community [14,15]. Sanitary facilities have been hypothesized as possible amplifiers and disseminators of the virus [16]; the pathogenic agent, most likely acquired from infected patients, may have released in their excreta (i.e., saliva, sputum, and feces) and then possibly becoming waterborne. Nonetheless, neither WHO nor the US Centers for Disease Control and Prevention (CDC) still don’t consider COVID-19 as waterborne and finding clues to support this claim throughout literature has not reached a clear conclusion.
The scientific community has recently witnessed an interest in shedding of virus into feces as well as the presence and persistence of SARS-CoV-2 in health care and municipal effluents and thus the potential of sewage to spread COVID-19 [15,17]. Outbreak investigations strongly suggest that infections in the hospital and the community may occur through this route. The presence of SARS-CoV-2 (either viable virus or associated viral debris) in feces indeed raises the potential for sewage analysis in order to inform epidemiological monitoring of COVID-19 as a complementary approach for current clinical surveillance techniques by providing information on the prevalence and spread of disease in a population [18]. The method as a relatively new approach in public health, based on raw wastewater fingerprinting to obtain qualitative and quantitative data within a given wastewater catchment, not only provides an early warning sign for disease outbreaks but also acts as a smart way of imposing preemptive quarantine [19].
There is wide-ranging research into sewage monitoring of SARS-CoV-2 often with two overarching objectives: (1) to detect the presence/prevalence of virus in a population; and (2) to assess infection risk to the public and sewage workers/operators from untreated/partially treated contaminated sewage and effluent as well as air surrounding sewage work facilities. As an affordable, convenient, and practical program, monitoring viral RNA in sewage can also be used to complement the current clinical surveillance by providing information on the prevalence and spread of disease in a population. The present review sets out to outline an overview of the current knowledge addressing the occurrence of SARS-CoV-2 in the feces of affected individuals and its dissemination in the public sewage system. The goal is to recognize key concepts, research gaps, and types of evidence within the following topics: (i) wastewater/effluent sampling strategy [20] methods for the detection and quantification of SARS-CoV-2 RNA in wastewater, particularly concerning an evolving field of wastewater-based epidemiology (WBE); and (ii) the potential ecological risks (secondary transmission risk) to the wider environment. We tried to highlight the areas where further research is needed and made further recommendations by interpreting current state of knowledge dealing with these topics.

2. Methods

A systematic review was carried out to synthesize our current knowledge of the presence of SARS-CoV-2 both in stool of confirmed patients and wastewater. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement was used to report the results of our review.
Our search was conducted across the Scopus, PubMed, and Web of Science databases for relevant literature with no restriction for publication date or language. The search included the keywords: wastewater OR sewage OR effluent OR fec* OR faec* OR diarrh* OR meconium* OR melena OR stool OR urine AND SARS-CoV-2 OR 2019 nCoV OR COVID-19. To give a comprehensive view of the topic, the literature search was extended to preprints using the medRxiv server (https://www.medrxiv.org/ (accessed on 28 December 2020)), with the same criteria. Searches were conducted on 28 December 2020. MeSH terms were applied when using the PubMed database in order to employ the thesaurus of medical vocabulary. Article inclusion required confirmation of infection via stool sampling, and/or confirmation of viral wastewater contamination.
A total of 3384 potentially appropriate articles was identified via the first review phase, decreasing to 2507 upon deduplication using the Mendeley Reference Manager Software (Figure 1). The screening phase was undertaken through an assessment of article title and abstract, resulting in 526 articles going forward for eligibility assessment. Full texts were independently and concomitantly analyzed by two researchers (FA and NM), again using developed inclusion/exclusion eligibility criteria. Discrepancies between pairs of reviewers were resolved through consensus or a third adjudicator. The primary inclusion criterion was the presence of RNA or infectious (viable) SARS-CoV-2 in feces or urine of a confirmed patient and/or wastewater. Overall, upon completion of the review process, 138 articles were included for data extraction and analysis (Table 1 and Table 2). Altogether, this systematic review aims at outlining the used protocols for virus sampling, sample preparation (i.e., concentration/enrichment procedures), and analysis (i.e., target gene regions) in feces, untreated wastewater, and effluents tested in the peer-reviewed literature in order to identify actual challenges, allowing us to suggest potentially appropriate procedures and solutions. In addition, the potential risks for wastewater operators, population size normalization, and practical use of wastewater surveillance are also presented with elaborated conclusions.

3. Results and Discussion

From the identified studies, 88 reported on the presence of SARS-CoV-2 in feces and urine, and the remaining 48 were concerned with presence of the virus in wastewater. The PRISMA flow diagram, presented in Figure 1, demonstrates the results of our review. Figure 2 summarizes the findings of our study. The obtained results are presented and critically discussed based on the collected information in five subsections. The first two subsections contain information regarding the presence of SARS-CoV-2 in feces and wastewater (Section 3.1 and Section 3.2). Detection and quantification of SARS-CoV-2 in wastewater coupled with their accompanied challenges, potential risks, and application of results have been outlined in the third, Section 3.3. The last two, Section 3.4 and Section 3.5, present knowledge gaps and concluding remarks derived from analyzing the outcomes.

3.1. Gastrointestinal Manifestations and Presence of SARS-CoV-2 in Feces

The fecal shedding of SARS-CoV-2 from not only symptomatic but also asymptomatic, pre-symptomatic, and post-symptomatic COVID-19 patients has been reported [11]. Nevertheless, few studies have specifically dealt with gastrointestinal malfunctions in infected individuals and the results are divergent. To go further in our understanding, it was previously reported that respiratory manifestations precede/tail gastrointestinal symptoms in human coronavirus strains, including SARS-CoV and MERS-CoV [151]. The most frequently addressed clinical manifestations, as can be seen in Table 1, were reported in 21 studies and are categorized as abdominal pain, diarrhea, and vomiting, with diarrhea as the most frequently reported gastrointestinal malfunction. A number of studies in patients with COVID-19 underplayed the relative importance of gastrointestinal symptoms and, notably, viral shedding in stool [46,152].
In an attempt to provide an explanation, Zang et al. [153] pointed to the inactivation of SARS-CoV-2 by simulated human colonic fluid. Inactivation of infectious viral particles by digestive enzymes has also been reported previously [154]. It has been universally demonstrated that the lysis of the viral envelope in enveloped viruses such as coronaviruses results in the loss of functional receptors required for infection of susceptible cells. Nonetheless, evidence regarding the effects of digestive enzymes on the survivability of enveloped viruses in gastrointestinal tract is not conclusive. Migration to throat tissues and passage through the stomach is conceivable given that SARS-CoV-2 can survive the extreme pH of the gastric tissues and infection could then expand into the intestines where Angiotensin-converting enzyme 2 (ACE2) levels are high [155]. ACE2 is a cellular receptor that is well expressed in the respiratory tract and interacts with the spike protein to facilitate entry of virus into the host cell [5,156]. The upper oesophagus and stratified epithelial cells as well as absorptive enterocytes of the ileum and the colon have also showed highly positive for ACE2 receptor, enumerating the gastrointestinal system as a potential transmission route for SARS-CoV-2 infection [5,157]. Some studies have also reported that human coronaviruses replicate in the gastrointestinal tract [157,158]. Wang et al. reported the occurrence of an increasing rate of diarrhea in confirmed COVID-19 patients [159]. A recent review on the pathogenesis, epidemiology, prevention, and management of diarrhea during COVID-19 infection also reported an incidence rate of diarrhea ranging from 2 to 50% of cases, with an overall percentage of diarrhea onset of 10.4% which may precede or tail respiratory symptoms [160]. In support of these findings, about 2 to 10% of hospitalized COVID-19 patients have been reported to have diarrhea [34,159,161,162,163,164]. Meanwhile, in an analysis of data from the Hong Kong cohort of patients with COVID-19, Cheung et al. [152] reported that 25.4% of affected individuals had diarrhea among other gastrointestinal symptoms. The same research in a meta-analysis of 60 studies, comprising 4243 patients, demonstrated that the prevalence of all gastrointestinal manifestations was 17.6%. These observations are in line with reported SARS-CoV pattern in 2003, where 16–73% of patients had chronic diarrhea during the first week of the illness while presenting pulmonary symptoms [165,166]. Thus, although some discrepancies between studies exist, they are consistent with those of official data from WHO pointing to that between 2 and 27% of COVID-19 infected individuals have diarrhea [167]. More studies of comparable quality are needed to conclude the rate of diarrhea in COVID-19 cases with severe, mild, or no symptoms.
In total, 88 studies dealing with the shedding of SARS-CoV-2 in stool were retrieved (Table 1). Although data on the subject is still in its infancy, there is a growing evidence for the presence of SARS-CoV-2 genetic material in human feces [168], detected mainly through reverse transcription polymerase chain reaction (RT-PCR) or reverse transcription quantitative real-time PCR (RT-qPCR) following RNA isolation [169]. Nevertheless, it is worth mentioning that there are small differences in protocols for virus isolation from feces between laboratories. Positive isolation of SARS-CoV-2 RNA from hospitalized patient’s stool samples or their rectal swabs was reported in 48% (31/65) [46], 53% (39/73) [80], and 67% (6/9) [64] of cases, while the virus RNA has also been detected in human feces of at least 82% (55/66) [79] of tested patients in a recent study in China. Nonetheless, few studies have investigated the fecal shedding in cases with mild symptoms or even asymptomatic individuals [170]. Preliminary estimates of asymptomatic infected individuals range from 17.9 to 30.8% [156]. Also, the persistence of fecal shedding in affected patients and its association with respiratory shedding largely remains elusive. It is somewhat surprising that virus RNA has been detected in stool samples from patients with COVID-19 even in samples collected after respiratory samples tested negative [51,80], demonstrating that shedding of virus RNA into feces lasts long after its clearance from pulmonary tract. Although it is unclear how long the fecal shedding continues, Jiang et al. have reported that SARS-CoV-2 was detectable in stool samples of an asymptomatic infected case for as many as 42 days [171]. Surprisingly, viral detection of SARS-CoV-2 in nasopharyngeal samples of infected case in this study was negative. Several studies on stool samples, in agreement with this finding, reported detection of SARS-CoV RNA from the fifth day, demonstrating a peak in viral titer at the 11th day which lasted till the 30th day [172]. It is encouraging to compare these records with that found by Leung [151] who reported that gastrointestinal symptoms were followed by fecal shedding of SARS-CoV in patients with SARS up to 73 days. Even though gastrointestinal manifestations in affected cases and particularly fecal shedding of virus may not be clinically significant, there is an underlying assumption that such an information may reflect the virus circulation among human populations at this stage. Further, the dynamics and duration of fecal shedding have important implications for normalization of data, which is discussed in Section 3.5. The main application of wastewater monitoring for SARS-CoV-2 is to infer infection rates in the population in a specific sewage catchment area. The measured viral load at the influent of each WWTP reflects a cumulative amount shed by affected individuals during the relevant window—approximately 22 days with an interquartile range between 17 and 31 days, as pointed out by Zheng et al. [62]. Thus, it can be deduced that the measured concentration accounts for ill individuals—those that have and have not been clinically identified—infected several days or weeks prior to the sampling event.
The discrepancy between studies on the presence of SARS-CoV-2 viral RNA may however be assigned to the limited resources in early reports for detection which were only provided to those patients with severe respiratory symptoms. Temporal dynamics and the persistence of viral shedding in stool as provided by ViralZone [173] has been summarized in Figure 3. This figure depicts data on the onset and duration that affected individuals can discharge the virus in their stool. It is evident that gastrointestinal infections precede the development of fever and respiratory symptoms in some infected individuals, particularly in patients admitted into intensive care.
Indeed, there is only limited and, of course, heterogeneous evidence for the amount and dynamics of SARS-CoV-2 shedding in human feces. It was reported that the concentration of enteric viral particles per gram of stool during diarrhea falls in the range of 1010-1012 [174]. Also, the literature already includes several reports of SARS viral loads, pointing towards 106.1 gene copies per gram (gc/g) of feces and 101.3 gc/mL of urine [175]. For instance, Woelfel and coworkers [176] in their study on a cluster of nine cases highlighted that SARS-CoV-2 RNA loads could be as high as 107 gc/g of stool following the onset of manifestations, which decreased to 103 copies/g three weeks after the emergence of symptoms. Likewise, 1.7 × 106–4.1 × 107 gc/mL appears in the Han et al. [57] report, while another recent study conducted in Europe reported 6.3 × 106–1.26 × 108 gc/g of stool [27]. In the context of these studies, Ahmed et al. [15] tried to translate this value to a log-uniform distribution from 2.56 to 7.67 as noticed during the periods of heaviest shedding among mild cases of COVID-19. These results provide further support to estimate the number of infected cases within the community via wastewater-based epidemiology, as a new paradigm in public-health assessment. In anal swabs of hospitalized cases of coronavirus disease, viral loads of 105 gc/swab for SARS-CoV-2 has also been reported [176].
Despite the extensive reports regarding fecal shedding of SARS-CoV-2 RNA from COVID-19 patients, none of the reported studies to date have provided information on the viability of infectious virions, capable of initiating disease in a susceptible individual, in stool samples possibly. This could partly be assigned to the difficulties in maintaining human viruses in vitro as well as the requirements for trained staff and specialized equipment. Nevertheless, the literature on this topic has only just started to increase. These studies typically use human or animal cell lines (e.g., Vero E6 cells, based on monkey kidney cell lines) to analyze qPCR positive samples through cell culture infectivity assay, integrated cell culture-qPCR (ICC-qPCR), and EMA/PMA-RT-qPCR which are then followed by genotyping [177]. Two studies found cultivable SARS-CoV-2 in fecal samples from hospitalized patients, reinforcing the scientific speculations on the serious consequences for transmission through exposure to feces [34,66]. Although Wang et al. [34] pointed toward detection of viable SARS-CoV-2 in stool samples, it is worth noting here that no evidence of viability was provided. However, in contrast to these findings, a more recent study has been unable to demonstrate the isolation of culturable virus from feces despite high viral RNA copies [64]. Although firm evidence is lacking, there are few case reports of SARS-CoV-2 shedding in urine [178,179,180]. There does, nevertheless, appear to be very little evidence to directly support such studies which thus remain largely speculative at this stage (which could be attributed to the plausible secondary contamination) until further research has been done.
In closing, although the literature on the occurrence and survival of virus in feces shows heterogeneous results, fecal–oral transmission of SARS-CoV-2 requires comprehensive and more nuanced interpretation. One of the issues that emerges from clearance of live SARS-CoV-2 virus or even viral RNA by feces of affected patients is potential enteric transmission to health professionals and those working with human excreta in the hospitals whose rate of infection is extremely high. The plausible effect of public toilets in the spread of disease, which may be places at risk of infection, particularly for patients having gastrointestinal symptoms, such as diarrhea, also remains purely speculative. In this essay, local authorities are recommended to consider disinfectant addition as the preferred emergency sanitation treatment and try to put forward the relative merits of the 0.5% or 2% chlorine solutions to disinfect human excreta, as has been suggested during the Ebola outbreak [181].

3.2. Occurrence of SARS-CoV-2 in Wastewater

Both viable SARS-CoV-2 and nonviable virus (and associate viral debris such as RNA fragments, mRNA, or capsid subunits) may found their way into sewage trough bodily excreta, including hand washing, saliva, sputum, vomit, feces, and possibly urine which are subsequently disposed of in wastewater. During the last seven months, considerable efforts have been devoted to detect SARS-Coronavirus-2 in sewage in several countries, particularly in high- and upper-middle-income communities. Details of these reports are summarized in Table 2. The imbalance between the number of studies in developed countries and those on the broad spectrum of under-developed and resource-limited communities clearly indicates that much work has yet to be accomplished. Gathering such comparative data would be the best way to ensure adoption of WBE worldwide for monitoring pandemics—not just for controlling Covid-19, but also for future epidemics.
Most of the studies on virus surveillance in wastewater have focused on the presence of viral fragments of SARS-CoV-2 RNA and little has been documented on viral titers or the potential infection risk in wastewater. The presence of SARS-CoV-2 in wastewater was first described by Medema et al. [17] in the Netherlands, six days before the first cases were officially announced. Nevertheless, Mallapaty et al. [182] provided a shred of evidence to support the presence of virus in wastewater at Amersfoort, the Netherlands, days before the first cases were reported. In parallel, Lodder and de Roda Husman [112] pointed towards the first report of SARS-CoV-2 detection at Amsterdam Airport Schiphol, the Netherlands, via human wastewater surveillance. They worked on 24-h 10 L composite samples exploiting quantitative RT-PCR methodology and provided the first evidence of virus presence in the wastewater four days after the first cases of COVID-19 were identified in the Netherlands [112]. This evidence was corroborated by two other studies in untreated wastewater in Italy [126] and Turkey [107]. The occurrence of SARS-CoV-2 RNA in the wastewater and sewage sludge was also reported even in a low-prevalence area in Spain [109], while another study reported the presence of corresponding virus in raw and partially-treated sewage in New York, USA [183].
Wu et al. [110], through monitoring a wastewater treatment plant in Massachusetts, USA, reported the concentration of SARS-CoV-2 in wastewater samples as ~104 copies/100 mL using a polyethylene glycol (PEG 8000) concentration method and CDC N1, N2 and N3 assays as has been used by Medema et al. [17]. One of the first reports of SARS-CoV-2 detection in public sewage was achieved in Brisbane, Australia [15]. This study reported on the positive detection of viral genome in untreated wastewater during COVID-19 epidemic peak. The study applied RT-qPCR to obtain quantification cycle (Cq) values and was followed up with WBE approach to estimate the prevalence of affected people in the catchment. The viral titers were estimated as 12 and 1.9 gc/100 mL of untreated wastewater based on the obtained Cq values in two studied samples (37.5 and 39, respectively). Although these Cq values are comparable to those that the virus has in the feces, there is a clear separation from clinical pharyngeal swab specimens [35]. This could be either attributed to the lower rate of viral shedding in stool or problems with the way the original data was collected (as discussed in detail in Section 3.1).
To summarize, infectious SARS-CoV-2 has not been detected in untreated or treated sewage [184]. However, RNA components of SARS-CoV-2 have been detected in untreated sewage and sludge in a number of countries and municipalities, with RNA signals, generally starting around the same time cases were first reported (February and March 2020) and increasing as the number of confirmed cases increased. Detection of viral genetic materials (RNA) alone, however, does not necessarily confirm the presence of viable infectious virus or a risk of infection. In general, RNA fragments are much more persistent in the sewage matrices than infectious virus, and there is no clear correlation between RNA load in sewage samples and infectiousness. However, tracing SARS-CoV-2 RNA ‘signature’ is at least a ‘smoking gun’, allowing local authorities to judge the possibility of infection hazard. We cannot, of course, exclude that exposure levels could be related to yet unrecognized or unmeasured confounders. Quantifying transmission risk, indeed, necessitates knowledge of the minimum infectious dose of the virus, which for SARS-CoV-2 has not been yet determined, as well as data on the amount of virus that individuals are exposed, too. An analysis of the data available in the literature already confirms the occurrence of SARS-CoV-2 RNA in untreated wastewater with maximum concentrations over 106 copies per liter. To further expand the investigations of the occurrence of SARS-CoV-2 RNA in sewage systems, several studies dealing with viral titers in treated effluent were retrieved (Table 2). Three of those reports specifically addressing detection of SARS-CoV-2 RNA in effluent announced viral loads up to 104 gc/100 mL [185]. The study in Paris detected the SARS-CoV-2 RNA in treated wastewater as well, with concentrations of up to approximately 105 copies per liter [106], forcing the scientific speculations on the serious consequences for the sake of public health.
Thus far, published data on the presence of SARS-CoV-2 in wastewater mainly come from municipal sewage, with limited data on virus presence in hospital effluents. SARS-CoV-2 is enveloped and thus less stable in the environment compared to non-enveloped human enteric viruses with known waterborne transmission (such as adenoviruses, norovirus, rotavirus, and hepatitis A virus) In support of these findings, viral fragments of SARS-CoV RNA were traced in 100% of untreated wastewater and 30% of disinfected effluent samples collected from a Chinese hospital during the SARS outbreak in 2004 [186].

