ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

COVID-19: Epidemiology, virology, and prevention

COVID-19: Epidemiology, virology, and prevention
Author:
Kenneth McIntosh, MD
Section Editor:
Martin S Hirsch, MD
Deputy Editor:
Allyson Bloom, MD
Literature review current through: Jan 2024.
This topic last updated: Jan 31, 2024.

INTRODUCTION — Coronaviruses are important human and animal pathogens. At the end of 2019, a novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was identified as the cause of a cluster of pneumonia cases in Wuhan, a city in the Hubei Province of China. It rapidly spread, resulting in an epidemic throughout China, followed by a global pandemic. In February 2020, the World Health Organization (WHO) designated the disease COVID-19, which stands for coronavirus disease 2019 [1]. The WHO declared an end to the COVID-19 global health emergency in May 2023, more than three years after its emergence.

This topic will discuss the virology, epidemiology, and prevention of COVID-19. The clinical features and diagnosis of COVID-19 are discussed in detail elsewhere. (See "COVID-19: Clinical features" and "COVID-19: Diagnosis".)

The management of COVID-19 is also discussed in detail elsewhere:

(See "COVID-19: Management in hospitalized adults".)

(See "COVID-19: Management of adults with acute illness in the outpatient setting".)

(See "COVID-19: Infection prevention for persons with SARS-CoV-2 infection".)

Issues related to COVID-19 in pregnant women and children are discussed elsewhere:

(See "COVID-19: Overview of pregnancy issues".)

(See "COVID-19: Clinical manifestations and diagnosis in children" and "COVID-19: Multisystem inflammatory syndrome in children (MIS-C) clinical features, evaluation, and diagnosis".)

See specific topic reviews for details on complications of COVID-19 and issues related to COVID-19 in other patient populations.

VIROLOGY

Coronavirus virology — Coronaviruses are enveloped positive-stranded RNA viruses. Full-genome sequencing and phylogenic analysis indicated that the coronavirus that causes COVID-19 is a betacoronavirus in the same subgenus as the severe acute respiratory syndrome (SARS) virus (as well as several bat coronaviruses), but in a different clade. The Coronavirus Study Group of the International Committee on Taxonomy of Viruses has proposed that this virus be designated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) [2]. The Middle East respiratory syndrome (MERS) virus, another betacoronavirus, appears more distantly related [3,4]. The closest RNA sequence similarity is to two bat coronaviruses, and it appears likely that bats are the primary source; whether COVID-19 virus is transmitted directly from bats or through some other mechanism (eg, through an intermediate host) is unknown [5]. (See "Coronaviruses", section on 'Virology'.)

The host receptor for SARS-CoV-2 cell entry is the same as for SARS-CoV, the angiotensin-converting enzyme 2 (ACE2) [6]. SARS-CoV-2 binds to ACE2 through the receptor-binding domain of its spike protein (figure 1). The cellular serine protease TMPRSS2 is also important for SARS-CoV-2 cell entry [7].

Variants of concern — Like other viruses, SARS-CoV-2 evolves over time. Most mutations in the SARS-CoV-2 genome have no impact on viral function. Certain variants have garnered widespread attention because of their rapid emergence within populations and evidence for transmission or clinical implications; these are considered variants of concern (table 1). Each variant has several designations based on the nomenclature used by distinct phylogenetic classification systems; the World Health Organization (WHO) has also designated labels for notable variants based on the Greek alphabet [8].

Omicron (B.1.1.529) and its sublineages — The Omicron variant was first reported from Botswana and very soon thereafter from South Africa in November 2021 (table 1). In South Africa, it was associated with an increase in regional infections, and it was promptly identified in multiple other countries, where it was similarly associated with sharp increases in reported infections [9-12]. Subsequently, Omicron sublineages with increasingly greater replication advantages emerged, replacing the previous predominant sublineage. The original Omicron variant was sublineage BA.1, followed by sublineage BA.2, which in turn was supplanted by BA.4 and BA.5 [13]. Other Omicron sublineages, such as BQ.1, BQ.11, BF.7, BA.2.75, XBB (including XBB.1, XBB.1.5, and XBB.1.6), and EG.5 (related to XBB.1.5) initially increased and subsequently waned in prevalence worldwide. Some Omicron sublineages, such as BA.2.86 (a distant descendant of BA.2) [14], were concerning for their immune-escape potential but did not become dominant among circulating variants.

In the United States, the proportion of variants circulating in different regions of the country can be found on the CDC variant tracker website.

Several Omicron sublineages had a replication advantage over the Delta variant and evaded infection- and vaccine-induced humoral immunity to a greater extent than prior variants. In general, they also appear to be associated with less severe disease than other variants:

Replication advantage – The emergence of each predominant Omicron sublineage had been associated with local increases in SARS-CoV-2 infections, suggesting a replication advantage over the prior prevailing variant or sublineage. Omicron (specifically the BA.1 sublineage) has also been associated with a higher secondary attack rate compared with Delta (in one study, 25 versus 19 percent) [15]. Another study of household contacts of patients with Omicron BA.1 infection suggested a secondary attack rate of 53 percent, which varied by vaccination status of the index patient and use of preventive measures in the household [16]. Data on secondary attack rates of other Omicron sublineages are lacking.

The replication advantage may be related, in part, to immune escape by Omicron sublineages, as discussed below. Whether Omicron sublineages are inherently more transmissible than the variants that preceded them is uncertain. Immune escape as well as increased affinity for the ACE2 receptor are proposed explanations for the replication advantage of the XBB.1.5 subvariant and its predecessors [17].

Immune evasion – Omicron variants may escape humoral immunity and have been associated with a higher risk of reinfection in individuals previously infected with a different strain. In a study evaluating national surveillance data from South Africa, the ratio of reinfections (repeat positive test at least 90 days after an earlier positive test) to primary infections was higher during the beginning of the case surge associated with the Omicron BA.1 variant compared with the surges associated with the Beta and Delta variants (0.25 versus 0.12 and 0.09) [18]. Similar findings were reported from a case-control study from Qatar, in which a history of prior infection was associated with an 85 to 90 percent lower risk of infection with Alpha, Delta, or Beta variants, but only a 56 percent lower risk with Omicron BA.1 [19]. These observations were further supported by findings from several laboratories, in which sera from individuals with prior infection or prior vaccination did not neutralize Omicron as well as other variants; in some cases, neutralizing activity against Omicron was undetectable in convalescent as well as post-vaccination sera [20-22]. Similarly, compared with Omicron BA.1, sublineages BA.2.12.1, BA.4, and BA.5 were not as well recognized by antibodies elicited by BA.1 or BA.2 infection or vaccination [23-25]. However, prior infection with one Omicron subvariant may still provide some protection against subsequent infection with certain other subvariants, even if in vitro studies suggest low antibody cross-reactivity [26-28]. As an example, the risk of reinfection with BA.4 and BA.5 was lower following BA.1 or BA.2 infection than following infection with a pre-Omicron variant [26,27]. The impact of vaccine-induced immunity on risk of Omicron infection is discussed elsewhere. (See "COVID-19: Vaccines", section on 'Benefits of vaccination'.)

Other data suggest that Omicron sublineages escape binding by bamlanivimab-etesevimab, casirivimab-imdevimab, and regdanvimab (a monoclonal antibody therapy available outside the United States), and thus these monoclonal antibodies are expected to be ineffective when these sublineages are circulating (table 1) [24,29-31]. Sotrovimab appears to bind to Omicron BA.1, but not to BA.2, BA.2.12.1, BA.4, or BA.5 [24,32]. Bebtelovimab and cilgavimab (a component of tixagevimab-cilgavimab) retain activity against these Omicron sublineages. However, emerging Omicron sublineages appear to escape neutralization by one or both of these monoclonal antibodies [33]. In particular, bebtelovimab and tixagevimab-cilgavimab activity against BQ.1, BQ.1.1, and XBB (and its derivatives XBB.1 and XBB.1.5) is greatly reduced [34].

Severity of disease – Observational data from multiple studies suggest that the risk of severe disease or death with Omicron infection is lower than with prior variants of concern [35-42]. An analysis from England estimated that the risk of hospital admission or death with Omicron was approximately one-third that with Delta, adjusted for age, sex, vaccination status, and prior infection [38]. Evidence also suggests that the risk of severe disease with BA.4 and BA.5 was similar to that with earlier Omicron sublineages [43-45].

Even if the individual risk for severe disease with Omicron is lower than with prior variants, the high number of associated cases can still result in a cumulative excess of COVID-19-associated hospitalizations and deaths compared with other variants [46,47].

The reduced risk for severe disease may reflect partial protection conferred by prior infection or vaccination. However, animal studies show lower viral levels in lung tissue and milder clinical features (eg, less weight loss) with Omicron compared with other variants; this provides further support that Omicron infection may be intrinsically less severe [48-50].

Impact on diagnostic testing – This is discussed in detail elsewhere. (See "COVID-19: Diagnosis", section on 'Impact of SARS-CoV-2 mutations/variants on test accuracy'.)

Others

Alpha (B.1.1.7 lineage) – This variant was first identified in the United Kingdom in late 2020 and subsequently became the globally dominant variant until the emergence of the Delta variant (table 1) [51-53]. Alpha was approximately 50 to 75 percent more transmissible than previously circulating strains [51,54-57]. Some [58,59], but not all, studies [60] suggested that the Alpha variant was associated with greater disease severity.

Beta (B.1.351 lineage) – This variant, also known as 20H/501Y.V2, was identified and predominated in South Africa in late 2020 (table 1) [61]. Although it was subsequently identified in other countries, including the United States, it did not become a globally dominant variant. The main concern with Beta variant was immune evasion: convalescent and post-vaccination plasma did not neutralize viral constructs with Beta spike protein as well as those with wild-type spike protein [62-65].

Gamma (P.1 lineage) This variant, also known as 20J/501Y.V3, was first identified in Japan in December 2020 and was prevalent in Brazil (table 1) [66]. Although it was subsequently identified in other countries, including the United States, it did not become a globally dominant variant. Several mutations in the variant raised concern about increased transmissibility and an impact on immunity [67].

Delta (B.1.617.2 lineage) — This lineage was first identified in India in December 2020 and had since been the most prevalent variant worldwide until emergence of the Omicron variant (table 1). Compared with the Alpha variant, the Delta variant was more transmissible [68,69] and was associated with a higher risk of severe disease and hospitalization [68,70-72]. Several studies suggest that vaccine effectiveness is slightly attenuated against symptomatic infection with Delta but remains high against severe disease and hospitalization.

EPIDEMIOLOGY

Geographic distribution and case counts — Since the first reports of cases from Wuhan, a city in the Hubei Province of China, at the end of 2019, cases have been reported in all continents. Globally, over 750 million confirmed cases of COVID-19 have been reported. A map highlighting reported cases throughout the world can be found here.

However, the reported case counts underestimate the overall burden of COVID-19, as only a fraction of acute infections are diagnosed and reported. Seroprevalence surveys in the United States and Europe have suggested that after accounting for potential false positives or negatives, the rate of prior exposure to SARS-CoV-2, as reflected by seropositivity, exceeds the incidence of reported cases by approximately 10-fold or more [73-76]. One study that used multiple data sources, including databases on case counts, COVID-19-related deaths, and seroprevalence, estimated that by November 2021, over 3 billion individuals, or 44 percent of the world’s population, had been infected with SARS-CoV-2 at least once [77]. Approximately one-third of the total cases were estimated to have occurred in South Asia (including India).

Transmission — Person-to-person spread is the main mode of SARS-CoV-2 transmission.

Person-to-person

Route of person-to-person transmission

Respiratory transmission as the primary route – Direct person-to-person respiratory transmission is the primary means of transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) [78]. It is thought to occur mainly through close-range contact (ie, within approximately six feet or two meters) via respiratory particles; virus released in the respiratory secretions when a person with infection coughs, sneezes, or talks can infect another person if it is inhaled or makes direct contact with the mucous membranes. Infection might also occur if a person's hands are contaminated by these secretions or by touching contaminated surfaces and then they touch their eyes, nose, or mouth, although contaminated surfaces are not a major route of transmission.

SARS-CoV-2 can also be transmitted longer distances through the airborne route (through inhalation of particles that remain in the air over time and distance), but the extent to which this mode of transmission has contributed to the pandemic is uncertain [79-82]. Scattered reports of SARS-CoV-2 outbreaks (eg, in a restaurant, on a bus) have highlighted the potential for longer distance airborne transmission in enclosed, poorly ventilated spaces [83-86]. Experimental studies have also supported the feasibility of airborne transmission [87-89]. Other studies have identified viral RNA in ventilation systems and in air samples of hospital rooms of patients with COVID-19, including patients with mild infection [90-94]; attempts to find viable virus in air and surface specimens in health care settings have only rarely been successful [93-97]. Nevertheless, the overall transmission and secondary attack rates of SARS-CoV-2 suggest that long-range airborne transmission is not a primary mode [81,82]. Recommendations on airborne precautions in the health care setting vary by location; airborne precautions are universally recommended when aerosol-generating procedures are performed. This is discussed in detail elsewhere. (See "COVID-19: Infection prevention for persons with SARS-CoV-2 infection", section on 'Aerosol-generating procedures/treatments'.)

Uncertain or minimal role of other person-to-person routes – SARS-CoV-2 has been detected in non-respiratory specimens, including stool, blood, ocular secretions, and semen, but the role of these sites in transmission is uncertain [98-105]. In particular, several reports have described detection of SARS-CoV-2 RNA from stool specimens, even after viral RNA could no longer be detected from upper respiratory specimens [101,102], and replicative virus has been cultured from stool in rare cases [99,106]. Scattered reports of clusters in a residential building and in a dense urban community with poor sanitation have suggested the possibility of transmission through aerosolization of virus from sewage drainage [107,108]. However, according to a joint WHO-China report, transmission through the fecal-oral route did not appear to be a significant factor in the spread of infection [109].

Detection of SARS-CoV-2 RNA in blood has also been reported in some but not all studies that have tested for it [98,99,102,110,111]. However, the likelihood of bloodborne transmission (eg, through blood products or needlesticks) appears low; respiratory viruses are generally not transmitted through the bloodborne route, and transfusion-transmitted infection has not been reported for SARS-CoV-2 or for the related Middle East respiratory syndrome coronavirus (MERS-CoV) or SARS-CoV [112]. (See "Blood donor screening: Laboratory testing", section on 'Emerging infectious disease agents'.)

There is also no evidence that SARS-CoV-2 can be transmitted through contact with non-mucous membrane sites (eg, abraded skin).

The risk of vertical transmission of SARS-CoV-2 is discussed elsewhere. (See "COVID-19: Overview of pregnancy issues", section on 'Risk of vertical transmission'.)

Viral shedding and period of infectiousness — The potential to transmit SARS-CoV-2 begins prior to the development of symptoms and is highest early in the course of illness; the risk of transmission decreases thereafter. Transmission after 10 days of illness is unlikely, particularly for otherwise immunocompetent patients with nonsevere infection.

Period of greatest infectiousness – Infected individuals are more likely to be contagious within the first 7 to 10 days of infection, when viral RNA levels from upper respiratory specimens are the highest and infectious virus is most likely detectable [113-121]. This is supported by data evaluating the duration of transmission risk. One modeling study, in which the mean serial interval between the onset of symptoms among 77 transmission pairs in China was 5.8 days, estimated that infectiousness peaked between two days before and one day after symptom onset and declined within seven days [116]. In another study that evaluated over 2500 close contacts of 100 patients with COVID-19 in Taiwan, all of the 22 secondary cases had their first exposure to the index case within six days of symptom onset; there were no infections documented in the 850 contacts whose exposure was after this interval [122].

