Skip to main content
F1000Research logoLink to F1000Research
. 2021 Nov 2;10:231. Originally published 2021 Mar 24. [Version 2] doi: 10.12688/f1000research.51592.2

SARS-CoV-2 and the role of orofecal transmission: a systematic review

Carl J Heneghan 1, Elizabeth A Spencer 1,a, Jon Brassey 2, Annette Plüddemann 1, Igho J Onakpoya 1, David H Evans 3, John M Conly 4, Tom Jefferson 1
PMCID: PMC8749895  PMID: 35035883

Version Changes

Revised. Amendments from Version 1

Thank you to both reviewers who offered suggestions to improve the manuscript; we have made additions and edits to include their suggestions. We have added a definition for viral load. We have added an explanation that these studies do not provide an estimate of infectious virus concentration and that it is not reasonable to deduce human transmission risk from PCR positive samples. We note that there may be overlap in included studies, but that we have no adequate means to identify this; we also explain that the reviews may also overlap. We note that limited data on sex or by age-group precluded additional investigation into these factors. We are grateful that the error in the table was noticed and we have deleted this unnecessary total number from the table. The issue of how many studies were available as preprints and how these varied by study type is interesting but beyond the scope of this review, not least because this changes over time. The next time we update this review we hope to have a better breakdown of those fully published, and hopefully a better bank of evidence to analyse.

Abstract

Background: Modes of transmission of SARS-CoV-2 are of key public health importance. SARS-CoV-2 has been detected in the feces of some COVID-19 patients, suggesting the possibility that the virus could, in addition to droplet and fomite transmission, be transmitted via the orofecal route.

Methods: This review is part of an Open Evidence Review on Transmission Dynamics of COVID-19. We conduct ongoing searches using WHO COVID-19 Database, LitCovid, medRxiv, and Google Scholar; assess study quality based on five criteria and report important findings. Where necessary, authors are contacted for further details on the content of their articles.

Results: We include searches up until 20 December 2020. We included 110 relevant studies: 76 primary observational studies or reports, and 35 reviews (one cohort study also included a review) examining the potential role of orofecal transmission of SARS-CoV-2. Of the observational studies, 37 were done in China. A total of 48 studies (n=9,081 patients) reported single cases, case series or cohort data on individuals with COVID-19 diagnosis or their contacts and 46 (96%) detected binary RT-PCR with 535 out of 1358 samples positive for SARS-CoV-2 (average 39.4%). The results suggest a long duration of fecal shedding, often recorded after respiratory samples tested negative, and symptoms of gastrointestinal disease were reported in several studies. Twenty-nine studies reported finding SARS-CoV-2 RNA in wastewater, river water or toilet areas. Six studies attempted viral culture from COVID-19 patients’ fecal samples: culture was successful in 3 of 6 studies, and one study demonstrated invasion of the virus into intestinal epithelial cells.

Conclusions: Varied observational and mechanistic evidence suggests SARS-CoV-2 can infect and be shed from the gastrointestinal tract, including some data demonstrating viral culture in fecal samples. To fully assess these risks, quantitative data on infectious virus in these settings and infectious dose are needed.

Keywords: Orofecal, transmission, COVID-19, SARS-CoV-2, systematic review

Introduction

Understanding how, when and in what types of settings SARS-CoV-2 spreads between people is critical to developing effective public health and infection prevention and control measures to break the chains of transmission. Current evidence suggests SARS-CoV-2 is primarily transmitted via respiratory droplets and fomites between infected individuals and others in close contact 1 .

SARS-CoV-2 has been shown to contaminate and survive on certain surfaces, and has also been detected in the feces of some patients, suggesting the possibility that SARS-CoV-2 could transmit via the orofecal route, including potentially via contamination of food. It is well recognized that coronaviruses are major pathogens in many mammalian species and predominantly target epithelial lining cells in the respiratory and gastrointestinal (GI) tracts. Many animal coronaviruses are transmitted by the fecal-oral route and there are several reports of intestinal disease associated with SARS-CoV-1 and other human coronaviruses. Main causes include lack of adequate sanitation and poor hygienic practices. Identifying infectious virus and quantifying viral load within human GI tissues, feces, and contaminated materials including fomites and sewage would help understand the potential for transmission. We aimed to systematically review the evidence on orofecal SARS-CoV-2 transmission. Terminology for this article can be found in Box 1.

Box 1. Terminology.

Orofecal: describes a route of transmission where the virus in fecal particles can pass from one person to the mouth of another.

Viral load:A measure of the number of viral particles present in an individual typically quantified by the number of virions, gene copies or equivalents, or amount of viral proteins present.

Cycle threshold: The number of cycles required for the fluorescent signal to cross the threshold. Ct levels are inversely proportional to the amount of target nucleic acid in the sample.

Methods

We are undertaking an open evidence review investigating factors and circumstances that impact on the transmission of SARS-CoV-2, based on our protocol (see Extended data: Appendix 1 2 ). For the original protocol, see https://www.cebm.net/evidence-synthesis/transmission-dynamics-of-covid-19/. In brief, this review aims to identify and evaluate relevant articles (peer-reviewed or awaiting peer review) that examine the mode of viral transmission and ecological variables influencing the mode of transmission. We conduct an ongoing search using WHO COVID-19 Database, LitCovid, medRxiv and Google Scholar for keywords and associated synonyms. Results are reviewed for relevance and for articles that looked particularly relevant forward citation matching was undertaken and relevant results were identified. Studies with modelling are only included if they report transmission outcome data and not predicted outcomes (see further details of the search strategy in the Extended data: Appendix 2 2 ). Searches are updated every two weeks.

We extracted data on the type of study, setting, sample source and methods, fecal PCR positive samples for SARS-CoV-2 RNA including cycle threshold (including methods), symptom chronology in relation to PCR testing and/or taking samples and viral culture. We tabulated the data and summarised data narratively by mode of sample. We assessed quality using a modified QUADAS-2 risk of bias tool 3 . We simplified the tool as the included studies were not designed as primary diagnostic accuracy studies and assessed study quality based on five criteria. Where necessary we write to authors of included studies for further details or clarification on the content of their articles. Meta-analyses were not performed, due to the variability of available data. The protocol was last updated on 1 December 2020 ( Version 3: 1 December 2020).

Results

This update includes searches up until 20 December 2020 (see Figure 1). We identified 110 relevant studies (see Extended data: Appendix 3 for references of included studies 2 ): 76 primary studies or reports, and 35 reviews, one of which also reported primary study results from a cohort study [Cheung K 2020].

Figure 1. Study flow diagram.

Figure 1.

Reviews

The included reviews summarised a range of observational studies including studies of detection of SARS-CoV-2 RNA in fecal samples of individuals testing positive for SARS-CoV-2 in respiratory samples, frequency of GI symptoms among those with COVID-19, and observations of SARS-CoV-2 RNA in toilets and wastewaters. The reviews included overlapping studies and must therefore not be considered as entirely additional information. Five followed systematic review methodology and reporting [Edwards 2020, Karia 2020, Pamplona 2020, Parasa 2020, Santos 2020]. The quality of the other reviews was low to moderate, with none assessing included study quality, and with reporting of methods often missing or very limited.

None of these reviews focussed on the infectiousness (and hence transmission potential) of SARS-CoV-2 identified in fecal or wastewater samples. A review on the potential for foodborne transmission of SARS-CoV-2 found no published studies of SARS-CoV-2 survival in or on food products. The totality of the reviews’ evidence shows that the SARS-CoV-2 RNA is commonly present in stool samples of COVID-19 patients but it is unknown if this represents primary invasion of enterocytes or simply saliva and sputum that has been swallowed and is transiting it way through the GI tract. The presence of viral RNA in the feces does not allow any conclusions to be drawn about infectiousness. The contribution of orofecal transmission to viral spread in the pandemic has not been established or quantified.

Primary studies

Quality of included studies. Overall the quality of the evidence was low to moderate mainly due to a lack of standardisation of techniques, omissions in reporting and a failure to account for biases in the research process (see Table 3; Figure 2). Sample sources were clear in two-thirds of studies (65.8%). Several studies mention the possibility of bias influencing their findings but did not use strategies (design or analysis) to deal with bias, and as such have been recorded as unclear risk of bias.

Figure 2. Risk of bias chart.

Figure 2.

Results. A total of 48 (n=9,081 patients) studies reported single cases, case series or cohort data on individuals with COVID-19 diagnosis or their contacts and 46 studies (96%) detected binary RT-PCR with 535 out of 1358 samples RT-PCR positive for SARS-CoV-2 (average 39.4%). All but five studies were in hospitalized patients; 31 were done in China; the others were in East Asia, South East Asia, South Asia, USA and Europe (see Table 1).

Table 1. Included study characteristics: primary studies.

Study ID cohorts
or case series
(n=48)
Setting Country Population/
environment
Patient
numbers
Fecal samples PCR-
positive n/d for
SARS-CoV-2 RNA
unless otherwise
stated
Live
culture
(positivity
rate)
Genome
sequencing
methods
Genome Sectioning /phylogeny
results where available
Chan 2020 Household China Family members 6 0/6 Not
attempted
Yes Two complete virus genomes
(HKU-SZ-002a and HKU-SZ-005b)
were sequenced & showed a novel
coronavirus that is most closely
related to those of a bat SARS-like
coronavirus. Exploratory WGS
so not confirming similarity with
previous studies.
Chen C 2020 Hospital China 121 adults, 22 children 133 22/133 Not
attempted
no
Chen W 2020 Hospital China Adult patients with
pneumonia
57 11/28 Not
attempted
no
Chen Y 2020 Hospital China Hospitalised adult
patients
42 28/42 Not
attempted
no
Cheung CCL 2020 Hospital Singapore Hospitalised adult
patients
2 N/A Not
attempted
no
Cheung K 2020 Public Hong Kong Cohort and systematic
review
52 9/52 Not
attempted
no
Cho 2020 Hospital
paediatric
department
S Korea Case study of
hospitalised infant
1 1/1 Not
attempted
no
Chu H 2020 Hospital China Case study of
breastfeeding woman
1 1/1 Not
attempted
no
COVID Ix Team Hospital and
community
USA Patients in six US states 12 7/10 Not
attempted
no
Ge 2020 Hospital Japan Case reports of one
hospitalised Covid-19
patient
1 1/1 Not
attempted
no
Han C 2020 Hospital China Patients diagnosed with
low severity Covid-19
206 12/22 Not
attempted
no
Hayee 2020 Hospital
outpatients
UK Patients (non-Covid-
19) attending for GI
endoscopy
6,208 N/A Not
attempted
no
Hoehl S 2020 Childcare
centres
Germany Children and staff of
childcare settings
1,197 1/5,907 Not
attempted
no
Holshue 2020 Hospital USA Case report 1 1/1 Not
attempted
no
Jeong 2020 Hospital Korea Specimens from 5
patients positive by
qPCR
5 5/5 yes (0/3) no
Jiehao 2020 Children's
hospital
China Children 10 5/6 Not
attempted
no
Kim J-M 2020 Hospital South Korea Hospitalised adult
patients
74 8/74 yes (0/13) no
Lescure 2020 Hospital France Case series of
hospitalised adult
patients
5 2/5 Not
attempted
no
Li 2020 Hospital China Case series of
hospitalised adult
patients
29 4/29 Not
attempted
no
Ling 2020 Hospital China 66 recovered patients 66 11/28 Not
attempted
no
Lo 2020 Hospital Macau,
China
Hospitalised adult
patients
10 10/10 Not
attempted
no
Nicastri 2020 Hospital Italy Community 1 1/1 Not
attempted
no
Pan 2020 Hospital China Hospitalised adult
patients
17 9/17 Not
attempted
no
Peng 2020 Hospital China Hospitalised adult
patients
9 2/9 Not
attempted
no
Qian 2020 Hospital China A case report of a
patient admitted
1 0/1 No, but
virions
observed
no
Senapati 2020 Hospital India Hospitalised adult
patients
12 12/12 Not
attempted
no
Tan 2020 Hospital Vietnam Hospitalised adult
patient
1 1/1 Not
attempted
no
Tang 2020 Community China Asymptomatic child 1 1/1 Not
attempted
no
Wang Q-X 2020 Hospital China Covid-19 patients with
-ve respiratory tract but
+ve fecal samples
5 5 Not
attempted
no
Wang S 2020 Hospital China Retrospective study of
17 hospitalised
Covid-19 patients
17 11 Not
attempted
no
Wang W, Xu Y 2020 Hospital China Patients in three
hospitals
205 1/6 yes (2/4) no
Wang X, Zheng J
2020
Hospital China Hospital Covid-19
patients
69 20/69 Not
attempted
no
Wang X, Zhou Y
2020
Hospital China Case reports on
readmitted adult
3 3/3 Not
attempted
no
Wolf 2020 Household Germany Family cluster 5 2/5 Not
attempted
no
Wölfel 2020 Hospital Germany Hospitalised adult
patients
9 9/9 yes (0/13) no
Wu Y 2020 Hospital China Hospitalised adult
patients
74 41/74 Not
attempted
no
Xiao F, Tang M 2020 Hospital China Hospitalised children
and adults
71 39/73 Not
attempted
no
Xiao F, Sun J 2020 Hospital China Hospitalised adult
patients
28 12/28 yes (2/3) Yes Obtained full-length viral
genome sequence (GenBank
accession no. MT123292) by using
next-generation sequencing.
The sequenced showed 5 nt
substitutions compared with the
original Wuhan strain (GenBank
accession no. NC045512.2)
Xing Y 2020 Paediatric
hospital
China Children 3 3/3 Not
attempted
no
Xu Y 2020 Hospital China Children 10 8/10 Not
attempted
no
Yang 2020 Hospital China Paediatric Covid-19
patients
35 17/35 Not
attempted
no
Young 2020 Hospital Singapore Hospitalised adult
patients
18 4/8 Not
attempted
no
Yuan 2020 Hospital China Children 78 37/78 Not
attempted
no
Zhang J 2020 Hospital China Patients with Covid-19
pneumonia.
14 14/14 Not
attempted
no
Zhang T 2020 Public
hospital
China Children post discharge 3 3/3 Not
attempted
no
Zhang W 2020 Pulmonary
hospital
China Hospitalised adult
patients
15 4/15 Not
attempted
no
Zhang Y, Chen C,
Zhu S 2020
Hospital China Adult severe pneumonia
case report
1 1 yes (1/1) Yes full-length genome sequence on
the one specimen using ABI 3130
Genetic Analyzer
Zhang Y, Chen C,
Song Y 2020
Hospital China PCR-confirmed Covid-19
patients with clinical
symptoms
258 93 [reports
same live
culture
as above
report]
Yes full-length genome sequence on
the one specimen using ABI 3130
Genetic Analyzer
Sewage 9081
Agrawal 2020 Wastewater
treatment
plants
Germany Two wastewater influent
sources
N/A Sewage Not
attempted
no
Ahmed 2020 Sewage Australia Wastewater N/A Sewage Not
attempted
no
Ampuero 2020 Two
wastewater
treatment
plants
Chile Wastewater N/A Sewage Not
attempted
no
Arora 2020 Wastewater India 14 wastewater
treatment plants
N/A Sewage Not
attempted
no
Betancourt 2020 Wastewater USA University campus
wastewater system
N/A Wastewater Not
attempted
no
Chavarria-Miro 2020 Sewage Spain Wastewater N/A Sewage Not
attempted
no
Curtis 2020 Wastewater USA Community wastewater
services
N/A Wastewater Not
attempted
no
Fongaro 2020 Sewage Brazil Urban sewage service N/A Sewage Not
attempted
no
Fernandez-de-Mera
2020
Wastewater Spain Village community and
wastewater
N/A Wastewater Not
attempted
no
Haramoto 2020 Wastewater
and river
water
Japan Wastewater and river
water
N/A Wastewater only
(river samples
negative)
Not
attempted
no
Hata 2020 Wastewater Japan Wastewater N/A Wastewater Not
attempted
no
Iglesias 2020 Urban raw
surface water
contaminated
with
wastewater
Argentina Raw surface water
samples in a low-
income urban
community
N/A Wastewater-
contaminated
surface water
Not
attempted
no
Izquierdo-Lara 2020 Sewage Belgium
& The
Netherlands
Sewage from a number
of sewage facilities
N/A Sewage Not
attempted
Yes To further investigate genetic
diversity of SARS-CoV-2, 55
wastewater samples from
13 different locations in the
Netherlands (48 samples) and 7
different locations in Belgium (7
samples) with Ct values of <36
were selected for whole genome
sequencing using Nanopore
sequencing.
La Rosa 2020b Sewage Italy Water environments N/A Sewage Not
attempted
no
Medema 2020 Sewage The
Netherlands
Public, cities and airport N/A Sewage Not
attempted
no
Neault 2020 Wastewater Canada Wastewater samples
from two urban
wastewater recovery
facilities
N/A Wastewater Not
attempted
no
Peccia 2020 Sewage USA Primary sewage sludge
sampled over time
N/A Sewage Not
attempted
no
Sharif 2020 Wastewater Pakistan Wastewater & drainage
samples from across
the country
N/A Wastewater Not
attempted
no
Shutler 2020 Waterborne 32 countries Faecal contaminated
water
N/A Sewage Not
attempted
no
Trottier 2020 Wastewater France Urban wastewater
treatment plant
N/A Wastewater Not
attempted
no
Wang J 2020 Hospital China Surfaces and sewage
samples
N/A Sewage yes (0/5) no
Wurtzer 2020 Community France Wastewater N/A Sewage Not
attempted
no
Zhao 2020 Wastewater
systems
China Municipal and hospital
wastewater systems
N/A influent, effluent,
sludge
Not
attempted
no
Toilets (n=4)
Del Brutto 2020 Community Guatemala Rural low-income village
community
362 N/A Not
attempted
no
Ding Z 2020 Infectious
diseases
hospital
China Surface samples N/A Toilet samples Not
attempted
no
Kang 2020 Building
plumbing
system
China Residential high-rise
apartment block
N/A Air samples around
drainage system
Not
attempted
no
Ong 2020 Hospital Singapore Sampling in the physical
areas around patients
3 Sewage toilet
samples
Not
attempted
no
Waterways (n=1)
Guerrero-Latorre 2020 Rivers Guatemala Urban streams in low
sanitation setting
N/A River samples Not
attempted
no

