Skip to main content
Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America logoLink to Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America
. 2021 Oct 15;75(1):e1195–e1201. doi: 10.1093/cid/ciab903

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Dose, Infection, and Disease Outcomes for Coronavirus Disease 2019 (COVID-19): A Review

Lisa M Brosseau 1,#,, Kevin Escandón 2,3,#, Angela K Ulrich 4,5, Angela L Rasmussen 6,7, Chad J Roy 8,9, Gregory J Bix 10,11,12, Saskia V Popescu 13,14, Kristine A Moore 15, Michael T Osterholm 16,17
PMCID: PMC8524637  PMID: 34651164

Abstract

The relationship between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) dose, infection, and coronavirus disease 2019 (COVID-19) outcomes remains poorly understood. This review summarizes the existing literature regarding this issue, identifies gaps in current knowledge, and suggests opportunities for future research. In humans, host characteristics, including age, sex, comorbidities, smoking, and pregnancy, are associated with severe COVID-19. Similarly, in animals, host factors are strong determinants of disease severity, although most animal infection models manifest clinically with mild to moderate respiratory disease. The influence of variants of concern as it relates to infectious dose, consequence of overall pathogenicity, and disease outcome in dose–response remains unknown. Epidemiologic data suggest a dose–response relationship for infection contrasting with limited and inconsistent surrogate-based evidence between dose and disease severity. Recommendations include the design of future infection studies in animal models to investigate inoculating dose on outcomes and the use of better proxies for dose in human epidemiology studies.

Keywords: SARS-CoV-2, COVID-19, infectious dose, disease severity, inoculum


This article reviews evidence on the relationship between SARS-CoV-2 dose, infection, and COVID-19 outcomes; identifies gaps in understanding; and suggests future research opportunities. While data suggest a dose–infection relationship, limited, inconsistent surrogate-based evidence exists for a dose–severity relationship.


The infection process and subsequent disease outcomes of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are complex and multifactorial. Recent studies in animals and humans address the impact of different exposure routes, the effect of dose on infection and disease outcomes, and the potential for greater transmissibility and more severe disease from emerging viral variants. However, critical gaps remain in elucidating the relationships between exposure, dose, infection, and severity of disease. We review the existing literature regarding SARS-CoV-2 dose, infection, and disease outcomes; identify knowledge gaps; and suggest opportunities for future research. We focus on evidence for a relationship between infectious dose and coronavirus disease 2019 (COVID-19) severity.

METHODS

Original studies, narrative and systematic reviews, and meta-analyses in peer-reviewed journals were identified using PubMed/MEDLINE. Keywords included “infection,” “SARS-CoV-2,” “COVID-19,” “animal,” “human,” “severity,” “comorbidity,” “infectious dose,” “dose,” “inoculum,” and “exposure.” Where available, earlier singular studies were excluded in favor of later reviews. We collected and synthesized existing studies on SARS-CoV-2 infectious dose and COVID-19 severity in animal and human studies. Preprints were considered in the absence of peer-reviewed evidence. Theoretical modeling studies were not considered.

SARS-CoV-2 INFECTIOUS DOSE AND VIRAL LOAD

Viral transmission and infection are complex, probabilistic processes. The major mode of SARS-CoV-2 transmission is inhalation of respiratory particles containing infectious virions [1]; contact transmission occurs less frequently [2]. Viral viability in respiratory particles is influenced by host physiology and biology and environmental conditions [3]. Exposure is a function of viable virus concentration and contact time (Figure 1A).

Figure 1.

Figure 1.

SARS-CoV-2 transmission, exposure, and infection. A, Viral transmission and infection are complex, probabilistic processes. Concentration of infectious respiratory particles, exposure duration, and environmental, viral, and host conditions are critical for an infectious dose leading to SARS-CoV-2 infection. B, Once an individual is infected with SARS-CoV-2, shedding of virus RNA and viable virus ensues. Viral transmission relies heavily on the viral kinetics around symptom onset. The detection of SARS-CoV-2 RNA exceeds the detection of culturable or replication-competent virus. Abbreviations: qRT-PCR, quantitative reverse transcriptase–polymerase chain reaction; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; TCID50, median tissue culture infectious dose.

SARS-CoV-2 infection is mediated primarily through the angiotensin-converting enzyme 2 (ACE2) receptor, found in a variety of tissues including the respiratory tract [4]. An “infectious dose” or inoculum, expressed as minimal infectious dose (the smallest quantity that leads to infection) or median infectious dose (ID50; the dose causing infection in 50% of those exposed), represents the amount of virus received by an uninfected person resulting in cell invasion, active viral replication, and production of infectious virus as well as shedding of detectable viral RNA (Figure 1B) [5]. Infectious dose is a function of ongoing virus viability, particle size and concentration, and breathing rate. Once inhaled and deposited in the respiratory tract, productive infection depends on the dose overcoming multiple factors, including mucus, ACE2 receptor distribution and expression, and innate antiviral immunity in target tissues [6, 7].

Viral RNA load, or “viral load,” which refers to the amount of SARS-CoV-2 nucleic acid in a sample, is measured by quantitative reverse transcriptase–polymerase chain reaction (qRT-PCR) and expressed as either cycle threshold (Ct) or RNA copies per volume. Viral load does not quantify infectious virus, which requires culture-based methods, nor does it correspond to infectious dose. Infectious virus capable of initiating infection, replicating, and producing progeny virus is expressed as plaque-forming units (PFU) or median tissue culture infectious dose (TCID50). There is some evidence linking higher viral loads with increased infectiousness [8–10], but the association is not linear. Furthermore, diagnostic assays typically do not discriminate between genomic and subgenomic RNA, which are only produced during productive viral replication. Thus, qRT-PCR metrics alone cannot be used to quantify or infer infectious virus or dose. There is some evidence of an association between viral load and COVID-19 severity [11–13], but this association can be highly variable depending on viral kinetics during infection course, timing of qRT-PCR testing, and presence of symptoms [14].