Most of these data were collected during the first year of the pandemic. Subsequent data on the Omicron variant suggest that the peak of viral RNA and greatest likelihood of infectious virus shedding may occur slightly later, at three to six days after symptom onset [123,124]. Nevertheless, the median duration that infectious Omicron virus was detectable in nasal specimens ranged from three to five days following diagnosis, and infectious virus was rarely detected more than 10 days after symptom onset, suggesting that transmission after this period remains unlikely with Omicron [125,126].

Prolonged viral RNA detection does not indicate prolonged infectiousness – The duration of viral RNA shedding is variable and may increase with age and the severity of illness [102,115,127-133]. In a review of 28 studies, the pooled median duration of viral RNA detection in respiratory specimens was 18 days following the onset of symptoms; in some individuals, viral RNA was detected from the respiratory tract several months after the initial infection [132]. Detectable viral RNA, however, does not necessarily indicate the presence of infectious virus, and there appears to be a threshold of viral RNA level below which infectiousness is unlikely.

As an example, in a study of nine patients with mild COVID-19, infectious virus was not detected from respiratory specimens when the viral RNA level was <106 copies/mL [115]. In other studies, infectious virus has only been detected in respiratory specimens with high concentrations of viral RNA. Such high viral RNA concentrations are reflected by lower numbers of reverse transcriptase polymerase chain reaction (RT-PCR) amplification cycles necessary for detection. Depending on the study, the cycle threshold (Ct) for specimen culture positivity may vary from <24 to ≤32 [134,135]. Isolation of infectious virus from upper respiratory specimens more than 10 days after illness onset has only rarely been documented in patients who had nonsevere infection and whose symptoms have resolved [115,134-139].

Occasional reports have described isolation of infectious virus from respiratory specimens for several months following symptom onset in immunocompromised patients [140-144]. Prolonged shedding of virus in fecal specimens has also been described [106]. Further data are needed to understand the frequency and clinical significance of these findings.

The relevance of virus and viral RNA detection to duration of infection control precautions is discussed elsewhere. (See "COVID-19: Infection prevention for persons with SARS-CoV-2 infection", section on 'Discontinuation of precautions'.)

Risk of transmission depends on exposure type — The risk of transmission from an individual with SARS-CoV-2 infection varies by the type and duration of exposure, use of preventive measures, and individual factors (eg, the amount of virus in respiratory secretions) [145]. Many individuals do not transmit SARS-CoV-2 to anyone else, and epidemiologic data suggest that the minority of index cases result in the majority of secondary infections [146-148].

The risk of transmission after contact with an individual with COVID-19 increases with the closeness and duration of contact and appears highest with prolonged contact in indoor settings. Thus, most secondary infections have been described in the following settings:

Among household contacts [149-152]. In a systematic review of 87 studies published through June 2021 that included over 1.2 million household contacts of individuals with SARS-CoV-2 infection in 30 countries, the overall secondary household attack rate was 18.9 percent (95% CI 16.2-22), although there was substantial heterogeneity across studies [152]. However, the attack rate increased over time with the emergence of more transmissible variants. As an example, in a subsequent systematic review of 58 studies published from June 2021 to March 2022, the secondary household attack rate during circulation of different variants was 36 percent (95% CI 33-39) for the Alpha variant, 29.7 percent (95% CI 23-37) for Delta, and 43 percent (95% CI 35-50) for Omicron. During Omicron prevalence, vaccination status of either the index case or household contact was not associated with a statistically significant difference in secondary attack rate [153]. However, other potential features that could impact household transmission rates (such as isolation from other household members) were not accounted for. (See 'Omicron (B.1.1.529) and its sublineages' above.)

Within households, spouses or significant others have the highest secondary infection rates [149]. Nevertheless, children and adolescents can also serve as index cases for secondary household infections [154-156]. (See "COVID-19: Clinical manifestations and diagnosis in children", section on 'Transmission'.)

In health care settings when personal protective equipment was not used (including hospitals [157] and long-term care facilities [158]).

In other congregate settings where individuals are residing or working in close quarters (eg, cruise ships [159], homeless shelters [160,161], detention facilities [162,163], college dormitories [164], and food processing facilities [165,166]).

Although transmission rates are highest in household and congregate settings, frequently reported clusters of cases after social or work gatherings also highlight the risk of transmission through close, non-household social contact [85,167-169]. As an example, epidemiologic analysis of a cluster of cases in the state of Illinois showed probable transmission through two family gatherings at which communal food was consumed, embraces were shared, and extended face-to-face conversations were exchanged with symptomatic individuals who were later confirmed to have COVID-19 [167]. Going to restaurants and other drinking or eating establishments has also been associated with a higher likelihood of infection, likely because of the difficulty with mask-wearing and distancing in such settings [170,171]. (See 'Wearing masks in the community' below.)

Superspreading events, in which large clusters of infections can been traced back to a single index case, are thought to be major drivers of the pandemic [145,146,172]. They have been mainly described following prolonged group exposure in an enclosed, usually crowded, indoor space. As an example, in an outbreak among a choir group, 33 confirmed and 20 probable cases were identified among 61 members who attended a practice session with a symptomatic index case [85]. This outbreak also highlighted the possibility of a high transmission risk through singing in close proximity.

Variable amounts of virus in respiratory secretions may contribute to the variable risk of transmission from different individuals [173,174]. In an observational study that included 282 individuals with COVID-19 who had undergone respiratory tract viral RNA quantification as part of a trial and 753 of their close contacts, transmission was identified from only 32 percent of index patients [173]. Higher respiratory tract RNA levels (taken at a median of four days after symptom onset) were independently associated with higher secondary attack rates.

Traveling with an individual with COVID-19 is also a high-risk exposure [175-178], as it generally results in close contact for a prolonged period. One study reported a 62 percent attack rate among passengers who shared a business class cabin with the index case during a 10-hour flight; almost all of the infected individuals (11 of 12) had been seated within six feet (two meters) of the index case [176]. An analysis from China looked at the risk among individuals who traveled by train and were exposed within three rows to individuals later confirmed to have COVID-19 [177]. The study identified 2334 primary and 234 secondary cases for an overall attack rate 0.32 percent. The risk of secondary infection was highest (3.5 percent) for individuals in seats adjacent to the index patient, and higher for those seated in the same row than for those in front or behind. The risk also increased over time of travel. This study could not account for the possibility that individuals seated next to one another could have been from the same household or shared other exposures.

The risk of transmission in outdoor settings appears to be substantially lower than indoors, although data are limited [179]. Nevertheless, close contact with an individual with COVID-19 remains a risk outdoors.

The risk of transmission with more indirect contact (eg, passing someone with infection on the street, handling items that were previously handled by someone with infection) is not well established and is likely very low. However, many individuals with COVID-19 do not report having had a specific close contact with COVID-19 in the weeks prior to diagnosis [180].

The risk of transmission from children with COVID-19 is discussed in detail elsewhere. (See "COVID-19: Clinical manifestations and diagnosis in children", section on 'Transmission'.)

Asymptomatic or presymptomatic transmission — Transmission of SARS-CoV-2 from individuals with infection but no symptoms (including those who later developed symptoms and thus were considered presymptomatic) has been well documented [181-187].

The biologic basis for this is supported by a study of a SARS-CoV-2 outbreak in a long-term care facility, in which infectious virus was cultured from RT-PCR-positive upper respiratory tract specimens in presymptomatic and asymptomatic patients as early as six days prior to the development of typical symptoms [188]. The levels and duration of viral RNA in the upper respiratory tract of asymptomatic patients are also similar to those of symptomatic patients [189].

The risk of transmission from an individual who is asymptomatic appears less than that from one who is symptomatic [150,155,190-193]. As an example, in an analysis of 628 COVID-19 cases and 3790 close contacts in Singapore, the risk of secondary infection was 3.85 times higher among contacts of a symptomatic individual compared with contacts of an asymptomatic individual [194]. Similarly, in an analysis of American passengers on a cruise ship that experienced a large SARS-CoV-2 outbreak, SARS-CoV-2 infection was diagnosed in 63 percent of those who shared a cabin with an individual with asymptomatic infection, compared with 81 percent of those who shared a cabin with a symptomatic individual and 18 percent of those without a cabin mate [192].

Nevertheless, asymptomatic or presymptomatic individuals are less likely to isolate themselves from other people, and the extent to which transmission from such individuals contributes to the pandemic is uncertain. A CDC modeling study estimated that 59 percent of transmission could be attributed to individuals without symptoms: 35 percent from presymptomatic individuals, and 24 percent from those who remained asymptomatic [195]. This estimate was based on several assumptions, including that 30 percent of infected individuals never develop symptoms and are 75 percent as infectious as those who do.

Incubation period — The time from exposure to infection is discussed in detail elsewhere. (See "COVID-19: Clinical features", section on 'Incubation period'.)

Environmental contamination — Virus present on contaminated surfaces may be another source of infection if susceptible individuals touch these surfaces and then transfer infectious virus to mucous membranes in the mouth, eyes, or nose. The frequency and relative importance of this type of transmission are uncertain, although contaminated surfaces are not thought to be a major source of transmission. It may be more likely a potential source of infection in settings where there is heavy viral contamination (eg, in an infected individual's household or in health care settings).

Extensive SARS-CoV-2 RNA contamination of environmental surfaces in hospital rooms and residential areas of patients with COVID-19 has been described [90,196,197]. In a study from Singapore, viral RNA was detected on nearly all surfaces tested (handles, light switches, bed and handrails, interior doors and windows, toilet bowl, sink basin) in the airborne infection isolation room of a patient with symptomatic mild COVID-19 prior to routine cleaning [90]. Viral RNA was not detected on similar surfaces in the rooms of two other symptomatic patients following routine cleaning (with sodium dichloroisocyanurate). Of note, viral RNA detection does not necessarily indicate the presence of infectious virus [115].

It is unknown how long SARS-CoV-2 can persist on surfaces [198-200]; other coronaviruses have been tested and may survive on inanimate surfaces for up to six to nine days without disinfection. In a study evaluating the survival of viruses dried on a plastic surface at room temperature, a specimen containing SARS-CoV (a virus closely related to SARS-CoV-2) had detectable infectivity at six but not nine days [199]. However, in a systematic review of similar studies, various disinfectants (including ethanol at concentrations between 62 and 71%) inactivated a number of coronaviruses related to SARS-CoV-2 within one minute [198]. Simulated sunlight has also been shown to inactivate SARS-CoV-2 over the course of 15 to 20 minutes in experimental conditions, with higher levels of ultraviolet-B (UVB) light associated with more rapid inactivation [201]. Based on data concerning other coronaviruses, duration of viral persistence on surfaces also likely depends on the ambient temperature, relative humidity, and the size of the initial inoculum [202].

These data highlight the importance of environmental disinfection in the home and health care setting. (See "COVID-19: Infection prevention for persons with SARS-CoV-2 infection", section on 'Environmental disinfection'.)

Risk of animal contact — SARS-CoV-2 infection is thought to have originally been transmitted to humans from an animal host, but the ongoing risk of transmission through animal contact is likely minimal. There is no evidence suggesting animals (including domesticated animals) are a major source of infection in humans.

SARS-CoV-2 infection has been described in animals in both natural and experimental settings. There have been rare reports of animals with SARS-CoV-2 infection (including asymptomatic infections in dogs and symptomatic infections in felines) following close contact with a human with COVID-19 [203-206]. Moreover, asymptomatic, experimentally infected domestic cats may transmit SARS-CoV-2 to cats they are caged with [207]. The risk of infection may vary by species. In one study evaluating infection in animals after intranasal viral inoculation, SARS-CoV-2 replicated efficiently in ferrets and cats; viral replication was also detected in dogs, but they appeared to be less susceptible overall to experimental infection [208]. Pigs and poultry were not susceptible to infection. Mink appear highly susceptible to SARS-CoV-2; outbreaks on mink farms have been reported in Europe and the United States, and in this setting, suspected cases of mink to human transmission have been described, including cases with SARS-CoV-2 variants that appear less susceptible to neutralizing antibodies to wild-type virus [209-211]. In view of these findings, mink on farms in both the Netherlands and Denmark have been, or are being, culled. Hamster-to-human transmission resulting in a large cluster of human cases has also been described [212].

Immune responses following infection — Protective SARS-CoV-2-specific antibodies and cell-mediated responses are induced following infection. Evidence suggests that some of these responses can be detected for at least a year following infection.

Humoral immunity – Following infection with SARS-CoV-2, the majority of patients develop detectable serum antibodies to the receptor-binding domain of the viral spike protein and associated neutralizing activity [114,115]. However, the magnitude of antibody response may be associated with severity of disease, and patients with mild infection may not mount detectable neutralizing antibodies [213,214]. When neutralizing antibodies are elicited, they generally decline over several months after infection, although studies have reported detectable neutralizing activity up to 12 months [215-221]. Other studies have also identified spike- and receptor-binding domain memory B cells that increased over the few months after infection as well as spike protein-specific plasma cells, and these findings suggest the potential for a long-term memory humoral response [215,217,218,222]. Although detectable antibodies and neutralizing activity have been associated with protection from subsequent infection [223-227], humoral responses after infection with one variant do not necessarily provide strong protection against other variants. (See 'Risk of reinfection' below.)

Cell-mediated immunity – Studies have also identified SARS-CoV-2-specific CD4 and CD8 T cell responses in patients who had recovered from COVID-19 and in individuals who had received COVID-19 vaccination, which suggest the potential for a durable T cell immune response [215,222,228,229].

Risk of reinfection — Following emergence of the Omicron variant and its sublineages, reinfections have been common. Prior infection with pre-Omicron variants provides poor long-term protection against subsequent Omicron infection; in one meta-analysis, it was estimated to reduce the risk by approximately 35 percent after 40 weeks [230]. Infection with one Omicron sublineage also does not prevent reinfection with a second Omicron sublineage; the risk depends on how closely the sublineages are related (and thus how likely one sublineage evades immunity from a different one), as well as the interval since the prior infection [231]. In a study of reinfections in the United States, by the end of 2022, when Omicron sublineage BQ.1/BQ.1.1 was circulating, half of reinfections occurred in individuals who were previously infected when a different Omicron sublineage was circulating [232].

The risk of severe disease with reinfection is generally less than with initial infections. As an example, in a study from Qatar, the odds of severe disease among 1304 individuals with reinfection was 0.12 compared with age-, sex-, and infection date-matched individuals with an initial infection [233]; there were no cases of critical illness or death among the reinfection group (compared with 28 and 7, respectively, in the initial infection group). Systematic reviews suggest that prior infection is associated with a 75 to 86 percent lower risk of severe illness or hospitalization with a subsequent infection [234,235]. Nevertheless, reinfections that are more severe than the initial infection as well as fatal reinfections can occur [236-238], and repeat infections add to the cumulative risk of severe disease or sequelae [239].

Simply having a positive SARS-CoV-2 viral test after recovery does not necessarily indicate reinfection; sequencing that demonstrates a different strain at the time of presumptive reinfection is necessary to make the distinction between reinfection and prolonged or intermittent viral RNA shedding following an initial infection. (See "COVID-19: Diagnosis", section on 'Diagnosis of reinfection' and 'Viral shedding and period of infectiousness' above.)

PREVENTION

Infection control in the health care setting — In locations where community transmission is widespread, preventive strategies for all individuals in a health care setting are warranted to reduce potential exposures. Additional measures are warranted for patients with suspected or confirmed COVID-19. Infection control in the health care setting is discussed in detail elsewhere. (See "COVID-19: Infection prevention for persons with SARS-CoV-2 infection", section on 'Infection prevention in the health care setting'.)

Personal preventive measures — In the setting of community transmission of SARS-CoV-2, the following general measures are recommended to prevent infection [240]:

Vaccination. (See "COVID-19: Vaccines", section on 'Dose and interval (for immunocompetent individuals)'.)