PCR testing for SARS-CoV-2 RNA in fecal samples in hospitalised COVID-19 patients. Of the 43 hospital studies, 41 (95%) detected binary RT-PCR, with 522 positive tests out of 1293 fecal samples (average 40.4%) from COVID-19 patients (see Table 4). These studies were mainly small case series, they included patients of a range of ages from infancy to elderly and with widely varying severity of disease, and the proportion of fecal samples varied from 1 (nine studies) to 258 [Zhang Y, Chen C], and the proportion testing positive for SARS-CoV-2 RNA varied from 14% to 100% across studies. One study that identified SARS-CoV-2 RNA in fecal samples among 39 of 73 hospitalised COVID-19 patients, also studied the gastric, duodenal and rectal epithelia of one patient using specimens collected via endoscopy [Xiao F, Tang M 2020].

Immunofluorescence data showed that ACE2 protein, proven to be a cell receptor for SARS-CoV-2, was abundantly expressed in the glandular cells of gastric, duodenal, and rectal epithelia, supporting entry of SARS-CoV-2 into the host cells. Intracellular staining of viral nucleocapsid protein in gastric, duodenal, and rectal epithelium showed that SARS-CoV-2 infects these GI glandular epithelial cells. Viral RNA was also detected in esophageal mucous tissue, but a lack of viral nucleocapsid protein staining in esophageal mucosa suggested low viral infection there. Viral nucleocapsid protein in rectal epithelial cells was detected in specimens from some additional COVID-19 patients, suggesting that infectious SARS-CoV-2 can survive the GI environment.

GI symptoms among COVID-19 patients

Reporting of GI symptoms among COVID-19 patients is frequent but not consistent within these studies (diagnosis is typically based on fever, respiratory symptoms and the results of PCR testing in respiratory swabs, so recording GI symptoms may not be routine). However, several observational studies report the presence of GI symptoms among COVID-19, including Chan 2020, Cheung 2020 and Han C 2020. GI symptoms do not necessarily correlate in severity or time with other COVID-19 disease symptoms.

Timing and duration of fecal shedding

Fecal shedding of SARS-CoV-2 has been reported throughout the disease course and also continuing after respiratory samples tested negative. A five-person family with a confirmed COVID-19 case was hospitalized and observed: the parents and two children aged two and five years became infected but the youngest child was not infected. These children shed infectious virus via the respiratory system, and this shedding observed in the nasopharynx cleared after five to 6 days; however, viral RNA was continuously detected in the children’s stool for more than four weeks [Wolf 2020]. Tang 2020 et al. reported an apparently asymptomatic (no fever or cough) 10-year-old child, from whom, 17 days after the last close contact with individuals testing positive for SARS-CoV-2, a fecal sample was positive for SARS-CoV-2 RNA. A retrospective study of 133 hospitalised COVID-19 patients identified 22 whose sputum or fecal samples tested positive after pharyngeal swabs became negative [Chen C 2020]. A study of 59 hospitalised COVID-19 patients reported that fecal discharge of SARS-CoV-2 RNA continued long after respiratory shedding had ceased [Cheung 2020]. Among fecal samples from 69 hospitalized COVID-19 patients, 20 tested PCR positive; SARS-CoV-2 RNA persisted for significantly longer in fecal samples than in oropharyngeal swabs [Wang X, Zheng J 2020].

The role of aerosol-generating procedures in relation to orofecal transmission. One study reported the results of PCR tests among participants attending for upper GI endoscopy at a UK hospital [Hayee 2020]. Only individuals testing PCR negative in a nasopharyngeal swab underwent their booked procedure. Post-procedure follow-up of 6,208 patients at one and two weeks identified no incident case of COVID-19.

Viral culture

Viral culture using fecal samples was attempted in 6 studies [Jeong 2020, Kim JM 2020, Wang W, Xu Y 2020; Wölfel 2020; Xiao F 2020 and Zhang Y 2020], three reported culture of SARS-CoV-2 from stool samples [Wang W, Xu Y 2020; Xiao F 2020, Zhang Y 2020], and three did not [Jeong 2020, Kim JM 2020 and Wölfel 2020].

Wang W 2020 et al. reported two of four viral culture samples were positive, however, no culture methods were reported; electron microscopy was performed suggesting the presence of the virus, but this observation does not show that the virus was viable. In Xiao F Sun J 2020, viral culture was attempted from an unreported number of specimens and cases and a cytopathic effect in Vero E cells was observed 2 days after a second-round passage. Zhang Y 2020 et al. reported isolating the virus from the stools of one severe hospitalised COVID-19 pneumonia case. The number of samples taken was unclear and while Vero cells were used for viral isolation from stool samples, culture methods were not described.

Additional evidence of SARS-CoV-2 replication activity was observed within the intestine: an inpatient for treatment of a rectal adenocarcinoma had samples taken from enteric sections, and the mucosa of rectum and ileum analysed [Qian 2020]. The rectal swab sample tested negative for SARS-CoV-2 RNA by PCR. However, typical coronavirus virions in rectal tissue were observed under electron microscopy with abundant lymphocytes and macrophages (some SARS-CoV-2 positive) infiltrating the lamina propria.

Methodological issues across these studies including variability in sample selection and methods of viral culture, reported in Table 5, mean the results may not be comparable and should be interpreted with caution.

Studies of wastewater and sewage. Twenty-two global studies investigated SARS-CoV-2 in wastewater or sewage, four looked at the role of toilets or sewage, and one reported observation in river water [Ahmed 2020, Ampuero 2020, Arora 2020, Chavarria-Miro 2020, Curtis 2020, Fongaro 2020, Fernandez-de-Mera 2020, Haramoto 2020, Hata 2020, Lara 2020, La Rosa 2020b, Medema 2020, Neault 2020, Ong 2020, Peccia 2020, Sharif 2020, Shutler 2020, Trottier 2020, Wang J 2020, Wang XW, Li J 2020, Wurtzer 2020, Zhao 2020].

All reported the detection of SARS-CoV-2 RNA and/or SARS-CoV-2 viral proteins; a number of studies suggested the potential of detection in sewage to be used as a public health monitoring system.

Wastewater surveillance may anticipate outbreaks: a Brazilian study identified SARS-CoV-2 RNA from sewage samples collected in November 2019 [Fongaro 2020]; a Spanish study detected sewage samples from 41 days prior to the declaration of the first COVID-19 case in Spain, and additionally in frozen samples dating back to 12 March 2019 [Chavarria-Miró 2020]. Virus concentrations found in wastewaters may correlate with prevalence in the local community. At very low community levels of circulation, sewage surveillance could detect SARS-CoV-2 RNA presence [La Rosa b 2020, Medema 2020].

Viral culture in sewage

A study of hospital surfaces and sewage areas in China reported no virus was detected by culture of surface swabs or sewage samples [Wang J 2020]. Another study in China tested hospital sewage and found SARS-CoV-2 RNA but no viral culture of SARS-CoV was present in the sewage in their assays [Wang XW, Li J 2020].

Toilets. An additional four studies investigated SARS-CoV-2 in toilets (Del Brutto 2020, Ding Z 2020, Kang 2020, Ong 2020]. A study in rural Guatemala reported the sole predictive variable was the use of the open latrines [Del Brutto 2020]. Ding 2020 et al. collected surface samples from a COVID-19 hospital in China reported toilet areas represented the most contaminated areas. Kang et al. reported the possibility of fecal aerosol spread but with little discussion of alternate pathways for transmission. They did not find any evidence of virus -laden bioaerosols. Ong and co-workers reported that 3 of 5 toilet sites (toilet bowl, sink, and door handle) in hospital settings were PCR-positive; anteroom and corridor samples were negative [Ong 2020].

River water. One study tested river water for SARS-CoV-2: taking samples from three urban river locations in a low sanitation urban context (i.e. highly impacted by raw sewage) in Quito, Ecuador, during a peak of COVID-19 cases. SARS-CoV-2 RNA was detected in all samples, at levels similar to those in wastewater from cities during outbreaks [Guerreo-Latorre 2020]. Evidence from reviews can be found in Table 2.

Table 2. Included study characteristics: reviews.