The incubation period for SARS-CoV-2 (from receipt of an infectious dose to symptom onset) is 2–14 days (median: 4–6 days) [15]. Peak shedding of viable virus leading to infectiousness occurs 1–3 days before symptom onset to 5 days after [5, 16]. Infectious virus is usually not shed beyond 8–10 days after symptom onset; viral RNA can be detected in clinical samples for days, weeks, or even months [17, 18]. Asymptomatic individuals can shed virus and may therefore have a role in transmission, but this is less understood as compared with symptomatic individuals [19]. Throughout SARS-CoV-2 infection, pathophysiological phenomena are complex and variable, resulting in a broad spectrum of symptoms and varying clinical course and outcomes.

COVID-19 SEVERITY

COVID-19 severity has been classified as asymptomatic, mild, moderate, severe, and critical [20, 21]. Proxies used to assess severity include mortality, intensive care unit (ICU) admission, hospitalization, and use of mechanical ventilation.

Several host factors are associated with disease outcomes. Older age (≥65 years), male sex, certain comorbidities (hypertension, diabetes mellitus, chronic obstructive pulmonary disease, obesity, cardiovascular disease, cerebrovascular disease, chronic kidney disease), immunocompromised status (eg, cancer), and smoking are associated with more severe morbidity, ICU admission, invasive mechanical ventilation, disease progression, and increased mortality [22–27]. Pregnancy is associated with an increased likelihood of hospitalization, ICU admission, and mechanical ventilation, but not mortality [28, 29]. In children, severe COVID-19 cases are less frequent than in adults but are not negligible [30]. Multisystem inflammatory syndrome in children and adults is a rare, life-threatening condition, for which risk factors remain largely unknown [31, 32]. Neither lack of comorbidities nor younger age guarantee mild or favorable disease outcomes, suggesting that unknown host factors may be significant determinants of COVID-19 severity. Evidence on post-acute sequelae of COVID-19 continues to accrue [33, 34].

One critical aspect in assessing severity is understanding that clinical course evolves over time, warranting the differentiation of presymptomatic from asymptomatic individuals, the latter estimated to represent 16–25% of SARS-CoV-2 infections [35–40].

SARS-CoV-2 VARIANTS

Certain viral characteristics are associated with the likelihood of infection and disease severity. Ongoing transmission of SARS-CoV-2 has led to the emergence of variants of concern (VOCs) with the potential for increased transmissibility, host immune evasion, or more severe outcomes [41, 42]. Human studies of the SARS-CoV-2 D614G mutant, globally dominant in 2020, consistently found no association with clinical outcomes compared with wild-type SARS-CoV-2 [43, 44]. More recently, 4 major circulating variants (Alpha, Beta, Gamma, Delta) have emerged in various locations and spread to other countries. Epidemiological data suggest that some of these VOCs may result in worse clinical outcomes.

Alpha variant (lineage B.1.1.7) has been associated with greater risk of hospitalization (40–63%) and death (28–71%) compared with non-VOCs [45–49]. One hospital-based study found no association between the Alpha variant and severe and fatal COVID-19 [50]. Studies have found that Gamma (lineage P.1) infections are more likely to result in hospitalization or death compared with non-Gamma infections [51, 52]. The Delta variant (lineage B.1.617.2) has emerged as a significant driver of waves of infection worldwide, and initial observations suggest that it may be associated with an increased risk of hospitalization as compared with other variants [53, 54]. A hamster study has shown that Delta is more pathogenic than a D614G-bearing B.1.1 isolate, independent of dose [55].

DATA FROM ANIMAL STUDIES

No single wild-type animal model replicates the full range of human disease from asymptomatic or mild symptoms to severe or fatal health outcomes. While viral RNA has been found in the lung, brain, liver, kidney, spleen, and gastrointestinal tract in nonhuman primates (NHPs), hamsters, and ferrets, there are no studies in any animal model that fully investigate the long-term effects of infection [56].

Nonhuman primates have emerged as reasonable higher-order species for COVID-19 pathogenesis, vaccine, and therapeutic studies. The COVID-19 rhesus macaque model uses a large mucosal dose, resulting in mild upper respiratory infection that resolves in 14 to 21 days [57–63], and models the milder form of the disease in humans. Hamsters and transgenic/transformed human ACE2 (hACE2) mice, in contrast, are the predominant rodent models in use [64–69] and progress to severe pathologies from experimental mucosal infection. Ferrets, which develop mild symptoms only in the upper respiratory system, have limited utility [70–72].

Most COVID-19 animal infection models require high doses—104 to 106 TCID50 or PFU—delivered to the upper mucosa to produce clinical disease. There are no reports in the literature of attempts to reliably determine a minimum infectious dose in the macaque COVID-19 model using any NHP species. Rodent studies (hamsters) show a correlation between viral load and dose; the ID50 for hamsters has been determined at approximately 5 TCID50 [73].

Route of exposure is a key factor in animal models of COVID-19 and can influence severity, kinetics, and sites of infection. Early studies in NHPs showed preferential infection when rhesus macaques received 106 TCID50 by conjunctival and intratracheal but not intragastric routes [74]. Although the conjunctival route resulted in infection, disease was milder compared with the intratracheal route. This contrasts with other NHP studies that did not find severity differences attributable to the route of exposure [62, 75]. Route of exposure influences clinical outcomes in rodent models as well. Aerosol exposure in hamsters accelerated and worsened clinical signs even if dosed at more than 1 log lower than mucosal exposure (1.5 × 103 vs 8 × 104 TCID50) [76]. More lung inflammation and disease symptoms were observed in hamsters receiving 105 PFU by intranasal than oral inoculation; oral and fecal shedding were similar by both routes [77]. This may be related more to viral fitness than infectious dose, as the Alpha variant outcompeted an earlier A-lineage variant in hamsters exposed to either virus by the aerosol route [78].