3.3. Detection and Quantification of SARS-CoV-2 in Wastewater

Detecting and enumerating of SARS-CoV-2 in the sewage requires a number of steps (Figure 4), each undoubtedly facing a number of challenges, many of which are often shared by existing reports [15,17,110]. Some of these challenges seriously affect the resultant outcome, as we will discuss in the following sections along with techniques developed to overcome them.

3.3.1. Challenges Involved in Obtaining Representative Samples

Given the heterogeneous nature of wastewater, this type of research is fraught with a statistically representative sampling of sewage. It is just as important to take a representative sample as it is to analyze the sample correctly. The question of what constitutes a representative sample has been addressed previously [120,187]. The limits of data variability are potentially influenced by many parameters, including the sampling procedure. Perhaps the greatest question remains whether the grab or composite sampling method should be performed. The grab sampling approach is likely to be utilized when either the level of viruses or the content of solids is relatively high, as is the case for raw wastewater. A composite sample, on the other hand, is a sample unit that is created by combining or pooling multiple sample units. Thus, if the study objective is to get a good estimate of the average concentration of infectious agents in a large volume of environmental matrix—like public raw sewage—individual replicates from a sampling point could be physically combined to create one sample unit that is sent for analysis. The advantage of this method is that it obtains values that are expected to more closely follow a normal distribution and hence meets the assumptions of standard statistical tests. The appropriate use of composite samples can indeed be a cost-effective way of producing precise and accurate estimates of average levels, among others [188]. Thus, the challenge is to effectively sample and encompass the variability in viral RNA, particularly in quantitative studies set out with the aim of sewage surveillance to estimate the infected people through the accurate detection and quantification of viral load by wastewater-based epidemiology. Except for individual variability in viral shedding coupled with viral persistence in the sewer network, this approach encounters huge challenges concerning variations in wastewater flow rates. Although it might not be a serious issue in large sewer systems, higher domestic water usage at peak times (e.g., mornings and evenings) [189] could result in reduced concentrations of viral load at these times because of dilution effects.
It is further important to bear in mind the possible bias in viral loads due to regional rainy days and melting snow again with a clear result of sewage dilution in combined sewers. Daily mean temperature and amount of rain therefore not only may affect the number of positive SARS-CoV-2 cases in the specific catchment area, but also gene copy numbers. In accordance with these hypothesis, Wurtz et al. pointed to two episodes of rain (September 19 to 22 and November 7 to 8) in their sewage monitoring for SARS-CoV-2. Surprisingly, they opined that the quantity of viruses did not fall during the first episode, but possibly with the second [190].
Retention time in the sewer network (i.e., the time a sample takes to travel in the sewer pipe) indeed may have a profound effect on the structure of the sample. Thus, monitoring across the network may assert the merit of priority or even better representation of the situation. A composite sampling approach, and not just one snapshot in time, thus will help to alleviate the problem and to obtain near real-time concentration of viruses detected in wastewater. Dealing with the limitations such as intermittent shedding of enteric pathogens by humans and the effects of dilution, Moore swab as a classic environmental surveillance tool, whereby a gauze pad is suspended in flowing wastewater to trap microorganisms (Figure 5), may regain appreciation in sewage fingerprinting [191]. The method is advantaged by its simplicity and affordability and is well suited to resource-limited communities. The use of Moore’s technique has been re-surfaced in recent conference proceedings describing environmental surveillance programs for enteric pathogens, such as Salmonella and Vibrio cholera [191]. Most recently, Rafiee et al. reported that the use of Moore swabs outperforms composite and grab sampling methods for SARS-CoV-2 monitoring in wastewater, providing promising results as to be the best sampling protocol to adopt when planning a sewage monitoring campaign particularly under WBE [192].
The sampling process is compounded by the virus’s degradation in a relatively short space of time. Thus, sample transport and storage need to be carefully controlled—usually keeping in a refrigerated container (or icebox) at a temperature ˂10 °C. The elapsed time allowed between sample collection and analysis ≤24 h. If sample analysis within the 24-h time limit is not possible, long-term storage of samples should be operated at −80 °C. Viruses are also in the throes of inactivation by the presence of chemicals, e.g., detergents and cleaning chemicals which are used copiously during a pandemic.

3.3.2. Challenges Involved in Concentration/Enrichment of SARS-CoV-2 in Wastewater

Given the potential low concentrations of virus titer in environmental matrices, sensitive detection of viral particles in such matrices like untreated wastewater and sewage effluent necessarily entails samples to be concentrated before they can be detected. In Nucleic-Acid-Based methods, the process may include a stage to physically concentrate the infectious particles (e.g., through filtration or precipitation), and an amplification stage (repeated replication of RNA to allow its detection). Wastewater samples are often centrifuged or filtered to eliminate debris. The process is followed by enrichment of viruses contained in sewage samples; the best available approaches at present include electronegative membrane filtration (0.45 µm) [15], skimmed milk flocculation [193], ultrafiltration [111,194,195] or polyethylene glycol precipitation [195,196], aluminum flocculation [109,197], or ultracentrifugation [106,113] enabling, as reported, 20 to 800 folds concentration. Meanwhile, except for Medema et al. [17] and Randazzo et al. [109] suggesting 3–50% viral recoveries, the literature is limited with respect to the percentage recovery of SARS-CoV-2 from sewage samples. This could be possibly due to the risks associated with handling the virus and requirements for a high-containment facility in order to fulfil biosafety guidelines. SARS-CoV-2 is classified as a hazard category 3 pathogen, which means that any work with the viable virus necessitates the use of biosafety level 3 (BSL-3) laboratories. Detection of RNA, however, could be operated in BSL-2 diagnostic laboratories which are more widely available and cost less to access. The CDC have recently released guidelines for testing environmental specimens to monitor SARS-CoV-2 in wastewater samples, clearly declaring that “Procedures that concentrate viruses, such as precipitation or membrane filtration, can be performed in a BSL-2 laboratory with unidirectional airflow and BSL-3 precautions” [198]. This same general result was concluded by Haramoto et al. [199] and Ye et al. [200], who pointed towards the inefficiency of standard virus concentration methods to recover enveloped viruses (e.g., SARS-CoV-2) from environmental matrices. The results thus remain highly controversial concerning reproducibility within and between studies. The use of appropriate process controls (e.g., spiking the sample with surrogate animal viruses of the same family or genus—such as heat-inactivated SARS-CoV-2 provided by ATCC or Pepper mild mottle virus (PMMoV)—having similar structure to the target pathogen before concentration) has been proposed by Randazzo et al. [109] and Farkas et al. [201] to estimate viral recoveries. The volume of sewage to be concentrated is another important parameter that still largely remains unknown but has a substantial impact on the results, which is discussed in Section 3.3.3 in more detail.
The tendency of viruses to biosolids paves the way for the partitioning of SARS-CoV-2 into sludge, particularly in a primary sedimentation tank because of its longer retention time. Except for a recent study conducted by Garham et al. [150], this issue has not been adequately addressed in the literature. Accordingly, monitoring primary sludge in a WWTP increases the chance of higher detection signals, as has been pointed out by Peccia et al. [202], who reported that the concentrations of SARS-CoV-2 RNA ranged from two to three orders of magnitude greater than the corresponding values previously reported in untreated wastewater. Nevertheless, it is important to note that such samples may inherently impose more inhibition in downstream analyses.

3.3.3. Challenges Involved with Laboratory Assays Dealing with SARS-CoV-2 in Wastewater

The resultant concentrated sample materials are then analyzed for the presence/enumeration of SARS CoV-2. This can follow two broad approaches targeting either (i) virus functional or structural motifs (e.g., RNA fragments, antigens, or other associated viral debris) or (ii) intact infectious virus particles [20].

3.3.4. Nucleic-Acid-Based Methods

Molecular approaches do not substitute for culture-based methods and address virus nucleic acids, which in SARS-CoV-2 is viral RNA. The efficiency and representativeness of genetic material—viral RNA—recovery from raw wastewater or treated effluent samples are fundamental concerns in molecular techniques. Today, RNA extraction is by and large carried out using commercial kits supplied by a variety of manufactures. The CDC have also recently published a report on Real Time RT-PCR Diagnostic Panel which deals with a list of RNA extraction kits that can be used for SARS-CoV-2 [198].
In addition to the limitations touched upon above regarding virus concentration/enrichment, the literature is somewhat contradictory with respect to the performance of PCR-based methods (PCR, RT-qPCR or allied approaches) designed for the detection of SARS-CoV-2 in sewage samples [15,17,109,110]. A specific segment of the viral RNA is targeted in these methods, paving the way for rapid, sensitive, and accurate strain-level detection of target(s) in real time RT-PCR assays [203]. Degradation products (such as RNA fragments) from the virus could also be selected as targets. As a quantitative nuclear-derived method, RT-PCR monitors the amplification of a targeted part of viral RNA using reverse transcriptase (to generate a DNA strand complementary to the RNA) and polymerase (to replicate the strand repeatedly) enzymes as far as it can be traced by fluorimetry [17]. This measurement is expressed as cycle threshold (Ct), a relative value that represents the cycle number (repeated PCR reactions) at which the amount of amplified DNA reaches the threshold level. Ct values can be alternatively normalized and expressed as gene copies (gc) per unit of volume (e.g., mL or L) or mass (e.g., g or Kg). Several questions remain unanswered at present. Accordingly, capability of detection and quantification are important performance characteristics of RT-PCR, as with any analytical technique. There has been considerable attention paid in recent works to unravel SARS-CoV-2 Detection and Quantitation Limits (LOD and LOQ) in wastewater, whereby early estimates put forward on LODs of a few thousand copies per liter with LOQs as reasonably 10–20,000 gc/L [204]. As of relative merits to cerebrate, an infected COVID-19 patient excrete approximately 108 to 1012 viral particles per day (see Section 3.1). However, more research on this topic needs to be undertaken. The ‘N1′ to ‘N3′ primer/probe sets each targeting a different region of the nucleocapsid (N) gene have been suggested by US CDC and their specificity against other viruses, including human coronaviruses, has been reported [205]. Corman et al. [206] in the Netherlands worked with a set against the RNA-dependent RNA polymerase [RdRp] gene, envelope [E] gene, and nucleocapsid [N] gene for detecting beta coronaviruses, including 2019-nCoV. Very recently, amplification of viral RNA targeting the ‘ORF1ab’ gene has also been reported in some studies concerning sewage surveillance to estimate the infection incidence within the population [75,119,207].
Although firm evidence is lacking, a combination of SARS-CoV-2 molecular tracing experiments in sewage samples put substantial differences for amplification of the viral RNA with different primer/probes. For instance, compared with the results for the sensitivity of primer/probe sets as ‘N1’ = ‘N3’ > ‘N2’ on SARS-CoV-2 RNA in clinical samples reported by US FDA, Medema et al. [17] observed a higher frequency of positive amplification targeting ‘N1’ and ‘N3’ genes, while ‘N2’ assay did not detect SARS-CoV-2 in wastewater samples. Moreover, ‘N1’ outperformed ‘N3’ and ‘E’ primers. In support of these findings, Rimoldi et al. [119] reported a distinct advantage of ‘ORF1ab’ gene over the ‘N’ and ‘E’ genes, which could be attributed to primer/probe sensitivity. To obtain a broader picture, Medema et al. [17] reported a positive signal for the ‘N2’ primer/probe set looking for a RT-qPCR signal between 40 and 45 cycles. It indicates that the role played by natural inhibitors in the gene amplification reaction of viruses might be solely responsible for increasing the Ct values, causing false-negative results [176]. It is expected that these findings will have important implications for using multiple primer/probe sets for future sewage surveillance projects.
Problems with the presence of inhibitory organic co-contaminants (e.g., humic and extracellular polymeric substances), which quite often afflict with the reverse transcription and polymerase enzymes, thus may invalidate the findings. Unfortunately, most concentration/enrichment methods could worsen the problem through the simultaneous concentrating and extracting organic compounds together with the targets [201]. Inhibition of RT-PCR reactions was reported by Medema et al. [17] in their initial trials. They stated that they were able to overcome these difficulties by the addition of BSA (Bovine Serum Albumin) to the reaction mixture. To the best of our knowledge, the report about assistance of another chemical or even process (which could be adsorption, ion exchange, etc.) to deeply remove inhibitory organic/inorganic compounds from sewage samples before PCR test targeting SARS-CoV-2 remains scarce. The use of polyethylene glycol (PEG) [110] and RNA magnetic beads [146,208], among others, has been previously reported to help get rid of inhibitors, which could be followed in downstream RNA targeting PCR. Addressing these problems calls out for a tremendous amount of research to be conducted to develop new robust methods, while at the same time concentrating the PCR targets. The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) based on Cas proteins [11,12] in combination with isothermal amplification and lateral flow assay has shown a promising potential in the detection of SARS-CoV-2 [209]. The isothermal PCR and visual observation make a faster alternative to the US CDC and SARS-CoV-2 real-time RT–PCR assay, with 95% positive and 100% negative predictive agreements, respectively [210].

3.3.5. Culture-Based Methods

It is important to bear in mind that the presence of viral RNA does not indicate the infectivity state of the target virus or a risk of infection. The virus genetic materials (including RNA fragments) are much more stable in the sewage than infectious virions. While, it can be inferred, with due care, that high counts of RNA provide an early sign of a potential hazard, there is no clear correlation between RNA concentration and infectiousness. With respect to its occurrence in sewage, viable SARS-CoV-2 probably enters mainly from shedding in the stool and to lesser extent from saliva and sputum. To assess whether there is a potential infection risk, sewage samples ideally need to show that the virus is infectious (viable). An assessment is further compounded by the lack of dose-response data, which means that it needs to come with details of whether the virus is present in adequate quantity in the particular location that has been monitored or sampled for transmission to take place. As of the time of writing, one of the reported studies was designed to detect live virus in sewage or deal with the potential infection hazard. The paucity of data on virus survival may have something to do with viable SARS-CoV-2 with regard to biosafety prerequisites (BSL-3 containment facilities, as mentioned previously) as well as the virus half-life in wastewater that seems to be very short [170]. Preliminary estimates of SARS-CoV-2 persistence in the water environment, as a metric for survivability and fate of SARS CoV-2 in wastewater matrices either municipal or hospital, are only a few days in the absence of disinfection measures [211]. However, some studies draw a different conclusion, suggesting that infectious viral titers may be able to survive longer than presumed [212,213,214]. It is, however, worth noting that the experimental conditions in which virus survivability has been examined coupled with the longer survival of the viral surrogates exploited in those studies seem to disprove this idea. According to a recent published report by Rimoldi et al. [119], SARS-CoV-2 remained infective on the time scale of a few hours. To obtain a clearer picture, they were unable to detect viable virus in wastewater samples just after 6–8 h from excretion of the viral particles in feces to the sewage sampling point. Indeed, the infectivity of virus is commonly confirmed in human or animal cell lines which not only is time-consuming (4–5 days for results) and resource-intensive, but also requires finding the appropriate host, as discussed before (Section 3.1). Another further concern in this regard is distinct susceptibility of cell lines exploited in culture-based assays which may not be infected as readily as respiratory tract cells which are the main targets of SARS-CoV-2 infection. This could be assigned to the plausible differences in the number of ACE2 receptors, the binding site for SARS-CoV-2.
As a result, it was not possible to draw a meaningful conclusion in relation to the sewage networks and role of WWTPs in the transmission of SARS-CoV-2. Nonetheless, the presence of enveloped viruses like coronaviruses, which undergo rapid inactivation outside the human host, have also been previously reported in wastewater [215]. Due to the lack of adequate evidence for the survival of SARS-CoV-2 in sewage, the infectious capacity of virus in sewer networks and wastewater treatment plants still needs more research that accounts for both viable and non-viable particles. It is worth bearing in mind that even if the presence of infectious virus is confirmed through culture-based techniques, it does not necessarily mean that it is present at a sufficiently high dose to pose a biohazard.
Interestingly, it was reported that the viral pathobiology still continues to be an evolving aspect that potentially shows virus genome is undergoing recurrent and independent mutations. The later demonstrates ongoing adaptation of SARS-CoV-2 to its novel human host [216]. The virus indeed is possibly in the throes of eventual mutations in the environmental compartments. Currently, the emergence of genomic diversity and mutations in virus come from symptomatic SARS-CoV-2 infected cases, while there are larger asymptomatic populations. Monitoring the mutations and evolution of virus in the sewage systems thus may provide a broad and accurate picture of viral mutation rates in the community level within the study area. Such information not only has the potential to inform on targets for drugs and vaccines, as has been previously highlighted, but also may subsequently assist in post-vaccination campaigns in which the virus mutation rate becomes a crucial factor in combating the pandemic.