Study (n= 33;
1 is a cohort
and SR so
appears in
both 1a and
1b)
Fulfils
systematic
review
methods
Research question (search date up
to)
No. included studies     (No.
participants)
Main results Key conclusions
Aboubakr 2020 no To present the available data on the
stability of coronaviruses (CoVs),
including SARS‐CoV‐2, from previous
reports to help understand its
environmental survival (search date
NR; study published June 2020)
Unclear (NA) SARS‐CoV‐2 and other human and animal CoVs have
short persistence on copper, latex and surfaces with
low porosity vs other surfaces like stainless steel,
plastics, glass & highly porous fabrics. SARS‐CoV‐2 is
associated with diarrhoea & is shed in faeces. Some
CoVs show persistence in human excrement, sewage
and waters for a few days. Possible faecal–oral,
foodborne & waterborne transmission of SARS‐CoV‐2
in regions using sewage‐polluted waters in irrigation &
with poor water treatment systems.
Possible faecal–oral, foodborne
& waterborne transmission of
SARS‐CoV‐2 is a concern for
regions using sewage‐polluted
waters in irrigation & with poor
water treatment systems.
Achak M 2020 no To review evidence on the presence of
SARS-CoV-2 in aqueous environments,
and to describe the knowledge on
detection and survival of SARS-CoV-2
in wastewater and hospital wastewater,
in order to understand the different
routes of SARS-CoV-2 transmission
(search date NR; study available online
24 October 2020).
NR SARS-CoV-2 RNA is identified in a range of water
environments including hospital wastewaters.
According to a few studies investigating the
deactivation of SARS-Co V-2 showed that chlorine-
based disinfectants are widely used for their broad
sterilization spectrum, high inactivation efficiency
and easy decomposition with little residue, as well as
represents the best economic solution. The complete
deactivation of SARS-CoV-2 can be achieved by
combination of other technologies (biological and/or
physical-chemical processes).
Authors suggest there is a
need to develop more secure,
efficient, economical disinfection
technologies in order to limit
the transmission of Covid-19
and to avoid other waves of the
pandemic of Covid-19 infections
Aguiar-Oliveira
M 2020
no To summarize the current global
experience on SARS-CoV-2 wastewater
based epidemiology in distinct
continents and viral detection in
polluted surface water. The advantages
and concerns of this strategy for
SARS-CoV-2 surveillance are discussed.
(Search date NR; article published 10
December 2020).
NR Viral RNA has been detected in the stool of Covid-19
patients and viable viruses had been isolated in some
of these samples. Thus, a putative role of SARS-CoV-2
fecal-oral transmission has been argued. SARS-CoV-
2 is shed in human excreta and further disposed in
the sewerage or in the environment, in poor basic
sanitation settings.
Outcomes suggest that
wastewater based epidemiology
is a valuable early warning alert
and a helpful complementary
surveillance tool to subside
public health response, to tailor
containment and mitigation
measures and to determine
target populations for testing.
In poor sanitation settings,
contaminated rivers could be
alternatively used as a source for
environmental surveillance.
Amirian 2020 no Current knowledge on the potential
for fecal transmission is briefly
reviewed and the possible implications
are discussed from a public health
perspective (search date NR; study
published April 2020)
Unclear (NR) A number of case series have reported the presence
of SARS-CoV-2 genetic material in the stool of some
COVID-19 patients.
If orofecal transmission is
established, it will impact public
health guidelines on sewage
exposure, nosocomial infections,
residential care facilities, and
food preparation.
Cahill 2020 no Are recreational waters a potential
transmission route for SARS-CoV-2
to humans? (search date NR; study
published October 2020)
Unclear (NR) SARS-CoV-2 has been detected in faeces and
wastewater in recent months. Wastewater is a potential
dissemination route for SARS-CoV-2 to recreational
waters. Limited data on the presence and viability of
SARS-CoV-2 in water bodies exists.
The risk of SARS-CoV-2 exposure
to recreational water users
is believed to be low. Further
research is required.
Cheung K 2020 no To review the evidence on
gastrointestinal symptoms and
detection of virus in stool in Covid-19.
(11 March 2020)
60 (4,243) Pooled prevalence of all GI symptoms was 17.6%
(95%CI 12.3 to 24.5); 11.8% of patients with nonsevere
COVID-19 had GI symptoms (95%CI 4.1 to 29.1),
and 17.1% of patients with severe COVID-19 had GI
symptoms (95% CI, 6.9 to 36.7). Pooled prevalence of
stool samples positive for virus RNA was 48.1% (95%
=CI 38.3 to 57.9); of these samples, 70.3% of those
collected after loss of virus from respiratory specimens
tested positive (95% CI 49.6 to 85.1).
Around 18% of Covid-19 patients
experienced GI symptoms.
Viral RNA can be identified in
stool samples of around half of
Covid-19 patients, and is often
detectable even after respiratory
samples test negative.
Chiappini 2020 no To give an overview of gastrointestinal
involvement in children with SARS‐
COV‐2 infection. (Search date NR;
article published 24 November 2020)
NR Diarrhea and vomiting have been reported in about
8%‐9% of cases, reaching more than 20% in some
studies. Fecal shedding in children has been reported
in 20%‐30% of children and has been observed in both
those with and those without overt GI involvement.
GI symptoms are common
findings in children with
SARS‐CoV‐2 infection. Fecal
shedding in asymptomatic
children and prolonged fecal
elimination, lasting several days
after negativization of RT‐PCR
on respiratory swabs, have been
reported with variable frequency
in children with Covid‐19.
Collivignarelli
2020
no To review the evidence on the
presence of SARS-CoV-2 in wastewater
and sewage sludge, the factors
affecting its inactivation and the main
proposed treatments. (search date NR;
preprint posted June 2020).
20 (NA) Literature on SARS-CoV-2 in wastewaters is currently
limited. For SARS-CoV-1, its resistance in wastewater
is limited, especially at temperatures above 20 °C, and the
virus has been easily removed with chlorine (> 0.5 mg
L-1 for 30 min).
Detection of SARS-CoV-2 in
wastewater might track the
epidemic trends: although
promising, an effective and wide
application of this approach
requires a deeper knowledge
of the amounts of viruses
excreted in faeces, and the
actual detectability of viral RNA in
sewage.
Cuicchi D 2020 no To collect the data available on SARS-
CoV-2 in the GI system and evaluate
whether the digestive system could
contribute to viral transmission (31
May 2020).
27 (671) 46.5% patients had a positive stool sample for SARS-
CoV-2 RNA; 63.9% remained positive after pharyngeal
swabs became negative; in 3 studies, in samples of
3 patients out of 8 examined, SARS-CoV-2 RNA was
found in GI tissues. Live virus in stool samples was
confirmed in two studies and not found in another
study. These results suggested that SARS-CoV-2 could
infect gastrointestinal epithelial cells and it may be
transmitted through the digestive tract.
These results suggested that
SARS-CoV-2 could infect GI
epithelial cells and it may
be transmitted through the
digestive tract.
Ding S 2020 no Is SARS-CoV-2 also an enteric
pathogen with potential fecal-oral
transmission? (search date NR; review
completed 23 April 2020)
NR SARS-CoV-2 is capable of infecting the gastrointestinal
tract and shedding in the environment for potential
human-to-human transmission; more reserach
in needed to understand the extent and relative
importance of these. .
Work is needed to examine the
full extent of GI and liver aspects
of COVID-19
Dona 2020 no To review evidence on GI symptoms
and the use of rectal swabs to detect
SARS-CoV-2 RNA, in children. (search
date NR; study published July 2020).
NR GI symptoms have been reported more commonly
among children than adults with Covid-19. SARS-CoV-
2 RNA has been detected in several studies of rectal
swabs in children.
Further evidence on GI
involvement and excretion of
SARS-CoV-2 in faeces is needed
to confirm fecal viral loads
regardless of enteric symptoms,
and to better explore viral RNA
detection across the disease
course.
Edwards 2020 yes To describe whether SARS-CoV-2 viral
loads (VLs) and cycle thresholds (CTs)
vary by sample type, disease severity
and symptoms duration (8 April 2020)
24 (173) Viral loads higher in saliva and sputum vs NP swabs; in
asymptomatics; & in severe COVID-19. Stool samples
positive for a longer period than other samples.
Diagnostic strategies should
consider these variations in viral
loads.
El-Wahab EWA
2020
no Summarize the ways in which SARS-
CoV-2 is transmitted and provide
scientific support for the prevention
and control of COVID-19 (31 July 2020).
302 (NR) The basic mechanisms of SARS-CoV-2 transmission
person-to-person contact through respiratory droplets,
or via indirect contact... Although SARS-CoV-2 has
been detected in non-respiratory specimens, including
stool, blood and breast milk, their role in transmission
remains uncertain.
Additional research is needed to
better understand transmission.
Gupta 2020 no To establish the incidence and timing
of positive faecal samples for SARS‐
CoV‐2 in patients with Covid‐19 (3 April
2020)
26 (NR) Persistent RNA shedding has been recorded in a
number of studies, even subsequent to respiratory
test negativity. Notably, in one study, 44/ 153 stool
specimens tested were PCR positive and live virus was
detected in 2 of 4 specimens culture.
There is a high incidence and
persistence of positive faecal
RT‐PCR tests for SARS‐CoV‐2 after
negative nasopharyngeal swabs
in patients with Covid‐19.
Han Z 2020 no To review published studies about
discharged patients testing positive again for SARS‐
CoV‐2 RNA. (27 April
2020)
12 (90) All studies were in China. These reports indicates
the presence of discharged patients who remain
asymptomatic but test PCR- positive; however, it is
unclear whether they are contagious
This review suggests the need
for parallel testing of different
samples e.g. fecal specimens,
from Covid‐19 patients before
and after they are discharged
from hospitals.
Ji B 2020 no To provide a comprehensive profile on
the transmission characteristics of the
coronaviruses in water, sludge, and air
environment, especially the water and
wastewater treatment systems. (Search
date NR; paper available online 27
October 2020).
NR Existing drinking water treatment protocols effectively
remove SARS-CoV-2 and to date, drinking water is safe.
SARS-CoV-2 is shed in the faeces and urine of infected
individuals and then enters wastewater. Wastewater
monitoring could be a strategic surveillance tool.
Intensive studies of SARS-CoV-2
activity and survival in water and
wastewater treatment are highly
desirable.
Jones 2020 no To critically evaluate the incidence of GI
symptoms, the quantity and infectivity
of SARS-CoV-2 in feces and
urine, and whether these pose an infection risk
in sanitary settings, sewage networks,
wastewater treatment plants, and the
wider environment e.g. rivers, lakes
and marine waters. (search date NR;
study published 20 December 2020)
48 (NA) SARS-CoV-2 RNA can be readily detected
in feces and occasionally urine. Severe GI dysfunction only occurs
in a small number of cases (11 ± 2%). Likelihood of
SARS-CoV-2 being transmitted via feces appears very
low. Likelihood of infection from sewage-contaminated
water or food is extremely low.
The likelihood of SARS-CoV-2
being transmitted via feces or
urine appears low due to the low
relative amounts of virus present in feces/urine.
Karia 2020 yes To review the sources of viral shedding
that have been reported to date and
compare the duration of shedding
from different sources and their
relation to clinical recovery (July 2020)
19 (1,433) Prolonged shedding observed in a range of specimens;
max duration to conversion 60 days; in several studies,
viral shedding from the GI tract was for a longer
duration and at a greater viral load than from the
respiratory tract.
Prolonged viral shedding
is important to consider
while discontinuing isolation
procedures and/or discharging
SARS-CoV-2 patients.
Kingsbury 2020 no What is international best practice
regarding reducing the likelihood
that food products or packaging
are vectors for COVID-19? In this
context, sources of COVID-19 may be
production or supply chain workers?
What is international best practice for
mitigation options to reduce transfer
of COVID-19 from workers to food
products? (28 April 2020)
> 77 (NR) Infectious virus has been found in faeces of some
infected people, raising the possibility of faecal-oral
transmission via contaminated vehicles such as food,
but there is no evidence for this having occurred.
Best practice for reducing the
risk of contamination of food
products or packaging continues
to be managing the risk of
SARS- CoV-2 infection amongst
workers.
La Rosa a no Scoping review (23 February 2020) 12 (N/A) Coronaviruses seems to have a low stability in the
environment and be very sensitive to oxidants such
as chlorine; coronaviruses appear to be inactivated
significantly faster in water than non-enveloped human
enteric viruses with known waterborne transmission;
temperature is an important factor influencing viral
survival: the titer of infectious virus declines more
rapidly at 23 to 25 °C than at 4 °C; there is not current
evidence that human coronaviruses are present in
surface or ground waters or are transmitted through
contaminated drinking-water.
Coroviridae have been isolated
in different types of liquids
from waste to surface water,
but in general they appear to
be unstable. Chlorination and
higher temperatures lead to
their inactivation. At the time
of search (Feb 2020) there was
not evidence of coronavirus
transmission through
contaminated water.
Mehraeen
2020
no To identify current evidence on
transmission modes of Covid-19. (April
2020).
36 (NR) The review identified five potential transmission modes
of COVID-19 including airborne, droplet, contact with
contaminated surfaces, oral and fecal secretions.
Furthermore, some studies have pointed out other
modes of virus transmission, such as person to person,
and direct contact with animals.
Droplet and contact with
contaminated surfaces were
the most frequent transmission
modes of COVID-19. Fecal
excretion, environmental
contamination, and fluid
pollution might contribute to a
viral transmission. The possibility
of fecal transmission of Covid-19
has implications, especially in
areas with poor sanitation.
Meyerowitz
2020
no To review the evidence on transmission
of SARS-CoV-2 (7 September 2020)
NR Strong evidence from case and cluster reports
indicates that respiratory transmission is dominant,
with proximity and ventilation being key determinants
of transmission risk. Although live virus has been
isolated from saliva and stool and viral RNA has been
isolated from semen and blood donations, there are
no reported cases of SARS-CoV-2 transmission via
fecal–oral, sexual, or bloodborne routes. To date, there
is 1 cluster of possible fecal–respiratory transmission.
Evidence indicates that
respiratory transmission is
dominant. Re: fecal aerosol
transmission, given how rarely
live virus has been isolated
from stool, the low levels of
replication-competent virus in
stool that might be aerosolized
from toilet flushing seem highly
unlikely to cause infection except
under unusual or extraordinary
circumstances.
Mohapatra S
2020
no To review evidence on the detection,
occurrence and fate of SARS-CoV-2
(& other enveloped viruses) during
primary, secondary, and tertiary
wastewater and water treatment
processes. (Search date NR; article
available online 6 October 2020).
NR SARS-CoV-2 contamination of water bodies may be
possible through orofecal route. SARS-CoV-2 RNA
has been detected in wastewater across the globe.
Coagulation-flocculation, filtration can remove SARS-
CoV-2 RNA and complete inactivation of SARS-CoV-2 is
possible through chlorination.
More research on the possibility
of faecal-oral transmission and
its possible fate and persistence
in various environmental
compartments is needed.
Morone 2020 yes To correlate the presence and
the relevant temporal patterns of
SARS-CoV-2 viral RNA in biological
specimens (stool, urine, blood, and
tears) of the transmission with clinical/
epidemiological features in patients
with Covid-19 (5 May 2020)
55 (1,348) Fecal positivity duration (median 19 days) was
significantly (p < 0.001) longer than respiratory tract
positivity (median 14 days). Limited data are available
about the other specimens.
Attention should be paid to
negativization criteria for COVID-
19, because patients could have
longer alternative viral shedding.
Pamplona 2020 yes To describe the evidence on GI
symptoms, enteric involvement and
fecal excretion of SARS-CoV-2 viral
RNA, and to discuss the possible fecal-
oral transmission pathway of Covid-19.
(12 April 2020)
33 (NR) High variabilty in GI symptom reporting, summary
estimate GI symptoms present in 16%, diarrhea in 8.1%,
nausea-vomiting in 12% and abdominal pain in 4% of
Covid-19 patients.
GI symptoms are common in
SARS CoV-2 infection at the time
of patient admission, sometimes
preceding respiratory symptoms,
and sometimes represent the
only clinical manifestation.
Parasa 2020 yes What are the incidence rates of
gastrointestinal symptoms among
patients with SARS-CoV-2 infection? (30
March 2020)
29 (4805) Pooled rates were 7.4% (95%CI 4.3% to 12.2%) of
patients reporting diarrhea and 4.6% (95%CI 2.6% to
8.0%) of patients reporting nausea or vomiting. Fecal
tests that were positive for SARS-CoV-2 were reported
in 8 studies, and viral RNA shedding was detected
in feces in 40.5% (95% CI, 27.4%-55.1%) of patients.
There was high level of heterogeneity (I2 = 94%), but no
statistically significant publication bias noted.
These findings suggest that
that 12% of patients with COVID-19
will manifest GI symptoms;
however, SAR-CoV-2 shedding
was observed in 40.5% of
patients with confirmed
SARS-CoV-2 infection.
Patel 2020 no To update the current literature on
transmission of SARS-CoV-2. (search
date NR; study published 7 July 2020)
NR The rationale behind its transmission potential is that
viral RNA has unexpectedly been detected in multiple
bodily fluids, with some samples having remained
positive for extended periods of time. Additionally, the
receptor by which the virus gains cellular entry, ACE2,
has been found to be expressed in different human
body systems, thereby potentiating its infection in those
locations.
Detection of viral RNA shedding
in multiple bodily fluids/samples
suggests the potential for modes
of transmission additional to
respiratory, such as bloodborne,
urinary, and fecal-respiratory.
Transmission by such means
remains controversial given the
limited supporting data for each
mode.
SAGE 2020 no To describe SARS-CoV-2 transmission
routes and environments (search date
NR; report available online 22 October
2020).
NR Orofecal-related results: SARS-CoV-2 RNA has been
found in stool samples and RNA shedding often persists
for longer than in respiratory samples; however,
isolation of live virus has rarely been successful from
stool or urine. The GI tract probably is also susceptible
to infection and may serve as a transmission portal
given the high concentration of ACE2 receptors in
the small bowel, however, no published reports have
described faecal-oral transmission. Positive RNA
samples have been found in samples. There is only
indirect evidence of orofecal transmission in this
pandemic.
More evidence is needed to
understand transmission.
Research should be embedded
into the public health responses
to the pandemic.
Santos 2020 yes To investigate differences in viral
shedding in respiratory and fecal
samples from children with Covid-19.
(19 April 2020)
4 (36) A higher proportion of children had viral shedding in
stools after 14 days of symptoms onset compared to
respiratory samples (RR= 3.2, 95%CI 1.2 to 8.9, I2 =
51%). Viral RNA shedding was longer in fecal samples
with a mean difference of approximately 9 days (Mean
Difference = 8.6, 95%CI 1.7 to 15.4, I2 = 77%) compared
with respiratory samples
SARS-CoV-2 shedding seems to
be present in feces for a longer
time than in the respiratory
tract of children. Although fecal
SARS-CoV-2 presence in feces do
not confirm its transmissibility,
the high and fast spread of the
COVID-19 disease worldwide
indicate other transmission
routes are also plausible
Sehmi 2020 no To establish if there is any evidence
of the presence of live SARS-CoV-2 in
feces of Covid-19 patients.n (6 June
2020)
4 (8) 1 study successfully isolated live SARS-CoV-2 virus in the
stool sample of a patient with severe Covid-19, using
Vero cell culture & electron microscopy. Another study
using similar methods isolated and demonstrated live
virus in 2 out of 3 patients who had tested PCR positive
in stool samples. The same authors in another report
mentioned their unpublished findings on successful
isolation of infectious SARS-CoV-2 from stool. In a
fourth study, ientification of live virus in stool was
attempted successfully in 2 out of 4 patients; neither of
these patients had GI symptoms.
Live SARS-CoV-2 virus is present
in fecal samples of Covid-19
patients, and therefore supports
the hypothesis that Covid-19
could potentially be transmitted
via the feco-oral route.
Sun S 2020 no To review fecal transmission of
Covid-19, the practices of open
defecation, and the resultant routes
of transmission of fecal pathogens.
Also, we highlight the open design
of common squat toilets and the
potential exposure to fecal droplets
and residues. (Search date NR; article
published 29 November 2020).
NR In communities practicing open defecation, poor hand
hygiene, contaminated shoes and objects, mechanical
vectors, and outdoor human activities could all
contribute to fecal transmission. Other risk factors
include squat pans with lidless designs and open
flushing mechanisms, in-cubicle open waste bins, and
the lack of water-sealing U-traps in squat toilets.
Fecal-associated transmission
has been identified as a
potential route of virus spread
in the Covid-19 pandemic, as
an increasing body of evidence
confirmed high viral loadings
and infectivity of SARS-CoV-2
in patients’ stools, including
asymptomatic, pre-symptomatic,
and convalescent individuals.
Tian 2020 no To report on the gastrointestinal
manifestations and pathological
findings of patients with COVID‐19,
and to discuss the possibility of faecal
transmission. (search date NR; study
published 31 March 2020)
NR (2023
patients)
With an incidence of 3 to 79%, GI symptoms included
anorexia 40 to 50%, diarrhoea 2% to 50%, vomiting 4 to
67%, nausea 1 to 29%, abdominal pain 2 to 6% and GI
bleeding 4 to 14%. Diarrhoea was observed before and
after diagnosis with a mean duration of 4.1 ± 2.5 days.
Adult and children patients can present with digestive
symptoms in the absence of respiratory symptoms.
Tran 2020 no To review evidence on the presence of
SARS-CoV-2 in water and wastewater
and what level of risk it may pose.
(search date NR; study published 1
October 2020)
NR SARS-CoV-2 has been detected in water and
wastewater. The suggested transmission route of
SARS-CoV-2 into water through stool and mask of
infected patients. Coronavirus is often inactivated
rapidly in water. Paper-based devices have been
suggested for detecting traces of SARS-CoV-2 in water.
Existing disinfection processes possibly sufficient to kill
SARS-CoV-2 in water.
The presence of SARS-CoV-2
RNA in river water and untreated
wastewater is confirmed, but
strong evidence of its survival
time in water environments is
missing. One study confirmed
lack of infectivity of SARS-CoV-2
in water based on absence of
cytopathic effect.
van Doorn
2020
no To critically assess the clinical relevance
of testing stool samples and anal
swabs and provide an overview of the
potential faecal‐oral transmission of
SARS‐CoV‐2 (7 July 2020)
95 (2,149) 934/2149 (43%) patients tested positive for SARS‐CoV‐2
in stool samples or anal swabs, with positive test results
up to 70 days after symptom onset. A meta‐analysis
executed with studies of at least 10 patients revealed
a pooled positive proportion of 51.8% (95% CI 43.8
‐ 59.7%). Positive faecal samples of 282/443 patients
(64%) remained positive for SARS‐CoV‐2 for a mean of
12.5 days, up to 33 days maximum, after respiratory
samples became negative for SARS‐CoV‐2. Viable
SARS‐CoV‐2 was found in 6/17 (35%) patients in whom
this was specifically investigated.
Viral shedding of SARS‐CoV‐2
in stool samples occurs in
a substantial proportion of
patients, making faecal‐oral
transmission plausible.
Furthermore, detection in
stool samples or anal swabs
can persist long after negative
respiratory testing.
Zhen-Dong
2020
no To assess epidemioogy and clinical
features of Covid-19 among Chinese
children.
37 (406) Asymptomatic infections and mild cases account for
44.8%, with only 7 cases of critical illness; laboratory
examination of lymphocyte counts is not reduced, as
it is for adults; chest CT findings are less severe than
those for adults. These presentations are the clinical
features of COVID-19 in children. Only 55 of the 406
cases were tested by anal swab for virus nucleic acid,
45 of which were positive, accounting for 81.8% of stool
samples.
There are more children than
adults with asymptomatic
infections, milder conditions,
faster recovery, and a better
prognosis. Some concealed
morbidity characteristics also
bring difficulties to the early
identification, prevention and
control of COVID-19.