Although no transmission studies have been reported in NHPs, rodents (hamsters) and ferrets have been shown to successfully transmit to naive animals by contact, fomites, and aerosol routes [70, 79–82]. Quantitative estimation of dose from infected to naive animals has not been done and is impossible to accomplish; thus, SARS-CoV-2 transmission studies rely on observation as a qualitative measure of dose.

Severe disease outcomes related to age, sex, and comorbidities have been replicated in NHPs [62, 75, 83], hamsters [84, 85], and some transgenic mice models [86–88]. The latter must be carefully interpreted, because transgenic hACE2 species may replicate infection or disease in a different manner than humans. Young (4–6 weeks) transgenic mice expressing hACE2 with a cytokeratin 18 promoter (K18-hACE2 mice) showed greater and more rapid weight loss in the high-dose (103 TCID50) versus the low-dose (104 TCID50) group and uniform lethality in the high-dose group. All mice shed SARS-CoV-2 and developed pulmonary pathology following SARS-CoV-2 inoculation, with limited dose-dependent differences [89]. Another study with K18-hACE2 mice that assessed doses at 103, 104, and 105 PFU showed 0, 50%, and 100% lethality, respectively, suggesting a median lethal dose (LD50) of 104 PFU [90].

Some studies in animals with inducible comorbidities (ie, diabetes, obesity, or compromised immunity) replicate serious health outcomes in young animals, as seen in younger humans with similar conditions. For example, among cyclophosphamide-treated hamsters (representing immunosuppression), those exposed intranasally to 102, 103, and 104 PFU showed significant weight loss compared with mock-challenged hamsters; viral shedding continued until cyclophosphamide treatment ended, after which all animals recovered. An intranasal dose of 104 PFU was lethal in all exposed recombination activating gene 2 (RAG2) knockout hamsters, which are unable to produce functional T or B cells [91].

DATA FROM EPIDEMIOLOGIC STUDIES

Observational epidemiologic studies can provide insight into the relationships between dose and infection and dose and disease outcomes, but many have methodologic limitations. For example, SARS-CoV-2 transmission is reported more frequently in indoor spaces, particularly when poorly ventilated or crowded, compared with outdoor settings [92, 93]. This suggests that infection is less likely in settings with lower virus concentrations, but, in most cases, no quantitative assessment of dose is reported. Instead, proxies have been used, such as the route and duration of exposure, proximity to an infectious source, number of contacts with infected sources, source infectiousness measured by infectious virus concentration, use of respiratory protection, and environment in which the exposure occurs. Proxies can have important limitations, however; for example, many cannot be directly or continuously observed in real time and rely instead on self-report of past behavior.

Studies of healthcare workers (HCWs) provide an opportunity to assess such proxies, given the risk of occupational exposure and the ability to control for confounding factors. While the strength of evidence for the likelihood of SARS-CoV-2 infection among HCWs and the dose received (often measured by directness or intensity of contact) trends toward a positive association, most studies to date are considered to be of low to moderate certainty due to limitations in methodology, such as recall bias, low participant numbers and participation rates, collinearity, and failure to control for confounding variables [94–103].

An ongoing review of SARS-CoV-2 infection rates and risk factors in HCWs consistently found a wide range of SARS-CoV-2 infection incidence (0.4–50%) and seropositivity prevalence (2–32%), thought to be due to differences among studies in locations, exposures, community infection rates, and control measures, among others [94–99]. Risk factors for higher rates of infection include being a nurse or working in hospital non-emergency wards [104], lack of personal protective equipment or adequate handwashing, direct patient contact or care for patients with COVID-19, and presence during intubation [94]. Each of these risk factors support an association between exposure and dose and infection. Race or ethnicity (Black, Asian, ethnic minority, Hispanic) has also been reported as a risk factor for HCW infections [101–103, 105], which may be due to job or community exposures.

Rates of hospitalization and severe disease range from 0–14% and 1–10%, respectively [100]. Mortality rates in HCWs are less than 1% [100]. Seroprevalence in HCWs ranges from 4% in Asia to 13–18% in North America [105, 106]. There is a paucity of evidence to suggest that dose is associated with disease severity among HCWs. Studies among US HCWs have shown that they may have less severe illness despite higher risk of unprotected or repeated exposure, with rates of severe disease significantly lower in HCWs (10%) than in all COVID-19–positive patients (29%); the same is true for mortality rates (0.3% vs 2.3%) [106]. Rather, age, a well-known, host-related risk factor, was associated with higher rates in HCWs over 50 years, with the highest rates in those over 70 years [107].

While infection and seropositivity rates suggest that HCWs are at greater risk than non-HCWs, these wide ranges illustrate the difficulty in using such measures to understand the nature of risk in the absence of information about community rates and exposures at and away from work. Most HCWs are young and healthy, which probably accounts for their low rates of severe disease and mortality. Higher fatality rates in HCWs over 50 years is consistent with disease outcomes in the general population, suggesting that dose is not associated with disease severity.

DISCUSSION

In this review, we sought to understand the relationships of dose to infection and disease severity by examining evidence from relevant animal models, clinical studies, and epidemiologic data. We found that there is some evidence of a relationship between dose and infection based on animal studies and human epidemiology but minimal data supporting a relationship between dose and disease severity. Instead, host responses and potentially viral genotype primarily determine disease outcomes.