3.4. Potential Risks for Wastewater Operators and Workers in the Vicinity of the Virus and the Environment

In general, raw wastewater contains, at any time, several pathogens and therefore poses certain health risks to whoever may come into direct contact with wastewater or be exposed to aerosolized droplets containing pathogenic virus, particularly near wastewater pumping stations and mechanical agitation. This is particularly important in less developed countries, where occupational exposure in WWTPs may warrant additional concern since the protocols of personal and collective protective equipment use is not as stringent as it is in the developed communities. Therefore, all such environments should have in place adequate health-safety procedures. The viral loads of SARS-CoV-2 in sewage at the entrance of the WWTPs are estimated to be 1.03 × 102 to 1.31 × 104 gc/mL (0.1 to 13.06 PFU/mL, respectively) [14]. Although detection of viral RNA in sewage does not necessarily mean that the sampled location contains infectious virus, the estimated risk of infection for workers based on quantitative microbiological risk assessment (QMRA), was less than WHO’s benchmark value of 10−3 for the moderate scenario, which has been highly advisable for workers in WWTPs. Nevertheless, given the lack of adequate evidence for the transmission of COVID-19 due to exposure to wastewater, WHO indicates that sewage has not yet played any role in the current pandemic [5]. In addition, evidence to date has demonstrated that the major infectious properties are destroyed during the wastewater treatment processes and the exposure to the virus is thus considered to be negligible compared to direct contacts between humans [184].
Finally, the potential ecological risks to the wider environment are also of great concern following poorly treated and untreated wastewater disposal or land-use of the raw/treated sludge. Some recent reports stressed that SARS-CoV-2 might have the ability to infect semiaquatic secondary animal vectors as well as several mammalian species [9,217,218,219]. Species close to outlets are in the throes of coming into direct contact with infectious virions from which it would likely become endemic in the secondary host, facilitating secondary transmission risk of virus. In addition, significant signals of selection and accelerated evolution in the ACE2 coding sequence across all mammals were found, and specific to the bat lineage [219]. Nevertheless, more studies are needed to address this concern properly.

3.5. Population Size Normalization

One of the greatest challenges in wastewater surveillance applications (e.g., WBE) for public-health studies is the accurate size estimation of contributing population to infer infection rates in a sewershed catchment. Uncertainty in connecting the observed viral signal in raw sewage to the contributing population could lead to a false-positive or -negative disease prevalence estimation in the community and can undermine WBE as a monitoring tool in situations such as post-vaccination. In this regard, both biomarker and census methods can be used, which should be independently collected and integrated with common units. Biomarkers are stable and human-related chemical or biological compounds whose quantity and quality can implicitly help with population estimation. The quantity of biomarkers would provide the basis for measured viral concentration and ensure that differences in viral loads could not be ascribed to population changes. Provided that observed viral concentrations are significantly high relative to the estimated population, a viral outbreak will be portended. Chemical biomarkers such as creatinine, cholesterol, coprostanol, nicotine, cortisol, androstenedione, ammonium, and the serotonin metabolite 5-hydroxyindoleacetic acid (5-HIAA) have been proposed as an endogenous or exogenous human substance to estimate real-time human population size from sewage samples [220]. Human mitochondrial DNA, human RNA, or CrAssphage (a benign virus that is common in the human gut) can be used in the same way [85,201]. Human nucleic acid has a great potential to act as a population biomarker due to its limited affinity to other species in wastewater, stability, constant excretion by humans, and the possibility of being quantifiable using the same pipelines and platforms as the viral nucleic acid of interest. For instance, Ling et al. [221] developed a new method to normalize data generated from sewage in view of recent developments in species-abundance distributions of microbial ecosystems. In their non-parametric model (Microbiome Census) metagenomic taxon-abundance of gut associated microbiome exhibited a good estimation of human population size in small communities.
Loading-based population proxies such as the copy numbers of crAssphage, pepper mild mosaic virus, and adenovirus are better than other human pathogenic viruses such as norovirus, due to demonstrating a strong seasonality in wastewater. Nevertheless, the lack of consensus among experts for accurate population estimation methods still remains as a major hurdle in adapting WBE for SARS-CoV2, and more research needs to be done on this front.

3.5.1. How Practical Is the Quantification of Virus in Wastewater Surveillance?

As discussed in previous sections, the limits of data variability are potentially influenced by sampling procedures (time, volume, and frequency), concentration/enrichment, detection, and quantification of virus in wastewater, as well as positive controls among others, so that detected amounts of RNA should be cautiously compared among studies. Thus, to obtain accurate results with statistical significance, applying standardized protocols is a must-have for sewage surveillance to ensure reproducibility and comparability of outcomes.
The joint research center of the European Union (JRC) has shown a correlation between the number of people infected in a sewershed and the viral load in wastewater [222]. Results also showed that the viral load in the sewage increases before the number of infected persons increases [22,223]. Therefore, the number of people infected in an area could be approximately estimated based on quantities of virus found in wastewater. However, it should be noted that each country might have a different testing procedure. In addition, while wastewater surveillance can reveal the presence of virus, it cannot tell whether this is from residents or from commuters (or tourists). Therefore, the wastewater surveillance estimates should be cross-checked by other sources of information to draw a more comprehensive conclusion.

3.5.2. How to Use and Interpret the Wastewater Surveillance?

Despite the challenges of wastewater surveillance for SARS-CoV2 detection in a community that have been mentioned before, it can still provide very useful information. However, it should be mentioned that this information should always be used as a complement to other epidemiological data when available. In the case of a lack of such reliable epidemiological data, wastewater surveillance can still be used in less favored conditions. Nevertheless, wastewater surveillance can be useful in three main categories [222,223]: As an early-warning tool to detect (re-)emergence of the pandemic in a specific sewage catchment area; As a management tool to identify the prevalence and trend of infection, as well as to determine low- or high-risk areas, for example, if surveillance data shows the absence of virus in wastewater, the corresponding community can be considered as of low risk; and As a safety factor in cases where testing of residents shows negative results but the virus is detected in wastewater, then further investigations should be prescribed—it can also be useful to evaluate the safety of tourists’ facilities with a controlled environment, such as cruise ships and other similar facilities.
In addition, wastewater surveillance has the potential for use as a tool for guiding and monitoring vaccination efforts [223,224]. To measure vaccine efficacy in real-world settings, there is a reliance on ongoing transmission of SARS-CoV-2, which is contradictory to public-health promotion efforts [225]. In contrast, the level of ongoing community transmission can be monitored by wastewater surveillance, which can provide insights into the impact of the vaccination programs. There are currently limited data on the efficacy of the vaccines to protect against different COVID-19 variants. Wastewater monitoring may provide evidence on the SARS-CoV-2 RNA viral load and viral shedding trends during the roll-out of vaccinations in communities. Furthermore, wastewater surveillance can provide an early-warning system for prioritizing hotspots where vaccination coverage is most urgently needed. Finally, targeted and untargeted sequencing of wastewater pathogens has the potential to track the spread of new specific sequence variants and identify mutations for which vaccinations may be needed to be modified [201,226].

4. Knowledge Gaps and Concluding Remarks

A comprehensive literature review was done to investigate the current state of knowledge and to identify the research gaps regarding the occurrence of SARS-CoV-2 in the feces of affected individuals and its dissemination in wastewater. In general, the current literature already confirms the occurrence of SARS-CoV-2 RNA in untreated wastewater with maximum concentrations over 106 copies per liter. While broad consensus has yet to form on the degree of risk, it is increasingly acknowledged that the presence of even SARS-CoV-2 genetic materials is of concern in both the raw and treated effluent and its potential to cause viral dissemination in workers in wastewater-treatment plants and in biota is clearly an issue which should inform environmental policy. Concern has also been raised about vegetables irrigated with untreated or partially treated wastewater, particularly in under-developed and developing countries. The lack of sewage separation (urban runoff from domestic effluents), which brings about combined sewer overflows in the events of heavy rainfalls, could also be a rationale for the occurrence of viral loads in water bodies [226]. Nevertheless, it should be recognized that there are still substantial knowledge gaps in the ever growing concern of potential infection hazard. An important aspect relating to the infection risk is that such samples ideally need to confirm that the virus is viable (infectious) and also demonstrate an adequate quantity for transmission to take place. Whilst this is a burgeoning area of research it needs to be made if individuals are likely in the throes of an infectious dose in the particular location where the sample was taken (e.g., toilet environments, sewer works, and WWTPs). These open questions warrant ad hoc investigations.
Based on the best available evidence at the present time, standardization of virus analysis in sewage is still needed. Areas that require the most improvements are sampling procedures, concentration/enrichment, detection, and quantification of virus in wastewater, as well as positive controls. Considering the quality of individual studies, problems with the protocols through which the original data was collected may invalidate these findings. Standardized protocols will indeed allow reproducibility and comparability of outcomes and would be reflected in the quality of data that are needed in policy and management interests. Testing in representative samples could also greatly attenuate the sources of uncertainty in model simulations, particularly in projects setting out to estimate numbers of infected people through wastewater-based epidemiology. Given the aforementioned differences in targeted procedures, conclusions on loading comparisons among studies and communities are difficult to draw. The presence of inhibitory organic co-contaminants in wastewater, which sometimes interferes with the PCR analysis, may invalidate the results. More research is thus needed regarding the assistance of other chemicals or processes (e.g., adsorption, ion exchange, etc.) to effectively remove inhibitory compounds from sewage samples before PCR test targeting SARS-CoV-2.
Another important knowledge gap to consider stems from the fact that the overwhelming majority of literature has focused on molecular methods to detect SARS-CoV-2. A confounding issue, however, is the infectious capacity of virus in feces and sewage systems since almost all studies just focused on viral genome detection/quantification. Nevertheless, given the lack of adequate evidence for the transmission of COVID-19 due to exposure to wastewater, it has been reported that sewage has not yet played any role in the current pandemic. In addition, evidence to date has demonstrated that the major infectious properties are destroyed in sewers and during wastewater treatment processes. The exposure to the virus is thus considered to be negligible compared to direct contacts between humans. A key research need is to investigate the correlation between PCR signal and infectious virus concentrations.
The nature of interactions between the virus in raw wastewater and other species (vertebrates and invertebrates) is yet to be fully elucidated since there are reports on secondary transmission of virus in animal vectors and several mammals. While by means of improved research techniques and announced progress in vaccine development, there is hope that the pace of disease will begin to slow, and studies looking into monitoring the traces of virus mutations in sewage as a broad picture of viral mutation rates in the community level within the study area particularly in post-vaccination campaigns should be encouraged. Genetic sequencing of virus in public sewage samples may help to explore the prevalence of different strains circulating within a specific catchment area, giving an indication of the evolution and spread of the disease. It is expected that the results of such research will have important implications for the sake of public health.
Results further showed that selecting the most accurate population estimation method for wastewater-based epidemiology studies is still a challenge. While census and biomarker studies have shown promising results, more research is needed to develop the best population size normalization method with respect to SARS-CoV-2. Although the number of people infected in an area could be approximately estimated based on quantities of virus found in the wastewaters, these estimates should be cross-checked by other sources of information to draw a more comprehensive conclusion. Furthermore, wastewater surveillance can be useful as an early-warning tool, a management tool, and/or a safety factor. Finally, wastewater surveillance has the potential to evaluate the effectiveness of vaccination programs, track the spread of specific sequence variants of SARS-CoV-2 and identify mutations for which vaccinations may be needed to be modified.