Table 3. Quality of included primary studies.

Study Study type Description of
methods with
sufficient detail
to replicate
Sample
sources clear
Analysis &
reporting
appropriate
Is bias dealt
with
Applicability
Agrawal 2020 Observational Yes Yes Yes Unclear Yes
Ahmed 2020 Observational Yes No Yes No Yes
Ampuero 2020 Observational Yes Yes Yes No Yes
Arora 2020 Observational Yes Yes Yes Unclear Yes
Betancourt 2020 Observational Yes Yes Yes Unclear Yes
Chan 2020 Observational Yes Yes Unclear No Unclear
Chavarria-Miro 2020 Observational Yes No Yes Unclear Unclear
Chen C 2020 Observational Yes Yes Not Applicable Not Applicable Unclear
Chen W 2020 Observational Yes Unclear Yes Unclear Yes
Chen Y 2020 Observational Yes Yes No No No
Cheung CCL 2020 Observational Yes Yes Yes Not Applicable Yes
Cheung K 2020 Observational Yes Yes Yes Unclear Yes
Cho 2020 Observational Yes Yes Yes No Yes
Chu H 2020 Observational Yes Yes Yes Not Applicable Yes
COVID Ix Team Observational Yes Yes Yes Unclear No
Curtis 2020 Observational Yes Yes Yes Unclear Unclear
Del Brutto 2020 Observational Yes Yes Yes Unclear Yes
Ding Z 2020 Observational Yes Not Applicable Yes Unclear Yes
Fernández‐de‐Mera 2020 Observational Yes Yes Unclear No Unclear
Fongaro 2020 Observational Yes Yes Yes Unclear Yes
Ge 2000 Observational Yes Yes Unclear Not Applicable Yes
Guerrero-Latorre 2020 Observational Yes Yes Yes Unclear Yes
Han C 2020 Observational Yes No Yes No No
Haramoto 2020 Observational Yes Yes Yes Not Applicable Yes
Hata 2020 Observational Yes Yes Yes Not Applicable Yes
Hayee 2020 Observational Yes Not Applicable Yes Unclear Yes
Hoehl M 2020 Observational Yes Yes Yes Unclear Yes
Holshue 2020 Observational Yes Yes Yes Unclear No
Iglesias 2020 Observational Yes Yes Yes Unclear Yes
Izquierdo-Lara 2020 Observational Unclear Yes Yes Unclear Yes
Jeong 2020 Observational Yes Yes Yes Unclear Yes
Jiehao 2020 Observational Yes Yes Yes Unclear No
Kang 2020 Observational Yes Yes Yes Unclear Yes
Kim J-M 2020 Observational Yes No Unclear No Unclear
La Rosa 2020 (b) Observational Yes Unclear Yes Not Applicable Not Applicable
Lescure 2020 Observational Yes Yes Yes Unclear No
Li 2020 Observational Unclear Yes Yes Unclear Unclear
Ling 2020 Observational Yes Unclear Yes Unclear No
Lo 2020 Observational Yes Yes Unclear Unclear No
Medema 2020 Observational Yes No Yes No Yes
Neault 2020 Observational Yes Unclear Yes Not Applicable Yes
Nicastri 2020 Observational Yes Yes Yes Unclear No
Ong 2020 Observational Yes No Unclear Unclear Yes
Pan 2020 Observational Unclear No Yes Unclear No
Peccia 2020 Observational Unclear Yes Yes Not Applicable Yes
Peng 2020 Observational No Yes Not Applicable No No
Qian 2020 Observational Yes Yes Yes Unclear Yes
Senapati 2020 Observational Unclear Yes Not Applicable Not Applicable No
Sharif 2020 Observational Yes Yes Yes Unclear Yes
Shutler 2020 Observational Yes No No Unclear No
Tan 2020 Observational Unclear Yes Yes Unclear No
Tang 2020 Observational Unclear Unclear Yes Unclear No
Trottier 2020 Observational Yes Yes Yes Not Applicable Yes
Wang J 2020b Observational Yes No Yes No Yes
Wang Q-X 2020 Observational Yes Yes Yes No Unclear
Wang S 2020 Observational Unclear Yes Yes No Unclear
Wang W 2020 Observational Unclear Yes Unclear Unclear Unclear
Wang X, Zhou Y 2020 Observational Yes Yes Unclear Unclear No
Wang X, Zheng J 2020 Observational Yes Yes Yes Unclear Yes
Wolf 2020 Observational Yes Yes Yes Unclear Yes
Wölfel 2020 Observational Unclear Unclear Unclear Yes Unclear
Wu Y 2020 Observational Yes No Yes No No
Wurtzer 2020 Observational Yes No Yes Unclear Yes
Xiao F & Tang M 2020 Observational Yes Yes Not Applicable No Unclear
Xiao F & Sun J 2020 Observational Yes No Not Applicable Not Applicable Unclear
Xing Y 2020 Observational Yes Unclear Unclear Unclear Yes
Xu Y 2020 Observational Yes Yes Not Applicable Not Applicable Unclear
Yang Y 2020 Observational Yes Yes Yes Unclear Yes
Young 2020 Observational Unclear Unclear Yes Unclear No
Yuan 2020 Observational Unclear Yes Yes Unclear No
Zhang J 2020 Observational Yes Unclear Unclear No Unclear
Zhang T 2020 Observational Yes Yes Yes Unclear Yes
Zhang W 2020 Observational Yes Yes Unclear Unclear Unclear
Zhang Y 2020 Observational Unclear No Yes Unclear No
Zhang Z, Chen C 2020 Observational No Yes Yes Unclear Yes
Zhao 2020 Observational Yes Yes Yes Not Applicable Yes

Table 4. Main findings of included primary studies: SARS-CoV-2 and the role of orofecal transmission (n=76).