Animal studies are the method by which the relationships between dose, infection, and disease severity will be further elucidated. Existing human clinical studies do not, in general, include or reveal information about the level of exposure or dose received. If the low median infectious dose of 5 TCID50 found in 1 hamster study is relevant to humans [73], it would suggest that human dose–response will be difficult to detect in nonexperimental settings.

Epidemiology data suggest an ill-defined dose–response relationship between SARS-CoV-2 and infection. Challenges include limited control for confounding factors, the potential for recall bias in retrospective studies, the potential for selection bias, low participation rates, inconsistencies across studies, and imprecise estimates of exposure and dose [103]. While human challenge studies and randomized controlled trials would provide the strongest evidence, researchers cannot ethically inoculate or randomize people to different SARS-CoV-2 doses. To quantify dose more fully, we recommend that future epidemiology studies assess and address as many of the proxies for dose as possible in reporting results. Disease severity should also be assessed objectively and consistently while recognizing that hospitalization rates and ICU admission are indicative of healthcare capacity, rather than disease pathogenesis. Future research must account for variables such as simultaneous interventions, changes in testing criteria over time, treatment improvements, and host risk factors for severity.

The complex interplay of host and virus is a much stronger determinant of disease severity than simply the size of the dose. Based on available evidence, once infection takes place, disease outcomes are a function of biological and physiologic response and defense mechanisms, which are host-dependent, and environmental factors, such as access to medical care and healthcare system capacity. Most animal models for SARS-CoV-2 replicate only mild to moderate respiratory disease outcomes; a few show more severe outcomes in aged animals. Studies in small-animal models suggest that host factors are a strong determinant of disease severity. This is also supported by species-specific differential COVID-19 severity across different animal models experimentally infected with the same dose via the same route [108–113]. While disease outcomes in humans range from asymptomatic to mild to severe, including death, the most compelling factors associated with disease severity are certain host factors, such as age, sex, smoking, pregnancy, and some comorbidities. It may be that less severe health outcomes are associated with lower doses, but there are few data to support that hypothesis in the human clinical studies conducted to date.

Few data from clinical or epidemiologic studies conducted to date support the hypothesis that lower dose is associated with less severe health outcomes, being limited by inferring viral dose from inappropriate surrogates, ecological approaches that cannot control for potentially relevant confounding factors, and failure to longitudinally follow subjects for misclassification of asymptomatic infections. Epidemiologic studies could be more robust by the use of better measurement tools, such as genotyping and environmental sampling, and through greater efforts to control for confounding and selection and information biases.

Thus, we conclude that, while there is an association between SARS-CoV-2 dose and infection, data do not support a relationship between dose and COVID-19 severity. Nonpharmaceutical interventions may limit the inoculum dose from an exposure, thereby reducing the risk of infection, but they are unlikely to individually have an impact on COVID-19 severity [114, 115].

Notes

Author contributions. All authors had full access to all the data in the study and take responsibility for the integrity and the accuracy of the data analysis. Concept and design: L. M. B., K. E., and A. K. U. Acquisition, analysis, or interpretation of data: all authors. Drafting of the manuscript: L. M. B., A. K. U., K. E., A. L. R., G. J. B., S. V. P., and C. J. R. Critical revision of the manuscript for important intellectual content and administrative, technical, or material support: all authors. Supervision: L. M. B., A. K. U., and M. T. O.

Potential conflicts of interest. The authors: No reported conflicts of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

Contributor Information

Lisa M Brosseau, Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA.

Kevin Escandón, School of Medicine, Universidad del Valle, Cali, Colombia; Grupo de Investigación en Virus Emergentes y Enfermedad (VIREM), Department of Microbiology, Universidad del Valle, Cali, Colombia.

Angela K Ulrich, Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA; Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA.

Angela L Rasmussen, Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; Georgetown Center for Global Health Science and Security, Washington, D.C., USA.

Chad J Roy, Tulane National Primate Research Center, Division of Microbiology, Covington, Louisiana, USA; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA.

Gregory J Bix, Clinical Neuroscience Research Center, Departments of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Tulane Brain Institute, Tulane University, New Orleans, Louisiana, USA; School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USAand.

Saskia V Popescu, Georgetown Center for Global Health Science and Security, Washington, D.C., USA; Biodefense Program, Schar School of Policy and Government, George Mason University, Arlington, Virginia, USA.

Kristine A Moore, Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA.

Michael T Osterholm, Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA; Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA.