Author Contributions

Conceptualization, M.R.; methodology, F.A.; validation, F.A., N.M. and M.R.; formal analysis, F.A.; investigation, F.A. and N.M.; data curation, F.A. and M.R.; writing—original draft preparation, M.R., F.A.; writing—review and editing, M.R., F.A., S.I., M.N.-A., H.D. and M.J.-r.; supervision, M.R. and M.N.-A.; project administration, M.R.; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Keeler, N.; Lingappa, J. Severe acute respiratory syndrome: Public health response and clinical practice update for an emerging disease. Curr. Opin. Pediatr. 2004, 16, 61–69. [Google Scholar] [CrossRef] [PubMed]
  2. Mehta, P.; McAuley, D.F.; Brown, M.; Sanchez, E.; Tattersall, R.S.; Manson, J.J.; Collaboration, H.A.S. COVID-19: Consider cytokine storm syndromes and immunosuppression. Lancet (London, England) 2020, 395, 1033. [Google Scholar] [CrossRef]
  3. Zhu, N.; Zhang, D.; Wang, W.; Li, X.; Yang, B.; Song, J.; Zhao, X.; Huang, B.; Shi, W.; Lu, R. A novel coronavirus from patients with pneumonia in China, 2019. N. Engl. J. Med. 2020, 382, 727–733. [Google Scholar] [CrossRef] [PubMed]
  4. Gorbalenya, A.; Baker, S.; Baric, R.; de Groot, R.; Drosten, C.; Gulyaeva, A.; Haagmans, B.; Lauber, C.; Leontovich, A.; Neuman, B. Coronaviridae Study Group of the International Committee on Taxonomy of Viruses. The species severe acute respiratory syndrome-related coronavirus: Classifying 2019-nCoV and naming it SARS-CoV-2. Nat. Microbiol. 2020, 5, 536. [Google Scholar]
  5. World Health Organization (WHO). Modes of Transmission of Virus Causing COVID-19: Implications for IPC Precaution Recommendations. 2020. Available online: https://www.who.int/news-room/commentaries/detail/modes-of-transmission-of-virus-causing-covid-19-implications-for-ipc-precaution-recommendations (accessed on 25 July 2021).
  6. World Health Organization (WHO). Director-General’s Opening Remarks at the Media Briefing on COVID-19—11 March 2020. 2020. Available online: https://www.who.int/director-general/speeches/detail/who-director-general-s-opening-remarks-at-the-media-briefing-on-covid-19---11-march-2020 (accessed on 11 March 2020).
  7. Johns Hopkins University and Medicine. Available online: https://coronavirus.jhu.edu/map.html (accessed on 12 January 2021).
  8. Ali, I.; Alharbi, O.M. COVID-19: Disease, management, treatment, and social impact. Sci. Total Environ. 2020, 728, 138861. [Google Scholar] [CrossRef]
  9. Kim, Y.-I.; Kim, S.-G.; Kim, S.-M.; Kim, E.-H.; Park, S.-J.; Yu, K.-M.; Chang, J.-H.; Kim, E.J.; Lee, S.; Casel, M.A.B. Infection and rapid transmission of SARS-CoV-2 in ferrets. Cell Host Microbe 2020, 27, 704–709. [Google Scholar] [CrossRef]
  10. Rothan, H.A.; Byrareddy, S.N. The epidemiology and pathogenesis of coronavirus disease (COVID-19) outbreak. J. Autoimmun. 2020, 109, 102433. [Google Scholar] [CrossRef]
  11. Chen, Y.; Chen, L.; Deng, Q.; Zhang, G.; Wu, K.; Ni, L.; Yang, Y.; Liu, B.; Wang, W.; Wei, C. The presence of SARS-CoV-2 RNA in the feces of COVID-19 patients. J. Med. Virol. 2020, 92, 833–840. [Google Scholar] [CrossRef] [Green Version]
  12. Peng, L.; Liu, J.; Xu, W.; Luo, Q.; Chen, D.; Lei, Z.; Huang, Z.; Li, X.; Deng, K.; Lin, B. SARS-CoV-2 can be detected in urine, blood, anal swabs, and oropharyngeal swabs specimens. J. Med. Virol. 2020, 92, 1676–1680. [Google Scholar] [CrossRef]
  13. Young, B.E.; Ong, S.W.X.; Kalimuddin, S.; Low, J.G.; Tan, S.Y.; Loh, J.; Ng, O.-T.; Marimuthu, K.; Ang, L.W.; Mak, T.M. Epidemiologic features and clinical course of patients infected with SARS-CoV-2 in Singapore. JAMA 2020, 323, 1488–1494. [Google Scholar] [CrossRef] [Green Version]
  14. Zaneti, R.N.; Girardi, V.; Spilki, F.R.; Mena, K.; Westphalen, A.P.C.; da Costa Colares, E.R.; Pozzebon, A.G.; Etchepare, R.G. Quantitative microbial risk assessment of SARS-CoV-2 for workers in wastewater treatment plants. medRxiv 2020. [Google Scholar] [CrossRef]
  15. Ahmed, W.; Angel, N.; Edson, J.; Bibby, K.; Bivins, A.; O’Brien, J.W.; Choi, P.M.; Kitajima, M.; Simpson, S.L.; Li, J. First confirmed detection of SARS-CoV-2 in untreated wastewater in Australia: A proof of concept for the wastewater surveillance of COVID-19 in the community. Sci. Total Environ. 2020, 728, 138764. [Google Scholar] [CrossRef] [PubMed]
  16. Ding, Z.; Qian, H.; Xu, B.; Huang, Y.; Miao, T.; Yen, H.-L.; Xiao, S.; Cui, L.; Wu, X.; Shao, W. Toilets dominate environmental detection of SARS-CoV-2 virus in a hospital. medRxiv 2020. [Google Scholar]
  17. Medema, G.; Heijnen, L.; Elsinga, G.; Italiaander, R.; Brouwer, A. Presence of SARS-Coronavirus-2 RNA in sewage and correlation with reported COVID-19 prevalence in the early stage of the epidemic in The Netherlands. Environ. Sci. Technol. Lett. 2020, 7, 511–516. [Google Scholar] [CrossRef]
  18. Bivins, A.; North, D.; Ahmad, A.; Ahmed, W.; Alm, E.; Been, F.; Bhattacharya, P.; Bijlsma, L.; Boehm, A.B.; Brown, J.; et al. Wastewater-based epidemiology: Global collaborative to maximize contributions in the fight against COVID-19. Environ. Sci. Technol. 2020, 54, 7754–7757. [Google Scholar] [CrossRef] [PubMed]
  19. Sims, J.T.; Krishnan, V.; Chang, C.-Y.; Engle, S.M.; Casalini, G.; Rodgers, G.H.; Bivi, N.; Nickoloff, B.J.; Konrad, R.J.; de Bono, S.; et al. Characterization of the cytokine storm reflects hyperinflammatory endothelial dysfunction in COVID-19. J. Allergy Clin. Immunol. 2021, 147, 107–111. [Google Scholar] [CrossRef]
  20. Hokajärvi, A.M.; Rytkönen, A.; Tiwari, A.; Kauppinen, A.; Oikarinen, S.; Lehto, K.M.; Kankaanpaa, A.; Gunnar, T.; Al-Hello, H.; Blomqvist, S.; et al. The detection and stability of the SARS-CoV-2 RNA biomarkers in wastewater influent in Helsinki, Finland. medRxiv 2020. [Google Scholar]
  21. Tan, X.; Huang, J.; Zhao, F.; Zhou, Y.; Li, J.Q.; Wang, X.Y. Clinical features of children with SARS-CoV-2 infection: An analysis of 13 cases from Changsha, China. Chin. J. Contemp. Pediatr. 2020, 22, 294. [Google Scholar]
  22. Cai, J.; Xu, J.; Lin, D.; Xu, L.; Qu, Z.; Zhang, Y.; Zhang, H.; Jia, R.; Wang, X.; Ge, Y. A Case Series of children with 2019 novel coronavirus infection: Clinical and epidemiological features. Clin. Infect. Dis. 2020, 71, 1547–1551. [Google Scholar] [CrossRef] [Green Version]
  23. Lei, Z.; Cao, H.; Jie, Y.; Huang, Z.; Guo, X.; Chen, J.; Peng, L.; Cao, H.; Dai, X.; Liu, J.; et al. A cross-sectional comparison of epidemiological and clinical features of patients with coronavirus disease (COVID-19) in Wuhan and outside Wuhan, China. Travel Med. Infect. Dis. 2020, 35, 101664. [Google Scholar] [CrossRef]
  24. Chan, J.F.-W.; Yuan, S.; Kok, K.-H.; To, K.K.-W.; Chu, H.; Yang, J.; Xing, F.; Liu, J.; Yip, C.C.-Y.; Poon, R.W.-S.; et al. A familial cluster of pneumonia associated with the 2019 novel coronavirus indicating person-to-person transmission: A study of a family cluster. Lancet 2020, 395, 514–523. [Google Scholar] [CrossRef] [Green Version]
  25. Yuan, Y.; Wang, N.; Ou, X. Caution should be exercised for the detection of SARS-CoV-2, especially in the elderly. J. Med. Virol. 2020, 92, 1641–1648. [Google Scholar] [CrossRef] [Green Version]
  26. Kujawski, S.A.; Wong, K.K.; Collins, J.P.; Epstein, L.; Killerby, M.E.; Midgley, C.M.; Abedi, G.R.; Ahmed, N.S.; Almendares, O.; Alvarez, F.N. First 12 patients with coronavirus disease 2019 (COVID-19) in the United States. medRxiv 2020. [Google Scholar]
  27. Lescure, F.-X.; Bouadma, L.; Nguyen, D.; Parisey, M.; Wicky, P.-H.; Behillil, S.; Gaymard, A.; Bouscambert-Duchamp, M.; Donati, F.; Le Hingrat, Q.; et al. Clinical and virological data of the first cases of COVID-19 in Europe: A case series. Lancet Infect. Dis. 2020, 20, 697–706. [Google Scholar] [CrossRef] [Green Version]
  28. Liu, W.; Wang, J.; Li, W.; Zhou, Z.; Liu, S.; Rong, Z. Clinical characteristics of 19 neonates born to mothers with COVID-19. Front. Med. 2020, 14, 193–198. [Google Scholar] [CrossRef] [Green Version]
  29. Xie, C.; Jiang, L.; Huang, G.; Pu, H.; Gong, B.; Lin, H.; Ma, S.; Chen, X.; Long, B.; Si, G.; et al. Comparison of different samples for 2019 novel coronavirus detection by nucleic acid amplification tests. Int. J. Infect. Dis. 2020, 93, 264–267. [Google Scholar] [CrossRef] [PubMed]
  30. Chen, L.; Lou, J.; Bai, Y.; Wang, M. COVID-19 disease with positive fecal and negative pharyngeal and sputum viral tests. Am. J. Gastroenterol. 2020, 115, 790. [Google Scholar] [CrossRef] [PubMed]
  31. Zhang, T.; Cui, X.; Zhao, X.; Wang, J.; Zheng, J.; Zheng, G.; Guo, W.; Cai, C.; He, S.; Xu, Y. Detectable SARS-CoV-2 viral RNA in feces of three children during recovery period of COVID-19 pneumonia. J. Med. Virol. 2020, 92, 909–914. [Google Scholar] [CrossRef] [PubMed]
  32. Liu, W.-J.W.; Yuan, C.; Yu, M.-L.; Li, P.; Yan, J.-B. Detection of novel coronavirus by RT-PCR in stool specimen from asymptomatic child, China. Emerg. Infect. Dis. 2020, 26, 1337. [Google Scholar]
  33. Li, J.; Zhang, L.; Liu, B.; Song, D. Case report: Viral shedding for 60 days in a woman with COVID-19. Am. J. Trop. Med. Hyg. 2020, 102, 1210–1213. [Google Scholar] [CrossRef]
  34. Wang, W.; Xu, Y.; Gao, R.; Lu, R.; Han, K.; Wu, G.; Tan, W. Detection of SARS-CoV-2 in different types of clinical specimens. JAMA 2020, 323, 1843–1844. [Google Scholar] [CrossRef] [Green Version]
  35. Wei, X.S.; Wang, X.; Niu, Y.R.; Ye, L.L.; Peng, W.B.; Wang, Z.H.; Yang, W.-B.; Yang, B.-H.; Zhang, J.-C.; Ma, W.-L.; et al. Diarrhea is associated with prolonged symptoms and viral carriage in COVID-19. Clin. Gastroenterol. Hepatol. 2020, 18, 1753–1759. [Google Scholar] [CrossRef] [PubMed]
  36. Han, C.; Duan, C.; Zhang, S.; Spiegel, B.; Shi, H.; Wang, W.; Zhang, L.; Lin, R.; Liu, J.; Ding, Z.; et al. Digestive symptoms in COVID-19 patients with mild disease severity: Clinical presentation, stool viral RNA testing and outcomes. Am. J. Gastroenterol. 2020, 115, 916–923. [Google Scholar] [CrossRef] [PubMed]
  37. Ma, X.; Su, L.; Zhang, Y.; Zhang, X.; Gai, Z.; Zhang, Z. Do children need a longer time to shed SARS-CoV-2 in stool than adults? J. Microbiol. Immunol. Infect. 2020, 53, 373–376. [Google Scholar] [CrossRef] [PubMed]
  38. Xing, Y.; Ni, W.; Wu, Q.; Li, W.; Li, G.; Wang, W.; Tong, J.; Song, X.; Wong, G.W.K.; Xing, Q. Dynamics of faecal SARS-CoV-2 in infected children during the convalescent phase. J. Infect. 2020, 81, 318. [Google Scholar] [CrossRef] [PubMed]
  39. Tan, Y.-P.; Tan, B.-Y.; Pan, J.; Wu, J.; Zeng, S.-Z.; Wei, H.-Y. Epidemiologic and clinical characteristics of 10 children with coronavirus disease 2019 in Changsha, China. J. Clin. Virol. 2020, 127, 104353. [Google Scholar] [CrossRef]
  40. Lo, I.L.; Lio, C.F.; Cheong, H.H.; Lei, C.I.; Cheong, T.H.; Zhong, X.; Tian, Y.; Sin, N.N. Evaluation of SARS-CoV-2 RNA shedding in clinical specimens and clinical characteristics of 10 patients with COVID-19 in Macau. Int. J. Biol. Sci. 2020, 16, 1698. [Google Scholar] [CrossRef] [Green Version]
  41. Xiao, F.; Sun, J.; Xu, Y.; Li, F.; Huang, X.; Li, H.; Zhao, J.; Huang, J.; Zhao, J. Infectious SARS-CoV-2 in feces of patient with severe COVID-19. Emerg. Infect. Dis. 2020, 26, 1920. [Google Scholar] [CrossRef]
  42. Hu, X.; Xing, Y.; Jia, J.; Ni, W.; Liang, J.; Zhao, D.; Song, X.; Gao, R.; Jiang, F. Factors associated with negative conversion of viral RNA in patients hospitalized with COVID-19. Sci. Total Environ. 2020, 728, 138812. [Google Scholar] [CrossRef]
  43. Effenberger, M.; Grabherr, F.; Mayr, L.; Schwaerzler, J.; Nairz, M.; Seifert, M.; Hilbe, R.; Seiwald, S.; Scholl-Buergi, S.; Fritsche, G.; et al. Faecal calprotectin indicates intestinal inflammation in COVID-19. Gut 2020, 69, 1543–1544. [Google Scholar] [CrossRef] [Green Version]
  44. Zhang, J.; Wang, S.; Xue, Y. Fecal specimen diagnosis 2019 novel coronavirus–infected pneumonia. J. Med. Virol. 2020, 92, 680–682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Holshue, M.L.; DeBolt, C.; Lindquist, S.; Lofy, K.H.; Wiesman, J.; Bruce, H.; Spitters, C.; Ericson, K.; Wilkerson, S.; Tural, A.; et al. First case of 2019 novel coronavirus in the United States. N. Engl. J. Med. 2020, 382, 929–936. [Google Scholar] [CrossRef] [PubMed]
  46. Lin, L.; Jiang, X.; Zhang, Z.; Huang, S.; Zhang, Z.; Fang, Z.; Gu, Z.; Gao, L.; Shi, H.; Mai, L.; et al. Gastrointestinal symptoms of 95 cases with SARS-CoV-2 infection. Gut 2020, 69, 997–1001. [Google Scholar] [CrossRef]
  47. Yun, H.; Sun, Z.; Wu, J.; Tang, A.; Hu, M.; Xiang, Z. Laboratory data analysis of novel coronavirus (COVID-19) screening in 2510 patients. Clin. Chim. Acta 2020, 507, 94–97. [Google Scholar] [CrossRef] [PubMed]
  48. Ling, Y.; Xu, S.-B.; Lin, Y.-X.; Tian, D.; Zhu, Z.-Q.; Dai, F.-H.; Wu, F.; Song, Z.-G.; Huang, W.; Chen, J. Persistence and clearance of viral RNA in 2019 novel coronavirus disease rehabilitation patients. Chin. Med. J. 133. [CrossRef]
  49. Li, Y.; Hu, Y.; Yu, Y.; Zhang, X.; Li, B.; Wu, J.; Li, J.; Wu, Y.; Xia, X.; Tang, H. Positive result of Sars-Cov-2 in faeces and sputum from discharged patient with COVID-19 in Yiwu, China. J. Med. Virol. 2020, 92, 1938–1947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Xing, Y.; Ni, W.; Wu, Q.; Li, W.; Li, G.; Tong, J.; Song, X.; Xing, Q. Prolonged presence of SARS-CoV-2 in feces of pediatric patients during the convalescent phase. medRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  51. Wu, Y.; Guo, C.; Tang, L.; Hong, Z.; Zhou, J.; Dong, X.; Yin, H.; Xiao, Q.; Tang, Y.; Qu, X.; et al. Prolonged presence of SARS-CoV-2 viral RNA in faecal samples. Lancet Gastroenterol. Hepatol. 2020, 5, 434–435. [Google Scholar] [CrossRef]
  52. Yu, F.; Yan, L.; Wang, N.; Yang, S.; Wang, L.; Tang, Y.; Gao, G.; Wang, S.; Ma, C.; Xie, R. Quantitative detection and viral load analysis of SARS-CoV-2 in infected patients. Clin. Infect. Dis. 2020, 71, 793–798. [Google Scholar] [CrossRef] [Green Version]
  53. Pei, G.; Zhang, Z.; Peng, J.; Liu, L.; Zhang, C.; Yu, C.; Ma, Z.; Huang, Y.; Liu, W.; Yao, Y.; et al. Renal involvement and early prognosis in patients with COVID-19 pneumonia. J. Am. Soc. Nephrol. 2020, 31, 1157–1165. [Google Scholar] [CrossRef]
  54. Hosoda, T.; Sakamoto, M.; Shimizu, H.; Okabe, N. SARS-CoV-2 enterocolitis with persisting to excrete the virus for approximately two weeks after recovering from diarrhea: A case report. Infect. Control. Hosp. Epidemiol. 2020, 41, 753–754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Chen, C.; Gao, G.; Xu, Y.; Pu, L.; Wang, Q.; Wang, L.; Wang, W.; Song, Y.; Chen, M.; Wang, L. SARS-CoV-2–positive sputum and feces after conversion of pharyngeal samples in patients with COVID-19. Ann. Intern. Med. 2020, 172, 832–834. [Google Scholar] [CrossRef] [PubMed]