Study Main findings of primary studies on orofecal transmission of SARS-CoV-2
Chan 2020 This very early study established the likelihood of person to person transmission of SARS-CoV-2, in hospital and family settings. The two faecal samples from
patients 3 and 4 who had preceding diarrhoea were negative on a multiplex PCR assay for common diarrhoeal viruses, bacteria, and parasites.
Chen C 2020 This retrospective study of 133 hospitalised COVID-19 patients identified 22 whose sputum or fecal samples tested positive, after their pharyngeal swabs
became negative.
Chen W 2020 SARS-CoV-2 RNA was readily detected in the blood (6/57 patients) and anal swabs (11/28 patients).
Chen Y 2020 Sixty seven percent (28/42) laboratory-confirmed hospitalised COVID-19 patients tested positive for SARS-CoV-2 RNA in stool specimens; this was not associated
with the presence of GI symptoms or severity of illness. Among them, 18 (64%) patients remained positive for viral RNA in the feces after the pharyngeal swabs
turned negative, for a duration of 6 to 10 days.
Cheung CCL 2020 This study used multiplex immunohistochemistry and unexpectedly detected SARS-CoV-2 viral antigens in intestinal and liver tissues, in surgical samples
obtained from two hospitalized patients who recovered from Covid-19. The presence of the virus was validated by RT-PCR and flow cytometry to detect SARS-
CoV-2-specific immunity in the tissues.
Cheung K 2020 This study analysed stool samples from a cohort of 59 patients with COVID-19 in Hong Kong during February 2020 and additionally did a meta-analysis of data
from 11 studies on the prevalence of GI symptoms and stool excretion of viruses. Fecal discharge continues long after respiratory shedding of COVID-19 has
ceased.
Cho 2020 This case study reports an infant with mild Covid-19, positive-to-negative nasal swab conversion occurred on the 21st day from the onset of symptoms, but stool
swab positivity persisted during the 6-week admission period and for 7 weeks during follow-up at an outpatient clinic after discharge.
Chu H 2020 Case report of a breastfeeding woman with a positive PCR test for SARS-CoV-2. The patient presented on 24 January 2020 with GI symptoms; later she
developed a fever. Her infant had been born 16 January 2020. She tested PCR positive in respiratory swabs. She had persistent SARS-CoV-2 RNA positivity in her
feces but negativity in her breastmilk. She bottle-fed her baby with her breastmilk after treatment. The baby appeared healthy and unaffected after a 1-month
follow up.
COVID Ix Team SARS-CoV-2 RNA was detected in at least one nasopharyngeal (NP) swab, 11/12 oropharyngeal (OP) swab and 7/10 in the stool in this case series describing the
first 12 US patients confirmed to have COVID-19 from 20 January to 5 February 2020.
Ge 2020 This case study of a hospitalised Covid-19 patient reported that the fecal samples remained PCR-positive for 22 days after their respiratory samples turned
negative.
Han C 2020 Among a group of hospitalised patients with low severity COVID-19, digestive symptoms were present in 57%. Patients with digestive symptoms were more
likely to be fecal virus-positive than those with respiratory symptoms.
Hayee 2020 This study reports PCR test results for outpatients attending for GI endoscopy at a UK hospital 30th April to 30th June 2020: 3/2,611 asymptomatic patients
tested positive for SARS-CoV-2 on nasopharyngeal swab testing pre-endoscopy. No cases of Covid-19 were detected for 14 days after the procedure.
Hoehl S 2020 Children and staff at 50 day-care centres in Germany were tested repeatedly over 12 weeks. Buccal mucosal swabs and anal swabs were taken (by parents) from
825 children aged 3 months to 8 years attending the day care centres and 372 staff members (swabs self-collected) of these settings, between 18 June and 10
September 2020. 7,366 buccal mucosa swabs and 5,907 anal swabs were analysed. No respiratory or GI shedding of SARS-CoV-2 was detected in any of the
children.

Two adult staff members at two different day care centers tested positive; one had symptoms.
Holshue 2020 Stool obtained from a single hospitalized Covid-19 case was positive for SARs-CO-V-2 on day 7 of the illness.
Jeong 2020 There was viable SARS-CoV-2 in saliva, urine, and stool from COVID-19 patients up until days 11 to 15 of the clinical course suggesting that viable SARS-CoV-2
can be secreted in various clinical samples as well as respiratory specimens.
Jiehao 2020 Prolonged virus shedding was observed in the respiratory tract and feces of children at the convalescent stage.
Kim J-M 2020 SARS-CoV-2 RNA was detected in serum, urine or stool samples in 20% of patients hospitalised with Covid-19: 13/129 stool samples obtained from 74 patients
tested positive. However, the virus could not be isolated from these samples and therefore the risk of transmission via these media is not established.
Lescure 2020 A case series of the first five identified Covid-19 cases in Europe; an early demonstration of the vastly increased risk for elderly versus younger people. Viral RNA
was detected in the stools of the two paucisymptomatic women.
Li 2020 This case series reported on 29 hospitalised mild-to-moderate severity Covid-19 patients in China. Fecal samples from 4 patients tested positive for SARS-CoV-2 RNA,
including one apparently asymptomatic case.
Ling 2020 From 292 confirmed cases with COVID-19 in the Shanghai region, 66 recovered patients were included. Clearance of viral RNA in patients’ stools was delayed
compared to oropharyngeal swabs.
Lo 2020 A report of the clinical and microbiological features of ten hospitalized Covid-19 patients in Brazil between 21 January and 16 February 2020 found that SARS-
CoV-2 can be shed in the stool.
Nicastri 2020 SARS-CoV-2 RNA was positive in stools, nasopharyngeal and oropharyngeal swabs at different time points in a case report.
Pan 2020 Stool samples from 9/17 confirmed patients were positive on RT-PCR analysis.
Peng 2020 Virus was found in urine, blood and in two anal swabs and oropharyngeal swabs of nine patients diagnosed with COVID-19.
Qian 2020 SARS-CoV-2 was detected in the rectum of a COVID-19 patient during the disease incubation period. There was direct evidence of replication of SARS-CoV-2 in
the intestine.
Senapati 2020 This pilot study in India found SARS-CoV-2 RNA in fecal samples from 12 symptomatic and asymptomatic COVID-19 patients.
Tan 2020 In a single case report, SARs-CoV-2 was detected in the throat and rectum of the patient with COVID‐19.
Tang 2020 An asymptomatic child was positive for a coronavirus by reverse transcription PCR in a stool specimen 17 days after the last virus exposure. The child was virus
positive in stool specimens for at least an additional 9 days.
Wang Q-X 2020 Case series of 5 individuals who had Covid-19 and whose respiratory samples were negative by PCR, but had positive fecal samples. Observed over 3 to 15 days,
no cases of reinfection occurred and all fecal samples became negative.
Wang S 2020 Retrospective case series using clinical records, laboratory results, and CT findings for 17 COVID-19 patients including fecal sample testing showed 11/17 had
positive PCR tests in fecal samples.
Wang W, Xu Y 2020 In this case series from China, 2 stool specimens out of 44 positives contained live virus, suggesting that orofecal transmission is possible. Transmission of the
virus by respiratory and extra respiratory routes may help explain the rapid spread of disease.
Wang X, Zheng J 2020 Among fecal samples from 69 hospitalized Covid-19 patients, 20 tested PCR positive. The duration of SARS-CoV-2 RNA persistence was significantly longer in
fecal samples than in oropharyngeal swabs.
Wang X, Zhou Y 2020 In three hospitalised cases, intestinal SARS-CoV-2 infection affected the disease course of Covid-19 and stool samples were positive for SARS-CoV-2 by RT-PCR.
Wolf 2020 A five person family with a confirmed Covid-19 case was hospitalized and observed: the parents and 2 children aged 2 and 5 years became infected but the
youngest child was not infected. These children shed infectious virus via the respiratory system, and this shedding observed in the nasopharynx cleared after 5
to 6 days; however, viral RNA was continuously detected in the children’s stool for more than 4 weeks
Wölfel 2020 A detailed virological analysis of 9 Covid-19 hospitalised patients that provide proof of active virus replication in tissues of the upper respiratory tract. Stool
samples were also positive for RNA, and in one case the course of RNA concentration in stools indicated replication in the GI tract. However, attempts to culture
live virus from fecal samples were unsuccessful in 0/13 samples (from 4 patients).
Wu Y 2020 In 98 hospitalized Covid-19 cases, patients’ faecal samples remained positive for SARS-CoV-2 for a mean of 11 days (maximum 5 weeks) after respiratory tract
samples became negative.
Xiao F, Tang M 2020 39 of 73 hospitalized Covid-19 patients aged 10 months to 78 years tested positive for SARS-CoV-2 in fecal samples. Gastric, duodenal and rectal epithelial
specimens collected via endoscopy of one patient were also studied:

Immunofluorescence data showed that ACE2 protein, proven to be a cell receptor for SARS-CoV-2, is abundantly expressed in the glandular cells of gastric,
duodenal, and rectal epithelia, supporting entry of SARS-CoV-2 into the host cells.