References

  • 1. Tang S, Mao Y, Jones RM, et al. Aerosol transmission of SARS-CoV-2? Evidence, prevention and control. Environ Int 2020; 144:106039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Centers for Disease Control and Prevention. Coronavirus disease 2019 (COVID-19). 2020. Available at: https://www.cdc.gov/coronavirus/2019-ncov/science/science-briefs/sars-cov-2-transmission.html. Accessed 16 May 2021. [Google Scholar]
  • 3. Leung NHL. Transmissibility and transmission of respiratory viruses. Nat Rev Microbiol 2021; 19:528–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. 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; 181:271–80, e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. 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:S170–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Lim YX, Ng YL, Tam JP, Liu DX.. Human coronaviruses: a review of virus–host interactions. Diseases 2016; 4:26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Romagnoli S, Peris A, De Gaudio AR, Geppetti P.. SARS-CoV-2 and COVID-19: from the bench to the bedside. Physiol Rev 2020; 100:1455–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. 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; 21:629–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Karan A, Klompas M, Tucker R, et al. The risk of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) transmission from patients with undiagnosed coronavirus disease 2019 (COVID-19) to roommates in a large academic medical center. Clin Infect Dis 2021. Available at: 10.1093/cid/ciab564. Accessed 5 October 2021. [DOI] [PubMed] [Google Scholar]
  • 10. Lee LYW, Rozmanowski S, Pang M, et al. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infectivity by viral load, S gene variants and demographic factors, and the utility of lateral flow devices to prevent transmission. Clin Infect Dis 2021. Available at: 10.1093/cid/ciab421. Accessed 5 October 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Pujadas E, Chaudhry F, McBride R, et al. SARS-CoV-2 viral load predicts COVID-19 mortality. Lancet Respir Med 2020; 8:e70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Westblade LF, Brar G, Pinheiro LC, et al. SARS-CoV-2 viral load predicts mortality in patients with and without cancer who are hospitalized with COVID-19. Cancer Cell 2020; 38:661–71, e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Trunfio M, Venuti F, Alladio F, et al. Diagnostic SARS-CoV-2 cycle threshold value predicts disease severity, survival, and six-month sequelae in COVID-19 symptomatic patients. Viruses 2021; 13:281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Escandón K, Rasmussen AL, Bogoch II, et al. COVID-19 false dichotomies and a comprehensive review of the evidence regarding public health, COVID-19 symptomatology, SARS-CoV-2 transmission, mask wearing, and reinfection. BMC Infect Dis 2021; 21:710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Alene M, Yismaw L, Assemie MA, Ketema DB, Gietaneh W, Birhan TY.. Serial interval and incubation period of COVID-19: a systematic review and meta-analysis. BMC Infect Dis 2021; 21:257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. He X, Lau EHY, Wu P, et al. Temporal dynamics in viral shedding and transmissibility of COVID-19. Nat Med 2020; 26:672–5. [DOI] [PubMed] [Google Scholar]
  • 17. Cevik M, Tate M, Lloyd O, Maraolo AE, Schafers J, Ho A.. 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:e13–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Walsh KA, Spillane S, Comber L, et al. The duration of infectiousness of individuals infected with SARS-CoV-2. J Infect 2020; 81:847–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Gao Z, Xu Y, Sun C, et al. A systematic review of asymptomatic infections with COVID-19. J Microbiol Immunol Infect 2021; 54:12–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. World Health Organization. COVID-19 clinical management: living guidance, 25 January 2021. Geneva, Switzerland: World Health Organization, 2021. Available at: https://apps.who.int/iris/handle/10665/338882. [Google Scholar]
  • 21. Marshall JC, Murthy S, Diaz J, et al. A minimal common outcome measure set for COVID-19 clinical research. Lancet Infect Dis 2020; 20:e192–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Zheng Z, Peng F, Xu B, et al. Risk factors of critical & mortal COVID-19 cases: a systematic literature review and meta-analysis. J Infect 2020; 81:e16–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Fang X, Li S, Yu H, et al. Epidemiological, comorbidity factors with severity and prognosis of COVID-19: a systematic review and meta-analysis. Aging (Albany NY) 2020; 12:12493–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Yang J, Zheng Y, Gou X, et al. Prevalence of comorbidities and its effects in patients infected with SARS-CoV-2: a systematic review and meta-analysis. Int J Infect Dis 2020; 94:91–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Ssentongo P, Ssentongo AE, Heilbrunn ES, Ba DM, Chinchilli VM.. Association of cardiovascular disease and 10 other pre-existing comorbidities with COVID-19 mortality: a systematic review and meta-analysis. PLoS One 2020; 15:e0238215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Földi M, Farkas N, Kiss S, et al. ; KETLAK Study Group. . Obesity is a risk factor for developing critical condition in COVID-19 patients: a systematic review and meta-analysis. Obes Rev 2020; 21:e13095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Gazzaz ZJ. Diabetes and COVID-19. Open Life Sci 2021; 16:297–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Ellington S. Characteristics of women of reproductive age with laboratory-confirmed SARS-CoV-2 infection by pregnancy status—United States, January 22–June 7, 2020. MMWR Morb Mortal Wkly Rep 2020; 769–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Allotey J, Stallings E, Bonet M, et al. Clinical manifestations, risk factors, and maternal and perinatal outcomes of coronavirus disease 2019 in pregnancy: living systematic review and meta-analysis. BMJ 2020; 370:m3320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Kim L. Hospitalization rates and characteristics of children aged 18 years hospitalized with laboratory-confirmed COVID-19—COVID-NET, 14 