Ensuring adequate ventilation of indoor spaces. This includes opening windows and doors, placing fans in front of windows to exhaust air to the outside, running heating/air conditioning fans continuously, and using portable high-efficiency particulate air (HEPA) filtration systems [241,242].

Hand washing and respiratory hygiene (eg, covering the cough or sneeze). Use of hand sanitizer that contains at least 60% alcohol is a reasonable alternative to hand washing if the hands are not visibly dirty. In one study, SARS-CoV-2 remained viable on the skin for about nine hours but was completely inactivated within 15 seconds of exposure to 80% alcohol [243].

If symptoms suggestive of COVID-19 (table 2) occur, staying home away from others and getting tested for SARS-CoV-2. (See "COVID-19: Diagnosis", section on 'Diagnostic approach'.)

Avoiding close contact with individuals who have or may have COVID-19. If levels of community transmission are high, avoiding crowds and close contact with other people outside of the household is also advised to reduce the risk of exposure. (See 'Social/physical distancing' below.)

Precautions following known exposure are discussed elsewhere. (See 'Post-exposure management' below.)

Wearing masks, depending on the level of community transmission and the individual risk for severe infection. (See 'Wearing masks in the community' below.)

These preventive measures are recommended for all individuals in locations where SARS-CoV-2 is circulating and are particularly emphasized for individuals who have immunocompromising conditions and are at higher risk for severe infection.

Wearing masks in the community

When to wear a mask and what type — Local guidelines on mask-wearing depend on the level of community transmission and vaccination rates.

WHO recommendations – The World Health Organization (WHO) recommends mask-wearing as part of a comprehensive approach to reducing SARS-CoV-2 transmission in either indoor or outdoor settings where there is widespread transmission and social distancing is difficult as well as indoor settings with poor ventilation (regardless of ability to distance) [244].

WHO recommends medical or nonmedical masks (including homemade multilayered masks) for most individuals and has issued standards for the ideal composition of a cloth mask to optimize fluid resistance and filtration efficiency [245]. However, it specifically recommends medical masks for individuals with symptoms consistent with COVID-19, for individuals at risk for severe COVID-19 (eg, individuals >60 years old or with high-risk underlying conditions) when in public settings where distancing is not feasible, and for household contacts of individuals with suspected or confirmed COVID-19 when in the same room [244].

CDC recommendations – In the United States, the Centers for Disease Control and Prevention (CDC) recommendations on masking depend on the estimated COVID-19 community levels, which reflect a combined measure of local case counts, new COVID-19 hospital admissions, and the percent of staffed inpatient beds occupied by patients with COVID-19 [246]. In locations with low community levels, the CDC suggests that mask wearing be optional; at medium levels, it advises individuals who are immunocompromised or otherwise at risk for severe disease to consider masking in public and advises their close contacts to wear masks; at high levels, the CDC recommends that all individuals wear masks in indoor public settings. All masking recommendations assume that strategies to achieve and maintain high rates of vaccination, including booster doses, are ongoing. The CDC also recommends that all individuals wear masks on public transportation (including taxis and ride-shares) and at transportation hubs (eg, airports, bus or ferry terminals, railway stations, seaports) [247]. Masking is also recommended for all persons who have suspected or documented COVID-19 or exposure to SARS-CoV-2, regardless of community level. Precautions for individuals with infection or exposure are discussed in detail elsewhere. (See "COVID-19: Infection prevention for persons with SARS-CoV-2 infection", section on 'Isolation at home' and 'Testing and masking precautions' below.)

In locations or situations where masks are recommended, the CDC advises individuals to wear the mask with the highest filtration efficacy that fits well and that one can wear reliably over the mouth and nose [248]. When fit tightly around the face, respirators (eg, N95) have the highest filtration efficacy, followed by disposable medical masks. In general, cloth masks have the lowest filtration efficacy, although cloth masks made of several layers of tightly woven fabric can approach the filtration efficacy of medical masks [249,250]. The importance of filtration efficacy increases in situations in which the risk of exposure is high (eg, prolonged close contact indoors or in vehicles with people outside the household, particularly if other people are unmasked) or for individuals who are at risk for severe COVID-19. Ultimately, however, consistent and correct use is the most important aspect of mask use, as incorrect use or poor fit diminishes the value of high filtration efficacy of the material. Strategies to improve mask fit include using a mask with an adjustable nose bridge, wearing a cloth mask over a disposable mask, knotting the ear loops of a medical mask to cinch the sides of the mask and secure it against the face, using masks with ties rather than ear loops, and using a mask brace [251]. Respirators and masks should not have exhalation valves. For individuals who opt to wear a respirator, KN95 and KF94 are advertised as meeting high filtration standards in China and South Korea, respectively, and are alternatives to the N95 respirator. People should be aware, however, that many marketed KN95 and KF94 respirators do not meet the advertised filtration standards; if used, KN95 or KF94 respirators that have been independently assessed for filtration efficiency should be chosen [252]. Detailed information on the types of recommended masks can be found on the CDC website.

When advising patients on the use of masks, clinicians should counsel them to avoid touching the eyes, nose, and mouth when putting on or removing the mask, to practice hand hygiene before and after handling the mask, and to launder cloth masks routinely. Clinicians should also emphasize that the mask does not diminish the importance of other preventive measures, such as vaccination. Patients can also be counseled that masks have not been associated with impairment in gas exchange, including among patients with underlying lung disease [253,254].

Rationale — The primary objective for wearing masks in the community is to prevent transmission from individuals with infection by containing their respiratory secretions. Masks can also reduce exposure to SARS-CoV-2 for the wearer.

Source control and transmission reduction – Multiple observational studies support the use of masks to provide source control and reduce transmission in the community [249,255-266]. In epidemiologic studies, government-issued mask mandates and high rates of self-reported mask wearing have each been associated with decreased community incidence rates and, in some cases, decreased COVID-19 hospitalization rates [262,267-269]; lifting of universal mask mandates has conversely been associated with increased case rates [270]. In a meta-analysis of six observational studies, mask-wearing was associated with a 53 percent reduction in the incidence of COVID-19 [264]. Modeling studies have also suggested that high adoption of mask-wearing by the general public can reduce transmission, even if masks are only moderately effective in containing infectious respiratory secretions [271,272].

Nevertheless, efficacy of masks has been difficult to demonstrate consistently in clinical trials. In a meta-analysis of six trials that did not demonstrate reductions in laboratory-confirmed influenza or SARS-CoV-2 infection with wearing medical masks in the community (risk ratio [RR] 1.01, 95% CI 0.72 to 1.42), only two of those trials evaluated SARS-CoV-2 transmission [273]. One of those was a cluster-randomized trial in Bangladesh, in which villages that received free masks as well as behavioral and social interventions to promote masks had increased mask use (40 versus 14 percent in control villages) and, among those who received medical masks, an associated 11 percent relative reduction in SARS-CoV-2 seroprevalence that was not statistically significant (adjusted RR 0.89, 95% CI 0.78-1.01) [274]. The other trial, from Denmark, is discussed below.

Prevent exposure – Mask-wearing in the community may protect the wearer; in several observational studies, consistent mask wearing, particularly with medical masks or respirators, has been associated with a lower risk of infection [275-278]. In a report of 382 service members who were surveyed about personal preventive strategies in the setting of a SARS-CoV-2 outbreak on a United States Navy aircraft carrier, self-report of wearing a face cover was independently associated with a lower likelihood of infection (odds ratio [OR] 0.3), as were avoiding common areas (OR 0.6) and observing social distancing (OR 0.5) [275]. In a retrospective analysis of 1060 individuals identified by contact tracing following clusters of infections in Thailand, wearing a mask all the time was associated with a lower odds of infection compared with not wearing a mask; there was no significant association between wearing a mask some of the time and infection rate [276]. In contrast, a randomized trial from Denmark did not identify a decreased rate of infection among individuals who were provided with surgical masks and advised to wear them when outside of the house for a month (1.8 versus 2.1 percent among individuals who were not given masks or the recommendation) [279]. However, the low rate of community transmission (as reflected by the low overall infection rate) may have made it difficult to detect a meaningful difference.

Filtration efficacy – Filtering facepiece respirators (FFR) have the highest filtration efficacy. In the United States, the prototypical FFR is the N95 respirator, which filters at least 95 percent of 0.3 micrometer particles. Medical masks have lower filtration efficacy, which depends on how closely the mask lies against the face. In one study, medical masks with ties versus ear loops filtered 72 and 38 percent of particles, respectively (approximately 0.02 to 3.00 micrometers) [280]. Other strategies to improve the fit of a medical mask, such as using a cloth mask over it or knotting the ear loops to eliminate gaps, also appear to increase filtration efficacy [281]. Studies on the filtration efficacy of fabrics suggest that certain fabrics (eg, tea towel fabric [termed dish towel fabric in the United States], cotton-polypropylene blends), particularly when double-layered, can approach the filtration efficacy of medical masks [249,282-284]. In an experimental model, universal masking with a three-ply cotton mask was shown to substantially reduce aerosol exposure [242]. Tight-weave fabric, two or more layers, and a tight fit are essential for adequate filtration.

Despite the variability in filtration efficacy of different masks (respirators, medical masks, cloth masks) in experimental settings, data on clinical efficacy differences in preventing transmission of SARS-CoV-2 are lacking.

Limited role for other face protection — Although eye protection is recommended in health care settings, the role of face shields or goggles in addition to masks to further reduce the risk of infection in the community is uncertain [285,286]. Data are mixed on whether wearing eyeglasses daily is associated with reduced risk of infection [287,288]; eyeglasses are generally considered insufficient for eye protection. (See "COVID-19: Infection prevention for persons with SARS-CoV-2 infection", section on 'Type of PPE'.)

Social/physical distancing — In locations where there are high levels of community transmission of SARS-CoV-2, individuals are advised to practice social or physical distancing by avoiding crowded spaces or otherwise maintaining distance from other people outside their household. The optimal distance is uncertain; the WHO recommends a minimum distance of three feet (one meter). The rationale is to minimize close-range contact with an individual with infection, which is thought to be the primary risk of exposure to SARS-CoV-2. (See 'Route of person-to-person transmission' above.)

Physical distancing is likely independently associated with a reduced risk of SARS-CoV-2 transmission [259,289-291]. In a meta-analysis of observational studies evaluating the relationship between physical distance and transmission of SARS-CoV-2, SARS-CoV, and Middle East respiratory syndrome coronavirus (MERS-CoV), proximity and risk of infection were closely associated, and the infection rate was higher with contact within three feet (one meter) compared with contact beyond that distance (12.8 versus 2.6 percent) [259]. A distance more than six feet (two meters) was associated with further reduction in transmission.

Screening in selected high-risk settings — Testing asymptomatic individuals without known exposure is not routinely warranted, although the practice may be useful in high-risk congregate settings, such as long-term care facilities, shelters, and correctional facilities, when community transmission of SARS-CoV-2 is prevalent.

Serial testing in congregate settings – Screening for SARS-CoV-2 infection with serial viral testing can quickly identify cases so that infected individuals can be isolated, contacts can be quarantined, and outbreaks can be prevented [292,293]. Both nucleic acid amplification tests (NAATs) and antigen tests have been used for serial screening. Although antigen tests are generally less sensitive than NAAT, modelling studies have suggested that if the frequency of testing is high enough, tests with lower sensitivity can be successfully used to reduce cumulative infection rates [294,295]. Accessibility and fast turnaround time are also important features of a useful screening test. (See "COVID-19: Diagnosis", section on 'For other screening purposes'.)

Testing prior to group events – Rapid testing with antigen tests prior to events (and only allowing individuals who test negative to enter) has been proposed as a strategy to reduce the risk of outbreaks. This is discussed in detail elsewhere. (See "COVID-19: Diagnosis", section on 'Antigen testing'.)

Testing-based screening strategies have the advantage of identifying asymptomatic or presymptomatic infections. Several studies have highlighted the limitations of symptom-based screening methods because of the high proportion of asymptomatic cases [296,297]. (See "COVID-19: Clinical features", section on 'Asymptomatic infections'.)

Other public health measures — Throughout the world, countries have employed various nonpharmaceutical interventions to reduce transmission. In addition to personal preventive measures (vaccination, hand and respiratory hygiene, ventilation, masking), transmission reduction strategies have included:

Social/physical distancing orders

Stay-at-home orders

School, venue, and nonessential business closure

Bans on public gatherings

Travel restriction with exit and/or entry screening

Aggressive case identification and isolation (separating individuals with infection from others)

Contact tracing and quarantine (separating individuals who have been exposed from others)

These measures have been associated with reductions in the incidence of SARS-CoV-2 infection, with epidemiologic studies showing reductions in cases, and in some situations, COVID-19-related deaths following implementation of these mitigation measures [264,298-307].

Implementation of these measures varies widely by country as well as over time, depending on regional rates of infection. Specific recommendations on global travel are available on the WHO website.

Vaccines — Vaccines to prevent SARS-CoV-2 infection are considered the most promising approach for curbing the pandemic [308]. COVID-19 vaccines are discussed in detail elsewhere. (See "COVID-19: Vaccines".)

Monoclonal antibodies ineffective for pre-exposure prophylaxis — COVID-19 vaccination is the optimal method of pre-exposure prophylaxis. The monoclonal antibody combination of tixagevimab-cilgavimab had previously been an effective option for pre-exposure prophylaxis in certain individuals expected to have suboptimal protection from vaccination or unable to receive vaccination [309-312]. However, it is not expected to be effective against circulating Omicron subvariants. The US Food and Drug Administration (FDA) adjusted the emergency use authorization (EUA) for tixagevimab-cilgavimab to be contingent on the prevalence of resistant variants being less than 90 percent; thus, it is no longer authorized in the United States [313].

POST-EXPOSURE MANAGEMENT — In areas where SARS-CoV-2 is prevalent, all residents should be encouraged to stay alert for symptoms and practice appropriate preventive measures to reduce the risk of infection. (See 'Personal preventive measures' above.)

Testing and masking precautions — Throughout the pandemic, identifying secondary infection in an exposed individual and reducing the risk of that individual exposing others before an infection is recognized have been consistent goals of preventive efforts.

In the United States, the Centers for Disease Control and Prevention (CDC) suggests the following for all individuals, regardless of vaccination history, who have had close contact with a person with suspected or confirmed SARS-CoV-2 infection in the community (including during the 48 hours prior to that patient developing symptoms and regardless of whether the individuals involved were wearing masks) [314]:

Wear well-fitting masks or respirators whenever around other people indoors for 10 days following exposure (day 0 is the day of exposure). Exposed individuals should avoid places where they cannot mask and should avoid contact, if possible, with other individuals at risk for severe infection.

Test for SARS-CoV-2 at least five full days following exposure (eg, on day 6) to identify new infections promptly [315]. Because the Omicron subvariants may have a shorter incubation period than earlier variants, testing as early as four days after exposure may be helpful to identify infections sooner. Individuals with a history of SARS-CoV-2 infection in the prior 30 days do not need to test if they are asymptomatic; those with infection in the prior 30 to 90 days should undergo antigen testing rather than NAAT. (See "COVID-19: Diagnosis", section on 'Selected asymptomatic individuals' and "COVID-19: Diagnosis", section on 'Diagnosis of reinfection'.)

Monitor for fever, cough, upper respiratory symptoms, and any other symptoms consistent with COVID-19 (table 2) following the exposure. Individuals who develop such signs or symptoms should get tested for SARS-CoV-2. While awaiting results, they should stay home, continue to mask, and maintain distance from other individuals, including those in their household. (See "COVID-19: Diagnosis", section on 'Symptomatic patients'.)

Anyone who tests positive likely has SARS-CoV-2 infection and should self-isolate. (See "COVID-19: Infection prevention for persons with SARS-CoV-2 infection", section on 'Isolation at home'.)