Intracellular staining of viral nucleocapsid protein in gastric, duodenal, and rectal epithelia showed that SARS-CoV-2 infects these GI glandular epithelial cells.
Viral RNA was also detected in esophageal mucous tissue, but the absence of viral nucleocapsid protein staining in esophageal mucosa indicates low viral
infection in esophageal mucosa.
Detection of some viral nucleocapsid protein in rectal epithelial cells was observed in some additional Covid-19 patients, suggesting that some infectious viral
particles may survive the GI environment.
Xiao F, Sun J 2020 This case series of 28 hospitalised patients for whom feces samples were available indicated that infectious virus was present in feces from two cases who also
tested positive for viral RNA by RT-PCR.
Xing Y 2020 Three children showed a prolonged presence of SARS‐CoV‐2 in feces after throat swabs were negative.
Xu Y 2020 This study of 10 children with COVID-19 found that symptoms among children were nonspecific and relatively mild; rectal swabs tested positive among 8/10
cases even once nasopharyngeal tests became negative.
Yang 2020 Viral shedding and immunological features of 35 hospitalized children with Covid-19 were analyzed. 14/35 of the children had no symptoms; CT scan showed
pneumonia in 32/35 children. Viral RNA was detected in fecal samples from 17/35. RNA was found in fecal samples up to 33 days after case detection.
Young 2020 SARS-CoV-2 Virus was detectable in the stool of 4 of 8 hospitalized patients.
Yuan 2020 A retrospective case note survey of 2,138 paediatric patients with suspected SARS-CoV-2 infection in Wuhan Children’s Hospital included PCR tests on both
throat swabs and anal swabs were available for 212 children. Viral loads detected on both throat and anal swabs available for 24 patients showed no significant
difference. The findings suggested that in some children, fecal shedding may be a sign of prolonged mildly asymptomatic infection and represent the final
phase of the disease.
Zhang J 2020 A small pilot sample of 14 hospitalised cases indicated agreement for the presence of COVID-19 between oropharyngeal samples and fecal samples.
Zhang T 2020 Three children with mild symptoms who were SARS‐CoV‐2 throat swab specimen negative on discharge from hospital were stool positive 10 days post-
discharge.
Zhang W 2020 A small study of hospitalised COVID-19 patients indicated that RNA of SARS-CoV-2 may be shed via multiple bodily routes, and highlights that it is found in anal
swabs sometimes when oral swabs show no viral RNA.
Zhang Y, Chen C, Zhu S 2020 A 2019-nCoV strain was isolated from a stool specimen of a laboratory-confirmed Covid-19 severe pneumonia case, who experienced onset on 16 January 2020
and was sampled on 1 February 2020 in China. The full-length genome sequence indicated that the virus had high-nucleotide similarity (99.98%) to that of the
first isolated novel coronavirus isolated from Wuhan. In the Vero cells, viral particles with typical morphology of a coronavirus could be observed under the
electron microscope.
Zhang Z, Chen C, Song Y 2020 Samples from 258 Covid-19 patients with clinical symptoms and positive PCR were collected: 93/258 stool samples were PCR positive; PCR-positivity in stool
did not correlate with GI symptoms and only suggestively correlated with disease severity. Viral load tended to be higher within respiratory swab samples.
Live SARS-CoV-2 was isolated from a stool specimen (Ct value 24) of a severe Covid-19 patient (date of onset 16 January 2020) (strain HLJ002/HLJ/CHN/2020).
The sequence of the full-length genome of strain HLJ002 indicated that the virus had high nucleotide similarity (99.98%) to the first SARS-CoV-2 (GenBank No.
NC_045512) strain isolated from Wuhan. In the Vero cells, the virus caused obvious CPE, and the virus particles with typical morphology of coronavirus could be
observed under the electron microscope. [NB it is not clear if additional cultures were attempted; we assume not.]
Sewage
Agrawal 2020 This study monitored the time course of the SARS-CoV-2 RNA concentration in raw sewage in the Frankfurt metropolitan area of Germany.
44 sewage samples were taken from three influent sources at two wastewater treatment plants, between April and August 2020. RT-qPCR was used to assess
the presence and quantity of SARS-CoV-2 RNA. The correlation of this with concurrent epidemiological surveillance data was examined. Temporal dynamics were
observed between different sampling points, indicating local dynamics in Covid-19 cases within the Frankfurt metropolitan area.
Ahmed 2020 Using samples collected between February and April 2020 from sewage treatment plants in Queensland, Australia, SARS-CoV-2 was detected by RT-qPCR assay,
confirmed by sequencing.
Ampuero 2020 SARS-CoV-2 RNA was detected in untreated and treated wastewater samples obtained from two treatment plants in Santiago, Chile, March to June 2020.
Arora 2020 Untreated (influent), biologically treated, and disinfected wastewater samples were collected from May to August 2020 in two North Indian states; SARS-CoV-2
RNA was detected in 16/56 samples.
Betancourt 2020 In a US university campus, wastewater from a student dormitory was tested for SARS-CoV-2 RNA. Baseline tests established no SARS-CoV-2 when the
students returned to campus; subsequently the virus’ RNA was detected in wastewater samples for that dormitory. The students were isolated and tested by
nasopharyngeal swab PCR to identify infected individuals. The study demonstrated surveillance using wastewater testing, leading to identifying and containing
an outbreak.
Chavarria-Miro 2020 Testing of 24-hour composite raw sewage samples from two large wastewater treatment plants in Spain showed that SARS-CoV-2 was detected in sewage 41
days (15 January 2020) before the declaration of the first COVID-19 case (25 February 2020) in Spain, and in frozen samples dating back to 12 March 2019. If
these results are confirmed, they suggest SARS CoV-2 has been circulating longer than first thought.
Curtis 2020 This study examined the variability of SARS-CoV-2 concentrations in wastewater grab samples collected every 2 hours for 72 hours compared with
corresponding 24-hour flow-weighted composite samples. The results suggest that grab samples may be sufficient to characterize SARS-CoV-2 concentrations,
but additional calculations using these data may be sensitive to grab sample variability and warrant the use of flow-weighted composite sampling.
Fongaro 2020 This study analysed human sewage in Florianopolis, Brazil from late October 2019 until the Brazil lockdown March 2020. SARS-CoV-2 was detected in two
samples collected independently on 27th November 2019 (5.49 ± 0.02 log genome copies/L).
Fernandez-de-Mera 2020 This study investigated how readily SARS‐CoV‐2 RNA could be detected in environmental samples collected from an isolated small rural community in Spain at a
time of a high COVID‐19 prevalence (6% of the population of 883 inhabitants). Surface samples and village wastewater samples were taken: a number of these
tested PCR-positive for SARS-CoV-2 RNA but two sewage samples tested negative.
Haramoto 2020 A study of the presence of SARS-CoV-2 RNA in wastewater and river water in a prefecture of Japan and compared two laboratory methods. Whilst 1 of 5
wastewater samples tested positive, no river samples tested positive for SARS-CoV-2 RNA.
Hata 2020 A study of wastewater samples over time in Japan reported that SARS-CoV-2 RNA detection frequency increased along with the number of reported cases, and
was detected even at low prevalence of <1.0 per 100,000 people. Further, the detection frequency remained high even after the increase in cases stopped.
Iglesias 2020 This study measured SARS-CoV-2 RNA from a surface water source in a low-income settlement in Buenos Aires, Argentina between June and September 2020.
Measurements of SARS-CoV-2 concentrations in surface water contaminated by sewage could be used to estimate changes in Covid-19 prevalence in the local
community..
Lara 2020 This study in Belgium and the Netherlands investigated the use of phylogenetic analysis in routine wastewater testing samples to evaluate the diversity of SARS-
CoV-2 at the community level, and compared these results with the virus diversity in patients. It showed that this method could approximate the diversity of
SARS-CoV-2 viruses circulating in a community.
La Rosa 2020 b An environmental surveillance study based on twelve influent sewage samples, collected between February and April 2020 from wastewater treatment plants in
Milan and Rome, Italy showed SARS-CoV-2 RNA fragments have been identified in sewage in Italy, and suggest a novel RT-PCR test for screening of waters.
Medema 2020 SARS-CoV-2 was detected in the sewage of five sites a week after the first COVID-19 case in the Netherlands. Even at low COVID-19 prevalence sewage
surveillance could be a sensitive tool to monitor the viral circulation.
Neault 2020 In this longitudinal study, the stochastic variability inherent to wastewater-based epidemiology was corrected for using multiple fecal content protein
biomarkers. These normalized SARS-CoV-2 protein data correlated well with public health SARS-CoV-2 prevalence metrics.
Ong 2020 The study ran from 24th January to 4th February 2020 and involved sampling in the physical areas around three COVID-19 patients at the Singapore dedicated
SARS-CoV-2 outbreak center. The toilet bowl (seat and inner surface) and sink samples were positive, suggesting that viral shedding in stool could be a potential
route of transmission. Post-cleaning samples were negative, suggesting that current decontamination measures were sufficient.
Peccia 2020 In an urban area of NE USA, this study of primary sewage sludge over time reported identifying SARS-CoV-2 RNA in all the samples. Adjusted for the time lag,
the virus RNA concentrations tracked the Covid-19 epidemiological curve. SARS-CoV-2 RNA concentrations were a leading indicator of community infection
ahead of compiled Covid-19 testing data and local hospital admissions.
Sharif 2020 78 wastewater samples collected from 38 districts across Pakistan, 74 wastewater samples from existing polio environmental surveillance sites, 3 from drains
of Covid-19 infected areas and 1 from Covid-19 quarantine center drainage, were tested for presence of SARs-CoV-2. 21 wastewater samples (27%) from 13
districts were positive by RT-qPCR. This surveillance system has potential to aid monitoring of the pandemic, but attention is needed on virus concentration and
detection assay to increase the sensitivity.
Shutler 2020 Combining in vitro data, pollution analysis and a virus survivability model, based on data from 39 countries, SARS-CoV-2 can remain stable within water for up
to 25 days. Country-specific risk of infection posed by faecal contaminated water is environment-dependent, with water flow and temperature as important
variables.
Trottier 2020 SARS-CoV-2 RNA was assessed in samples from the inflow point of the main waste water treatment plant of Montpellier, France, spring 2020. Samples were
collected 4 days before the end of lockdown (7 May 2020) up to 70 days post-lockdown (20 July 2020). Increased amounts of SARS-CoV-2 RNA were observed
from mid-June on, whereas the number of new Covid-19 cases recorded in the area started increasing a fortnight later.
Wang J 2020 The study reports the presence of SARS-Cov-2 in the hospital environment, surfaces, sewage, and the staff PPE in isolation wards in a Covid-19 hospital in China.
SARS-Cov-2 RNA were positive from inlets of the sewage disinfection pool and negative from the outlet of the last sewage disinfection pool but no viable virus
was detected by culture.
Wang XW 2020 No live SARS-CoV was found in any sewage samples from two hospitals receiving COVID-19 patients. SARS-CoV RNA was detected in sewage concentrates of two
hospitals receiving SARS patients prior to disinfection, and occasionally after disinfection.
Wurtzer 2020 An increase of SARS-CoV-2 genome units in raw wastewaters in and around Paris, France accurately followed the increase of human COVID-19 cases observed
at the regional level.
Zhao 2020 Wastewater, sludge, surface water, ground water, and soil samples of municipal and hospital wastewater systems and related environments in Wuhan during
the Covid-19 middle and low risk periods were tested for SARS-CoV-2 RNA and the viral copies quantified using RT-qPCR. During the middle risk period, 1
influent sample and 3 secondary treatment effluents, 2 influent samples from wastewater system of a Covid-19 hospital were SARS-CoV-2 RNA positive. 1 sludge
sample collected from a Covid-19 hospital 4 during a low risk period, was positive for SARS-CoV-2 RNA.
Toilet and or
Sewage
Del Brutto 2020 SARS-CoV-2 prevalence and incidence were assessed in a rural Guatemalan village setting using serology. One month after baseline testing, 362 of 370 initially
seronegative individuals were re-tested to assess incidence of seroconversion and associated risk factors. Twenty-eight of them (7.7%) became seropositive.
The overall incidence rate ratio was 7.4 per 100 person months of potential virus exposure (95%CI 4.7 to 10.2). The only covariate significantly associated with
seroconversion was the use of an open latrine.
Ding Z 2020 This study randomly sampled in rooms and areas in the COVID-19 designated infectious diseases hospital Nanjing, China. 4/107 surface samples tested
positive: two ward door door-handles, one bathroom toilet toilet-seat cover and one bathroom door door-handle. Three were weakly positive from a bathroom
toilet seat, one bathroom washbasin tap lever and one bathroom ceiling exhaust louvre. 1/46 corridor air samples tested weakly positive.
Kang 2020 An outbreak of 9 confirmed cases of Covid-19 between 26 January 2020 and 13 February 2020 in 3 vertically aligned flats in a high-rise building in Guangzhou,
China, during a period of social distancing, was investigated. There were 9 infected individuals, 193 other residents of the building, and 24 members of the
building's management staff. The researchers collected environmental samples and measured the drainage airflow dispersion of a tracer gas in the block
to investigate the potential for a fecal aerosol transmission route of SARS-CoV-2. No evidence was found for transmission via the elevator or elsewhere. The
families lived in 3 vertically aligned flats connected by drainage pipes in the master bathrooms. Both the observed infections and the locations of positive
environmental samples are consistent with the vertical spread of virus-laden aerosols via these stacks and vents. After tracer gas was released though one flat’s
toilet it was found in all 5 other flats monitored, supporting the possibility of fecal aerosol spread.
Ong 2020 Between 24 January and 4 February 2020, 3 patients in airborne infection isolation rooms with anterooms and bathrooms had surface environmental samples
taken at 26 sites, and air samples. Samples taken after cleaning for two patients were all negative. For one patient (samples taken pre-cleaning): 13/15 room
sites including air outlet fans, 3/5 toilet sites (toilet bowl, sink, and door handle) were PCR positive. Anteroom and corridor samples were negative. This patient
had upper respiratory tract involvement with no pneumonia and had 2 positive stool samples for SARS-CoV-2 on RT-PCR despite not having diarrhea.
Waterways
Guerrero-Latorre
2020
This study assessed the presence of SARS-COV-2 in urban streams from a low sanitation context i.e. highly impacted by sewage. Three river locations along the
urban rivers of Quito, Ecuador were sampled on the 5 June 2020 during a peak of Covid-19 cases. SARS-CoV-2 RNA was detected in all samples, at levels similar
to those in wastewater from cities during outbreaks.

Table 5. Viral culture using fecal samples (n=6 studies).

Study (n=6) Method Viral culture of fecal
samples
n successful/n attempted
Notes Methodological issues
Jeong 2020 Specimens were used to inoculate Vero cells; these were
cultured in Eagle's minimal essential medium with 8%
heat-inactivated fetal bovine serum and antibiotics. Cells
were monitored daily for 4 days to examine the cytopathic
effects. To confirm virus isolation, RT-PCR was performed
on supernatants from infected cell cultures.
0/3 The same study was able to isolate
viable SARS-CoV-2 from naso/
oropharyngeal swabs and saliva of
COVID-19 patients, as well as nasal
washes of ferrets inoculated with
patient urine or stool.
CPE, not plaque assays were cited
as evidence of virus replication.
The low RNA levels, late disease
stage, and high antibody render
it unlikely this study resulted in
cultured virus.
Kim JM 2020 For the cell inoculation, cells were cultured from the
CaCo-2 cell line (derived from human epithelial colorectal
adenocarcinoma cells) in Dulbecco modified Eagle
medium supplemented with 20% fetal bovine serum and
1% penicillin, and were incubated at 37°C, 5% CO2. The
cells were cultured for 5 days and then harvested.

To evaluate the viral replication process, RNA from the
secondary inoculation cell culture supernatant was
extracted and assessed for the presence of SARS-CoV-2
using real-time RT-PCR.
0/13 Collection of samples was reported
as date of hospital admission (not
reported in relation to date of
symptom onset).
Enough genomes by PCR to grow
virus if it was there. Concerned
they couldn’t detect any virus
replication. Some should have
been positive, but an unusual,
indirect assay was used.
Wang W, Xu Y
2020
No details 2/4 Electron microscopy was performed
to detect live virus.
Electron microscopy can identify
virions but does not demonstrate
viral viability.
Wölfel 2020 Vero E6 cells were seeded on a 24-well plate at 3.5 × 10^5
cells/mL in Dulbecco modified Eagle medium containing
1% sodium pyruvate, 1% nonessential amino acids, 1%
l-glutamine, and 10% fetal calf serum one day prior to
inoculation. Cells were observed daily for cytopathogenic
effects for 6 days. Every 2 days or upon observation of
cytopathogenic effects, 50 μl of cell culture supernatant
was subjected to viral RNA extraction and SARS-CoV-2
specific real-time RT-PCR using the SARS-2-CoV E assay.
0/13 Culture of the virus was attempted
on multiple occasions from 13 fecal
samples from 4 patients with high
viral loads between day 6 and day 12
of infection.
Had positive controls showing an
RNA increase in culture, absence
in fecal samples suggests no
infectious virus. Robust study,
clearly illustrating the problem
detecting infectious material after
seroconversion.
Xiao F, Sun J
2020
The cytopathic effect in Vero E cells was observed 2 days
after a second-round passage. Negatively stained culture
supernatant was visualized by transmission electron
microscopy. Viral particles that were visible were spherical
and had distinct surface spike protein projections,
consistent with a previously published SARS-CoV2
image. Viral nucleic acid was collected from cell culture
supernatant, whole genome sequencing identified SARS-
CoV-2.
2 cases Virus culture was attempted from an
unreported number of specimens
and cases, successful in two cases.
The CPE shown in the figure
is not very convincing; it
does not show plaques. Not
immunostained, no evidence by
PCR of growth.
Zhang Y 2020 Vero cells were used for viral isolation from stool samples
(no other methods described). Full-length genome
sequence on the one specimen was identified using ABI
3130 Genetic Analyzer. Genome sequence indicated
high nucleotide similarity (99.98%) to first isolated novel
coronavirus from Wuhan.
1 Isolated the virus from the stools of
one severe hospitalised COVID-19
pneumonia case. The number of
samples taken is unclear.
No evidence of infectious
material; EM is not proof of this.