  56. Ng, S.C.; Tilg, H. COVID-19 and the gastrointestinal tract: More than meets the eye. Gut 2020, 69, 973–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Han, M.S.; Seong, M.W.; Heo, E.Y.; Park, J.H.; Kim, N.; Shin, S.; Cho, S.I.; Park, S.S.; Choi, E.H. Sequential analysis of viral load in a neonate and her mother infected with SARS-CoV-2. Clin. Infect. Dis. 2020, 71, 2236–2239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Paoli, D.; Pallotti, F.; Colangelo, S.; Basilico, F.; Mazzuti, L.; Turriziani, O.; Antonelli, G.; Lenzi, A.; Lombardo, F. Study of SARS-CoV-2 in semen and urine samples of a volunteer with positive naso-pharyngeal swab. J. Endocrinol. Investig. 2020, 43, 1819–1822. [Google Scholar] [CrossRef]
  59. To, K.K.-W.; Tsang, O.T.-Y.; Leung, W.-S.; Tam, A.R.; Wu, T.-C.; Lung, D.C.; Yip, C.C.-Y.; Cai, J.-P.; Chan, J.M.-C.; Chik, T.S.-H. Temporal profiles of viral load in posterior oropharyngeal saliva samples and serum antibody responses during infection by SARS-CoV-2: An observational cohort study. Lancet Infect. Dis. 2020, 20, 565–574. [Google Scholar] [CrossRef] [Green Version]
  60. Su, L.; Ma, X.; Yu, H.; Zhang, Z.; Bian, P.; Han, Y.; Sun, J.; Liu, Y.; Yang, C.; Geng, J.; et al. The different clinical characteristics of corona virus disease cases between children and their families in China—The character of children with COVID-19. Emerg. Microbes Infect. 2020, 9, 707–713. [Google Scholar] [CrossRef] [Green Version]
  61. Peng, Z.; Wang, J.; Mo, Y.; Duan, W.; Xiang, G.; Yi, M.; Bo, L.; Sh, Y. Unlikely SARS-CoV-2 vertical transmission from mother to child: A case report. J. Infect. Public Health 2020, 13, 818–820. [Google Scholar] [CrossRef]
  62. Zheng, S.; Fan, J.; Yu, F.; Feng, B.; Lou, B.; Zou, Q.; Xie, G.; Lin, S.; Wang, R.; Yang, X. Viral load dynamics and disease severity in patients infected with SARS-CoV-2 in Zhejiang province, China, January–March 2020: Retrospective cohort study. BMJ 2020, 369, m1443. [Google Scholar] [CrossRef] [Green Version]
  63. Pan, Y.; Zhang, D.; Yang, P.; Poon, L.L.; Wang, Q. Viral load of SARS-CoV-2 in clinical samples. Lancet Infect. Dis. 2020, 20, 411–412. [Google Scholar] [CrossRef]
  64. Wölfel, R.; Corman, V.M.; Guggemos, W.; Seilmaier, M.; Zange, S.; Müller, M.A.; Niemeyer, D.; Jones, T.C.; Vollmar, P.; Rothe, C.; et al. Virological assessment of hospitalized patients with COVID-2019. Nat. Cell Biol. 2020, 581, 465–469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Wang, J.; Feng, H.; Zhang, S.; Ni, Z.; Ni, L.; Chen, Y.; Zhuo, L.; Zhong, Z.; Qu, T. SARS-CoV-2 RNA detection of hospital isolation wards hygiene monitoring during the Coronavirus Disease 2019 outbreak in a Chinese hospital. Int. J. Infect. Dis. 2020, 94, 103–106. [Google Scholar] [CrossRef] [PubMed]
  66. Xing, Y.; Ni, W.; Wu, Q.; Li, W.-J.; Li, G.-J.; Wang, W.-D.; Tong, J.-N.; Song, X.-F.; Wong, G.W.-K.; Xing, Q.-S. Prolonged viral shedding in feces of pediatric patients with coronavirus disease 2019. J. Microbiol. Immunol. Infect. 2020, 53, 473–480. [Google Scholar] [CrossRef]
  67. Liu, W.-D.; Chang, S.-Y.; Wang, J.-T.; Tsai, M.-J.; Hung, C.-C.; Hsu, C.-L.; Chang, S.-C. Prolonged virus shedding even after seroconversion in a patient with COVID-19. J. Infect. 2020, 81, 318–356. [Google Scholar] [CrossRef] [PubMed]
  68. Cao, Q.; Chen, Y.-C.; Chen, C.-L.; Chiu, C.-H. SARS-CoV-2 infection in children: Transmission dynamics and clinical characteristics. J. Formos. Med. Assoc. 2020, 119, 670. [Google Scholar] [CrossRef]
  69. Liu, F.; Cai, Z.-B.; Huang, J.-S.; Yu, W.-Y.; Niu, H.-Y.; Zhang, Y.; Sui, D.-M.; Wang, F.; Xue, L.-Z.; Xu, A.-F. Positive SARS-CoV-2 RNA recurs repeatedly in a case recovered from COVID-19: Dynamic results from 108 days of follow-up. Pathog. Dis. 2020, 78, 31. [Google Scholar] [CrossRef] [PubMed]
  70. Hase, R.; Kurita, T.; Muranaka, E.; Sasazawa, H.; Mito, H.; Yano, Y. A case of imported COVID-19 diagnosed by PCR-positive lower respiratory specimen but with PCR-negative throat swabs. Infect. Dis. 2020, 52, 423–426. [Google Scholar] [CrossRef] [Green Version]
  71. Zuo, T.; Zhang, F.; Lui, G.C.; Yeoh, Y.K.; Li, A.Y.; Zhan, H.; Wan, Y.; Chung, A.C.; Cheung, C.P.; Chen, N.; et al. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology 2020, 159, 944–955. [Google Scholar] [CrossRef] [PubMed]
  72. Mao, L.-J.; Xu, J.; Xu, Z.-H.; Xia, X.-P.; Li, B.; He, J.-G.; Zhao, P.; Pan, J.-W.; Zhang, D.; Su, Y.; et al. A child with household transmitted COVID-19. BMC Infect. Dis. 2020, 20, 1–5. [Google Scholar] [CrossRef]
  73. Walpole, S.C.; McHugh, R.; Samuel, J.; Schmid, M.L. COVID-19 presenting as severe, persistent abdominal pain and causing late respiratory compromise in a 33-year-old man. BMJ Case Rep. 2020, 13, e236030. [Google Scholar] [CrossRef]
  74. Mesoraca, A.; Margiotti, K.; Viola, A.; Cima, A.; Sparacino, D.; Giorlandino, C. Evaluation of SARS-CoV-2 viral RNA in fecal samples. Virol. J. 2020, 17, 1–3. [Google Scholar] [CrossRef]
  75. Kumar, M.; Patel, A.K.; Shah, A.V.; Raval, J.; Rajpara, N.; Joshi, M.; Joshi, C.G. First proof of the capability of wastewater surveillance for COVID-19 in India through detection of genetic material of SARS-CoV-2. Sci. Total Environ. 2020, 141326. [Google Scholar] [CrossRef]
  76. Jeong, H.W.; Kim, S.-M.; Kim, H.-S.; Kim, Y.-I.; Kim, J.H.; Cho, J.Y.; Kim, S.-H.; Kang, H.; Kim, S.-G.; Park, S.-J. Viable SARS-CoV-2 in various specimens from COVID-19 patients. Clin. Microbiol. Infect. 2020, 26, 1520–1524. [Google Scholar] [CrossRef] [PubMed]
  77. Perchetti, G.A.; Nalla, A.K.; Huang, M.-L.; Zhu, H.; Wei, Y.; Stensland, L.; Loprieno, M.A.; Jerome, K.R.; Greninger, A.L. Validation of SARS-CoV-2 detection across multiple specimen types. J. Clin. Virol. 2020, 28, 104438. [Google Scholar] [CrossRef]
  78. Wang, X.; Zhou, Y.; Jiang, N.; Zhou, Q.; Ma, W.-L. Persistence of intestinal SARS-CoV-2 infection in patients with COVID-19 leads to re-admission after pneumonia resolved. Int. J. Infect. Dis. 2020, 95, 433–435. [Google Scholar] [CrossRef] [PubMed]
  79. Yu, Y.; Li, Y.; Hu, Y.; Li, B.; Xu, J. Breastfed 13 month-old infant of a mother with COVID-19 pneumonia: A case report. Int. Breastfeed. J. 2020, 15, 68. [Google Scholar] [CrossRef]
  80. Xiao, F.; Tang, M.; Zheng, X.; Liu, Y.; Li, X.; Shan, H. Evidence for gastrointestinal infection of SARS-CoV-2. Gastroenterology 2020, 158, 1831–1833. [Google Scholar] [CrossRef]
  81. Du, W.; Yu, J.; Liu, X.; Chen, H.; Lin, L.; Li, Q. Persistence of SARS-CoV-2 virus RNA in feces: A case series of children. J. Infect. Public Health 2020, 13, 926–931. [Google Scholar] [CrossRef]
  82. Kirtsman, M.; Diambomba, Y.; Poutanen, S.M.; Malinowski, A.K.; Vlachodimitropoulou, E.; Parks, W.T.; Erdman, L.; Morris, S.K.; Shah, P.S. Probable congenital SARS-CoV-2 infection in a neonate born to a woman with active SARS-CoV-2 infec-tion. CMAJ 2020, 192, E647–E650. [Google Scholar] [CrossRef]
  83. Sun, J.; Xiao, J.; Sun, R.; Tang, X.; Liang, C.; Lin, H.; Zeng, L.; Hu, J.; Yuan, R.; Zhou, P. Prolonged persistence of SARS-CoV-2 RNA in body fluids. Emerg. Infect. Dis. 2020, 26, 1834. [Google Scholar] [CrossRef] [PubMed]
  84. Fu, B.; Qian, K.; Fu, X. SARS-CoV-2-induced vomiting as onset symptom in a patient with COVID-19. Dig. Dis. Sci. 2020, 65, 1568–1570. [Google Scholar] [CrossRef]
  85. Park, S.-k.; Lee, C.-W.; Park, D.-I.; Woo, H.-Y.; Cheong, H.S.; Shin, H.C.; Ahn, K.; Kwon, M.-J.; Joo, E.-J. Detection of SARS-CoV-2 in fecal samples from patients with asymptomatic and mild COVID-19 in Korea. Clin. Gastroenterol. Hepatol. 2020, 19, 1387–1394. [Google Scholar] [CrossRef]
  86. Kim, J.-M.; Kim, H.M.; Lee, E.; Jo, H.J.; Yoon, Y.; Lee, N.-J.; Son, J.; Lee, Y.-J.; Kim, M.S.; Lee, Y.-P.; et al. Detection and isolation of SARS-CoV-2 in serum, urine, and stool specimens of COVID-19 patients from the Republic of Korea. Osong Public Health Res. Perspect. 2020, 11, 112–117. [Google Scholar] [CrossRef] [PubMed]
  87. Mansour, A.; Atoui, R.; Kanso, K.; Mohsen, R.; Fares, Y.; Fares, J. First case of an infant with COVID-19 in the Middle East. Cureus 2020, 12, e7520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Rigamonti, E.; Salera, D.; Gheorghiu, A.C.; Fratila, C.; Gianella, P. The many faces of interstitial pneumonia: A case of presumed SARS-CoV-2 infection. Swiss Med. Wkly. 2020, 150, 2728. [Google Scholar] [CrossRef]
  89. Cho, S.-M.; Ha, G.Y. A case of COVID-19 in a 45-day-old infant with persistent fecal virus shedding for more than 12 weeks. Yonsei Med. J. 2020, 61, 901–903. [Google Scholar] [CrossRef]
  90. Binder, R.A.; Alarja, N.A.; Robie, E.R.; Kochek, K.E.; Xiu, L.; Rocha-Melogno, L.; Abdelgadir, A.; Goli, S.V.; Farrell, A.S.; Coleman, K.K.; et al. Environmental and aerosolized severe acute respiratory syndrome coronavirus 2 among hospitalized coronavirus disease 2019 Patients. J. Infect. Dis. 2020, 222, 1798–1806. [Google Scholar] [CrossRef] [PubMed]
  91. Han, M.S.; Seong, M.-W.; Kim, N.; Shin, S.; Cho, S.I.; Park, H.; Kim, T.S.; Park, S.S.; Choi, E.H. Viral RNA load in mildly symptomatic and asymptomatic children with COVID-19, Seoul. Emerg. Infect. Dis. 2020, 26, 2497–2499. [Google Scholar] [CrossRef]
  92. Colavita, F.; Lapa, D.; Carletti, F.; Lalle, E.; Messina, F.; Rueca, M.; Matusali, G.; Meschi, S.; Bordi, L.; Marsella, P.; et al. Virological characterization of the first 2 COVID-19 patients diagnosed in Italy: Phylogenetic analysis, virus shedding profile from different body sites, and antibody response kinetics. Open Forum Infect. Dis. 2020, 7, ofaa403. [Google Scholar] [CrossRef] [PubMed]
  93. Slaats, M.A.; Versteylen, M.; Gast, K.B.; Munnink, B.B.O.; Pas, S.D.; Bentvelsen, R.G.; van Beek, R. Case report of a neonate with high viral SARSCoV-2 loads and long-term virus shedding. J. Infect. Public Health 2020, 13, 1878–1884. [Google Scholar] [CrossRef]
  94. Italiano, J.; Bush, R.; Acharya, R.; Upadhyay, K. Persistent viral shedding despite seroconversion in a kidney transplant recipient with severe extrapulmonary COVID-19. BMJ Case Rep. 2020, 13, e239612. [Google Scholar] [CrossRef]
  95. Scutari, R.; Piermatteo, L.; Ciancio Manuelli, M.; Iannetta, M.; Salpini, R.; Bertoli, A.; Alteri, C.; Saccomandi, P.; Bellocchi, M.C.; Malagnino, V. Long-term SARS-CoV-2 infection associated with viral dissemination in different body fluids including bile in two patients with acute cholecystitis. Life 2020, 10, 302. [Google Scholar] [CrossRef]
  96. Hinojosa-Velasco, A.; de Oca, P.V.B.-M.; García-Sosa, L.E.; Mendoza-Durán, J.G.; Pérez-Méndez, M.J.; Dávila-González, E.; Ramírez-Hernández, D.G.; García-Mena, J.; Zárate-Segura, P.; Reyes-Ruiz, J.M. A case report of newborn infant with severe COVID-19 in Mexico: Detection of SARS-CoV-2 in human breast milk and stool. Int. J. Infect. Dis. 2020, 100, 21–24. [Google Scholar] [CrossRef]
  97. Zhang, Y.; Chen, C.; Song, Y.; Zhu, S.; Wang, D.; Zhang, H.; Han, G.; Weng, Y.; Xu, J.; Xu, J. Excretion of SARS-CoV-2 through faecal specimens. Emerg. Microbes Infect. 2020, 1, 28. [Google Scholar] [CrossRef]
  98. Wang, X.; Zheng, J.; Guo, L.; Yao, H.; Wang, L.; Xia, X.; Zhang, W. Fecal viral shedding in COVID-19 patients: Clinical significance, viral load dynamics and survival analysis. Virus Res. 2020, 289, 198147. [Google Scholar] [CrossRef] [PubMed]
  99. Novazzi, F.; Cassaniti, I.; Piralla, A.; Di Sabatino, A.; Bruno, R.; Baldanti, F.; Force, S.M.P.C.T.; Mondelli, M.; Brunetti, E.; Di Matteo, A. Detection of the SARS-CoV-2 in different biologic specimens from positive patients with COVID-19, in Northern Italy. Pediatr. Allergy Immunol. 2020, 31, 72–74. [Google Scholar] [CrossRef]
  100. Chen, S.; Si, J.; Tang, W.; Zhang, A.; Pan, L.; An, M.; Zhang, H.; Xue, S.; Wu, K.; Chen, S. An asymptomatic SARS-CoV-2-infected infant with persistent fecal viral RNA shedding in a family cluster: A rare case report. Front. Med. 2020, 7, 648. [Google Scholar] [CrossRef] [PubMed]
  101. Khoury, N.C.; Russi, T.J. A case of gastrointestinal-predominant COVID-19 demonstrates value of stool PCR test. J. Med. Virol. 2020. [Google Scholar] [CrossRef] [PubMed]
  102. Chen, G.-Q.; Luo, W.-T.; Zhao, C.-H.; Li, C.-N.; Hong, Z.-S.; Xia, J.-Y.; Liu, X. Comparison of clinical characteristics between fecal/perianal swab nucleic acid-positive and-negative pa-tients with COVID-19. J. Infect. Dev. Ctries. 2020, 14, 847–852. [Google Scholar] [CrossRef] [PubMed]
  103. Barth, R.E.; De Regt, M.J. Persistence of viral RNA in stool samples from patients recovering from COVID-19. Br. Med. J. 2020, 369, m1724. [Google Scholar] [CrossRef]
  104. Fumian, T.M.; Malta, F.C.; dos Santos, D.R.L.; Pauvolid-Corrêa, A.; Fialho, A.M.; Leite, J.P.G.; Miagostovich, M.P. SARS-CoV-2 RNA detection in stool samples from acute gastroenteritis cases, Brazil. J. Med. Virol. 2021, 93, 2543–2547. [Google Scholar] [CrossRef] [PubMed]
  105. Chu, H.; Li, J.; Yan, J.; Bai, T.; Schnabl, B.; Zou, L.; Yang, L.; Hou, X. Persistent SARS-CoV-2 RNA positive in feces but negative in breastmilk: A case report of COVID-19 in a breastfeeding patient. Front. Med. 2020, 7, 866. [Google Scholar] [CrossRef]
  106. Wurtzer, S.; Marechal, V.; Mouchel, J.M.; Moulin, L. Time course quantitative detection of SARS-CoV-2 in Parisian wastewaters correlates with COVID-19 confirmed cases. medRxiv 2020. [Google Scholar]
  107. Kocamemi, B.A.; Kurt, H.; Hacioglu, S.; Yarali, C.; Saatci, A.M.; Pakdemirli, B. First data-set on SARS-CoV-2 detection for Istanbul wastewaters in Turkey. medRxiv 2020. [Google Scholar]
  108. Rimoldi, S.G.; Stefani, F.; Gigantiello, A.; Polesello, S.; Comandatore, F.; Mileto, D.; Maresca, M.; Longobardi, C.; Mancon, A.; Romeri, F. Presence and infectivity of SARS-CoV-2 virus in wastewaters and rivers. Sci. Total Environ. 2020, 744, 140911. [Google Scholar] [CrossRef]
  109. Randazzo, W.; Truchado, P.; Cuevas-Ferrando, E.; Simón, P.; Allende, A.; Sánchez, G. SARS-CoV-2 RNA in wastewater anticipated COVID-19 occurrence in a low prevalence area. Water Res. 2020, 181, 115942. [Google Scholar] [CrossRef]
  110. Wu, F.; Zhang, J.; Xiao, A.; Gu, X.; Lee, W.L.; Armas, F.; Kauffman, K.; Hanage, W.; Matus, M.; Ghaeli, N.; et al. SARS-CoV-2 titers in wastewater are higher than expected from clinically confirmed cases. medRxiv 2020. [Google Scholar] [CrossRef] [PubMed]