Recommendations on post-exposure precautions are intended to balance the risk of infection over time (which is based in part on the incubation period for SARS-CoV-2) with the community burdens and adherence challenges associated with strategies to avoid ongoing exposure. As the impact of infection on individuals and health care systems has declined over the course of the pandemic along with rising rates of immunity and expanded therapeutic options, these recommendations have evolved from a 14-day post-exposure quarantine (ie, staying at home, away from others, for the duration of the incubation period) to shorter quarantine periods, to relying on masking to mitigate potential transmission. There are limited data informing the risk of transmission with these various approaches.

Management of health care workers with a documented exposure is discussed in detail elsewhere. (See "COVID-19: Infection prevention for persons with SARS-CoV-2 infection".)

No role for post-exposure prophylaxis

Monoclonal antibodies ineffective against circulating variants – In the United States, the Food and Drug Administration (FDA) had issued an emergency use authorization (EUA) to use the monoclonal antibody combinations casirivimab-imdevimab or bamlanivimab-etesevimab to prevent SARS-CoV-2 infection in select individuals over 12 years of age [316,317]. However, these combinations do not neutralize the Omicron subvariants and thus are likely ineffective for post-exposure prophylaxis in regions where these variants predominate, which is the case throughout the United States. (See 'Omicron (B.1.1.529) and its sublineages' above.)

Thus, we defer using post-exposure prophylaxis with monoclonal antibodies during the Omicron subvariant surges. If other variants emerge that are susceptible to casirivimab-imdevimab or bamlanivimab-etesevimab, post-exposure prophylaxis may be useful for individuals who are at high risk for progression (table 3) and are either unvaccinated or expected to have suboptimal immune response to vaccination (table 4).

In a trial performed before the emergence of the Omicron variant, administration of casirivimab-imdevimab to household contacts of individuals with SARS-CoV-2 infection (within 96 hours of the index case’s positive test) reduced the risk of symptomatic COVID-19 (1.5 versus 7.8 percent with placebo, adjusted OR 0.17, 95% CI 0.09-0.33) and the risk of any SARS-CoV-2 infection (4.8 versus 14.2 percent with placebo, adjusted OR 0.31, 95% CI 0.21-0.46) [318]. The efficacy of bamlanivimab-etesevimab was extrapolated from an earlier trial of nursing home residents and staff, in which bamlanivimab alone reduced the risk of subsequent COVID-19 [319]. Casirivimab-imdevimab and bamlanivimab-etesevimab are expected to retain activity against the Delta variant but, as above, not the Omicron variant.

We recommend against using other agents for post-exposure prophylaxis outside a clinical trial. Specifically, another monoclonal antibody combination, tixagevimab-cilgavimab is not authorized for post-exposure prophylaxis and, in an unpublished trial, did not result in a significant reduction in COVID-19 rates compared with placebo when given within eight days of exposure [309].

Other agents ineffective – Data from placebo-controlled randomized trials indicate that hydroxychloroquine is not effective in preventing infection [319-324]; the World Health Organization specifically recommends against using hydroxychloroquine to prevent COVID-19 [325]. Ivermectin has also been proposed as a potential prophylactic agent, but it has only been evaluated in low-quality unpublished studies [326], and clinical evidence supporting its use is lacking. Furthermore, although ivermectin has demonstrated activity against SARS-CoV-2 in vitro, plasma levels high enough for antiviral activity cannot be achieved with safe drug doses [327].

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: COVID-19 – Index of guideline topics".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topics (see "Patient education: COVID-19 overview (The Basics)" and "Patient education: COVID-19 vaccines (The Basics)" and "Patient education: COVID-19 and pregnancy (The Basics)" and "Patient education: COVID-19 and children (The Basics)" and "Patient education: Long COVID (The Basics)")

SUMMARY AND RECOMMENDATIONS

Burden of disease Since the first reports of coronavirus disease 2019 (COVID-19) and identification of the novel coronavirus that causes it, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), more than 750 million confirmed cases have been reported worldwide, although reported cases grossly underestimate the true burden of infection. (See 'Epidemiology' above.)

Variants of concern Several variants of SARS-CoV-2 have emerged that are notable because of the potential for increased transmissibility. Omicron variant sublineages are associated with a higher risk of reinfection in individuals previously infected with other variants and breakthrough infection in vaccinated individuals, but they are also associated with less severe disease. (See 'Variants of concern' above.)

Modes of transmission Direct person-to-person transmission is the primary means of SARS-CoV-2 transmission. It is thought to occur mainly through close-range contact via respiratory particles; virus released in respiratory secretions when a person with infection coughs, sneezes, or talks can infect another person if it is inhaled or makes direct contact with the mucous membranes. SARS-CoV-2 can also be transmitted over longer distances, particularly in enclosed, poorly ventilated spaces. (See 'Route of person-to-person transmission' above and 'Environmental contamination' above.)

Period of infectiousness Individuals with SARS-CoV-2 infection are most infectious in the earlier stages of infection (starting a few days prior to the development of symptoms). Transmission after 7 to 10 days of illness is unlikely, particularly for otherwise immunocompetent patients with nonsevere infection. Prolonged viral RNA shedding after symptom resolution is not clearly associated with prolonged infectiousness. (See 'Viral shedding and period of infectiousness' above.)

Personal preventive measures In settings where there is community transmission of SARS-CoV-2, personal measures to reduce the risk of transmission include vaccination, hand and respiratory hygiene, improving indoor ventilation and avoiding poorly ventilated crowded areas, being vigilant for signs and symptoms of COVID-19, and avoiding close contact with ill individuals. In the United States, recommendations on mask-wearing depend on the COVID-19 community levels. (See 'Personal preventive measures' above and 'Wearing masks in the community' above and 'Social/physical distancing' above and 'Other public health measures' above.)

Post-exposure precautions Individuals who have close contact with someone known or suspected to have COVID-19 should monitor for symptoms, wear well-fitting masks whenever around other people, and get tested for SARS-CoV-2. (See 'Post-exposure management' above.)

Vaccines COVID-19 vaccines are an essential element of prevention and are discussed in detail elsewhere. (See "COVID-19: Vaccines".)

Monoclonal antibodies not effective for pre-exposure prophylaxis – In the United States, the monoclonal antibody combination tixagevimab-cilgavimab had previously received emergency use authorization (EUA) for pre-exposure prophylaxis in individuals who are expected to have suboptimal response to vaccination; however, tixagevimab-cilgavimab is not expected to be effective against the most prevalent Omicron subvariants and thus is no longer authorized. (See 'Monoclonal antibodies ineffective for pre-exposure prophylaxis' above.)

Public health guidance Guidance has been issued by the WHO and the United States Centers for Disease Control and Prevention (CDC), as well as other expert organizations. These are updated on an ongoing basis. Links to these guidelines can be found elsewhere. (See 'Society guideline links' above.)