Discussion

The evidence from 110 relevant studies supports a potential role of orofecal transmission of SARS-CoV-2. Fecal shedding of SARS-CoV-2 RNA has been reported in 96% of the included observational studies, often for relatively long durations. Three studies reported the culture of SARS-CoV-2 using fecal samples, but requisite methods to confirm viral growth were lacking. One study demonstrated viral isolation from rectal tissue of a COVID-19 patient. Studies in hospitals show the presence of SARS-CoV-2 RNA at and around toilets and toilet rooms; there is evidence that disinfectant cleaning leaves no SARS-CoV-2 RNA detectable. Many studies report identifying SARS-CoV-2 RNA in sewage and wastewaters, but viral culture from such sources has not been demonstrated, so there is no evidence of infection risk from those sources; however, detection of SARS-CoV-2 RNA in sewage and wastewaters can be useful as a surveillance tool.

Experimental models of the human intestinal epithelium show that SARS-CoV-2 can infect this tissue and replicate, supporting the rationale for the human GI tract as a possible transmission route 46 ). Zang 2020 demonstrated that human enterocytes express high ACE2 receptor levels, supporting viral invasion at these sites 4 . Zang 2020 and Lamers 2020 et al. showed that SARS-CoV-2 productively infected human small intestinal organoids 4, 5 . Zhou J 2020 et al. and co-workers showed active replication of SARS-CoV-2 in human intestinal organoids, and isolated infectious virus from the stool specimen of a patient with diarrheal COVID-19 6 . Yao et al. investigated the mutation spectrum, replication dynamics, and infectivity of 11 patient-derived SARS-CoV-2 isolates in diverse cell lines; the authors report that "three of our viral isolates were extracted from stool samples (two of which were very potent) indicating that viable SARS-CoV-2 particles could be found in stool samples" 7 .

These studies do not provide an estimate of infectious virus concentration; nor has the infectious dose for humans been established, so at this point it is unwarranted to deduce a human transmission risk based on this small number of virus-positive fecal samples.

MERS-CoV has been shown to infect human primary intestinal epithelial cells, small intestine explants and intestinal organoids 8 . MERS-CoV has been detected in 42% of milk samples collected from lactating camels where it can survive for a prolonged period. A study of human primary intestinal epithelial cells and small intestine explants of MERS-CoV patterns identified the viral replication intermediates in stool specimens. MERS-CoV was found to be resistant to fed-state gastrointestinal fluids but less tolerant to the high acidic fasted-state gastric fluid.

Prolonged excretion of coronaviruses in feces was first observed in 1977 9 . In the SARS-CoV-1 outbreak in 2002-03, a significant portion of patients had enteric involvement. In the Toronto outbreak in 2003, 6% of 144 patients had diarrhoea on presentation 10 . Among 138 patients with SARS in Hong Kong, 20% presented with watery diarrhoea and 38% had symptoms of diarrhoea during the illness. Intestinal biopsy specimens showed the presence of active viral replication, and SARS-CoV RNA was detected in the stool of some patients for more than ten weeks after symptom onset 11 . A retrospective study on specimens from 154 patients in Hong Kong with laboratory-confirmed SARS found the viral load to be the highest in stool specimens 12 . Up to 70% of 75 patients in a community outbreak in Hong Kong developed watery diarrhoea 13 . This outbreak was linked to a faulty sewage system in the Amoy Gardens apartment complex, further suggesting orofecal transmission might be a route for transmission 14 .

The human gastrointestinal tract can act as a primary infection site for SARS-CoV. Ding et al. used a monoclonal antibody specific for the SARS‐CoV nucleoprotein, and probes for the RNA polymerase gene fragment in four patients who died from SARS‐CoV-1 15 . Virus was detected in the stomach, small intestine, distal convoluted renal tubule, sweat gland, parathyroid, pituitary, pancreas, adrenal, liver and cerebrum. The authors discussed that viruses in contaminated food and water may enter the human body through epithelial cells covering the surface of the gastrointestinal tract, although there was no direct evidence to show that food‐borne transmission had occurred. A study from the sewage of two hospitals receiving SARS patients in Beijing found no infectious SARS-CoV contamination in any of the samples collected but did detect the nucleic acid in the sewage from the two hospitals before disinfection - providing further evidence that SARS-CoV-1 can be excreted by feces into the sewage system 16 .

Transmission of coronaviruses via the feces is established among animals: feline coronavirus, for instance, is typically shed in feces of healthy cats and transmitted by the orofecal route to other cats 17 . Pigs are also infected by the transmissible gastroenteritis coronavirus via the orofecal 18 . Bat coronavirus infects the gastrointestinal and respiratory tracts of bats, seemingly without causing disease 19 . Transmission following exposure to camel feces has also been considered biologically plausible 20 .

There is evidence that SARS-CoV-2 can survive adverse conditions in the gastrointestinal system. It has been identified in endoscopic specimens of the oesophagus, stomach, duodenum, and rectum of COVID-19 patients; substantial amounts of SARS-CoV-2 RNA have been consistently detected in stool specimens, and evidence suggests that SARS-CoV-2 can survive the adverse conditions in the gastrointestinal system. Heavy glycosylation of the large spike S protein has been shown to lead to resistance to the proteases, the low pH and bile salts found in the gastrointestinal system. Some gastric processes may actually facilitate viral entry into the enterocytes: in bovine coronavirus, one specific site on the S glycoprotein has to be cleaved by an intracellular protease or trypsin to activate viral infectivity and cell fusion 21 .

Evidence of ingestion, penetration of enterocytes and excretion of live SARS-CoV-2 is possible; however, this working hypothesis requires testing by conducting case-control studies during the investigation of outbreaks, following a set protocol. For such investigations, cases of COVID-19 (categorised by symptom presence and severity) either fecally excreting virions or not (cases and contacts) and controls would be healthy matches. Exposure to potentially fecally contaminated materials and protective measures taken would be elicited at interview. To minimise the play of recall and ascertainment bias, interviewers should be blind to fecal excretion status and the interview should take place as soon as possible after the event. Viability of fecal isolates and their possible pathogenicity should be tested in outbreaks.

Strengths and limitations. This review is limited by the quality of included studies: many were small and did not provide a protocol that established a priori methods. Studies were often poorly reported and often did not take biases into account. Reporting is often heterogeneous and essential information such as symptom onset and cycle threshold values were often missing. We do not have information on publication bias, but the current urgency to understand SARS-CoV-2 may have an impact on research, with unknown implications and a tendency to publish those studies with positive results. We note the possibility that some cases may be reported in more than one publication, but there is no adequate method by which to identify this. It is likely we missed some studies, but we plan to keep updating this review. Furthermore, our judgments of quality are to some extended subjective and open to disagreement. This does not, however, undermine our overall assessment of the quality of the included studies. We perceive that standardization of methods in this area would improve the quality of the research. Some of these limitations increase uncertainties and prevent firm conclusions being drawn; however, this body of research provides largely consistent evidence on the main conclusions that SARS-CoV-2 is excreted fecally, is found in sewage and can be cultured from fecal samples.

Conclusion

Observational and mechanistic evidence as well as established animal orofecal transmission of coronaviruses suggests SARS-CoV-2 can infect and be shed from the human gastrointestinal tract. However, quantitative data on infectiousness and the consequent likelihood of transmission from orofecal contamination is not available. Whilst SARS-CoV-2 RNA is observed in sewage and wastewaters, there is no evidence of infectiousness in these sources and a transmission risk from these sources is considered unlikely based on the reviewed studies. To properly assess these risks, quantitative data on infectious virus are needed, along with information on likely infectious dose in humans.

Data availability

Underlying data

All data underlying the results are available as part of the article and no additional source data are required.

Extended data

Figshare: Extended data for SARS-CoV-2 and the role of orofecal transmission: a systematic review, https://doi.org/10.6084/m9.figshare.14247470.v1 2 .

This project contains the following extended data:

  • -

    Appendix 1: Updated protocol

  • -

    Appendix 2: Search strategy

  • -

    Appendix 3: References to included studies.

Reporting guidelines

Figshare: PRISMA checklist for ‘SARS-CoV-2 and the role of orofecal transmission: a systematic review’, https://doi.org/10.6084/m9.figshare.14247470.v1 2 .

Data are available under the terms of the Creative Commons Attribution 4.0 International license (CC-BY 4.0).

Acknowledgements

A previous version of this review is available on medRxiv: https://doi.org/10.1101/2020.08.04.20168054

Funding Statement

This study was commissioned and paid for by the World Health Organization (WHO), WHO registration No2020/1077093. Copyright in the original work on which this article is based belongs to WHO. The authors have been given permission to publish this article. The authors alone are responsible for the views expressed in the publication, which do not necessarily represent views, decisions or policies of the World Health Organization. CH, ES and AP also receive funding support from the NIHR SPCR Evidence Synthesis Working Group project 390.

The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

[version 2; peer review: 2 approved]

References

  • 1. World Health Organisation.Accessed 2 January 2021. Reference Source [Google Scholar]
  • 2. Heneghan C, Spencer E, Brassey J, et al. : Extended data for SARS-CoV-2 and the role of orofecal transmission: a systematic review. figshare. Dataset.2021. 10.6084/m9.figshare.14247470.v1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Whiting PF, Rutjes AW, Westwood ME, et al. : QUADAS-2 Group. QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med. 2011;155(8):529–36. 10.7326/0003-4819-155-8-201110180-00009 [DOI] [PubMed] [Google Scholar]
  • 4. Lamers MM, Beumer J, van der Vaart J, et al. : SARS-CoV-2 productively infects human gut enterocytes. Science. 2020;369(6499):50–54. 10.1126/science.abc1669 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Zang R, Castro MFG, McCune BT, et al. : TMPRSS2 and TMPRSS4 promote SARS-CoV-2 infection of human small intestinal enterocytes. Sci Immunol. 2020;5(47):eabc3582. 10.1126/sciimmunol.abc3582 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Zhou J, Li C, Liu X, et al. : Infection of bat and human intestinal organoids by SARS-CoV-2. Nat Med. 2020;26(7):1077–1083. 10.1038/s41591-020-0912-6 [DOI] [PubMed] [Google Scholar]
  • 7. Yao H, Lu X, Chen C, et al. : Patient-derived mutations impact pathogenicity of SARS-CoV-2. Reference Source [Google Scholar]
  • 8. Zhou J, Li C, Zhao G, et al. : Human intestinal tract serves as an alternative infection route for Middle East respiratory syndrome coronavirus. Sci Adv. 2017;3(11):eaao4966. 10.1126/sciadv.aao4966 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Caul EO, Egglestone SI: Further studies on human enteric coronaviruses. Arch Virol. 1977;54(1–2):107–17. 10.1007/BF01314383 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Booth CM, Matukas LM, Tomlinson GA, et al. : Clinical features and short-term outcomes of 144 patients with SARS in the greater Toronto area. JAMA. 2003;289(21):2801–2809. 10.1001/jama.289.21.JOC30885 [DOI] [PubMed] [Google Scholar]
  • 11. Leung WK, To KF, Chan PK, et al. : Enteric involvement of severe acute respiratory syndrome-associated coronavirus infection. Gastroenterology. 2003;125(4):1011–7. 10.1016/s0016-5085(03)01215-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Hung IF, Cheng VC, Wu AK, et al. : Viral loads in clinical specimens and SARS manifestations. Emerg Infect Dis. 2004;10(9):1550–1557. 10.3201/eid1009.040058 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Peiris JS, Chu CM, Cheng VC, et al. : Clinical progression and viral load in a community outbreak of coronavirus-associated SARS pneumonia: a prospective study. Lancet. 2003;361(9371):1767–72. 10.1016/s0140-6736(03)13412-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. WHO environmental health team reports on Amoy gardens. Reference Source [Google Scholar]
  • 15. Ding Y, He L, Zhang Q, et al. : Organ distribution of severe acute respiratory syndrome (SARS) associated coronavirus (SARS-CoV) in SARS patients: implications for pathogenesis and virus transmission pathways. J Pathol. 2004;203(2):622–630. 10.1002/path.1560 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Wang XW, Li JS, Guo TK, et al. : Concentration and detection of SARS coronavirus in sewage from Xiao Tang Shan Hospital and the 309th Hospital.[published correction appears in J Virol Methods. 2005 Dec; 130(1–2): 210]. J Virol Methods. 2005;128(1-2):156–161. 10.1016/j.jviromet.2005.03.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Hartmann K: Feline infectious peritonitis. Vet Clin North Am Small Anim Pract. 2005;35(1):39–79, vi. 10.1016/j.cvsm.2004.10.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Zhang Q, Yoo D: Immune evasion of porcine enteric coronaviruses and viral modulation of antiviral innate signaling. Virus Res. 2016;226:128–141. 10.1016/j.virusres.2016.05.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Fan Y, Zhao K, Shi ZL, et al. : Bat Coronaviruses in China. Viruses. 2019;11(3):210. 10.3390/v11030210 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Killerby ME, Biggs HM, Midgley CM, et al. : Middle East Respiratory Syndrome Coronavirus Transmission. Emerg Infect Dis. 2020;26(2):191–198. 10.3201/eid2602.190697 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Holmes KV: Enteric infections with coronaviruses and toroviruses. Novartis Found Symp. 2001;238:258–69; discussion 269-75. 10.1002/0470846534.ch16 [DOI] [PubMed] [Google Scholar]
F1000Res. 2022 Jan 7. doi: 10.5256/f1000research.78110.r98695

Reviewer response for version 2

Mark D Sobsey 1,2

I have read the revised article and the response of the manuscript authors. I agree with the authors' response and the changes in the manuscript. The authors now make clear that so far, there is no evidence for the presence of infectious SARS-CoV-2 in sewage, other wastewater and environmental waters. I support this position. In my judgement, I now consider the article acceptable without reservations. 