states, March 1–July 25, 2020. MMWR Morb Mortal Wkly Rep 2020: 69:1081–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Morris SB, Schwartz NG, Patel P, et al. Case series of multisystem inflammatory syndrome in adults associated with SARS-CoV-2 infection—United Kingdom and United States, March–August 2020. MMWR Morb Mortal Wkly Rep 2020; 69:1450–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Nakra NA, Blumberg DA, Herrera-Guerra A, Lakshminrusimha S.. Multi-system inflammatory syndrome in children (MIS-C) following SARS-CoV-2 infection: review of clinical presentation, hypothetical pathogenesis, and proposed management. Children (Basel) 2020; 7:69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Cabrera Martimbianco AL, Pacheco RL, Bagattini ÂM, Riera R.. Frequency, signs and symptoms, and criteria adopted for long COVID-19: a systematic review. Int J Clin Pract 2021; 75:e14357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Sudre CH, Murray B, Varsavsky T, et al. Attributes and predictors of long COVID. Nat Med 2021; 27:626–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. He J, Guo Y, Mao R, Zhang J.. Proportion of asymptomatic coronavirus disease 2019: a systematic review and meta-analysis. J Med Virol 2021; 93:820–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Byambasuren O, Cardona M, Bell K, Clark J, McLaws M-L, Glasziou P.. Estimating the extent of asymptomatic COVID-19 and its potential for community transmission: systematic review and meta-analysis. J Assoc Med Microbiol Infect Dis Can 2020; 5:223–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Beale S, Hayward A, Shallcross L, Aldridge RW, Fragaszy E.. A rapid review and meta-analysis of the asymptomatic proportion of PCR-confirmed SARS-CoV-2 infections in community settings. Wellcome Open Res 2020; 5:266. [Google Scholar]
  • 39. Kronbichler A, Kresse D, Yoon S, Lee KH, Effenberger M, Shin JI.. Asymptomatic patients as a source of COVID-19 infections: a systematic review and meta-analysis. Int J Infect Dis 2020; 98:180–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Alene M, Yismaw L, Assemie MA, et al. Magnitude of asymptomatic COVID-19 cases throughout the course of infection: a systematic review and meta-analysis. PLoS One 2021; 16:e0249090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Lauring AS, Hodcroft EB.. Genetic variants of SARS-CoV-2—what do they mean? JAMA 2021; 325:529–31. [DOI] [PubMed] [Google Scholar]
  • 42. Walensky RP, Walke HT, Fauci AS.. SARS-CoV-2 variants of concern in the United States—challenges and opportunities. JAMA 2021; 325:1037–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Korber B, Fischer WM, Gnanakaran S, et al. Tracking changes in SARS-CoV-2 spike: evidence that D614G increases infectivity of the COVID-19 virus. Cell 2020; 182:812–27, e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Volz E, Hill V, McCrone JT, et al. Evaluating the effects of SARS-CoV-2 spike mutation D614G on transmissibility and pathogenicity. Cell 2021; 184:64–75, e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Iacobucci G. Covid-19: new UK variant may be linked to increased death rate, early data indicate. BMJ 2021; 372:n230. [DOI] [PubMed] [Google Scholar]
  • 46. Davies NG, Jarvis CI, Edmunds WJ, Jewell NP, Diaz-Ordaz K, Keogh RH; CMMID COVID-19 Working Group. . Increased mortality in community-tested cases of SARS-CoV-2 lineage B.1.1.7. Nature 2021; 593:270–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Challen R, Brooks-Pollock E, Read JM, Dyson L, Tsaneva-Atanasova K, Danon L.. Risk of mortality in patients infected with SARS-CoV-2 variant of concern 202012/1: matched cohort study. BMJ 2021; 372:n579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Grint DJ, Wing K, Williamson E, et al. Case fatality risk of the SARS-CoV-2 variant of concern B.1.1.7 in England, 16 November to 5 February. Eurosurveillance 2021; 26:2100256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Patone M, Thomas K, Hatch R, et al. Mortality and critical care unit admission associated with the SARS-CoV-2 lineage B.1.1.7 in England: an observational cohort study. Lancet Infect Dis 2021. Available at: 10.1016/S1473-3099(21)00318-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. 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; 21:1246–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Faria NR, Mellan TA, Whittaker C, et al. Genomics and epidemiology of the P.1 SARS-CoV-2 lineage in Manaus, Brazil. Science 2021; 372:815–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. de Oliveira MHS, Lippi G, Henry BM.. Sudden rise in COVID-19 case fatality among young and middle-aged adults in the south of Brazil after identification of the novel B.1.1.28.1 (P.1) SARS-CoV-2 strain: analysis of data from the state of Parana. medRxiv [preprint]. March 26, 2021. [cited 2021 June 30]. Available from: 10.1101/2021.03.24.21254046. [DOI] [Google Scholar]
  • 53. Ong SWX, Chiew CJ, Ang LW, et al. Clinical and virological features of SARS-CoV-2 variants of concern: a retrospective cohort study comparing B.1.1.7 (Alpha), B.1.315 (Beta), and B.1.617.2 (Delta). Rochester, NY: Social Science Research Network, 2021. Available at: https://papers.ssrn.com/abstract=3861566. Accessed 3 October 2021. [Google Scholar]
  • 54. Sheikh A, McMenamin J, Taylor B, Robertson C.. SARS-CoV-2 Delta VOC in Scotland: demographics, risk of hospital admission, and vaccine effectiveness. Lancet 2021; 397:2461–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Saito A, Irie T, Suzuki R, et al. SARS-CoV-2 spike P681R mutation, a hallmark of the Delta variant, enhances viral fusogenicity and pathogenicity. bioRxiv [preprint]. July 19, 2021. [cited 2021 Oct 3]. Available from: 10.1101/2021.06.17.448820. [DOI] [Google Scholar]