  1. World Health Organization. Director-General's remarks at the media briefing on 2019-nCoV on 11 February 2020. Available at: http://www.who.int/dg/speeches/detail/who-director-general-s-remarks-at-the-media-briefing-on-2019-ncov-on-11-february-2020 (Accessed on February 12, 2020).
  2. Coronaviridae Study Group of the International Committee on Taxonomy of Viruses. The species Severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2. Nat Microbiol 2020; 5:536.
  3. Zhu N, Zhang D, Wang W, et al. A Novel Coronavirus from Patients with Pneumonia in China, 2019. N Engl J Med 2020; 382:727.
  4. Lu R, Zhao X, Li J, et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet 2020; 395:565.
  5. Perlman S. Another Decade, Another Coronavirus. N Engl J Med 2020; 382:760.
  6. Zhou P, Yang XL, Wang XG, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020; 579:270.
  7. Hoffmann M, Kleine-Weber H, Schroeder S, et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020; 181:271.
  8. https://www.who.int/en/activities/tracking-SARS-CoV-2-variants/ (Accessed on June 07, 2021).
  9. The National Institute for Communicable Diseases, South Africa. Frequently asked questions for the B.1.1.529 mutated SARS-CoV-2 lineage in South Africa. https://www.nicd.ac.za/frequently-asked-questions-for-the-b-1-1-529-mutated-sars-cov-2-lineage-in-south-africa/ (Accessed on November 29, 2021).
  10. European Centre for Disease Prevention and Control. Threat Assessment Brief: Implications of the emergence and spread of the SARS-CoV-2 B.1.1. 529 variant of concern (Omicron) for the EU/EEA. https://www.ecdc.europa.eu/en/publications-data/threat-assessment-brief-emergence-sars-cov-2-variant-b.1.1.529 (Accessed on November 29, 2021).
  11. World Health Organization. Enhancing response to Omicron SARS-CoV-2 variant. https://www.who.int/publications/m/item/enhancing-readiness-for-omicron-(b.1.1.529)-technical-brief-and-priority-actions-for-member-states (Accessed on November 29, 2021).
  12. Centers for Disease Control and Prevention. New SARS-CoV-2 Variant of Concern Identified: Omicron (B.1.1.529) Variant. https://emergency.cdc.gov/han/2021/han00459.asp?ACSTrackingID=USCDC_511-DM71221&ACSTrackingLabel=HAN%20459%20-%20General%20Public&deliveryName=USCDC_511-DM71221 (Accessed on December 03, 2021).
  13. Implications of the emergence and spread of the SARS-CoV-2 variants of concern BA.4 and BA.5 for the EU/EEA. European Centre for Disease Prevention and Control. Available at: https://www.ecdc.europa.eu/en/news-events/implications-emergence-spread-sars-cov-2-variants-concern-ba4-and-ba5 (Accessed on July 04, 2022).
  14. UK Health Security Agency. Risk assessment for SARS-CoV-2 variant V-23AUG-01 (or BA.2.86). https://www.gov.uk/government/publications/investigation-of-sars-cov-2-variants-of-concern-variant-risk-assessments/risk-assessment-for-sars-cov-2-variant-v-23aug-01-or-ba286 (Accessed on August 23, 2023).
  15. Jørgensen SB, Nygård K, Kacelnik O, Telle K. Secondary Attack Rates for Omicron and Delta Variants of SARS-CoV-2 in Norwegian Households. JAMA 2022; 327:1610.
  16. Baker JM, Nakayama JY, O'Hegarty M, et al. SARS-CoV-2 B.1.1.529 (Omicron) Variant Transmission Within Households - Four U.S. Jurisdictions, November 2021-February 2022. MMWR Morb Mortal Wkly Rep 2022; 71:341.
  17. Yao C, Song W, Wang L, et al. Enhanced transmissibility of XBB.1.5 is contributed by both strong ACE2 binding and antibody evasion. UNPUBLISHED. https://www.biorxiv.org/content/10.1101/2023.01.03.522427v1.full.pdf (Accessed on January 04, 2023).
  18. Pulliam JRC, van Schalkwyk C, Govender N, et al. Increased risk of SARS-CoV-2 reinfection associated with emergence of Omicron in South Africa. Science 2022; 376:eabn4947.
  19. Altarawneh HN, Chemaitelly H, Hasan MR, et al. Protection against the Omicron Variant from Previous SARS-CoV-2 Infection. N Engl J Med 2022; 386:1288.
  20. Rössler A, Riepler L, Bante D, et al. SARS-CoV-2 Omicron Variant Neutralization in Serum from Vaccinated and Convalescent Persons. N Engl J Med 2022; 386:698.
  21. Schmidt F, Muecksch F, Weisblum Y, et al. Plasma neutralization properties of the SARS-CoV-2 Omicron variant. medRxiv 2021.
  22. Sheward DJ, Kim C, Ehling RA, et al. Neutralisation sensitivity of the SARS-CoV-2 omicron (B.1.1.529) variant: a cross-sectional study. Lancet Infect Dis 2022; 22:813.
  23. Hachmann NP, Miller J, Collier AY, et al. Neutralization Escape by SARS-CoV-2 Omicron Subvariants BA.2.12.1, BA.4, and BA.5. N Engl J Med 2022; 387:86.
  24. Cao Y, Yisimayi A, Jian F, et al. BA.2.12.1, BA.4 and BA.5 escape antibodies elicited by Omicron infection. Nature 2022; 608:593.
  25. Wang Q, Guo Y, Iketani S, et al. Antibody evasion by SARS-CoV-2 Omicron subvariants BA.2.12.1, BA.4 and BA.5. Nature 2022; 608:603.
  26. Altarawneh HN, Chemaitelly H, Ayoub HH, et al. Protective Effect of Previous SARS-CoV-2 Infection against Omicron BA.4 and BA.5 Subvariants. N Engl J Med 2022; 387:1620.
  27. Malato J, Ribeiro RM, Leite PP, et al. Risk of BA.5 Infection among Persons Exposed to Previous SARS-CoV-2 Variants. N Engl J Med 2022; 387:953.
  28. Carazo S, Skowronski DM, Brisson M, et al. Protection against omicron (B.1.1.529) BA.2 reinfection conferred by primary omicron BA.1 or pre-omicron SARS-CoV-2 infection among health-care workers with and without mRNA vaccination: a test-negative case-control study. Lancet Infect Dis 2023; 23:45.
  29. Cao Y, Wang J, Jian F, et al. Omicron escapes the majority of existing SARS-CoV-2 neutralizing antibodies. Nature 2022; 602:657.
  30. VanBlargan LA, Errico JM, Halfmann PJ, et al. An infectious SARS-CoV-2 B.1.1.529 Omicron virus escapes neutralization by therapeutic monoclonal antibodies. Nat Med 2022; 28:490.
  31. Wilhelm A, Widera M, Grikscheit K, et al. Limited neutralisation of the SARS-CoV-2 Omicron subvariants BA.1 and BA.2 by convalescent and vaccine serum and monoclonal antibodies. EBioMedicine 2022; 82:104158.
  32. Takashita E, Kinoshita N, Yamayoshi S, et al. Efficacy of Antiviral Agents against the SARS-CoV-2 Omicron Subvariant BA.2. N Engl J Med 2022; 386:1475.
  33. Takashita E, Yamayoshi S, Fukushi S, et al. Efficacy of Antiviral Agents against the Omicron Subvariant BA.2.75. N Engl J Med 2022; 387:1236.
  34. Cao Y, Jian F, Wang J, et al. Imprinted SARS-CoV-2 humoral immunity induces convergent Omicron RBD evolution. UNPUBLISHED. https://www.biorxiv.org/content/10.1101/2022.09.15.507787v3.full.pdf (Accessed on October 23, 2022).
  35. Maslo C, Friedland R, Toubkin M, et al. Characteristics and Outcomes of Hospitalized Patients in South Africa During the COVID-19 Omicron Wave Compared With Previous Waves. JAMA 2022; 327:583.
  36. Wolter N, Jassat W, Walaza S, et al. Early assessment of the clinical severity of the SARS-CoV-2 omicron variant in South Africa: a data linkage study. Lancet 2022; 399:437.
  37. Ulloa AC, Buchan SA, Daneman N, Brown KA. Estimates of SARS-CoV-2 Omicron Variant Severity in Ontario, Canada. JAMA 2022; 327:1286.
  38. Nyberg T, Ferguson NM, Nash SG, et al. Comparative analysis of the risks of hospitalisation and death associated with SARS-CoV-2 omicron (B.1.1.529) and delta (B.1.617.2) variants in England: a cohort study. Lancet 2022; 399:1303.
  39. Bouzid D, Visseaux B, Kassasseya C, et al. Comparison of Patients Infected With Delta Versus Omicron COVID-19 Variants Presenting to Paris Emergency Departments : A Retrospective Cohort Study. Ann Intern Med 2022; 175:831.
  40. Ward IL, Bermingham C, Ayoubkhani D, et al. Risk of covid-19 related deaths for SARS-CoV-2 omicron (B.1.1.529) compared with delta (B.1.617.2): retrospective cohort study. BMJ 2022; 378:e070695.
  41. Jassat W, Abdool Karim SS, Mudara C, et al. Clinical severity of COVID-19 in patients admitted to hospital during the omicron wave in South Africa: a retrospective observational study. Lancet Glob Health 2022; 10:e961.
  42. Adjei S, Hong K, Molinari NM, et al. Mortality Risk Among Patients Hospitalized Primarily for COVID-19 During the Omicron and Delta Variant Pandemic Periods - United States, April 2020-June 2022. MMWR Morb Mortal Wkly Rep 2022; 71:1182.
  43. Tegally H, Moir M, Everatt J, et al. Emergence of SARS-CoV-2 Omicron lineages BA.4 and BA.5 in South Africa. Nat Med 2022; 28:1785.
  44. UK Health Security Agency. SARS-CoV-2 variants of concern and variants under investigation in England.Technical briefing 43 https://assets.publishing.service.gov.uk/government/uploads/system/uploads/attachment_data/file/1086494/Technical-Briefing-43-28.06.22.pdf (Accessed on July 05, 2022).
  45. Davies MA, Morden E, Rosseau P, et al. Outcomes of laboratory-confirmed SARS-CoV-2 infection during resurgence driven by Omicron lineages BA.4 and BA.5 compared with previous waves in the Western Cape Province, South Africa. medRxiv 2022.
  46. Iuliano AD, Brunkard JM, Boehmer TK, et al. Trends in Disease Severity and Health Care Utilization During the Early Omicron Variant Period Compared with Previous SARS-CoV-2 High Transmission Periods - United States, December 2020-January 2022. MMWR Morb Mortal Wkly Rep 2022; 71:146.
  47. Faust JS, Du C, Liang C, et al. Excess Mortality in Massachusetts During the Delta and Omicron Waves of COVID-19. JAMA 2022; 328:74.
  48. Bentley EG, Kirby A, Sharma P, et al. SARS-CoV-2 Omicron-B.1.1.529 Variant leads to less severe disease than Pango B and Delta variants strains in a mouse model of severe COVID-19. UNPUBLISHED. https://www.biorxiv.org/content/10.1101/2021.12.26.474085v2 (Accessed on January 05, 2022).
  49. McMahan K, Giffin V, Tostanoski LH, et al. Reduced pathogenicity of the SARS-CoV-2 omicron variant in hamsters. Med 2022; 3:262.
  50. Diamond M, Halfmann P, Maemura T, et al. The SARS-CoV-2 B.1.1.529 Omicron virus causes attenuated infection and disease in mice and hamsters. Res Sq 2021.
  51. European Centre for Disease Prevention and Control. Rapid increase of a SARS-CoV-2 variant with multiple spike protein mutations observed in the United Kingdom, December 2020. https://www.ecdc.europa.eu/sites/default/files/documents/SARS-CoV-2-variant-multiple-spike-protein-mutations-United-Kingdom.pdf (Accessed on December 21, 2020).
  52. NERVTAG meetingon SARS-CoV-2 variant under investigation VUI-202012/01 https://app.box.com/s/3lkcbxepqixkg4mv640dpvvg978ixjtf/file/756963730457 (Accessed on January 08, 2021).
  53. NERVTAG/SPI-M Extraordinary meetingon SARS-CoV-2 variant of concern 202012/01 (variant B.1.1.7) https://app.box.com/s/3lkcbxepqixkg4mv640dpvvg978ixjtf/file/756964987830 (Accessed on January 08, 2021).
  54. New and Emerging Respiratory Virus Threats Advisory Group. NERVTAG meeting on SARS-CoV-2 variant under investigation VUI-202012/01. https://www.gov.uk/government/groups/new-and-emerging-respiratory-virus-threats-advisory-group#meetings (Accessed on December 21, 2020).
  55. Davies NG, Abbott S, Barnard RC, et al. Estimated transmissibility and impact of SARS-CoV-2 lineage B.1.1.7 in England. Science 2021; 372.
  56. Volz E, Mishra S, Chand M, et al. Assessing transmissibility of SARS-CoV-2 lineage B.1.1.7 in England. Nature 2021; 593:266.
  57. Public Health England. Investigation of novel SARS-CoV-2 variant: Variant of Concern 202012/01. Technical briefing 5. https://assets.publishing.service.gov.uk/government/uploads/system/uploads/attachment_data/file/957504/Variant_of_Concern_VOC_202012_01_Technical_Briefing_5_England.pdf (Accessed on February 05, 2021).
  58. Challen R, Brooks-Pollock E, Read JM, et al. Risk of mortality in patients infected with SARS-CoV-2 variant of concern 202012/1: matched cohort study. BMJ 2021; 372:n579.
  59. Davies NG, Jarvis CI, CMMID COVID-19 Working Group, et al. Increased mortality in community-tested cases of SARS-CoV-2 lineage B.1.1.7. Nature 2021; 593:270.
  60. Frampton D, Rampling T, Cross A, et al. Genomic characteristics and clinical effect of the emergent SARS-CoV-2 B.1.1.7 lineage in London, UK: a whole-genome sequencing and hospital-based cohort study. Lancet Infect Dis 2021; 21:1246.
  61. Tegally H, Wilkinson E, Giovanetti M, et al. Detection of a SARS-CoV-2 variant of concern in South Africa. Nature 2021; 592:438.
  62. Wu K, Werner AP, Moliva JI, et al. mRNA-1273 vaccine induces neutralizing antibodies against spike mutants from global SARS-CoV-2 variants. bioRxiv 2021.
  63. Greaney AJ, Loes AN, Crawford KHD, et al. Comprehensive mapping of mutations in the SARS-CoV-2 receptor-binding domain that affect recognition by polyclonal human plasma antibodies. Cell Host Microbe 2021; 29:463.
  64. Wibmer CK, Ayres F, Hermanus T, et al. SARS-CoV-2 501Y.V2 escapes neutralization by South African COVID-19 donor plasma. Nat Med 2021; 27:622.
  65. Xie X, Liu Y, Liu J, et al. Neutralization of SARS-CoV-2 spike 69/70 deletion, E484K and N501Y variants by BNT162b2 vaccine-elicited sera. Nat Med 2021; 27:620.
  66. Virological. Genomic characterisation of an emergent SARS-CoV-2 lineage in Manaus: preliminary findings. https://virological.org/t/genomic-characterisation-of-an-emergent-sars-cov-2-lineage-in-manaus-preliminary-findings/586 (Accessed on January 19, 2021).
  67. Faria NR, Mellan TA, Whittaker C, et al. Genomics and epidemiology of the P.1 SARS-CoV-2 lineage in Manaus, Brazil. Science 2021; 372:815.
  68. https://assets.publishing.service.gov.uk/government/uploads/system/uploads/attachment_data/file/991343/Variants_of_Concern_VOC_Technical_Briefing_14.pdf (Accessed on June 07, 2021).
  69. Dougherty K, Mannell M, Naqvi O, et al. SARS-CoV-2 B.1.617.2 (Delta) Variant COVID-19 Outbreak Associated with a Gymnastics Facility - Oklahoma, April-May 2021. MMWR Morb Mortal Wkly Rep 2021; 70:1004.
  70. Sheikh A, McMenamin J, Taylor B, et al. SARS-CoV-2 Delta VOC in Scotland: demographics, risk of hospital admission, and vaccine effectiveness. Lancet 2021; 397:2461.
  71. Twohig KA, Nyberg T, Zaidi A, et al. Hospital admission and emergency care attendance risk for SARS-CoV-2 delta (B.1.617.2) compared with alpha (B.1.1.7) variants of concern: a cohort study. Lancet Infect Dis 2022; 22:35.
  72. Fisman DN, Tuite AR. Evaluation of the relative virulence of novel SARS-CoV-2 variants: a retrospective cohort study in Ontario, Canada. CMAJ 2021; 193:E1619.
  73. Stringhini S, Wisniak A, Piumatti G, et al. Seroprevalence of anti-SARS-CoV-2 IgG antibodies in Geneva, Switzerland (SEROCoV-POP): a population-based study. Lancet 2020; 396:313.
  74. Centers for Disease Control and Prevention. Commercial Laboratory Seroprevalence Survey Data. https://www.cdc.gov/coronavirus/2019-ncov/cases-updates/commercial-lab-surveys.html (Accessed on July 06, 2020).
  75. Havers FP, Reed C, Lim T, et al. Seroprevalence of Antibodies to SARS-CoV-2 in 10 Sites in the United States, March 23-May 12, 2020. JAMA Intern Med 2020.
  76. Clarke KEN, Jones JM, Deng Y, et al. Seroprevalence of Infection-Induced SARS-CoV-2 Antibodies - United States, September 2021-February 2022. MMWR Morb Mortal Wkly Rep 2022; 71:606.
  77. COVID-19 Cumulative Infection Collaborators. Estimating global, regional, and national daily and cumulative infections with SARS-CoV-2 through Nov 14, 2021: a statistical analysis. Lancet 2022; 399:2351.
  78. Meyerowitz EA, Richterman A, Gandhi RT, Sax PE. Transmission of SARS-CoV-2: A Review of Viral, Host, and Environmental Factors. Ann Intern Med 2021; 174:69.
  79. Morawska L, Milton DK. It Is Time to Address Airborne Transmission of Coronavirus Disease 2019 (COVID-19). Clin Infect Dis 2020; 71:2311.
  80. World Health Organization. Transmission of SARS-CoV-2: Implications for infection prevention precautions. https://www.who.int/publications/i/item/modes-of-transmission-of-virus-causing-covid-19-implications-for-ipc-precaution-recommendations (Accessed on July 10, 2020).
  81. Klompas M, Baker MA, Rhee C. Airborne Transmission of SARS-CoV-2: Theoretical Considerations and Available Evidence. JAMA 2020.
  82. Chagla Z, Hota S, Khan S, et al. Re: It Is Time to Address Airborne Transmission of COVID-19. Clin Infect Dis 2021; 73:e3981.
  83. Duval D, Palmer JC, Tudge I, et al. Long distance airborne transmission of SARS-CoV-2: rapid systematic review. BMJ 2022; 377:e068743.
  84. Lu J, Gu J, Li K, et al. COVID-19 Outbreak Associated with Air Conditioning in Restaurant, Guangzhou, China, 2020. Emerg Infect Dis 2020; 26:1628.
  85. Hamner L, Dubbel P, Capron I, et al. High SARS-CoV-2 Attack Rate Following Exposure at a Choir Practice - Skagit County, Washington, March 2020. MMWR Morb Mortal Wkly Rep 2020; 69:606.
  86. Shen Y, Li C, Dong H, et al. Community Outbreak Investigation of SARS-CoV-2 Transmission Among Bus Riders in Eastern China. JAMA Intern Med 2020; 180:1665.
  87. Bahl P, Doolan C, de Silva C, et al. Airborne or Droplet Precautions for Health Workers Treating Coronavirus Disease 2019? J Infect Dis 2022; 225:1561.
  88. Bourouiba L. Turbulent Gas Clouds and Respiratory Pathogen Emissions: Potential Implications for Reducing Transmission of COVID-19. JAMA 2020; 323:1837.
  89. Stadnytskyi V, Bax CE, Bax A, Anfinrud P. The airborne lifetime of small speech droplets and their potential importance in SARS-CoV-2 transmission. Proc Natl Acad Sci U S A 2020; 117:11875.
  90. Ong SWX, Tan YK, Chia PY, et al. Air, Surface Environmental, and Personal Protective Equipment Contamination by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) From a Symptomatic Patient. JAMA 2020; 323:1610.
  91. Guo ZD, Wang ZY, Zhang SF, et al. Aerosol and Surface Distribution of Severe Acute Respiratory Syndrome Coronavirus 2 in Hospital Wards, Wuhan, China, 2020. Emerg Infect Dis 2020; 26:1583.
  92. Liu Y, Ning Z, Chen Y, et al. Aerodynamic analysis of SARS-CoV-2 in two Wuhan hospitals. Nature 2020; 582:557.
  93. Zhou J, Otter JA, Price JR, et al. Investigating Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Surface and Air Contamination in an Acute Healthcare Setting During the Peak of the Coronavirus Disease 2019 (COVID-19) Pandemic in London. Clin Infect Dis 2021; 73:e1870.
  94. Santarpia JL, Rivera DN, Herrera VL, et al. Aerosol and surface contamination of SARS-CoV-2 observed in quarantine and isolation care. Sci Rep 2020; 10:12732.
  95. Lednicky JA, Lauzard M, Fan ZH, et al. Viable SARS-CoV-2 in the air of a hospital room with COVID-19 patients. Int J Infect Dis 2020; 100:476.
  96. Ben-Shmuel A, Brosh-Nissimov T, Glinert I, et al. Detection and infectivity potential of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) environmental contamination in isolation units and quarantine facilities. Clin Microbiol Infect 2020; 26:1658.
  97. Birgand G, Peiffer-Smadja N, Fournier S, et al. Assessment of Air Contamination by SARS-CoV-2 in Hospital Settings. JAMA Netw Open 2020; 3:e2033232.
  98. Chen W, Lan Y, Yuan X, et al. Detectable 2019-nCoV viral RNA in blood is a strong indicator for the further clinical severity. Emerg Microbes Infect 2020; 9:469.
  99. Wang W, Xu Y, Gao R, et al. Detection of SARS-CoV-2 in Different Types of Clinical Specimens. JAMA 2020; 323:1843.
  100. Colavita F, Lapa D, Carletti F, et al. SARS-CoV-2 Isolation From Ocular Secretions of a Patient With COVID-19 in Italy With Prolonged Viral RNA Detection. Ann Intern Med 2020; 173:242.
  101. Cheung KS, Hung IFN, Chan PPY, et al. Gastrointestinal Manifestations of SARS-CoV-2 Infection and Virus Load in Fecal Samples From a Hong Kong Cohort: Systematic Review and Meta-analysis. Gastroenterology 2020; 159:81.
  102. Zheng S, Fan J, Yu F, et al. Viral load dynamics and disease severity in patients infected with SARS-CoV-2 in Zhejiang province, China, January-March 2020: retrospective cohort study. BMJ 2020; 369:m1443.
  103. Li D, Jin M, Bao P, et al. Clinical Characteristics and Results of Semen Tests Among Men With Coronavirus Disease 2019. JAMA Netw Open 2020; 3:e208292.
  104. Pham TD, Huang C, Wirz OF, et al. SARS-CoV-2 RNAemia in a Healthy Blood Donor 40 Days After Respiratory Illness Resolution. Ann Intern Med 2020; 173:853.
  105. Azzolini C, Donati S, Premi E, et al. SARS-CoV-2 on Ocular Surfaces in a Cohort of Patients With COVID-19 From the Lombardy Region, Italy. JAMA Ophthalmol 2021; 139:956.
  106. Xiao F, Sun J, Xu Y, et al. Infectious SARS-CoV-2 in Feces of Patient with Severe COVID-19. Emerg Infect Dis 2020; 26:1920.
  107. Kang M, Wei J, Yuan J, et al. Probable Evidence of Fecal Aerosol Transmission of SARS-CoV-2 in a High-Rise Building. Ann Intern Med 2020; 173:974.
  108. Yuan J, Chen Z, Gong C, et al. Sewage as a Possible Transmission Vehicle During a Coronavirus Disease 2019 Outbreak in a Densely Populated Community: Guangzhou, China, April 2020. Clin Infect Dis 2021; 73:e1487.
  109. Report of the WHO-China Joint Mission on Coronavirus DIsease 2019 (COVID-2019). February 16-24, 2020. http://www.who.int/docs/default-source/coronaviruse/who-china-joint-mission-on-covid-19-final-report.pdf (Accessed on March 04, 2020).
  110. Yu F, Yan L, Wang N, et al. Quantitative Detection and Viral Load Analysis of SARS-CoV-2 in Infected Patients. Clin Infect Dis 2020; 71:793.
  111. Xu D, Zhou F, Sun W, et al. Relationship Between Serum Severe Acute Respiratory Syndrome Coronavirus 2 Nucleic Acid and Organ Damage in Coronavirus 2019 Patients: A Cohort Study. Clin Infect Dis 2021; 73:68.
  112. AABB. AABB’s Coronavirus Resources. http://www.aabb.org/advocacy/regulatorygovernment/Pages/AABB-Coronavirus-Resources.aspx (Accessed on April 21, 2020).
  113. Zou L, Ruan F, Huang M, et al. SARS-CoV-2 Viral Load in Upper Respiratory Specimens of Infected Patients. N Engl J Med 2020; 382:1177.
  114. To KK, Tsang OT, Leung WS, et al. Temporal profiles of viral load in posterior oropharyngeal saliva samples and serum antibody responses during infection by SARS-CoV-2: an observational cohort study. Lancet Infect Dis 2020; 20:565.
  115. Wölfel R, Corman VM, Guggemos W, et al. Virological assessment of hospitalized patients with COVID-2019. Nature 2020; 581:465.
  116. He X, Lau EHY, Wu P, et al. Temporal dynamics in viral shedding and transmissibility of COVID-19. Nat Med 2020; 26:672.
  117. COVID-19 Investigation Team. Clinical and virologic characteristics of the first 12 patients with coronavirus disease 2019 (COVID-19) in the United States. Nat Med 2020; 26:861.
  118. Jones TC, Biele G, Mühlemann B, et al. Estimating infectiousness throughout SARS-CoV-2 infection course. Science 2021; 373.
  119. Ge Y, Martinez L, Sun S, et al. COVID-19 Transmission Dynamics Among Close Contacts of Index Patients With COVID-19: A Population-Based Cohort Study in Zhejiang Province, China. JAMA Intern Med 2021; 181:1343.
  120. Ke R, Martinez PP, Smith RL, et al. Daily longitudinal sampling of SARS-CoV-2 infection reveals substantial heterogeneity in infectiousness. Nat Microbiol 2022; 7:640.
  121. Killingley B, Mann AJ, Kalinova M, et al. Safety, tolerability and viral kinetics during SARS-CoV-2 human challenge in young adults. Nat Med 2022; 28:1031.
  122. Cheng HY, Jian SW, Liu DP, et al. Contact Tracing Assessment of COVID-19 Transmission Dynamics in Taiwan and Risk at Different Exposure Periods Before and After Symptom Onset. JAMA Intern Med 2020; 180:1156.
  123. Japan National Institute of Infectious Diseases Disease Control and Prevention Center, National Center for Global Health and Medicine. Active epidemiological investigation on SARS-CoV-2 infection caused by Omicron variant (Pango lineage B.1.1.529) in Japan: preliminary report on infectious period. https://www.niid.go.jp/niid/en/2019-ncov-e/10884-covid19-66-en.html (Accessed on January 24, 2022).
  124. Hay J, Kissler S, Fauver JR, et al. Viral dynamics and duration of PCR positivity of the SARS-CoV-2 Omicron variant. UNPUBLISHED. https://nrs.harvard.edu/URN-3:HUL.INSTREPOS:37370587 (Accessed on January 26, 2022).
  125. Bouton TC, Atarere J, Turcinovic J, et al. Viral Dynamics of Omicron and Delta Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Variants With Implications for Timing of Release from Isolation: A Longitudinal Cohort Study. Clin Infect Dis 2023; 76:e227.
  126. Boucau J, Marino C, Regan J, et al. Duration of Shedding of Culturable Virus in SARS-CoV-2 Omicron (BA.1) Infection. N Engl J Med 2022; 387:275.
  127. Liu Y, Yan LM, Wan L, et al. Viral dynamics in mild and severe cases of COVID-19. Lancet Infect Dis 2020; 20:656.
  128. Zhou F, Yu T, Du R, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet 2020; 395:1054.
  129. Xu K, Chen Y, Yuan J, et al. Factors Associated With Prolonged Viral RNA Shedding in Patients with Coronavirus Disease 2019 (COVID-19). Clin Infect Dis 2020; 71:799.
  130. Van Vinh Chau N, Lam VT, Dung NT, et al. The Natural History and Transmission Potential of Asymptomatic Severe Acute Respiratory Syndrome Coronavirus 2 Infection. Clin Infect Dis 2020; 71:2679.
  131. Xiao AT, Tong YX, Zhang S. Profile of RT-PCR for SARS-CoV-2: A Preliminary Study From 56 COVID-19 Patients. Clin Infect Dis 2020; 71:2249.
  132. Fontana LM, Villamagna AH, Sikka MK, McGregor JC. Understanding viral shedding of severe acute respiratory coronavirus virus 2 (SARS-CoV-2): Review of current literature. Infect Control Hosp Epidemiol 2021; 42:659.
  133. Cevik M, Tate M, Lloyd O, et al. SARS-CoV-2, SARS-CoV, and MERS-CoV viral load dynamics, duration of viral shedding, and infectiousness: a systematic review and meta-analysis. Lancet Microbe 2021; 2:e13.
  134. Bullard J, Dust K, Funk D, et al. Predicting Infectious Severe Acute Respiratory Syndrome Coronavirus 2 From Diagnostic Samples. Clin Infect Dis 2020; 71:2663.
  135. Basile K, McPhie K, Carter I, et al. Cell-based Culture Informs Infectivity and Safe De-Isolation Assessments in Patients with Coronavirus Disease 2019. Clin Infect Dis 2021; 73:e2952.
  136. Centers for Disease Control and Prevention. Symptom-Based Strategy to Discontinue Isolation for Persons with COVID-19: Decision Memo. https://www.cdc.gov/coronavirus/2019-ncov/community/strategy-discontinue-isolation.html (Accessed on May 04, 2020).
  137. Liu WD, Chang SY, Wang JT, et al. Prolonged virus shedding even after seroconversion in a patient with COVID-19. J Infect 2020; 81:318.
  138. Perera RAPM, Tso E, Tsang OTY, et al. SARS-CoV-2 Virus Culture and Subgenomic RNA for Respiratory Specimens from Patients with Mild Coronavirus Disease. Emerg Infect Dis 2020; 26:2701.
  139. Kim MC, Cui C, Shin KR, et al. Duration of Culturable SARS-CoV-2 in Hospitalized Patients with Covid-19. N Engl J Med 2021; 384:671.
  140. Avanzato VA, Matson MJ, Seifert SN, et al. Case Study: Prolonged Infectious SARS-CoV-2 Shedding from an Asymptomatic Immunocompromised Individual with Cancer. Cell 2020; 183:1901.
  141. Choi B, Choudhary MC, Regan J, et al. Persistence and Evolution of SARS-CoV-2 in an Immunocompromised Host. N Engl J Med 2020; 383:2291.
  142. Aydillo T, Gonzalez-Reiche AS, Aslam S, et al. Shedding of Viable SARS-CoV-2 after Immunosuppressive Therapy for Cancer. N Engl J Med 2020; 383:2586.
  143. Tarhini H, Recoing A, Bridier-Nahmias A, et al. Long-Term Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infectiousness Among Three Immunocompromised Patients: From Prolonged Viral Shedding to SARS-CoV-2 Superinfection. J Infect Dis 2021; 223:1522.
  144. Baang JH, Smith C, Mirabelli C, et al. Prolonged Severe Acute Respiratory Syndrome Coronavirus 2 Replication in an Immunocompromised Patient. J Infect Dis 2021; 223:23.
  145. Cevik M, Marcus JL, Buckee C, Smith TC. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Transmission Dynamics Should Inform Policy. Clin Infect Dis 2021; 73:S170.
  146. Adam DC, Wu P, Wong JY, et al. Clustering and superspreading potential of SARS-CoV-2 infections in Hong Kong. Nat Med 2020; 26:1714.
  147. Laxminarayan R, Wahl B, Dudala SR, et al. Epidemiology and transmission dynamics of COVID-19 in two Indian states. Science 2020; 370:691.
  148. Sun K, Wang W, Gao L, et al. Transmission heterogeneities, kinetics, and controllability of SARS-CoV-2. Science 2021; 371.
  149. Fung HF, Martinez L, Alarid-Escudero F, et al. The Household Secondary Attack Rate of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2): A Rapid Review. Clin Infect Dis 2021; 73:S138.
  150. Madewell ZJ, Yang Y, Longini IM Jr, et al. Household Transmission of SARS-CoV-2: A Systematic Review and Meta-analysis. JAMA Netw Open 2020; 3:e2031756.
  151. Pollán M, Pérez-Gómez B, Pastor-Barriuso R, et al. Prevalence of SARS-CoV-2 in Spain (ENE-COVID): a nationwide, population-based seroepidemiological study. Lancet 2020; 396:535.
  152. Madewell ZJ, Yang Y, Longini IM Jr, et al. Factors Associated With Household Transmission of SARS-CoV-2: An Updated Systematic Review and Meta-analysis. JAMA Netw Open 2021; 4:e2122240.
  153. Madewell ZJ, Yang Y, Longini IM Jr, et al. Household Secondary Attack Rates of SARS-CoV-2 by Variant and Vaccination Status: An Updated Systematic Review and Meta-analysis. JAMA Netw Open 2022; 5:e229317.
  154. Grijalva CG, Rolfes MA, Zhu Y, et al. Transmission of SARS-COV-2 Infections in Households - Tennessee and Wisconsin, April-September 2020. MMWR Morb Mortal Wkly Rep 2020; 69:1631.
  155. Li F, Li YY, Liu MJ, et al. Household transmission of SARS-CoV-2 and risk factors for susceptibility and infectivity in Wuhan: a retrospective observational study. Lancet Infect Dis 2021; 21:617.
  156. Chu VT, Yousaf AR, Chang K, et al. Household Transmission of SARS-CoV-2 from Children and Adolescents. N Engl J Med 2021; 385:954.
  157. Wang D, Hu B, Hu C, et al. Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. JAMA 2020; 323:1061.
  158. McMichael TM, Clark S, Pogosjans S, et al. COVID-19 in a Long-Term Care Facility - King County, Washington, February 27-March 9, 2020. MMWR Morb Mortal Wkly Rep 2020; 69:339.
  159. Kakimoto K, Kamiya H, Yamagishi T, et al. Initial Investigation of Transmission of COVID-19 Among Crew Members During Quarantine of a Cruise Ship - Yokohama, Japan, February 2020. MMWR Morb Mortal Wkly Rep 2020; 69:312.
  160. Mosites E, Parker EM, Clarke KEN, et al. Assessment of SARS-CoV-2 Infection Prevalence in Homeless Shelters - Four U.S. Cities, March 27-April 15, 2020. MMWR Morb Mortal Wkly Rep 2020; 69:521.
  161. Baggett TP, Keyes H, Sporn N, Gaeta JM. Prevalence of SARS-CoV-2 Infection in Residents of a Large Homeless Shelter in Boston. JAMA 2020; 323:2191.
  162. Wallace M, Hagan L, Curran KG, et al. COVID-19 in Correctional and Detention Facilities - United States, February-April 2020. MMWR Morb Mortal Wkly Rep 2020; 69:587.
  163. Saloner B, Parish K, Ward JA, et al. COVID-19 Cases and Deaths in Federal and State Prisons. JAMA 2020; 324:602.
  164. Wilson E, Donovan CV, Campbell M, et al. Multiple COVID-19 Clusters on a University Campus - North Carolina, August 2020. MMWR Morb Mortal Wkly Rep 2020; 69:1416.
  165. Steinberg J, Kennedy ED, Basler C, et al. COVID-19 Outbreak Among Employees at a Meat Processing Facility - South Dakota, March-April 2020. MMWR Morb Mortal Wkly Rep 2020; 69:1015.
  166. Dyal JW, Grant MP, Broadwater K, et al. COVID-19 Among Workers in Meat and Poultry Processing Facilities - 19 States, April 2020. MMWR Morb Mortal Wkly Rep 2020; 69.
  167. Ghinai I, Woods S, Ritger KA, et al. Community Transmission of SARS-CoV-2 at Two Family Gatherings - Chicago, Illinois, February-March 2020. MMWR Morb Mortal Wkly Rep 2020; 69:446.
  168. Pung R, Chiew CJ, Young BE, et al. Investigation of three clusters of COVID-19 in Singapore: implications for surveillance and response measures. Lancet 2020; 395:1039.
  169. Mahale P, Rothfuss C, Bly S, et al. Multiple COVID-19 Outbreaks Linked to a Wedding Reception in Rural Maine - August 7-September 14, 2020. MMWR Morb Mortal Wkly Rep 2020; 69:1686.
  170. Fisher KA, Tenforde MW, Feldstein LR, et al. Community and Close Contact Exposures Associated with COVID-19 Among Symptomatic Adults ≥18 Years in 11 Outpatient Health Care Facilities - United States, July 2020. MMWR Morb Mortal Wkly Rep 2020; 69:1258.
  171. Tenforde MW, Fisher KA, Patel MM. Identifying COVID-19 Risk Through Observational Studies to Inform Control Measures. JAMA 2021; 325:1464.
  172. Chang S, Pierson E, Koh PW, et al. Mobility network models of COVID-19 explain inequities and inform reopening. Nature 2021; 589:82.
  173. Marks M, Millat-Martinez P, Ouchi D, et al. Transmission of COVID-19 in 282 clusters in Catalonia, Spain: a cohort study. Lancet Infect Dis 2021; 21:629.
  174. Zhou J, Singanayagam A, Goonawardane N, et al. Viral emissions into the air and environment after SARS-CoV-2 human challenge: a phase 1, open label, first-in-human study. Lancet Microbe 2023; 4:e579.
  175. Bi Q, Wu Y, Mei S, et al. Epidemiology and transmission of COVID-19 in 391 cases and 1286 of their close contacts in Shenzhen, China: a retrospective cohort study. Lancet Infect Dis 2020; 20:911.
  176. Khanh NC, Thai PQ, Quach HL, et al. Transmission of SARS-CoV 2 During Long-Haul Flight. Emerg Infect Dis 2020; 26:2617.
  177. Hu M, Lin H, Wang J, et al. Risk of Coronavirus Disease 2019 Transmission in Train Passengers: an Epidemiological and Modeling Study. Clin Infect Dis 2021; 72:604.
  178. Hu M, Wang J, Lin H, et al. Risk of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Transmission Among Air Passengers in China. Clin Infect Dis 2022; 75:e234.
  179. Bulfone TC, Malekinejad M, Rutherford GW, Razani N. Outdoor Transmission of SARS-CoV-2 and Other Respiratory Viruses: A Systematic Review. J Infect Dis 2021; 223:550.
  180. Tenforde MW, Billig Rose E, Lindsell CJ, et al. Characteristics of Adult Outpatients and Inpatients with COVID-19 - 11 Academic Medical Centers, United States, March-May 2020. MMWR Morb Mortal Wkly Rep 2020; 69:841.
  181. Rothe C, Schunk M, Sothmann P, et al. Transmission of 2019-nCoV Infection from an Asymptomatic Contact in Germany. N Engl J Med 2020; 382:970.
  182. Yu P, Zhu J, Zhang Z, Han Y. A Familial Cluster of Infection Associated With the 2019 Novel Coronavirus Indicating Possible Person-to-Person Transmission During the Incubation Period. J Infect Dis 2020; 221:1757.
  183. Bai Y, Yao L, Wei T, et al. Presumed Asymptomatic Carrier Transmission of COVID-19. JAMA 2020; 323:1406.
  184. Hu Z, Song C, Xu C, et al. Clinical characteristics of 24 asymptomatic infections with COVID-19 screened among close contacts in Nanjing, China. Sci China Life Sci 2020; 63:706.
  185. Qian G, Yang N, Ma AHY, et al. COVID-19 Transmission Within a Family Cluster by Presymptomatic Carriers in China. Clin Infect Dis 2020; 71:861.
  186. Böhmer MM, Buchholz U, Corman VM, et al. Investigation of a COVID-19 outbreak in Germany resulting from a single travel-associated primary case: a case series. Lancet Infect Dis 2020; 20:920.
  187. Wang Y, He Y, Tong J, et al. Characterization of an Asymptomatic Cohort of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infected Individuals Outside of Wuhan, China. Clin Infect Dis 2020; 71:2132.
  188. Arons MM, Hatfield KM, Reddy SC, et al. Presymptomatic SARS-CoV-2 Infections and Transmission in a Skilled Nursing Facility. N Engl J Med 2020; 382:2081.
  189. Lee S, Kim T, Lee E, et al. Clinical Course and Molecular Viral Shedding Among Asymptomatic and Symptomatic Patients With SARS-CoV-2 Infection in a Community Treatment Center in the Republic of Korea. JAMA Intern Med 2020; 180:1447.
  190. Wei WE, Li Z, Chiew CJ, et al. Presymptomatic Transmission of SARS-CoV-2 - Singapore, January 23-March 16, 2020. MMWR Morb Mortal Wkly Rep 2020; 69:411.
  191. Buitrago-Garcia D, Egli-Gany D, Counotte MJ, et al. Occurrence and transmission potential of asymptomatic and presymptomatic SARS-CoV-2 infections: A living systematic review and meta-analysis. PLoS Med 2020; 17:e1003346.
  192. Plucinski MM, Wallace M, Uehara A, et al. Coronavirus Disease 2019 (COVID-19) in Americans Aboard the Diamond Princess Cruise Ship. Clin Infect Dis 2021; 72:e448.
  193. Luo L, Liu D, Liao X, et al. Contact Settings and Risk for Transmission in 3410 Close Contacts of Patients With COVID-19 in Guangzhou, China : A Prospective Cohort Study. Ann Intern Med 2020; 173:879.
  194. Sayampanathan AA, Heng CS, Pin PH, et al. Infectivity of asymptomatic versus symptomatic COVID-19. Lancet 2021; 397:93.
  195. Johansson MA, Quandelacy TM, Kada S, et al. SARS-CoV-2 Transmission From People Without COVID-19 Symptoms. JAMA Netw Open 2021; 4:e2035057.
  196. Yung CF, Kam KQ, Wong MSY, et al. Environment and Personal Protective Equipment Tests for SARS-CoV-2 in the Isolation Room of an Infant With Infection. Ann Intern Med 2020; 173:240.
  197. Yamagishi T, Ohnishi M, Matsunaga N, et al. Environmental Sampling for Severe Acute Respiratory Syndrome Coronavirus 2 During a COVID-19 Outbreak on the Diamond Princess Cruise Ship. J Infect Dis 2020; 222:1098.
  198. Kampf G, Todt D, Pfaender S, Steinmann E. Persistence of coronaviruses on inanimate surfaces and their inactivation with biocidal agents. J Hosp Infect 2020; 104:246.
  199. Rabenau HF, Cinatl J, Morgenstern B, et al. Stability and inactivation of SARS coronavirus. Med Microbiol Immunol 2005; 194:1.
  200. van Doremalen N, Bushmaker T, Morris DH, et al. Aerosol and Surface Stability of SARS-CoV-2 as Compared with SARS-CoV-1. N Engl J Med 2020; 382:1564.
  201. Ratnesar-Shumate S, Williams G, Green B, et al. Simulated Sunlight Rapidly Inactivates SARS-CoV-2 on Surfaces. J Infect Dis 2020; 222:214.
  202. Otter JA, Donskey C, Yezli S, et al. Transmission of SARS and MERS coronaviruses and influenza virus in healthcare settings: the possible role of dry surface contamination. J Hosp Infect 2016; 92:235.
  203. World Organization for Animal Health. Questions and Answers on the 2019 Coronavirus Disease (COVID-19), section on Surveillance and events in animals. https://www.oie.int/en/scientific-expertise/specific-information-and-recommendations/questions-and-answers-on-2019novel-coronavirus/ (Accessed on April 13, 2020).
  204. Sit THC, Brackman CJ, Ip SM, et al. Infection of dogs with SARS-CoV-2. Nature 2020; 586:776.
  205. Newman A, Smith D, Ghai RR, et al. First Reported Cases of SARS-CoV-2 Infection in Companion Animals - New York, March-April 2020. MMWR Morb Mortal Wkly Rep 2020; 69:710.
  206. McAloose D, Laverack M, Wang L, et al. From People to Panthera: Natural SARS-CoV-2 Infection in Tigers and Lions at the Bronx Zoo. mBio 2020; 11.
  207. Halfmann PJ, Hatta M, Chiba S, et al. Transmission of SARS-CoV-2 in Domestic Cats. N Engl J Med 2020; 383:592.
  208. Shi J, Wen Z, Zhong G, et al. Susceptibility of ferrets, cats, dogs, and other domesticated animals to SARS-coronavirus 2. Science 2020; 368:1016.
  209. Oude Munnink BB, Sikkema RS, Nieuwenhuijse DF, et al. Transmission of SARS-CoV-2 on mink farms between humans and mink and back to humans. Science 2021; 371:172.
  210. European Ministry of Africulture, Nature and Food Quality. SARS-CoV-2 infections of mink in the Netherlands. https://ec.europa.eu/food/sites/food/files/animals/docs/reg-com_ahw_20200618_covid_mink_nld.pdf (Accessed on July 24, 2020).
  211. SARS-CoV-2 mink-associated variant strain – Denmark. https://www.who.int/csr/don/06-november-2020-mink-associated-sars-cov2-denmark/en/ (Accessed on November 10, 2020).
  212. Yen HL, Sit THC, Brackman CJ, et al. Transmission of SARS-CoV-2 delta variant (AY.127) from pet hamsters to humans, leading to onward human-to-human transmission: a case study. Lancet 2022; 399:1070.
  213. Rijkers G, Murk JL, Wintermans B, et al. Differences in Antibody Kinetics and Functionality Between Severe and Mild Severe Acute Respiratory Syndrome Coronavirus 2 Infections. J Infect Dis 2020; 222:1265.
  214. Lynch KL, Whitman JD, Lacanienta NP, et al. Magnitude and kinetics of anti-SARS-CoV-2 antibody responses and their relationship to disease severity. Clin Infect Dis 2020.
  215. Dan JM, Mateus J, Kato Y, et al. Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection. Science 2021; 371.
  216. Crawford KHD, Dingens AS, Eguia R, et al. Dynamics of Neutralizing Antibody Titers in the Months After Severe Acute Respiratory Syndrome Coronavirus 2 Infection. J Infect Dis 2021; 223:197.
  217. Wang Z, Muecksch F, Schaefer-Babajew D, et al. Naturally enhanced neutralizing breadth against SARS-CoV-2 one year after infection. Nature 2021; 595:426.
  218. Turner JS, Kim W, Kalaidina E, et al. SARS-CoV-2 infection induces long-lived bone marrow plasma cells in humans. Nature 2021; 595:421.
  219. Yao L, Wang GL, Shen Y, et al. Persistence of Antibody and Cellular Immune Responses in Coronavirus Disease 2019 Patients Over Nine Months After Infection. J Infect Dis 2021; 224:586.
  220. Wajnberg A, Amanat F, Firpo A, et al. Robust neutralizing antibodies to SARS-CoV-2 infection persist for months. Science 2020; 370:1227.
  221. Holmer HK, Mackey K, Fiordalisi CV, Helfand M. Major Update 2: Antibody Response and Risk for Reinfection After SARS-CoV-2 Infection-Final Update of a Living, Rapid Review. Ann Intern Med 2023; 176:85.
  222. Rodda LB, Netland J, Shehata L, et al. Functional SARS-CoV-2-Specific Immune Memory Persists after Mild COVID-19. Cell 2021; 184:169.
  223. Khoury DS, Cromer D, Reynaldi A, et al. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med 2021; 27:1205.
  224. Lumley SF, O'Donnell D, Stoesser NE, et al. Antibody Status and Incidence of SARS-CoV-2 Infection in Health Care Workers. N Engl J Med 2021; 384:533.
  225. Harvey RA, Rassen JA, Kabelac CA, et al. Association of SARS-CoV-2 Seropositive Antibody Test With Risk of Future Infection. JAMA Intern Med 2021; 181:672.
  226. Hall VJ, Foulkes S, Charlett A, et al. SARS-CoV-2 infection rates of antibody-positive compared with antibody-negative health-care workers in England: a large, multicentre, prospective cohort study (SIREN). Lancet 2021; 397:1459.
  227. Letizia AG, Ge Y, Vangeti S, et al. SARS-CoV-2 seropositivity and subsequent infection risk in healthy young adults: a prospective cohort study. Lancet Respir Med 2021; 9:712.
  228. Zhu FC, Li YH, Guan XH, et al. Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: a dose-escalation, open-label, non-randomised, first-in-human trial. Lancet 2020; 395:1845.
  229. Grifoni A, Weiskopf D, Ramirez SI, et al. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 2020; 181:1489.
  230. COVID-19 Forecasting Team. Past SARS-CoV-2 infection protection against re-infection: a systematic review and meta-analysis. Lancet 2023; 401:833.
  231. Chemaitelly H, Tang P, Coyle P, et al. Protection against Reinfection with the Omicron BA.2.75 Subvariant. N Engl J Med 2023; 388:665.
  232. Ma KC, Dorabawila V, León TM, et al. Trends in Laboratory-Confirmed SARS-CoV-2 Reinfections and Associated Hospitalizations and Deaths Among Adults Aged ≥18 Years - 18 U.S. Jurisdictions, September 2021-December 2022. MMWR Morb Mortal Wkly Rep 2023; 72:683.
  233. Abu-Raddad LJ, Chemaitelly H, Bertollini R, National Study Group for COVID-19 Epidemiology. Severity of SARS-CoV-2 Reinfections as Compared with Primary Infections. N Engl J Med 2021; 385:2487.
  234. Bobrovitz N, Ware H, Ma X, et al. Protective effectiveness of previous SARS-CoV-2 infection and hybrid immunity against the omicron variant and severe disease: a systematic review and meta-regression. Lancet Infect Dis 2023; 23:556.
  235. Deng J, Ma Y, Liu Q, et al. Severity and Outcomes of SARS-CoV-2 Reinfection Compared with Primary Infection: A Systematic Review and Meta-Analysis. Int J Environ Res Public Health 2023; 20.
  236. Mulder M, van der Vegt DSJM, Oude Munnink BB, et al. Reinfection of Severe Acute Respiratory Syndrome Coronavirus 2 in an Immunocompromised Patient: A Case Report. Clin Infect Dis 2021; 73:e2841.
  237. BNO News. COVID-19 reinfection tracker. https://bnonews.com/index.php/2020/08/covid-19-reinfection-tracker/ (Accessed on March 31, 2021).
  238. Lawandi A, Warner S, Sun J, et al. Suspected Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-COV-2) Reinfections: Incidence, Predictors, and Healthcare Use Among Patients at 238 US Healthcare Facilities, 1 June 2020 to 28 February 2021. Clin Infect Dis 2022; 74:1489.
  239. Bowe B, Xie Y, Al-Aly Z. Acute and postacute sequelae associated with SARS-CoV-2 reinfection. Nat Med 2022; 28:2398.
  240. CDC. How to Protect Yourself and Others. https://www.cdc.gov/coronavirus/2019-ncov/prevent-getting-sick/prevention.html (Accessed on August 12, 2022).
  241. Centers for Disease Control and Prevention. Ventilation in Buildings. https://www.cdc.gov/coronavirus/2019-ncov/community/ventilation.html (Accessed on December 17, 2020).
  242. Lindsley WG, Derk RC, Coyle JP, et al. Efficacy of Portable Air Cleaners and Masking for Reducing Indoor Exposure to Simulated Exhaled SARS-CoV-2 Aerosols - United States, 2021. MMWR Morb Mortal Wkly Rep 2021; 70:972.
  243. Hirose R, Ikegaya H, Naito Y, et al. Survival of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and Influenza Virus on Human Skin: Importance of Hand Hygiene in Coronavirus Disease 2019 (COVID-19). Clin Infect Dis 2021; 73:e4329.
  244. World Health Organization. COVID-19 infection prevention and control living guideline: mask use in community settings, 22 December 2021. https://www.who.int/publications/i/item/WHO-2019-nCoV-IPC_masks-2021.1 (Accessed on January 25, 2022).
  245. World Health Organization. Advice on the use of masks in the context of COVID-19. https://www.who.int/publications/i/item/advice-on-the-use-of-masks-in-the-community-during-home-care-and-in-healthcare-settings-in-the-context-of-the-novel-coronavirus-(2019-ncov)-outbreak (Accessed on June 08, 2020).
  246. Centers for Disease Control and Prevention. COVID-19 Community Levels. https://www.cdc.gov/coronavirus/2019-ncov/science/community-levels.html#anchor_47145 (Accessed on February 28, 2022).
  247. Centers for Disease Control and Prevention. Federal Register Notice: Wearing of face masks while on conveyances and at transportation hubs. https://www.cdc.gov/quarantine/masks/mask-travel-guidance.html (Accessed on February 05, 2021).
  248. CDC. Types of masks and respirators. https://www.cdc.gov/coronavirus/2019-ncov/prevent-getting-sick/types-of-masks.html (Accessed on January 25, 2022).
  249. Clase CM, Fu EL, Joseph M, et al. Cloth Masks May Prevent Transmission of COVID-19: An Evidence-Based, Risk-Based Approach. Ann Intern Med 2020; 173:489.