  111. Nemudryi, A.; Nemudraia, A.; Wiegand, T.; Surya, K.; Buyukyoruk, M.; Cicha, C. Temporal detection and phylogenetic assessment of SARS-CoV-2 in municipal wastewater. medRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  112. Lodder, W.; de Roda Husman, A.M. SARS-CoV-2 in wastewater: Potential health risk, but also data source. Lancet Gastroenterol. Hepatol. 2020, 5, 533–534. [Google Scholar] [CrossRef]
  113. Wurtzer, S.; Marechal, V.; Mouchel, J.M.; Maday, Y.; Teyssou, R.; Richard, E.; Almayrac, J.L.; Moulin, L. Evaluation of lockdown impact on SARS-CoV-2 dynamics through viral genome quantification in Paris wastewaters. medRxiv 2020. [Google Scholar] [CrossRef]
  114. Manupati, H.; Kiran, U.; Kuncha, S.K.; Kopperi, H.; Gokulan, C.; Mishra, R.K. Comprehensive surveillance of SARS-CoV-2 spread using wastewater-based epidemiology studies. medRxiv 2020. [Google Scholar] [CrossRef]
  115. Curtis, K.; Keeling, D.; Yetka, K.; Larson, A.; Gonzalez, R. Wastewater SARS-CoV-2 concentration and loading variability from grab and 24-hour composite samples. medRxiv 2020. [Google Scholar] [CrossRef]
  116. Hong, P.; Rachmadi, A.T.; Mantilla-Calderon, D.; Alkahtani, M.; Bashwari, Y.M.; Al Qarni, H.; Zhou, J. How early into the outbreak can surveillance of SARS-CoV-2 in wastewater tell us? medRxiv 2020. [Google Scholar] [CrossRef]
  117. Green, H.; Wilder, M.; Middleton, F.A.; Collins, M.; Fenty, A.; Gentile, K.; Kmush, B.; Zeng, T.; Larsen, D.A. Quantification of SARS-CoV-2 and cross-assembly phage (crAssphage) from wastewater to monitor coronavirus transmission within communities. medRxiv 2020. [Google Scholar] [CrossRef]
  118. Sharif, S. Detection of SARs-CoV-2 in wastewater, using the existing environmental surveillance network: An epidemiological gateway to an early warning for COVID-19 in communities. medRxiv 2020. [Google Scholar] [CrossRef]
  119. Rimoldi, S.G.; Stefani, F.; Gigantiello, A.; Polesello, S.; Comandatore, F.; Mileto, D.; Maresca, M.; Longobardi, C.; Mancon, A.; Romeri, F. Presence and vitality of SARS-CoV-2 virus in wastewaters and rivers. medRxiv 2020. [Google Scholar] [CrossRef]
  120. Medema, G.; Heijnen, L.; Elsinga, G.; Italiaander, R.; Brouwer, A. Presence of SARS-Coronavirus-2 in sewage. medRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  121. Prado, T.; Fumian, T.M.; Mannarino, C.F.; Maranhão, A.G.; Siqueira, M.M. Preliminary results of SARS-CoV-2 detection in sewerage system in Niterói municipality, Rio de Janeiro, Brazil. Memórias Inst. Oswaldo Cruz 2020, 115. [Google Scholar] [CrossRef] [PubMed]
  122. Sherchan, S.P.; Shahin, S.; Ward, L.M.; Tandukar, S.; Aw, T.G.; Schmitz, B.; Ahmed, W.; Kitajima, M. First detection of SARS-CoV-2 RNA in wastewater in North America: A study in Louisiana, USA. Sci. Total Environ. 2020, 743, 140621. [Google Scholar] [CrossRef]
  123. Trottier, J.; Darques, R.; Mouheb, N.A.; Partiot, E.; Bakhache, W.; Deffieu, M.S.; Gaudin, R. Post-lockdown detection of SARS-CoV-2 RNA in the wastewater of Montpellier, France. One Health 2020, 100157. [Google Scholar] [CrossRef] [PubMed]
  124. Mlejnkova, H.; Sovova, K.; Vasickova, P.; Ocenaskova, V.; Jasikova, L.; Juranova, E. Preliminary study of Sars-Cov-2 occurrence in wastewater in the Czech Republic. Int. J. Environ. Res. Public Health 2020, 17, 5508. [Google Scholar] [CrossRef]
  125. Haramoto, E.; Malla, B.; Thakali, O.; Kitajima, M. First environmental surveillance for the presence of SARS-CoV-2 RNA in wastewater and river water in Japan. medRxiv 2020. [Google Scholar] [CrossRef]
  126. La Rosa, G.; Iaconelli, M.; Mancini, P.; Ferraro, G.B.; Veneri, C.; Bonadonna, L.; Lucentini, L.; Suffredini, E. First detection of SARS-CoV-2 in untreated wastewaters in Italy. Sci. Total Environ. 2020, 736, 139652. [Google Scholar] [CrossRef]
  127. Gonzalez, R.; Curtis, K.; Bivins, A.; Bibby, K.; Weir, M.H.; Yetka, K.; Thompson, H.; Keeling, D.; Mitchell, J.; Gonzalez, D. COVID-19 surveillance in Southeastern Virginia using wastewater-based epidemiology. Water Res. 2020, 186, 116296. [Google Scholar] [CrossRef]
  128. Miyani, B.; Fonoll, X.; Norton, J.; Mehrotra, A.; Xagoraraki, I. SARS-CoV-2 in Detroit wastewater. J. Environ. Eng. 2020, 146, 06020004. [Google Scholar] [CrossRef]
  129. Westhaus, S.; Weber, F.-A.; Schiwy, S.; Linnemann, V.; Brinkmann, M.; Widera, M.; Greve, C.; Janke, A.; Hollert, H.; Wintgens, T. Detection of SARS-CoV-2 in raw and treated wastewater in Germany—Suitability for COVID-19 surveil-lance and potential transmission risks. Sci. Total Environ. 2020, 751, 141750. [Google Scholar] [CrossRef]
  130. D’Aoust, P.M.; Mercier, E.; Montpetit, D.; Jia, J.-J.; Alexandrov, I.; Neault, N.; Baig, A.T.; Mayne, J.; Zhang, X.; Alain, T. Quantitative analysis of SARS-CoV-2 RNA from wastewater solids in communities with low COVID-19 incidence and prevalence. Water Res. 2020, 188, 116560. [Google Scholar] [CrossRef]
  131. Hata, A.; Hara-Yamamura, H.; Meuchi, Y.; Imai, S.; Honda, R. Detection of SARS-CoV-2 in wastewater in Japan during a COVID-19 outbreak. Sci. Total Environ. 2020, 143578. [Google Scholar] [CrossRef]
  132. Jafferali, M.H.; Khatami, K.; Atasoy, M.; Birgersson, M.; Williams, C.; Cetecioglu, Z. Benchmarking virus concentration methods for quantification of SARS-CoV-2 in raw wastewater. Sci. Total Environ. 2020, 142939. [Google Scholar] [CrossRef] [PubMed]
  133. Hasan, S.W.; Ibrahim, Y.; Daou, M.; Kannout, H.; Jan, N.; Lopes, A.; Alsafar, H.; Yousef, A.F. Detection and quantification of SARS-CoV-2 RNA in wastewater and treated effluents: Surveillance of COVID-19 epidemic in the United Arab Emirates. Sci. Total Environ. 2020, 142929. [Google Scholar] [CrossRef] [PubMed]
  134. Baldovin, T.; Amoruso, I.; Fonzo, M.; Buja, A.; Baldo, V.; Cocchio, S.; Bertoncello, C. SARS-CoV-2 RNA detection and persistence in wastewater samples: An experimental network for COVID-19 environmental surveillance in Padua, Veneto Region (NE Italy). Sci. Total Environ. 2020, 760, 143329. [Google Scholar] [CrossRef] [PubMed]
  135. Feng, B.; Xu, K.; Gu, S.; Zheng, S.; Zou, Q.; Xu, Y.; Yu, L.; Lou, F.; Yu, F.; Jin, T. Multi-route transmission potential of SARS-CoV-2 in healthcare facilities. J. Hazard. Mater. 2020, 402, 123771. [Google Scholar] [CrossRef]
  136. Kumar, M.; Kuroda, K.; Patel, A.K.; Patel, N.; Bhattacharya, P.; Joshi, M.; Joshi, C.G. Decay of SARS-CoV-2 RNA along the wastewater treatment outfitted with Upflow Anaerobic Sludge Blanket (UASB) system evaluated through two sample concentration techniques. Sci. Total Environ. 2020, 754, 142329. [Google Scholar] [CrossRef]
  137. Saguti, F.; Magnil, E.; Enache, L.; Churqui, M.P.; Johansson, A.; Lumley, D.; Davidsson, F.; Dotevall, L.; Mattsson, A.; Trybala, E. Surveillance of wastewater revealed peaks of SARS-CoV-2 preceding those of hospitalized patients with COVID-19. Water Res. 2020, 189, 116620. [Google Scholar] [CrossRef] [PubMed]
  138. Gallardo-Escárate, C.; Valenzuela-Muñoz, V.; Núñez-Acuña, G.; Valenzuela-Miranda, D.; Benaventel, B.P.; Sáez-Vera, C.; Urrutia, H.; Novoa, B.; Figueras, A.; Roberts, S.; et al. The wastewater microbiome: A novel insight for COVID-19 surveillance. Sci. Total Environ. 2021, 764, 142867. [Google Scholar] [CrossRef] [PubMed]
  139. Gonçalves, J.; Koritnik, T.; Mioč, V.; Trkov, M.; Bolješič, M.; Berginc, N.; Prosenc, K.; Kotar, T.; Paragi, M. Detection of SARS-CoV-2 RNA in hospital wastewater from a low COVID-19 disease prevalence area. Sci. Total Environ. 2020, 755, 143226. [Google Scholar] [CrossRef] [PubMed]
  140. Martin, J.; Klapsa, D.; Wilton, T.; Zambon, M.; Bentley, E.; Bujaki, E.; Fritzsche, M.; Mate, R.; Majumdar, M. Tracking SARS-CoV-2 in sewage: Evidence of changes in virus variant predominance during COVID-19 pandemic. Viruses 2020, 12, 1144. [Google Scholar] [CrossRef]
  141. Peccia, J.; Zulli, A.; Brackney, D.E.; Grubaugh, N.D.; Kaplan, E.H.; Casanovas-Massana, A.; Ko, A.I.; Malik, A.A.; Wang, D.; Wang, M. Measurement of SARS-CoV-2 RNA in wastewater tracks community infection dynamics. Nat. Biotechnol. 2020, 2020. 38, 1164–1167. [Google Scholar] [CrossRef]
  142. Albastaki, A.; Naji, M.; Lootah, R.; Almeheiri, R.; Almulla, H.; Almarri, I.; Alreyami, A.; Aden, A.; Alghafri, R. First confirmed detection of SARS-COV-2 in untreated municipal and aircraft wastewater in Dubai, UAE: The use of wastewater based epidemiology as an early warning tool to monitor the prevalence of COVID-19. Sci. Total Environ. 2021, 760, 143350. [Google Scholar] [CrossRef]
  143. Agrawal, S.; Orschler, L.; Lackner, S. Long-term monitoring of SARS-CoV-2 in wastewater of the Frankfurt metropolitan area in Southern Germany. medRxiv 2020. [Google Scholar]
  144. Abu-Ali, H.; Yaniv, K.; Bar-Zeev, E.; Chaudhury, S.; Shaga, M.; Lakkakula, S.; Ronen, Z.; Kushmaro, A.; Nir, O. Tracking SARS-CoV-2 RNA through the wastewater treatment process. medRxiv 2020. [Google Scholar]
  145. Kocamemi, B.A.; Kurt, H.; Sait, A.; Kadi, H.; Sarac, F.; Aydin, I.; Saatci, A.M.; Pakdemirli, B. Nationwide SARS-CoV-2 Surveillance Study for Sewage and Sludges of Wastewater Treatment Plants in Turkey. medRxiv 2020. [Google Scholar]
  146. AL, P.G.; Sweeney, C.; Hayes, E.; Trueman, B.; Huang, Y.; Jamieson, R.; Rand, J.; Gagnon, G.; Stoddart, A. Development and optimization of a new method for direct extraction of SARS-CoV-2 RNA from municipal wastewater using magnetic beads. medRxiv 2020. [Google Scholar]
  147. Iglesias, N.G.; Gebhard, L.G.; Carballeda, J.M.; Aiello, I.; Recalde, E.; Terny, G.; Ambrosolio, S.; L’Arco, G.; Konfino, J.; Brardinelli, J.I. SARS-CoV-2 surveillance in untreated wastewater: First detection in a low-resource community in Buenos Aires, Argentina. medRxiv 2020. [Google Scholar]
  148. Wu, F.; Xiao, A.; Zhang, J.; Moniz, K.; Endo, N.; Armas, F.; Bonneau, R.; Brown, M.A.; Bushman, M.; Chai, P.R. SARS-CoV-2 titers in wastewater foreshadow dynamics and clinical presentation of new COVID-19 cases. medRxiv 2020. [Google Scholar]
  149. Lara, R.W.I.; Elsinga, G.; Heijnen, L.; Munnink, B.B.O.; Schapendonk, C.M.; Nieuwenhuijse, D.; Kon, M.; Lu, L.; Aarestrup, F.M.; Lycett, S. Monitoring SARS-CoV-2 circulation and diversity through community wastewater sequencing. medRxiv 2020. [Google Scholar]
  150. Graham, K.E.; Loeb, S.K.; Wolfe, M.K.; Catoe, D.; Sinnott-Armstrong, N.; Kim, S.; Yamahara, K.M.; Sassoubre, L.M.; Mendoza Grijalva, L.M.; Roldan-Hernandez, L. SARS-CoV-2 RNA in wastewater settled solids is associated with COVID-19 cases in a large urban sewershed. Environ. Sci. Technol. 2020, 55, 488–498. [Google Scholar] [CrossRef] [PubMed]
  151. Leung, W.K.; To, K.F.; Chan, P.K.; Chan, H.L.; Wu, A.K.; Lee, N.; Yuen, K.Y.; Sung, J.J.Y. Enteric involvement of severe acute respiratory syndrome-associated coronavirus infection. Gastroenterology 2003, 125, 1011–1017. [Google Scholar] [CrossRef] [Green Version]
  152. Cheung, K.S.; Hung, I.F.; Chan, P.P.; Lung, K.C.; Tso, E.; Liu, R.; Ng, Y.Y.; Chu, M.Y.; Chung, T.W.H.; Tam, A.R.; et al. Gastrointestinal manifestations of SARS-CoV-2 infection and virus load in fecal samples from the Hong Kong cohort and systematic review and meta-analysis. Gastroenterology 2020, 159, 81–95. [Google Scholar] [CrossRef]
  153. Zang, R.; Castro, M.F.G.; McCune, B.T.; Zeng, Q.; Rothlauf, P.W.; Sonnek, N.M.; Liu, Z.; Brulois, K.F.; Wang, X.; Greenberg, H.B.; et al. TMPRSS2 and TMPRSS4 promote SARS-CoV-2 infection of human small intestinal enterocytes. Sci. Immunol. 2020, 5, 47. [Google Scholar] [CrossRef] [PubMed]
  154. Wigginton, K.; Ye, Y.; Ellenberg, R.M. Emerging investigators series: The source and fate of pandemic viruses in the urban water cycle. Environ. Sci. Water Res. Technol. 2015, 1, 735–746. [Google Scholar] [CrossRef]
  155. Garvin, M.R.; Alvarez, C.; Miller, J.I.; Prates, E.T.; Walker, A.; Amos, B.K.; Mast, A.E.; Justice, A.; Aronow, B.; Jacobson, D. A mechanistic model and therapeutic interventions for COVID-19 involving a RAS-mediated bradykinin storm. eLife 2020, 9. [Google Scholar] [CrossRef]
  156. Mizumoto, K.; Kagaya, K.; Zarebski, A.; Chowell, G. Estimating the asymptomatic proportion of coronavirus disease 2019 (COVID-19) cases on board the Diamond Princess cruise ship, Yokohama, Japan, 2020. Eurosurveillance 2020, 25, 2000180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Zhang, H.; Kang, Z.; Gong, H.; Xu, D.; Wang, J.; Li, Z.; Cui, X.; Xiao, J.; Meng, T.; Zhou, W.; et al. The digestive system is a potential route of 2019-nCov infection: A bioinformatics analysis based on single-cell transcriptomes. bioRxiv 2020. [Google Scholar]
  158. Zhou, J.; Li, C.; Zhao, G.; Chu, H.; Wang, D.; Yan, H.N.H.; Poon, V.K.-M.; Wen, L.; Wong, B.H.-Y.; Zhao, X.; et al. Human intestinal tract serves as an alternative infection route for Middle East respiratory syndrome coronavirus. Sci. Adv. 2017, 3, eaao4966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Wang, D.; Hu, B.; Hu, C.; Zhu, F.; Liu, X.; Zhang, J.; Wang, B.; Xiang, H.; Cheng, Z.; Xiong, Y.; et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus–infected pneumonia in Wuhan, China. JAMA 2020, 323, 1061. [Google Scholar] [CrossRef]
  160. D’Amico, F.; Baumgart, D.C.; Danese, S.; Peyrin-Biroulet, L. Diarrhea during COVID-19 infection: Pathogenesis, epidemiology, prevention, and management. Clin. Gastroenterol. Hepatol. 2020, 18, 1663–1672. [Google Scholar] [CrossRef]
  161. Chen, N.; Zhou, M.; Dong, X.; Qu, J.; Gong, F.; Han, Y.; Qiu, 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. Lancet 2020, 395, 507–513. [Google Scholar] [CrossRef] [Green Version]
  162. Chen, Y.; Wei, Q.; Li, R.; Gao, H.; Zhu, H.; Deng, W.; Bao, L.; Tong, W.; Cong, Z.; Jiang, H.; et al. Protection of Rhesus Macaque from SARS-coronavirus challenge by recombinant adenovirus vaccine. bioRxiv 2020. [Google Scholar]
  163. Gao, Q.Y.; Chen, Y.X.; Fang, J.Y. 2019 Novel coronavirus infection and gastrointestinal tract. J. Dig. Dis. 2020, 21, 125–126. [Google Scholar] [CrossRef] [Green Version]
  164. Song, Y.; Liu, P.; Shi, X.L.; Chu, Y.L.; Zhang, J.; Xia, J.; Gao, X.Z.; Qu, T.; Wang, M.Y. SARS-CoV-2 induced diarrhoea as onset symptom in patient with COVID-19. Gut 2020, 69, 1143–1144. [Google Scholar] [CrossRef] [Green Version]
  165. Organization (WHO). WHO issues consensus document on the epidemiology of SARS. Wkly. Epidemiol. Rec. Relev. Épidémiol. Hebd. 2003, 78, 373–375. [Google Scholar]