Furthermore, the authors and other readers of the manuscript may be interested to know that I recently published an invited review article in the Journal of Water and Health, a journal of the international Water Association, that is entitled: "Absence of virological and epidemiological evidence that SARS-CoV-2 poses COVID-19 risks from environmental fecal waste, wastewater and water exposures' (Sobsey, 2021 1 ).

My JWH article further supports the assessments and conclusions of the authors of this F1000 Research article.

Are the rationale for, and objectives of, the Systematic Review clearly stated?

Yes

Is the statistical analysis and its interpretation appropriate?

Partly

Are sufficient details of the methods and analysis provided to allow replication by others?

Partly

Are the conclusions drawn adequately supported by the results presented in the review?

Partly

Reviewer Expertise:

NA

I confirm that I have read this submission and believe that I have an appropriate level of expertise to confirm that it is of an acceptable scientific standard.

References

  • 1. : Absence of virological and epidemiological evidence that SARS-CoV-2 poses COVID-19 risks from environmental fecal waste, wastewater and water exposures. Journal of Water and Health .2021; 10.2166/wh.2021.182 10.2166/wh.2021.182 [DOI] [PubMed] [Google Scholar]
F1000Res. 2021 Nov 12. doi: 10.5256/f1000research.78110.r98696

Reviewer response for version 2

Jennifer Audsley 1

Thanks to the authors for the responses to my queries. The issues have been addressed satisfactorily.

Are the rationale for, and objectives of, the Systematic Review clearly stated?

Yes

Is the statistical analysis and its interpretation appropriate?

Partly

Are sufficient details of the methods and analysis provided to allow replication by others?

Yes

Are the conclusions drawn adequately supported by the results presented in the review?

Partly

Reviewer Expertise:

I am a Clinical Research Fellow with a major research interest in HIV-Hepatitis co-infection, and since 2020 also COVID-19 infection

I confirm that I have read this submission and believe that I have an appropriate level of expertise to confirm that it is of an acceptable scientific standard.

F1000Res. 2021 Aug 12. doi: 10.5256/f1000research.54778.r89625

Reviewer response for version 1

Jennifer Audsley 1

Heneghan et al. present a systematic review of SARS-CoV-2 and the role of orofecal transmission. They included 110 relevant studies (including 35 reviews) from searches up until 20 th December 2020. Potential orofecal transmission of SARS-CoV-2 is an important public health issue. They found the observational evidence suggested SARS-CoV-2 infects and is shed from the gastrointestinal tract (GIT) and with further testing could allow the development of appropriate public health measures. This systematic review has followed the PRISMA guidelines including developing a review protocol and completing the PRISMA checklist. All underlying data is available and extended data has been uploaded onto Figshare. This review is very well-written and the references relevant. The authors have undertaken this review thoroughly, with the largely low-moderate data available. There are, however, some points that should be addressed to strengthen the review and conclusions.

  • The aim was to systematically review evidence on orofecal SARS-CoV-2 transmission while the conclusion focuses on SARS-CoV-2 fecal excretion and presence in sewage, which is not evidence of transmission. Detection of SARS-CoV-2 in stool samples and in ecological settings is important but it should be noted that was not the aim of this review. Viral culture data from six studies is included but this is a small minority of studies, and as the authors note due to methodological issues should be interpreted with caution.

  • Examining the ecological studies and review is very worthwhile but none address the issues of orofecal SARS-CoV-2 transmission. Perhaps ecological factors are a secondary outcome?

  • While the authors do note in the results that some reviews included overlapping studies, there is no figure/percentage given for this. How much overlap in total is there, is the overlap greater for cohort/case series or ecological studies? What is the quality of the overlapping studies – are they over-representing the low-quality data?

  • What is the breakdown for included reviews between cohort/case series and ecological studies? This is provided for primary studies in Figure 1 but I can’t find it for the reviews.

  • Have you considered a sub analysis of the studies in children?

  • Is gender reported routinely across the studies?

  • How many/percentage of the studies are only on MedRxiv? Consider including breakdown by primary/review, cohort-case series/ecological. This could be a source of bias.

Minor:

  • Table 1 pages 10, 11 & 12 – total patient number from the cohort/case series (9081) appears in the “sewage” header rows.

Are the rationale for, and objectives of, the Systematic Review clearly stated?

Yes

Is the statistical analysis and its interpretation appropriate?

Partly

Are sufficient details of the methods and analysis provided to allow replication by others?

Yes

Are the conclusions drawn adequately supported by the results presented in the review?

Partly

Reviewer Expertise:

I am a Clinical Research Fellow with a major research interest in HIV-Hepatitis co-infection, and since 2020 also COVID-19 infection

I confirm that I have read this submission and believe that I have an appropriate level of expertise to confirm that it is of an acceptable scientific standard, however I have significant reservations, as outlined above.

F1000Res. 2021 Sep 30.
Elizabeth Spencer 1

Thank you for these helpful suggestions to improve the review. Please see our changes made, noted below. 

1. Added: typically quantified by the number of virions, gene copies or equivalents, or by antigen indicators of viral load.

2. Added to the Discussion: these studies do not provide an estimate of infectious virus concentration; nor has the infectious dose for humans been established, so at this point it is unwarranted to deduce a human transmission risk based on this small number of virus-positive fecal samples.

3. Changed Discussion sentence: Many studies report identifying SARS-CoV-2 RNA in sewage and wastewaters, but viral culture from such sources has not been demonstrated, so there is no evidence of infection risk from those sources; however, detection of SARS-CoV-2 RNA in sewage and wastewaters can be useful as a surveillance tool.

4. Added: However, quantitative data on infectiousness and the consequent likelihood of transmission from orofecal contamination is not available. Whilst SARS-CoV-2 RNA is observed in sewage and wastewaters, there is no evidence of infectiousness in these sources and a transmission risk from these sources is therefore unlikely. To properly assess these risks, quantitative data on infectious virus are needed, along with information on likely infectious dose in humans.

5. Added to Discussion: We note the possibility that some cases may be reported in more than one publication, but there is no adequate means by which to identify this.

6. Unclear reporting in the reviews did not allow full analysis of their included studies. To do this we could revisit the full text of their included studies, but this is beyond the scope of this review. Inclusion of these reviews represents a level of interest in the field but necessarily they will overlap in scope and therefore we do not analyse their findings in detail.

7. Thank you for the suggestion to further sub analyse the data; at this stage there are insufficient data for such sub analyses including among children.

8. Unfortunately there are several studies in which sex of participants is not reported and so we were limited by this. 

9. This was a very interesting suggestion to sub-analyse according to preprint publication and examine possible publication biases; this is unfortunately beyond the scope of this review, not least because this changes over time. The next time we update this review we hope to have a better breakdown of those fully published, and hopefully a better bank of evidence to analyse.

10. Thank you for noting the anomaly in the table, we have checked and rectified and hopefully this will all be in the right place now. 

Thank you for these comments.

F1000Res. 2021 Apr 7. doi: 10.5256/f1000research.54778.r82122

Reviewer response for version 1

Mark D Sobsey 1,2

This article is of generally high quality, but improvements are suggested in some areas, including in the conclusions. Starting from the beginning of the article here are some points that should be clarified or improved:

  1. In Box 1. Terminology, Viral load is defined in a rather vague and insufficiently quantitative way. Viral load can be quantified on the basis of virions (physical particles (as seen by electron microscopy), gene copies or equivalents based on RNA units or CT values, antigen units such viral proteins quantified in different ways, including immunofluorescence intensity observed in infected tissues and as infectious units for replication competent virus based on titers in susceptible cell cultures or experimental animals. There is value in focusing on quantified "viral load" in human gastrointestinal tissues, intestinal contents and fecally contaminated materials such as fomites, wastewater (e.g., sewage) and water (such as recreational, agricultural irrigation and drinking waters). These nuances in "viral load" are important in interpreting and communicating the transmission-relevant results reported in the research studies reviewed..

  2. The data found in the study and provided in Table 1 indicate that only a few studies (5 according to table 1) attempted to detect infectious or replication-competent SARS-CoV-2 in either feces, sewage or a related sample, of which only 3 reported the presence of infectious virus. Further, infectious virus results are reported as "yes" or "no" (presence or absence) per sample. The data for infectious virus are not quantified in typical units such as plaque-forming units or TCID-50 units. Therefore, the concentrations of the detected viruses are unknown. Such lack of quantitative data on infectious virus concentration makes it impossible to determine if sufficient infectious virus is present to pose a transmission risk from human exposure. This point should be mentioned in the overall presentation of the infectious virus results. Furthermore, we do not know the infectivity dose-response relationship of SARS-CoV-2 for human hosts, such an estimate of 50% infectious dose. Unless we know what virus concentration is in samples such as feces or sewage/wastewater and unless we know what the likelihood of human infection is from exposure to known concentrations of infectious virus, it is probably unjustified to suggest that the presence of an unknown concentration of infectious virus in a very small number of reported virus-positive fecal samples poses a human health risk from exposure. While infectious virus presence in a few samples is noteworthy, the lack of quantitative infectivity data makes it impossible to judge that there is plausible human health exposure risk from this infectious virus-positive fecal matter of unknown concentration. The best one can say is that the risk of infection or other health effects from exposure is not zero, but is also not quantifiable due to lack of data on virus concentration in feces and lack of knowledge of human infectivity dose-response for infection or other health endpoints. 

  3. For sewage and water, the lack of data on virus infectivity in such samples makes it impossible to determine if these samples pose risks of human infection from exposure. Although some data on concentrations of viral RNA are reported based on estimated gene copies or CT values, the quantitative relationship of virus infectious units to RNA concentration units is highly variable and therefore uncertain This is especially so for samples that have been in the environment for unknown and variable periods of time since the feces were shed from a human host. This is due to the variable and difficult-to-quantify effects of exposure time, temperature, microbial activity, predation and other environmental stressors on virus survival. These factors are not mentioned in the paper, even though we know they are important for determining the quality and validity of the results for infectious virus presence and concentrations. The last sentence of the first paragraph of the Discussion section does not adequately address the issue of lack of evidence of virus culture in wastewater and sewage and its implications for human health risks from sewage. Instead it sidesteps to the potential value using SARS-CoV-2 RNA in sewage and wastewater as a surveillance tool. This seems to miss the main point of assessing the evidence for human health risk from exposure to sewage or wastewater.In the "strengths and limitations section of the Discussion section, the authors state "however, this body of research provides largely consistent evidence on the main conclusions that SARS-CoV-2 is excreted fecally, is found in sewage and can be cultured from fecal samples." While there there is some evidence that unknown but probably low concentrations of infectious SARS-CoV-2 have been occasionally found in excreted feces, they have not been found in sewage or wastewater. The wording of the above sentence conflates feces with wastewater and sewage, even though there is only limited and poor evidence for infectious virus in the latter and no evidence for it in sewage or wastewater. I recommend that the sentence be revised to avoid conflating these rather separate exposure sources of feces versus sewage and wastewater.

  4. The Conclusion sentence is short and limited in scope. It does not adequately address the issue of whether sufficient infectious SARS-CoV-2 is or is not likely to be present in feces or wastewater and sewage to pose human health risks from exposure. The available evidence seems to indicate that such risks are not possible to assess or quantify without better and more rigorous and as well as more quantitative data that is still not available.

Are the rationale for, and objectives of, the Systematic Review clearly stated?

Yes

Is the statistical analysis and its interpretation appropriate?

Partly

Are sufficient details of the methods and analysis provided to allow replication by others?

Partly

Are the conclusions drawn adequately supported by the results presented in the review?

Partly

Reviewer Expertise:

I am an environmental and public health virologist and microbiologist with expertise in infection protection and control and water-sanitation and hygiene, as well as in infectious disease epidemiology in the context of environmental exposure risks and disease transmission of human pathogens .

I confirm that I have read this submission and believe that I have an appropriate level of expertise to confirm that it is of an acceptable scientific standard, however I have significant reservations, as outlined above.

F1000Res. 2021 Sep 30.
Elizabeth Spencer 1

Thank you for these helpful suggestions to improve the review. Please see our changes made, noted below. 

1. Added: typically quantified by the number of virions, gene copies or equivalents, or by antigen indicators of viral load.

2. Added to the Discussion: these studies do not provide an estimate of infectious virus concentration; nor has the infectious dose for humans been established, so at this point it is unwarranted to deduce a human transmission risk based on this small number of virus-positive fecal samples.

3. Changed Discussion sentence: Many studies report identifying SARS-CoV-2 RNA in sewage and wastewaters, but viral culture from such sources has not been demonstrated, so there is no evidence of infection risk from those sources; however, detection of SARS-CoV-2 RNA in sewage and wastewaters can be useful as a surveillance tool.

4. Added: However, quantitative data on infectiousness and the consequent likelihood of transmission from orofecal contamination is not available. Whilst SARS-CoV-2 RNA is observed in sewage and wastewaters, there is no evidence of infectiousness in these sources and a transmission risk from these sources is therefore unlikely. To properly assess these risks, quantitative data on infectious virus are needed, along with information on likely infectious dose in humans.

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Data Availability Statement

    Underlying data

    All data underlying the results are available as part of the article and no additional source data are required.

    Extended data

    Figshare: Extended data for SARS-CoV-2 and the role of orofecal transmission: a systematic review, https://doi.org/10.6084/m9.figshare.14247470.v1 2 .

    This project contains the following extended data:

    • -

      Appendix 1: Updated protocol

    • -

      Appendix 2: Search strategy

    • -

      Appendix 3: References to included studies.

    Reporting guidelines

    Figshare: PRISMA checklist for ‘SARS-CoV-2 and the role of orofecal transmission: a systematic review’, https://doi.org/10.6084/m9.figshare.14247470.v1 2 .

    Data are available under the terms of the Creative Commons Attribution 4.0 International license (CC-BY 4.0).


    Articles from F1000Research are provided here courtesy of F1000 Research Ltd

    RESOURCES