  • 56. Winkler MS, Skirecki T, Brunkhorst FM, et al. Bridging animal and clinical research during SARS-CoV-2 pandemic: a new-old challenge. EBioMedicine 2021; 66:103291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Li D, Edwards RJ, Manne K, et al. In vitro and in vivo functions of SARS-CoV-2 infection-enhancing and neutralizing antibodies. Cell 2021; 184:4203–19.e32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Ren W, Sun H, Gao GF, et al. Recombinant SARS-CoV-2 spike S1-Fc fusion protein induced high levels of neutralizing responses in nonhuman primates. Vaccine 2020; 38:5653–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Jones BE, Brown-Augsburger PL, Corbett KS, et al. The neutralizing antibody, LY-CoV555, protects against SARS-CoV-2 infection in nonhuman primates. Sci Transl Med 2021; 13:eabf1906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Klasse PJ, Nixon DF, Moore JP.. Immunogenicity of clinically relevant SARS-CoV-2 vaccines in nonhuman primates and humans. Sci Adv 2021; 7:eabe8065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Maisonnasse P, Guedj J, Contreras V, et al. Hydroxychloroquine use against SARS-CoV-2 infection in non-human primates. Nature 2020; 585:584–7. [DOI] [PubMed] [Google Scholar]
  • 62. Blair RV, Vaccari M, Doyle-Meyers LA, et al. Acute respiratory distress in aged, SARS-CoV-2–infected African green monkeys but not rhesus macaques. Am J Pathol 2021; 191:274–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Fahlberg MD, Blair RV, Doyle-Meyers LA, et al. Cellular events of acute, resolving or progressive COVID-19 in SARS-CoV-2 infected non-human primates. Nat Commun 2020; 11:6078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Chan JF-W, Zhang AJ, Yuan S, et al. Simulation of the clinical and pathological manifestations of coronavirus disease 2019 (COVID-19) in a Golden Syrian hamster model: implications for disease pathogenesis and transmissibility. Clin Infect Dis 2020; 71:2428–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Luan J, Lu Y, Jin X, Zhang L.. Spike protein recognition of mammalian ACE2 predicts the host range and an optimized ACE2 for SARS-CoV-2 infection. Biochem Biophys Res Commun 2020; 526:165–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Plante JA, Liu Y, Liu J, et al. Spike mutation D614G alters SARS-CoV-2 fitness. Nature 2021; 592:116–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Schäfer A, Muecksch F, Lorenzi JCC, et al. Antibody potency, effector function, and combinations in protection and therapy for SARS-CoV-2 infection in vivo. J Exp Med 2021; 218:e20201993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Zabaleta N, Dai W, Bhatt U, et al. An AAV-based, room-temperature-stable, single-dose COVID-19 vaccine provides durable immunogenicity and protection in non-human primates. Cell Host Microbe 2021; 29:1437–53, e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Martinez DR, SchäferA, Leist SR, et al. Prevention and therapy of SARS-CoV-2 and the B.1.351 variant in mice. Cell Rep 2021; 36:109450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Kim Y-I, Kim S-G, Kim S-M, et al. Infection and rapid transmission of SARS-CoV-2 in ferrets. Cell Host Microbe 2020; 27:704–9, e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Shi J, Wen Z, Zhong G, et al. Susceptibility of ferrets, cats, dogs, and other domesticated animals to SARS–coronavirus 2. Science 2020; 368:1016–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Francis ME, Richardson B, Goncin U, et al. Sex and age bias viral burden and interferon responses during SARS-CoV-2 infection in ferrets. Sci Rep 2021; 11:14536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Rosenke K, Meade-White K, Letko M, et al. Defining the Syrian hamster as a highly susceptible preclinical model for SARS-CoV-2 infection. Emerg Microbes Infect 2020; 9:2673–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Deng W, Bao L, Gao H, et al. Ocular conjunctival inoculation of SARS-CoV-2 can cause mild COVID-19 in rhesus macaques. Nat Commun 2020; 11:4400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Hartman AL, Nambulli S, McMillen CM, et al. SARS-CoV-2 infection of African green monkeys results in mild respiratory disease discernible by PET/CT imaging and shedding of infectious virus from both respiratory and gastrointestinal tracts. PLoS Pathog 2020; 16:e1008903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Port JR, Yinda CK, Owusu IO, et al. SARS-CoV-2 disease severity and transmission efficiency is increased for airborne compared to fomite exposure in Syrian hamsters. Nat Commun 2021; 12:4985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Lee AC-Y, Zhang AJ, Chan JF-W, et al. Oral SARS-CoV-2 inoculation establishes subclinical respiratory infection with virus shedding in golden Syrian hamsters. Cell Rep Med 2020; 1:100121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Port JR, Yinda CK, Avanzato VA, et al. Increased aerosol transmission for B.1.1.7 (alpha variant) over lineage A variant of SARS-CoV-2. bioRxiv [preprint]. July 26, 2021. [cited 2021 Oct 3]. Available from: 10.1101/2021.07.26.453518. [DOI] [Google Scholar]
  • 79. Kutter JS, de Meulder D, Bestebroer TM, et al. SARS-CoV and SARS-CoV-2 are transmitted through the air between ferrets over more than one meter distance. Nat Commun 2021; 12:1653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Richard M, Kok A, de Meulder D, et al. SARS-CoV-2 is transmitted via contact and via the air between ferrets. Nat Commun 2020; 11:3496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Sia SF, Yan L-M, Chin AWH, et al. Pathogenesis and transmission of SARS-CoV-2 in golden hamsters. Nature 2020; 583:834–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Chan JF-W, 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; 71:2139–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Yu P, Qi F, Xu Y, et al. Age-related rhesus macaque models of COVID-19. Animal Model Exp Med 2020; 3:93–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Osterrieder N, Bertzbach LD, Dietert K, et al. Age-dependent progression of SARS-CoV-2 infection in Syrian hamsters. Viruses 2020; 12:779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Yuan L, Zhu H, Zhou M, et al. Gender associates with both susceptibility to infection and pathogenesis of SARS-CoV-2 in Syrian hamster. Signal Transduct Target Ther 2021; 6:136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Dinnon KH, Leist SR, Schäfer A, et al. A mouse-adapted model of SARS-CoV-2 to test COVID-19 countermeasures. Nature 2020; 586:560–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Golden JW, Cline CR, Zeng X, et al. Human angiotensin-converting enzyme 2 transgenic mice infected with SARS-CoV-2 develop severe and