  166. Yeo, C.; Kaushal, S.; Yeo, D. Enteric involvement of coronaviruses: Is faecal-oral transmission of SARS-CoV-2 possible? Lancet Gastroenterol. Hepatol. 2020, 5, 335–337. [Google Scholar] [CrossRef] [Green Version]
  167. World Health Organization (WHO). Water, Sanitation, Hygiene and Waste Management for COVID-19. Technical Brief, 19 March 2020. 2020. Available online: https://apps.who.int/iris/bitstream/handle/10665/331305/WHO-2019-NcOV-IPC_WASH-2020.1-eng.pdf (accessed on 19 March 2020).
  168. Gu, J.; Han, B.; Wang, J. COVID-19: Gastrointestinal manifestations and potential fecal-oral transmission. Gastroenterology 2020, 158, 1518–1519. [Google Scholar] [CrossRef]
  169. Treibel, T.; Manisty, C.; Burton, M.; McKnight, A.; Lambourne, J.; Augusto, J.; Couto-Parada, X.; Cutino-Moguel, T.; Noursadeghi, M.; Moon, J.C. COVID-19: PCR screening of asymptomatic health-care workers at London hospital. Lancet 2020, 395, 1608–1610. [Google Scholar] [CrossRef]
  170. Kitajima, M.; Ahmed, W.; Bibby, K.; Carducci, A.; Gerba, C.P.; Hamilton, K.A.; Haramoto, E.; Rose, J.B. SARS-CoV-2 in wastewater: State of the knowledge and research needs. Sci. Total Environ. 2020, 739, 139076. [Google Scholar] [CrossRef]
  171. Jiang, X.; Luo, M.; Zou, Z.; Wang, X.; Chen, C.; Qiu, J. Asymptomatic SARS-CoV-2 infected case with viral detection positive in stool but negative in nasopharyngeal samples lasts for 42 days. J. Med. Virol. 2020, 92, 1807–1809. [Google Scholar] [CrossRef]
  172. Amirian, E.S. Potential fecal transmission of SARS-CoV-2: Current evidence and implications for public health. Int. J. Infect. Dis. 2020, 95, 363–370. [Google Scholar] [CrossRef] [PubMed]
  173. Viralzone. COVID-19 and Treatment. 2020. Available online: https://viralzone.expasy.org/9116 (accessed on 22 March 2020).
  174. Haas, C.N.; Rose, J.B.; Gerba, C.P. Quantitative Microbial Risk Assessment; John Wiley and Sons: Hoboken, NJ, USA, 2014. [Google Scholar]
  175. Hung, I.F.N.; Cheng, V.; Wu, A.; Tang, B.; Chan, K.; Chu, C.M.; Wong, M.; Hui, W.; Poon, L.; Tse, D.; et al. Viral loads in clinical specimens and SARS manifestations. Emerg. Infect. Dis. 2004, 10, 1550–1557. [Google Scholar] [CrossRef]
  176. Woelfel, R.; Corman, V.M.; Guggemos, W.; Seilmaier, M.; Zange, S.; Mueller, M.A.; Niemeyer, D.; Kelly, T.C.J.; Vollmar, P.; Rothe, C.; et al. Clinical presentation and virological assessment of hospitalized cases of coronavirus disease 2019 in a travel-associated transmission cluster. medRxiv 2020. [Google Scholar]
  177. Borchardt, M.A.; Spencer, S.K.; Kieke, B.A.; Lambertini, E.; Loge, F.J. Viruses in nondisinfected drinking water from municipal wells and community incidence of acute gastrointestinal illness. Environ. Health Perspect. 2012, 120, 1272–1279. [Google Scholar] [CrossRef] [Green Version]
  178. Sun, J.; Zhu, A.; Li, H.; Zheng, K.; Zhuang, Z.; Chen, Z.; Shi, S.; Zhang, Z.; Chen, S.; Liu, X.; et al. Isolation of infectious SARS-CoV-2 from urine of a COVID-19 patient. Emerg. Microbes Infect. 2020, 9, 991–993. [Google Scholar] [CrossRef] [PubMed]
  179. Jones, D.L.; Baluja, M.Q.; Graham, D.W.; Corbishley, A.; McDonald, J.E.; Malham, S.K.; Hillary, L.S.; Connor, T.R.; Gaze, W.H.; Moura, I.B.; et al. Shedding of SARS-CoV-2 in feces and urine and its potential role in person-to-person transmission and the environment-based spread of COVID-19. Sci. Total Environ. 2020, 749, 141364. [Google Scholar] [CrossRef]
  180. Yao, H.; Zhang, N.; Zhang, R.; Duan, M.; Xie, T.; Pan, J.; Peng, E.; Huang, J.; Zhang, Y.; Xu, X.; et al. Severity detection for the coronavirus disease 2019 (COVID-19) patients using a machine learning model based on the blood and urine tests. Front. Cell Dev. Biol. 2020, 8, 683. [Google Scholar] [CrossRef] [PubMed]
  181. Trajano Gomes da Silva, D.; Dias, E.; Ebdon, J.; Taylor, H. Assessment of recommendation for the containment and disinfection of human excreta in cholera treatment centers. Water 2019, 11, 188. [Google Scholar] [CrossRef] [Green Version]
  182. Mallapaty, S. How sewage could reveal true scale of coronavirus outbreak. Nat. Cell Biol. 2020, 580, 176–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Sayess, R.; Hychka, K.C.; Rahm, B.G. Presence of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the Virus Causing COVID-19, in Raw and Partially Treated Sewage. 2020. Available online: https://wri.cals.cornell.edu/sites/wri.cals.cornell.edu/files/shared/Final_Draft_051720.pdf (accessed on 2 February 2020).
  184. World Health Organization. Water, Sanitation, Hygiene and Waste Management for SARS-CoV-2, the Virus that Causes COVID-19. Available online: https://www.who.int/publications/i/item/WHO-2019-nCoV-IPC-WASH-2020.4 (accessed on 24 May 2021).
  185. Zhang, D.; Ling, H.; Huang, X.; Li, J.; Li, W.; Yi, C.; Zhang, T.; Jiang, Y.; Deng, S.; Zhang, X.; et al. Potential spreading risks and disinfection challenges of medical wastewater by the presence of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) viral RNA in septic tanks of Fangfang hospital. Sci. Total Environ. 2020, 741, 140445. [Google Scholar] [CrossRef]
  186. Wang, X.W.; Li, J.; Guo, T.; Zhen, B.; Kong, Q.; Yi, B.; Li, Z.; Song, N.; Jin, M.; Xiao, W.; et al. Concentration and detection of SARS coronavirus in sewage from Xiao Tang Shan Hospital and the 309th Hospital of the Chinese People’s Liberation Army. Water Sci. Technol. 2005, 52, 213–221. [Google Scholar] [CrossRef] [PubMed]
  187. Brock, T.D. How sensitive is the light microscope for observations on microorganisms in natural habitats? Microb. Ecol. 1984, 10, 297–300. [Google Scholar] [CrossRef]
  188. Yates, M.V. Manual of Environmental Microbiology; John Wiley and Sons: Hoboken, NJ, USA, 2020. [Google Scholar]
  189. Metcalf, L.; Eddy, H.P.; Tchobanoglous, G. Wastewater Engineering: Treatment, Disposal and Reuse; McGraw-Hill: New York, NY, USA, 1979; Volume 4. [Google Scholar]
  190. Wurtz, N.; Lacoste, A.; Jardot, P.; Delache, A.; Fontaine, X.; Verlande, M.; Annessi, A.; Giraud-Gatineau, A.; Chaudet, H.; Fournier, P.-E.; et al. Viral RNA in city wastewater as a key indicator of COVID-19 recrudescence and containment measures effectiveness. Front. Microbiol. 2021, 12, 664477. [Google Scholar] [CrossRef]
  191. Sikorski, M.J.; Levine, M.M. Reviving the “Moore swab”: A classic environmental surveillance tool involving filtration of flowing surface water and sewage water to recover Typhoidal Salmonella bacteria. Appl. Environ. Microbiol. 2020, 86. [Google Scholar] [CrossRef]
  192. Rafiee, M.; Isazadeh, S.; Mohseni-Bandpei, A.; Mohebbi, S.R.; Jahangiri-Rad, M.; Eslami, A.; Dabiri, H.; Roostaei, K.; Tanhaei, M.; Amereh, F. Moore swab performs equal to composite and outperforms grab sampling for SARS-CoV-2 monitoring in wastewater. Sci. Total Environ. 2021, 790, 148205. [Google Scholar] [CrossRef]
  193. Guerrero-Latorre, L.; Ballesteros, I.; Villacrés-Granda, I.; Granda, M.G.; Freire-Paspuel, B.; Ríos-Touma, B. SARS-CoV-2 in river water: Implications in low sanitation countries. Sci. Total. Environ. 2020, 743, 140832. [Google Scholar] [CrossRef] [PubMed]
  194. Balboa, S.; Mauricio-Iglesias, M.; Rodriguez, S.; Martínez-Lamas, L.; Vasallo, F.J.; Regueiro, B.; Lema, J.M. The fate of SARS-CoV-2 in wastewater treatment plants points out the sludge line as a suitable spot for in-cidence monitoring. medRxiv 2020. [Google Scholar]
  195. Or, I.B.; Yaniv, K.; Shagan, M.; Ozer, E.; Erster, O.; Mendelson, E.; Mannasse, B.; Shirazi, R.; Winter, E.; Nir, O.; et al. Regressing SARS-CoV-2 sewage measurements onto COVID-19 burden in the population: A proof-of-concept for quantitative environmental surveillance. medRxiv 2020. [Google Scholar]
  196. Kocamemi, B.A.; Kurt, H.; Sait, A.; Sarac, F.; Saatci, A.M.; Pakdemirli, B. SARS-CoV-2 detection in Istanbul wastewater treatment plant sludges. medRxiv 2020. [Google Scholar]
  197. Randazzo, W.; Cuevas-Ferrando, E.; Sanjuán, R.; Domingo-Calap, P.; Sánchez, G. Metropolitan wastewater analysis for COVID-19 epidemiological surveillance. Int. J. Hyg. Environ. Health 2020, 230, 113621. [Google Scholar] [CrossRef]
  198. Control, C.f.D.; Prevention. Interim Laboratory Biosafety Guidelines for Handling and Processing Specimens Associated with Coronavirus Disease 2019 (COVID-19). Available online: https://www.cdc.gov/coronavirus/2019-nCoV/lab/lab-biosafety-guidelines and https://www.archive.is/wip/5Ii97; (accessed on 31 March 2020).
  199. Haramoto, E.; Kitajima, M.; Katayama, H.; Ito, T.; Ohgaki, S. Development of virus concentration methods for detection of koi herpesvirus in water. J. Fish. Dis. 2009, 32, 297–300. [Google Scholar] [CrossRef]
  200. Ye, Y.; Ellenberg, R.M.; Graham, K.E.; Wigginton, K.R. Survivability, partitioning and recovery of enveloped viruses in untreated municipal wastewater. Environ. Sci. Technol. 2016, 50, 5077–5085. [Google Scholar] [CrossRef]
  201. Farkas, K.; Hillary, L.S.; Malham, S.K.; McDonald, J.E.; Jones, D.L. Wastewater and public health: The potential of wastewater surveillance for monitoring COVID-19. Curr. Opin. Environ. Sci. Heal. 2020, 17, 14–20. [Google Scholar] [CrossRef]
  202. Peccia, J.; Zulli, A.; Brackney, D.E.; Grubaugh, N.D.; Kaplan, E.H.; Casanovas-Massana, A.; Ko, A.I.; Malik, A.A.; Wang, D.; Wang, M.; et al. SARS-CoV-2 RNA concentrations in primary municipal sewage sludge as a leading indicator of COVID-19 outbreak dynamics. medRxiv 2020. [Google Scholar]
  203. Corman, V.; Bleicker, T.; Brünink, S.; Drosten, C.; Landt, O.; Koopmans, M. Diagnostic Detection of Wuhan Coronavirus 2019 by Real-Time RT-PCR; World Health Organization: Geneva, Switzerland, 2020. [Google Scholar]
  204. Arnaout, R.; Lee, R.A.; Lee, G.R.; Callahan, C.; Yen, C.F.; Smith, K.P.; Arora, R.; Kirby, J.E. SARS-CoV2 testing: The limit of detection matters. bioRxiv 2020. [Google Scholar]
  205. Centers for Disease Control and Prevention. Division of Viral Diseases, 2019-Novel Coronavirus (2019-nCoV) Real-Time rRT-PCR Panel Primers and Probes; Department of Health and Human Services: Washington, DC, USA, 2020.
  206. Corman, V.M.; Landt, O.; Kaiser, M.; Molenkamp, R.; Meijer, A.; Chu, D.K.; Bleicker, T.; Brünink, S.; Schneider, J.; Schmidt, M.L.; et al. Detection of 2019 novel coronavirus (2019-nCoV) by real-time RT-PCR. Eurosurveillance 2020, 25, 2000045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Arora, S.; Nag, A.; Sethi, J.; Rajvanshi, J.; Saxena, S.; Shrivastava, S.K.; Gupta, A.B. Sewage surveillance for the presence of SARS-CoV-2 genome as a useful wastewater based epidemiology (WBE) tracking tool in India. medRxiv 2020. [Google Scholar] [CrossRef]
  208. Kalikiri, M.K.; Hasan, M.R.; Mirza, F.; Xaba, T.; Tang, P.; Lorenz, S. High-throughput extraction of SARS-CoV-2 RNA from nasopharyngeal swabs using solid-phase reverse immobilization beads. medRxiv 2020. [Google Scholar]
  209. Wang, X.; Xiong, E.; Tian, T.; Cheng, M.; Lin, W.; Wang, H.; Zhang, G.; Sun, J.; Zhou, X. Clustered regularly interspaced short palindromic repeats/Cas9-mediated lateral flow nucleic acid assay. ACS Nano 2020, 14, 2497–2508. [Google Scholar] [CrossRef]
  210. Broughton, J.P.; Deng, X.; Yu, G.; Fasching, C.L.; Servellita, V.; Singh, J.; Miao, X.; Streithorst, J.A.; Granados, A.; Sotomayor-Gonzalez, A. CRISPR–Cas12-based detection of SARS-CoV-2. Nat. Biotechnol. 2020, 38, 870–874. [Google Scholar] [CrossRef] [Green Version]
  211. Kampf, G.; Todt, D.; Pfaender, S.; Steinmann, E. Persistence of coronaviruses on inanimate surfaces and their inactivation with biocidal agents. J. Hosp. Infect. 2020, 104, 246–251. [Google Scholar] [CrossRef] [Green Version]
  212. Casanova, L.; Rutala, W.A.; Weber, D.J.; Sobsey, M.D. Survival of surrogate coronaviruses in water. Water Res. 2009, 43, 1893–1898. [Google Scholar] [CrossRef]
  213. Gundy, P.M.; Gerba, C.P.; Pepper, I.L. Survival of coronaviruses in water and wastewater. Food Environ. Virol. 2008, 1, 10–14. [Google Scholar] [CrossRef] [Green Version]
  214. Wang, X.-W.; Li, J.-S.; Jin, M.; Zhen, B.; Kong, Q.-X.; Song, N.; Xiao, W.-J.; Yin, J.; Wei, W.; Wang, G.-J.; et al. Study on the resistance of severe acute respiratory syndrome-associated coronavirus. J. Virol. Methods 2005, 126, 171–177. [Google Scholar] [CrossRef]
  215. Radin, D. New trends in food- and waterborne viral outbreaks. Arch. Biol. Sci. 2014, 66, 1–9. [Google Scholar] [CrossRef]
  216. Sreenivasan, C.C.; Thomas, M.; Wang, D.; Li, F. Susceptibility of livestock and companion animals to COVID-19. J. Med. Virol. 2020, 93, 1351–1360. [Google Scholar] [CrossRef]
  217. Gollakner, R.; Capua, I. Is COVID-19 the first pandemic that evolves into a panzootic? Vet. Ital. 2020, 56, 11–12. [Google Scholar]
  218. Oreshkova, N.; Molenaar, R.-J.; Vreman, S.; Harders, F.; Munnink, B.B.O.; Honing, R.W.H.-v.; Gerhards, N.; Tolsma, P.; Bouwstra, R.; Sikkema, R. SARS-CoV2 infection in farmed mink, Netherlands, April and May 2020. bioRxiv 2020. [Google Scholar]
  219. Damas, J.; Hughes, G.M.; Keough, K.C.; Painter, C.A.; Persky, N.S.; Corbo, M.; Hiller, M.; Koepfli, K.-P.; Pfenning, A.R.; Zhao, H. Broad host range of SARS-CoV-2 predicted by comparative and structural analysis of ACE2 in vertebrates. bioRxiv 2020. [Google Scholar] [CrossRef]
  220. Rico, M.; Andrés-Costa, M.J.; Pico, Y. Estimating population size in wastewater-based epidemiology. Valencia metropolitan area as a case study. J. Hazard. Mater. 2017, 323, 156–165. [Google Scholar] [CrossRef] [PubMed]
  221. Ling, F.; Chen, L.; Zhang, L.; Yu, X.; Duvallet, C.; Isazadeh, S.; Dai, C.; Park, S.; Frois-Moniz, K.; Duarte, F. Microbial species abundance distributions guide human population size estimation from sewage meta-genomes. bioRxiv 2020. [Google Scholar]
  222. Donia, A.; Hassan, S.-U.; Zhang, X.; Al-Madboly, L.; Bokhari, H. COVID-19 Crisis Creates Opportunity towards Global Monitoring & Surveillance. Pathogens 2021, 10, 256. [Google Scholar] [PubMed]
  223. Gawlik, B.M.; Tavazzi, S.; Mariani, G.; Skejo, H.; Sponar, M.; Higgins, T.; Medema, G.; Wintgens, T. SARS-CoV-2 Surveillance Employing Sewers towards a Sentinel System, Feasibility assessment of an EU Approach, EUR 30684 EN; Publications Office of the European Union: Luxembourg, 2021. [Google Scholar]
  224. Smith, T.; Cassell, G.; Bhatnagar, A. Wastewater surveillance can have a second act in COVID-19 vaccine distribution. JAMA Health Forum 2021, 2, e201616. [Google Scholar] [CrossRef]