  250. Bahl P, Bhattacharjee S, de Silva C, et al. Face coverings and mask to minimise droplet dispersion and aerosolisation: a video case study. Thorax 2020; 75:1024.
  251. Centers for Disease Control and Prevention. Improve the Fit and Filtration of Your Mask to Reduce the Spread of COVID-19. https://www.cdc.gov/coronavirus/2019-ncov/prevent-getting-sick/mask-fit-and-filtration.html (Accessed on February 11, 2021).
  252. Centers for Disease Control and Prevention. NPPTL Respirator Assessments to Support the COVID-19 Response. https://www.cdc.gov/niosh/npptl/respirators/testing/NonNIOSHresults.html (Accessed on January 11, 2022).
  253. Samannan R, Holt G, Calderon-Candelario R, et al. Effect of Face Masks on Gas Exchange in Healthy Persons and Patients with Chronic Obstructive Pulmonary Disease. Ann Am Thorac Soc 2021; 18:541.
  254. Chan NC, Li K, Hirsh J. Peripheral Oxygen Saturation in Older Persons Wearing Nonmedical Face Masks in Community Settings. JAMA 2020; 324:2323.
  255. Wang Y, Tian H, Zhang L, et al. Reduction of secondary transmission of SARS-CoV-2 in households by face mask use, disinfection and social distancing: a cohort study in Beijing, China. BMJ Glob Health 2020; 5.
  256. Leung NHL, Chu DKW, Shiu EYC, et al. Respiratory virus shedding in exhaled breath and efficacy of face masks. Nat Med 2020; 26:676.
  257. Chan JF, Yuan S, Zhang AJ, et al. Surgical Mask Partition Reduces the Risk of Noncontact Transmission in a Golden Syrian Hamster Model for Coronavirus Disease 2019 (COVID-19). Clin Infect Dis 2020; 71:2139.
  258. Liang M, Gao L, Cheng C, et al. Efficacy of face mask in preventing respiratory virus transmission: A systematic review and meta-analysis. Travel Med Infect Dis 2020; 36:101751.
  259. Chu DK, Akl EA, Duda S, et al. Physical distancing, face masks, and eye protection to prevent person-to-person transmission of SARS-CoV-2 and COVID-19: a systematic review and meta-analysis. Lancet 2020; 395:1973.
  260. Wang X, Ferro EG, Zhou G, et al. Association Between Universal Masking in a Health Care System and SARS-CoV-2 Positivity Among Health Care Workers. JAMA 2020; 324:703.
  261. Czypionka T, Greenhalgh T, Bassler D, Bryant MB. Masks and Face Coverings for the Lay Public : A Narrative Update. Ann Intern Med 2021; 174:511.
  262. Rader B, White LF, Burns MR, et al. Mask-wearing and control of SARS-CoV-2 transmission in the USA: a cross-sectional study. Lancet Digit Health 2021; 3:e148.
  263. Hendrix MJ, Walde C, Findley K, Trotman R. Absence of Apparent Transmission of SARS-CoV-2 from Two Stylists After Exposure at a Hair Salon with a Universal Face Covering Policy - Springfield, Missouri, May 2020. MMWR Morb Mortal Wkly Rep 2020; 69:930.
  264. Talic S, Shah S, Wild H, et al. Effectiveness of public health measures in reducing the incidence of covid-19, SARS-CoV-2 transmission, and covid-19 mortality: systematic review and meta-analysis. BMJ 2021; 375:e068302.
  265. Donovan CV, Rose C, Lewis KN, et al. SARS-CoV-2 Incidence in K-12 School Districts with Mask-Required Versus Mask-Optional Policies - Arkansas, August-October 2021. MMWR Morb Mortal Wkly Rep 2022; 71:384.
  266. Chou R, Dana T, Jungbauer R. Update Alert 8: Masks for Prevention of Respiratory Virus Infections, Including SARS-CoV-2, in Health Care and Community Settings. Ann Intern Med 2022; 175:W108.
  267. Van Dyke ME, Rogers TM, Pevzner E, et al. Trends in County-Level COVID-19 Incidence in Counties With and Without a Mask Mandate - Kansas, June 1-August 23, 2020. MMWR Morb Mortal Wkly Rep 2020; 69:1777.
  268. Joo H, Miller GF, Sunshine G, et al. Decline in COVID-19 Hospitalization Growth Rates Associated with Statewide Mask Mandates - 10 States, March-October 2020. MMWR Morb Mortal Wkly Rep 2021; 70:212.
  269. Guy GP Jr, Lee FC, Sunshine G, et al. Association of State-Issued Mask Mandates and Allowing On-Premises Restaurant Dining with County-Level COVID-19 Case and Death Growth Rates - United States, March 1-December 31, 2020. MMWR Morb Mortal Wkly Rep 2021; 70:350.
  270. Cowger TL, Murray EJ, Clarke J, et al. Lifting Universal Masking in Schools - Covid-19 Incidence among Students and Staff. N Engl J Med 2022; 387:1935.
  271. Stutt ROJH, Retkute R, Bradley M, et al. A modelling framework to assess the likely effectiveness of facemasks in combination with 'lock-down' in managing the COVID-19 pandemic. Proc Math Phys Eng Sci 2020; 476:20200376.
  272. Ngonghala CN, Iboi E, Eikenberry S, et al. Mathematical assessment of the impact of non-pharmaceutical interventions on curtailing the 2019 novel Coronavirus. Math Biosci 2020; 325:108364.
  273. Jefferson T, Dooley L, Ferroni E, et al. Physical interventions to interrupt or reduce the spread of respiratory viruses. Cochrane Database Syst Rev 2023; 1:CD006207.
  274. Abaluck J, Kwong LH, Styczynski A, et al. Impact of community masking on COVID-19: A cluster-randomized trial in Bangladesh. Science 2022; 375:eabi9069.
  275. Payne DC, Smith-Jeffcoat SE, Nowak G, et al. SARS-CoV-2 Infections and Serologic Responses from a Sample of U.S. Navy Service Members - USS Theodore Roosevelt, April 2020. MMWR Morb Mortal Wkly Rep 2020; 69:714.
  276. Doung-Ngern P, Suphanchaimat R, Panjangampatthana A, et al. Case-Control Study of Use of Personal Protective Measures and Risk for SARS-CoV 2 Infection, Thailand. Emerg Infect Dis 2020; 26:2607.
  277. Freedman DO, Wilder-Smith A. In-flight transmission of SARS-CoV-2: a review of the attack rates and available data on the efficacy of face masks. J Travel Med 2020; 27.
  278. Andrejko KL, Pry JM, Myers JF, et al. Effectiveness of Face Mask or Respirator Use in Indoor Public Settings for Prevention of SARS-CoV-2 Infection - California, February-December 2021. MMWR Morb Mortal Wkly Rep 2022; 71:212.
  279. Bundgaard H, Bundgaard JS, Raaschou-Pedersen DET, et al. Effectiveness of Adding a Mask Recommendation to Other Public Health Measures to Prevent SARS-CoV-2 Infection in Danish Mask Wearers : A Randomized Controlled Trial. Ann Intern Med 2021; 174:335.
  280. Sickbert-Bennett EE, Samet JM, Clapp PW, et al. Filtration Efficiency of Hospital Face Mask Alternatives Available for Use During the COVID-19 Pandemic. JAMA Intern Med 2020; 180:1607.
  281. Brooks JT, Beezhold DH, Noti JD, et al. Maximizing Fit for Cloth and Medical Procedure Masks to Improve Performance and Reduce SARS-CoV-2 Transmission and Exposure, 2021. MMWR Morb Mortal Wkly Rep 2021; 70:254.
  282. Fischer EP, Fischer MC, Grass D, et al. Low-cost measurement of face mask efficacy for filtering expelled droplets during speech. Sci Adv 2020; 6.
  283. Clase CM, Fu EL, Ashur A, et al. Forgotten Technology in the COVID-19 Pandemic: Filtration Properties of Cloth and Cloth Masks-A Narrative Review. Mayo Clin Proc 2020; 95:2204.
  284. Clapp PW, Sickbert-Bennett EE, Samet JM, et al. Evaluation of Cloth Masks and Modified Procedure Masks as Personal Protective Equipment for the Public During the COVID-19 Pandemic. JAMA Intern Med 2021; 181:463.
  285. Marra AR, Edmond MB, Popescu SV, Perencevich EN. Examining the need for eye protection for coronavirus disease 2019 (COVID-19) prevention in the community. Infect Control Hosp Epidemiol 2021; 42:646.
  286. Perencevich EN, Diekema DJ, Edmond MB. Moving Personal Protective Equipment Into the Community: Face Shields and Containment of COVID-19. JAMA 2020; 323:2252.
  287. Zeng W, Wang X, Li J, et al. Association of Daily Wear of Eyeglasses With Susceptibility to Coronavirus Disease 2019 Infection. JAMA Ophthalmol 2020; 138:1196.
  288. Fretheim A, Elgersma IH, Helleve A, et al. Effect of Wearing Glasses on Risk of Infection With SARS-CoV-2 in the Community: A Randomized Clinical Trial. JAMA Netw Open 2022; 5:e2244495.
  289. Islam N, Sharp SJ, Chowell G, et al. Physical distancing interventions and incidence of coronavirus disease 2019: natural experiment in 149 countries. BMJ 2020; 370:m2743.
  290. Rubin D, Huang J, Fisher BT, et al. Association of Social Distancing, Population Density, and Temperature With the Instantaneous Reproduction Number of SARS-CoV-2 in Counties Across the United States. JAMA Netw Open 2020; 3:e2016099.
  291. Tsai AC, Harling G, Reynolds Z, et al. Coronavirus Disease 2019 (COVID-19) Transmission in the United States Before Versus After Relaxation of Statewide Social Distancing Measures. Clin Infect Dis 2021; 73:S120.
  292. World Health Organization. Preventing and managing COVID-19 across long-term care services: Web annex. 2020. Available at: https://www.who.int/publications/i/item/WHO-2019-nCoV-Policy_Brief-Long-term_Care-web-annex-2020.1 (Accessed on September 02, 2020).
  293. Denny TN, Andrews L, Bonsignori M, et al. Implementation of a Pooled Surveillance Testing Program for Asymptomatic SARS-CoV-2 Infections on a College Campus - Duke University, Durham, North Carolina, August 2-October 11, 2020. MMWR Morb Mortal Wkly Rep 2020; 69:1743.
  294. Paltiel AD, Zheng A, Walensky RP. Assessment of SARS-CoV-2 Screening Strategies to Permit the Safe Reopening of College Campuses in the United States. JAMA Netw Open 2020; 3:e2016818.
  295. Larremore DB, Wilder B, Lester E, et al. Test sensitivity is secondary to frequency and turnaround time for COVID-19 screening. Sci Adv 2021; 7.
  296. Dollard P, Griffin I, Berro A, et al. Risk Assessment and Management of COVID-19 Among Travelers Arriving at Designated U.S. Airports, January 17-September 13, 2020. MMWR Morb Mortal Wkly Rep 2020; 69:1681.
  297. Ng OT, Marimuthu K, Koh V, et al. SARS-CoV-2 seroprevalence and transmission risk factors among high-risk close contacts: a retrospective cohort study. Lancet Infect Dis 2021; 21:333.
  298. Pan A, Liu L, Wang C, et al. Association of Public Health Interventions With the Epidemiology of the COVID-19 Outbreak in Wuhan, China. JAMA 2020; 323:1915.
  299. Tian H, Liu Y, Li Y, et al. An investigation of transmission control measures during the first 50 days of the COVID-19 epidemic in China. Science 2020; 368:638.
  300. Lyu W, Wehby GL. Comparison of Estimated Rates of Coronavirus Disease 2019 (COVID-19) in Border Counties in Iowa Without a Stay-at-Home Order and Border Counties in Illinois With a Stay-at-Home Order. JAMA Netw Open 2020; 3:e2011102.
  301. Jüni P, Rothenbühler M, Bobos P, et al. Impact of climate and public health interventions on the COVID-19 pandemic: a prospective cohort study. CMAJ 2020; 192:E566.
  302. Sen S, Karaca-Mandic P, Georgiou A. Association of Stay-at-Home Orders With COVID-19 Hospitalizations in 4 States. JAMA 2020; 323:2522.
  303. Flaxman S, Mishra S, Gandy A, et al. Estimating the effects of non-pharmaceutical interventions on COVID-19 in Europe. Nature 2020; 584:257.
  304. Hsiang S, Allen D, Annan-Phan S, et al. The effect of large-scale anti-contagion policies on the COVID-19 pandemic. Nature 2020; 584:262.
  305. Marriott D, Beresford R, Mirdad F, et al. Concomitant Marked Decline in Prevalence of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and Other Respiratory Viruses Among Symptomatic Patients Following Public Health Interventions in Australia: Data from St Vincent's Hospital and Associated Screening Clinics, Sydney, NSW. Clin Infect Dis 2021; 72:e649.
  306. Fuller JA, Hakim A, Victory KR, et al. Mitigation Policies and COVID-19-Associated Mortality - 37 European Countries, January 23-June 30, 2020. MMWR Morb Mortal Wkly Rep 2021; 70:58.
  307. Ahlers M, Aralis H, Tang W, et al. Non-pharmaceutical interventions and covid-19 burden in the United States: retrospective, observational cohort study. BMJ Med 2022; 1:e000030.
  308. World Health Organization. https://www.who.int/publications-detail/an-international-randomised-trial-of-candidate-vaccines-against-covid-19 (Accessed on April 22, 2020).
  309. FACT SHEET FOR HEALTHCARE PROVIDERS: EMERGENCY USE AUTHORIZATION FOR EVUSHELD(tixagevimab co-packaged with cilgavimab). https://www.fda.gov/media/154701/download (Accessed on January 04, 2023).
  310. Levin MJ, Ustianowski A, De Wit S, et al. Intramuscular AZD7442 (Tixagevimab-Cilgavimab) for Prevention of Covid-19. N Engl J Med 2022; 386:2188.
  311. Al Jurdi A, Morena L, Cote M, et al. Tixagevimab/cilgavimab pre-exposure prophylaxis is associated with lower breakthrough infection risk in vaccinated solid organ transplant recipients during the omicron wave. Am J Transplant 2022; 22:3130.
  312. Kertes J, Shapiro Ben David S, Engel-Zohar N, et al. Association Between AZD7442 (Tixagevimab-Cilgavimab) Administration and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection, Hospitalization, and Mortality. Clin Infect Dis 2023; 76:e126.
  313. EVUSHELD Emergency Use Authorization (EUA) Update. AstraZeneca. Available at: https://www.fda.gov/media/164833/download (Accessed on January 26, 2023).
  314. CDC. What to Do If You Were Exposed to COVID-19. https://www.cdc.gov/coronavirus/2019-ncov/your-health/if-you-were-exposed.html (Accessed on August 12, 2022).
  315. Massetti GM, Jackson BR, Brooks JT, et al. Summary of Guidance for Minimizing the Impact of COVID-19 on Individual Persons, Communities, and Health Care Systems - United States, August 2022. MMWR Morb Mortal Wkly Rep 2022; 71:1057.
  316. FACT SHEET FOR HEALTH CARE PROVIDERS. EMERGENCY USE AUTHORIZATION (EUA) OF REGEN-COVTM (casirivimab and imdevimab) https://www.fda.gov/media/145611/download (Accessed on August 03, 2021).
  317. FACT SHEET FOR HEALTH CARE PROVIDERS EMERGENCY USE AUTHORIZATION (EUA) OF BAMLANIVIMAB AND ETESEVIMAB https://www.fda.gov/media/145802/download (Accessed on October 01, 2021).
  318. O'Brien MP, Forleo-Neto E, Musser BJ, et al. Subcutaneous REGEN-COV Antibody Combination to Prevent Covid-19. N Engl J Med 2021; 385:1184.
  319. Abella BS, Jolkovsky EL, Biney BT, et al. Efficacy and Safety of Hydroxychloroquine vs Placebo for Pre-exposure SARS-CoV-2 Prophylaxis Among Health Care Workers: A Randomized Clinical Trial. JAMA Intern Med 2021; 181:195.
  320. Boulware DR, Pullen MF, Bangdiwala AS, et al. A Randomized Trial of Hydroxychloroquine as Postexposure Prophylaxis for Covid-19. N Engl J Med 2020; 383:517.
  321. Rajasingham R, Bangdiwala AS, Nicol MR, et al. Hydroxychloroquine as Pre-exposure Prophylaxis for Coronavirus Disease 2019 (COVID-19) in Healthcare Workers: A Randomized Trial. Clin Infect Dis 2021; 72:e835.
  322. Mitjà O, Corbacho-Monné M, Ubals M, et al. A Cluster-Randomized Trial of Hydroxychloroquine for Prevention of Covid-19. N Engl J Med 2021; 384:417.
  323. Barnabas RV, Brown ER, Bershteyn A, et al. Hydroxychloroquine as Postexposure Prophylaxis to Prevent Severe Acute Respiratory Syndrome Coronavirus 2 Infection : A Randomized Trial. Ann Intern Med 2021; 174:344.
  324. Bartoszko JJ, Siemieniuk RAC, Kum E, et al. Prophylaxis against covid-19: living systematic review and network meta-analysis. BMJ 2021; 373:n949.
  325. Lamontagne F, Stegemann M, Agarwal A, et al. A living WHO guideline on drugs to prevent covid-19. BMJ 2021; 372:n526.
  326. Shouman W. Prophylactic Ivermectin in COVID-19 Contacts. Zagazig University. https://clinicaltrials.gov/ct2/show/study/NCT04422561 (Accessed on December 17, 2020).
  327. Heidary F, Gharebaghi R. Ivermectin: a systematic review from antiviral effects to COVID-19 complementary regimen. J Antibiot (Tokyo) 2020; 73:593.
Topic 126981 Version 207.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