fatal respiratory disease. JCI Insight 2020; 5:e142032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Johansen MD, Irving A, Montagutelli X, et al. Animal and translational models of SARS-CoV-2 infection and COVID-19. Mucosal Immunol 2020; 13:877–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Yinda CK, Port JR, Bushmaker T, et al. K18-hACE2 mice develop respiratory disease resembling severe COVID-19. PLoS Pathog 2021; 17:e1009195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Zheng J, Wong L-YR, Li K, et al. COVID-19 treatments and pathogenesis including anosmia in K18-hACE2 mice. Nature 2021; 589:603–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Brocato RL, Principe LM, Kim RK, et al. Disruption of adaptive immunity enhances disease in SARS-CoV-2-infected Syrian hamsters. J Virol 2020; 94:e01683–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Leclerc Q, Fuller N, Knight L, Funk S, Knight G.. What settings have been linked to SARS-CoV-2 transmission clusters? [version 2; peer review: 2 approved]. Wellcome Open Res 2020; 5:83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Madewell ZJ, Yang Y, Longini IM Jr, Halloran ME, Dean NE.. Household transmission of SARS-CoV-2: a systematic review and meta-analysis. JAMA Network Open 2020; 3:e2031756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Chou R, Dana T, Buckley DI, Selph S, Fu R, Totten AM.. Epidemiology of and risk factors for coronavirus infection in health care workers. Ann Intern Med 2020; 173:120–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Chou R, Dana T, Buckley DI, Selph S, Fu R, Totten AM.. Update alert: epidemiology of and risk factors for coronavirus infection in health care workers. Ann Intern Med 2020; 173:W46–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Chou R, Dana T, Buckley DI, Selph S, Fu R, Totten AM.. Update alert 2: epidemiology of and risk factors for coronavirus infection in health care workers. Ann Intern Med 2020; 173:W77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Chou R, Dana T, Buckley DI, Selph S, Fu R, Totten AM.. Update alert 3: epidemiology of and risk factors for coronavirus infection in health care workers. Ann Intern Med 2020; 173:W123–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Chou R, Dana T, Buckley DI, Selph S, Fu R, Totten AM.. Update alert 4: epidemiology of and risk factors for coronavirus infection in health care workers. Ann Intern Med 2020; 173:143–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Chou R, Dana T, Buckley DI, Selph S, Fu R, Totten AM.. Update alert 5: epidemiology of and risk factors for coronavirus infection in health care workers. Ann Intern Med 2020; 173:W154–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Chou R, Dana T, Selph S, Totten AM, Buckley DI, Fu R.. Update alert 6: epidemiology of and risk factors for coronavirus infection in health care workers. Ann Intern Med 2021; 174:W18–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Chou R, Dana T, Selph S, Totten AM, Buckley DI, Fu R.. Update alert 7: epidemiology of and risk factors for coronavirus infection in health care workers. Ann Intern Med 2021; 174:W45–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Chou R, Dana T, Buckley DI, Selph S, Fu R, Totten AM.. Update alert 8: epidemiology of and risk factors for coronavirus infection in health care workers. Ann Intern Med 2021; 174:W48–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Chou R, Dana T, Buckley DI, Selph S, Fu R, Totten AM.. Update alert 9: epidemiology of and risk factors for coronavirus infection in health care workers. Ann Intern Med 2021; 174:W63–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Gómez-Ochoa SA, Franco OH, Rojas LZ, et al. COVID-19 in health-care workers: a living systematic review and meta-analysis of prevalence, risk factors, clinical characteristics, and outcomes. Am J Epidemiol 2021; 190:161–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Galanis P, Vraka I, Fragkou D, Bilali A, Kaitelidou D.. Seroprevalence of SARS-CoV-2 antibodies and associated factors in healthcare workers: a systematic review and meta-analysis. J Hosp Infect 2021; 108:120–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Sahu AK, Amrithanand VT, Mathew R, Aggarwal P, Nayer J, Bhoi S.. COVID-19 in health care workers—a systematic review and meta-analysis. Am J Emerg Med 2020; 38:1727–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Bandyopadhyay S, Baticulon RE, Kadhum M, et al. Infection and mortality of healthcare workers worldwide from COVID-19: a systematic review. BMJ Global Health 2020; 5:e003097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Bao L, Deng W, Huang B, et al. The pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nature 2020; 583:830–3. [DOI] [PubMed] [Google Scholar]
  • 109. Hassan AO, Case JB, Winkler ES, et al. A SARS-CoV-2 infection model in mice demonstrates protection by neutralizing antibodies. Cell 2020; 182:744–53, e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Jiang R-D, Liu M-Q, Chen Y, et al. Pathogenesis of SARS-CoV-2 in transgenic mice expressing human angiotensin-converting enzyme 2. Cell 2020; 182:50–8, e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Winkler ES, Bailey AL, Kafai NM, et al. SARS-CoV-2 infection of human ACE2-transgenic mice causes severe lung inflammation and impaired function. Nat Immunol 2020; 21:1327–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Moreau GB, Burgess SL, Sturek JM, Donlan AN, Petri WA, Mann BJ.. Evaluation of K18-hACE2 mice as a model of SARS-CoV-2 infection. Am J Trop Med Hyg 2020; 103:1215–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Golden JW, Cline CR, Zeng X, et al. Human angiotensin-converting enzyme 2 transgenic mice infected with SARS-CoV-2 develop severe and fatal respiratory disease. JCI Insight 2020; 5:e142032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Rasmussen AL, Escandón K, Popescu SV.. Facial masking for Covid-19. N Engl J Med 2020; 383:2092. [DOI] [PubMed] [Google Scholar]
  • 115. Brosseau LM, Roy CJ, Osterholm M.. Facial masking for Covid-19. N Engl J Med 2020; 383:2092–3. [DOI] [PubMed] [Google Scholar]

Articles from Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America are provided here courtesy of Oxford University Press

RESOURCES