  225. Street, R.A.; Mathee, A.; Johnson, R.; Muller, C.; Louw, J.; Mdhluli, M.; Gelderblom, H.C.; Gray, G.E. COVID-19 vaccine roll-out in South Africa: The added value of wastewater surveillance for SARS. S. Afr. Med. J. 2021, 111, 524–525. [Google Scholar] [CrossRef]
  226. Hodgson, S.; Mansatta, K.; Mallett, G.; Harris, V.; Emary, K.; Pollard, A. What defines an efficacious COVID-19 vaccine? A review of the challenges assessing the clinical efficacy of vaccines against SARS-CoV-2. Lancet Infect. Dis. 2021, 21, e26–e35. [Google Scholar] [CrossRef]
Figure 1. Flow chart for the systematic literature search.
Figure 1. Flow chart for the systematic literature search.
Pathogens 10 00946 g001
Figure 2. Fecal shedding of SARS-CoV-2.
Figure 2. Fecal shedding of SARS-CoV-2.
Pathogens 10 00946 g002
Figure 3. Timeline for SARS-CoV-2 shedding and detection (adapted from [173]).
Figure 3. Timeline for SARS-CoV-2 shedding and detection (adapted from [173]).
Pathogens 10 00946 g003
Figure 4. Steps in detecting and enumerating of SARS-CoV-2 in sewage.
Figure 4. Steps in detecting and enumerating of SARS-CoV-2 in sewage.
Pathogens 10 00946 g004
Figure 5. The Moore swab [192].
Figure 5. The Moore swab [192].
Pathogens 10 00946 g005
Table 1. Selected data extraction fields employed in the literature review protocol.
Table 1. Selected data extraction fields employed in the literature review protocol.
StudyCountryPositive Stool Samples/Total (%)DiarrheaDuration of SARS-CoV-2 RNA Positive (Day)Duration of Persistent Positive Faecal RT PCR after Negative NP RT-PCR (Day)Same Pathogens Found in UrineViral LoadCt
TAN Xin et al. [21]China10/13 (77)NANA12NANANA
Cai Jiehao et al. [22]China5/6 (83)0/6NA18–30NoNANA
Ziying Lei et al. [23]China4/7 (57.1)5/20 (25)4–165-6NANANA
Jasper Fuk-Woo Chan et al. [24]China0/7NANANA0/7NANA
Yajun Yuan et al. [25]China6/6 (100)NA0-28NANANANA
Stephanie A. Kujawski et al. [26]United States7/10 (70)3/10 (30), all had viral RNA detected in stoolNANANoNA24.1–39.4
Francois-Xavier Lescure et al. [27]France2/5 (40)NANA10No6·8–8.1 log10 copiesNA
Wei Liu et al. [28]China0/19 (0)NANANANoNANA
Chunbao Xie et al. [29]China8/9 (88)0/9NANANoNANA
Lijuan Chen et al. [30]China1/1 (100)NANANANANANA
Tongqiang Zhang et al. [31]China3/3 (100)NA10–13NANANANA
An Tang et al. [32]China1/1 (100)NA1710NANAORF1ab: 32.6; Youjiang nucleoprotein gene: 33.7
Juan Liu et al. [33]China4/4 (100)NA8–11NANANA31.7–36.6
Wenling Wang et al. [34]China44/153 (29)NANANA0NA31.4
Xiao-Shan Wei et al. [35]China28/84 (33)26/84 (31)NANANANANA
Chaoqun Han et al. [36]China12/22 (54.5)
respiratory only: 3/5 (60), digestive + respiratory: 1/7 (14.3), and digestive only 8/10 (80)
117/206 (56)NANANANANA
Xiang Ma et al. [37]China8/27 (29.6)NA28NANANANA
Yuhan Xing et al. [38]China3/3 (100)NA288NANANA
Yu-pin Tan et al. [39]China3/10 (30)NA6–1011NANANA
Barnaby Edward Young et al. [13]China4/8 (50)3/17 (17)NANA0/10NA29–36
Iek Long Lo et al. [40] China10/10 (100)8/10 (80)2–179-110/10NANA
Fei Xiao et al. [41]China39/73 (53.49)26/39 (66)1–12NANANANA
Xiaowen Hu et al. [42]China0/59NANANA0/59NANA
Maria Effenberger et al. [43]Austria12/40 (30)22/40 (55)NANANANANA
JingCheng Zhang et al. [44]China5/14 (35.7)0/141–31NANANA
Michelle L. Holshue et al. [45]United States1/1 (100)1/1 (100)NANANANA36–38
Lu Lin et al. [46]China31/65 (47.7)23/56 (39.65)NANANANANA
Hu Yun et al. [47]China8/32 (25)NANANANANANA
Yun Ling et al. [48]China55/66 (82)NANANA4/58 (7)NANA
Youjiang Li et al. [49]China5/13 (38)1/13 (7)5–1515NANANA
Yuhan Xing et al. [50]China3/3 (10)NA1428NANANA
Yongjian Wu et al. [51]China41/74 (55)NA2811NANANA
Fengting Yu et al. [52]China80/116 (69)3/78 (4)NANANA17,429 ± 6920 copies/test34–38
Guangchang Pei et al. [53]ChinaNA108/333 (32.4)NANA0/251NANA
Liang Peng et al. [12]China2/9 (22)1/9 (11)NANA1/9 (11)3.22 × 102–5.42 × 104NA
Tomohiro Hosoda et al. [54]Japan1/1 (100)NA15NANA200 copies/wellNA
Chen Chen et al. [55]China22/133 (16)NANA13NANANA
Siew C Ng et al. [56]China21/21 (100)NANANANA2 × 9–7 × 1 log10 copies/mLNA
Mi Seon Han et al. [57]Korea2/2 (100)NA5–1718- 1.7 × 106–4.1 × 107 copies/mLNA
D. Paoli et al. [58]Italy0/1 (0)NANANA0/1NANA
Kelvin Kai-Wang To et al. [59]China4/23 (17)2/23 (8)NANANANANA
Liang Su et al. [60]China5/9 (55)NANANANANANA
Yifei Chen MD et al. [11]China28/42 (67)7/427–136–10NANANA
ZhoujiePeng et al. [61]China0/1 (0)NANANANANANA
Shufa Zheng et al. [62]China59/96 (59.3)10/96 (10)7–2821–28NA3.8–5 log10 copies/mLNA
Yang Pan et al. [63]China2/2 (100)NA5–6NANA104 to 107 copies/mLNA
Roman Wölfel et al. [64]Germany6/9 (67)NANANANA7.00 × 106 copies/mLNA
Jie Wang et al. [65]China1/2 (50)NANANANANA21.28
Yu-Han Xing et al. [66] China 2/3 (33.3)NA98-20NANANA
Wang-Da Liu et al. [67]China1/1 (100)NANANANANANA
Qing Cao et al. [68]China1/1 (100)NA15NA0/1NANA
Fang Liu et al. [69]China0/1 (0)NANANANANANA
Ryota Hase et al. [70]Japan1/1 (100)1/1 (100)5NANANANA
Tao Zuo et al [71]China7/15 (46)1/15 (6)NA6NA>3.2 × 104 copies/mLNA
Li-juan Mao, et al [72]China1/1 (100)0/1 (0)NANANANANA
Sarah Catherine Walpole et al [73]UK1/1 (100)NANANANANANA
Alvaro Mesoraca
et al [74]
Italy6/15 (40)NANANANANANA
Divjot S. Kumar et al. [75]Colombia0/1 (0)1/1 (100)NANANANANA
Hye Won Jeong et al. [76]Korea3/5 (60)NANANANA1.08 ± 0.16–2.09 ± 0.85 log10 copies/mL in urine 1.17 ± 0.32 log10 copies/mL in stoolNA
Garrett A.Perchetti et al. [77]United States20/20 (100)NANANANANA37.1–37.8
Xiaorong Wang et al. [78]China3/3 (100)NANA40NANANA
Yuanyuan Yu et al. [79]China1/2 (50)NANANANANANA
Fei Xiao et al. [80]China12/28 (42)NANANANANANA
Wenjun Du et al. [81]China7/10 (70)NA934NANANA
Maksim Kirtsman MDCM et al. [82]Canada1/2 (50)NANANANANANA
Jiufeng Sun et al. [83]China171/490 (34) NANANANANANA
Bao Fu et al. [84]China1/1 (100)NANANANANANA
Soo-kyung Park et al. [85]Korea2/46 (4) case
12/213 (5) sample
7/46 (15)50NANANA29.94
Jeong-Min Kim et al. [86]Korea15/74 (20)NANANANA79 ± 30 copy/μL for urine and 3176 ± 7208 copy/μL for stoolNA
Amani Mansour et al. [87]Beirut0/1 (0)NANANANANANA
Rigamonti Elia et al. [88]Switzerland0/1 (0)NANANANANANA
Seung-Man et al. [89]Korea(1/1) (100)(1/1) (100)421210/1NA33
Binder et al. [90]United States3/12(25)NANANANANANA
Han et al. [91]South Korea10/12(83)NA2172/12 (17)
3.82–7.55 log10 copies/mL
4.1–10.27 log10 copies/mLNA
Colavita et al. [92]Italy2/2 (100)NA15–1730/2NA30.1–38.59
Slaats et al. [93]The Netherlands1/1 (100)NA4223NANANA
Italiano et al. [94]United States1/1 (100)1/1 (100)2170/1NANA
Scutari et al. [95]Italy2/2 (100)NANANA1/2 (50)6–52 copies/mL for NNA
Hinojosa-Velasco et al. [96]Mexico1/1 (100)NANANANANANA
Zhang et al. [97]China93/258 (36)47/82 (57.3)28–35 NANANACT: 19–30
Wang et al. [98]China20/69 (28)NA259NANACT: 25–27
Npvazzi et al. [99]Italy11/107 (10.3) NANANA1/85 (1.2)4.1 × 106 copies/mlNA
Chen et al. [100]China1/4 (25)NA100NANANANA
Khoury et al. [101]United States1/1 (100)NANANANANANA
Chen et al. [102]China52/97 (53.61)6/97 (6.19)NA14.13 ± 8.61NANANA
Barth et al. [103]China(50)NANANANANANA
Fumian et al. [104]United States33/121 (28)NANANANANANA
Chu et al. [105]China1/1 (100)NANANANANANA
Table 2. Selected data extraction fields employed in the literature review protocol.
Table 2. Selected data extraction fields employed in the literature review protocol.
StudyCountrySite of SamplingSample SizeVirus Concentration MethodVirus Detection MethodQuantitative Data
Wurtzer S et al. [106]Francethree wastewater treatment plantsNAcentrifugation and filtration stepRT-qPCR5.104 to 3.106 GU/L
Warish Ahmed et al. [15]Australiasuburban pumping station andurban wastewater treatment plants100–200 mLdirect RNA extraction from electronegative membranes and ultrafiltrationRT-qPCR1.9–12 copies/100 mL
Bilge Alpaslan Kocamemi et al. [107]Turkeywastewater treatment plants and manholes250 mLAmicon® Ultra-15 with 10 KDa cutoff ultrafiltration unitsRT-qPCR2.89 × 103–9.33 × 104 (Virus titer/liter)
Sara Giordana Rimoldi et al. [108]Italyrivers and wastewater treatment plantsNAglass fiber filtrationRT-PCR-cell cultureNA
Gertjan Medema et al. [17]The Netherlandswastewater treatment plant250 mLcentrifugation and filtration stepRT-PCRNA
Walter Randazzo et al. [109]SpainWWTPsNAcentrifugation and filtration stepRT-qPCR5.38 ± 0.21 log genomic copies/L
Wu FQ et al. [110]United Statesurban wastewaterNAcentrifugation and filtration stepqPCR6 log units
Artem Nemudryi et al. [111]United Statespre-treated wastewater500 mLfiltrationRT-qPCRNA
Willemijn Lodder et al. [112]The Netherlandsairport wastewater10 LNART-PCRNA
Wurtzer S et al. [113]France wastewater treatment plant11 mLultracentrifugationRT-PCRNA
Manupati Hemalatha [114]Indiaraw sewage100–500 mLfiltrationRT-qPCR30,818–266,360 copies/L
Kyle Curtis
[115]
Englandraw and treated wastewater50 mL of raw wastewater 200 mL of treated final effluentelectronegative filtrationRT-ddPCR159 copies/100 mL
Pei-Ying Hong
[116]
Saudi Arabiahospital wastewater250 to 500 mLHA membrane and filtrationqPCR6.89-log copies/L
Hyatt Green
[117]
Syracusewastewater treatment plants, influent pump stations, or interceptor lines1.9 LultracentrifugationRT-qPCR42.7 genomes/mL
Salmaan Sharif
[118]
Pakistanraw sewage500 mLcentrifugedRT-qPCRNA
Rimoldi et al. [119]Italyraw and treated wastewaterNAfiltrationRT-PCRNA
Medema et al. [120]The Netherlandssewage250 mLultrafiltration RT-PCR2.6–2.2 × 103
Prado et al. [121]Brazilraw sewageNANART-qPCRCt: 36.3–39.8.
SP Sherchan et al. [122]United Statesuntreated and treated wastewater1 Lultrafiltration, and an adsorption–elution method using electronegative membranesRT-qPCR 3.2 log10 copies/L from the N1 and 2.5 and 3.0 log10 copies/L from the N2 assay
Kumar et al. [75]Indiawastewater treatment plant50 mLfiltration and poly ethylene glycol (PEG) methodsPCR5.6 × 10 3.5 × 102 copies/L
Trottier et al. [123]Francewastewater upstream of the main wastewater treatment plant50 mLcentrifugation and 40 μm cell strainer RT-qPCRNA
Mlejnkova et al. [124]Czech Republicwastewater treatment plant500 mL flocculation using beef extract solutionRT-qPCRCq: 34–40
Haramoto et al. [125]Japaninfluent and secondary-treated wastewater1 Lelectronegative membranevortex (EMV) prior to the filtrationRT-qPCR1.4 × 102−2.5 × 103 copies/L
Rosa et al. [126]Italyraw sewage250 mLPEG dextran RT-PCRNA
Gonzalez et al. [127] United Stateswastewater treatment plant1 LfiltrationRT-ddPCR101–104 copies/100 mL
Miyani et al. [128]United Stateswastewater treatment plant1 LNanoCeram electropositive cartridge filtersRT-PCR104–105 genomic copies/L
Westhaus et al. [129]Germanywastewater treatment plant45 mLcentrifugation and filtration stepsRT-PCR3–20 gene equivalents/mL in the inflow, and 2.7–37 gene equivalents in the effluent
D’Aoust et al. [130]CanadaInfluent post grit solids and primary clarified sludge in two water resource recovery facilities6 L1.5 µm glass fiber filter followed by a 0.45 µm GF6 mixed cellulose ester filterRT-qPCR and RT-ddPCR1.7 × 103 to 3.8 × 105 copies/L
Hata et al. [131]Japanwastewater treatment plants80 mLpolyethylene glycol precipitationTaqMan-based qRT-PCR1.2 × 104–3.5 × 104 copies/L
Jafferali et al. [132]Swedenuntreated municipal wastewaters NAultrafiltration, double ultrafiltration, adsorption-extraction, centrifugation combined with adsorption-extractionRT-qPCRCq: 35.71–38.15
Hasan et al. [133]United Arab Emiratesinfluents and treated effluents of wastewater treatment plants and untreated wastewater250 mLultrafiltrationRT-qPCR2.86 × 102–3.4 × 104 copies/L
Baldovin et al. [134]Italymunicipal wastewater treatment plant1000 mLultrafiltrationRT-qPCR4.8-4.9 log10 gc/L
Feng et al. [135]Chinaflushing in the toilet and in sewage pipes15 mL NART-qPCR3092 copies/mL in sewage/wastewater
Kumar et al. [136]Indiawastewater treatment plantsNAfiltration and polyethylene glycol precipitationRT-qPCRCt: 22–37
Saguti et al. [137]Swedeninfluent and effluent wastewater treatment plant 500 mLadsorption to milk powder and to Nano-Ceram filterRT-qPCR0.14–16 log10/L
Gallardo-Escárate et al. [138]Southern Chileuntreated wastewater10 Lfiltration and centrifugationRT-PCR4 × 103–7 × 103 Genome Unit/L
Gonçalves et al. [139]Sloveniahospital wastewater1 LfiltrationRT-PCRCt: 29.65–37.03 for RdRP, 33.61–38.65 for E
Martin et al. [140]Englandsewage plant1 Lfiltration–centrifugationRT-qPCR3.50 and 4.20 Log10 gc/L
Peccia et al. [141]United Statesprimary sewage sludge40 mL-qRT-PCR1.7 × 103 mL−1 to 4.6 × 105 mL−1
Albastaki et al. [142]United Arab Emirateseffluent of wastewater treatment plant1 Lfiltration–centrifugationRT–PCRCt: 32–36
Agrawal et al. [143]Germanywastewater treatment plantsNAfiltrationRT-PCR3 × 1013–2 × 1014 copies/day
Abu Ali et al. [144]Israelwastewater treatment plantsNAultrafiltrationRT-qPCR7 × 103 copies/mL
Kocamem et al. [145]Turkeyinfluent, effluent, primary and waste activated sludge250 mLfiltration–centrifugation then PEGRT-qPCR2.6 × 102–8.2 × 106 titers/L
Al et al. [146]Canadasewer shed site and influent100 mLcentrifugationRT-qPCR6.6 × 104–1.4 × 104 GU L-1
Iglesias et al. [147]Argentinaraw water samples500 mLcentrifugation and PEGRT-qPCRNA
Wu et al. [148]Chinaraw sewageNAfiltrationRT-PCR<90 copies/mL
Lara et al. [149]The Netherlandssewage100–200 mLultrafiltrationRT-qPCRCt: 32.5–34.6
Garham et al. [150]United Statesinfluent and primary settled solids50 mLPEG precipitationddPCR and RT-QPCR730–4250 cp/g for N1
Hokajärvi et al. [20]Finlandinfluent of wastewater treatment plant2–5 LultrafiltrationRT-qPCR8.1 log10 copies 100 mL−1
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Amereh, F.; Negahban-Azar, M.; Isazadeh, S.; Dabiri, H.; Masihi, N.; Jahangiri-rad, M.; Rafiee, M. Sewage Systems Surveillance for SARS-CoV-2: Identification of Knowledge Gaps, Emerging Threats, and Future Research Needs. Pathogens 2021, 10, 946. https://doi.org/10.3390/pathogens10080946

AMA Style

Amereh F, Negahban-Azar M, Isazadeh S, Dabiri H, Masihi N, Jahangiri-rad M, Rafiee M. Sewage Systems Surveillance for SARS-CoV-2: Identification of Knowledge Gaps, Emerging Threats, and Future Research Needs. Pathogens. 2021; 10(8):946. https://doi.org/10.3390/pathogens10080946

Chicago/Turabian Style

Amereh, Fatemeh, Masoud Negahban-Azar, Siavash Isazadeh, Hossein Dabiri, Najmeh Masihi, Mahsa Jahangiri-rad, and Mohammad Rafiee. 2021. "Sewage Systems Surveillance for SARS-CoV-2: Identification of Knowledge Gaps, Emerging Threats, and Future Research Needs" Pathogens 10, no. 8: 946. https://doi.org/10.3390/pathogens10080946

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop