Chapter 1

COVID Overview

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

SymptomsCopy Link!

Common SymptomsCopy Link!

Updated Date: May, 2020
Literature Review:
Gallery View, Grid View
Tool:
CDC Symptom Self-Checker

This section covers symptom prevalence, click here for Triage Based on Symptom Questionnaires.

Many patients are asymptomatic. Among patients with symptoms, most present with an influenza-like illness (fevers, myalgias, respiratory symptoms), but many do not present with this classic combination. Some may present with less-usual findings such as perniosis (COVID toes) or anosmia. These ranges are pulled from the following articles, and symptom prevalence varies greatly depending on testing and survey methodology (Arentz et al; Chen et al; Guan et al; Li et al; Wu et al; Zhou et al; WHO-China Joint Mission on COVID-19; Young et al; Yan et al; Jiang et al; Huang et al; Tostmann et al).

  1. Fever, 44-94%
  1. We recommend using >= 38°C to define fever, taking into account the patient’s age, immune status, medications (steroids, chemotherapy, etc.), and recent use of fever-reducing medications.
  2. Children are less likely to have fever or cough (Bialek et al).
  1. Cough, 68-83%
  2. Anosmia and/or ageusia (loss of sense of taste and/or smell) ~70%
  3. Upper respiratory symptoms (sore throat, dripping nose, nasal or sinus congestion), 5-61%
  4. Shortness of breath, 11- 40%
  5. Fatigue, 23-38%
  6. Muscle aches 11-63%
  7. Headache 8-14%
  8. Confusion 9%
  9. Gastrointestinal symptoms (nausea, vomiting, diarrhea), 3-17%

Clinical CourseCopy Link!

Literature Review: University of Washington Literature Report (Clinical Characteristics)

Incubation and Window PeriodCopy Link!

Updated Date: December 19, 2020

Incubation period is the time from exposure to symptom onset. Latency period is the time from exposure to infectiousness (or viral detection, depending on the definition). COVID-19 has a relatively long incubation period, and typically at least 2 days of infectivity before symptoms develop.

IncubationCopy Link!

Time from exposure to symptom onset: mean and median 5 days (common range 2-7 days). (Li et al; Guan et al; Velavan et al; Chan et al; Nie et al).

  • 97.5% of exposed cases will develop symptoms within 11 days and 99% within 14 days. Over 95% of cases develop symptoms within 13 days of infection (Nie et al).
  • Incubation periods of up to 24 days are shown in some reports (Nie et al).

Window PeriodCopy Link!

Samples taken before symptom onset have high false negative rates, as modeled by (Kurcirka et al). 68% false negatives one day before symptoms, compared to 38% false negatives on the first day of symptoms, based on serial testing. They estimated the window period between exposure and detectability of SARS-CoV-2 RNA on nasopharyngeal sampling at 3-5 days, with peak sensitivity 8 days after exposure or 3 days after symptom-onset in their model. As with incubation, individual cases may show longer delays. Asymptomatic patients should still be tested in certain circumstances, but a negative result does not rule out infection.

Duration and Time CourseCopy Link!

Updated Date: May 2020
Literature Review (Clinical Course):
Gallery View, Grid View

Median duration of common symptoms (median in survivors only), drawn from (Zhou et al; Young et al):

  1. Fever: 12 days
  2. Shortness of breath: 13 days
  3. Cough: 19 days

Time course from symptom onset to complications (Zhou et al, Feldstein et al):

  1. Multi-System Inflammatory Syndrome in Children (MIS-C): 6 days (range 4-8 days).
  2. Sepsis: Median onset 9 days (range 7-13 days)
  3. Acute Respiratory Distress Syndrome (ARDS): median onset 12 days (range 7-15 days)
  4. Need for Mechanical Ventilation: Median onset 10 days (range 3-12.5 days)
  5. Acute Cardiac Injury: Median onset 15 days (range 10-17 days)
  6. Acute Kidney Injury: Median onset 15 days (range 13-19.5 days)
  7. Secondary Infection: Median onset 17 days (range 13-19 days)
  8. Death: Median 18.5 days, interquartile range 15-22 days (Zhou et al)
  1. Illness severity has been noted to have two peaks at ~14 days and ~22 days (Ruan et al)

SeverityCopy Link!

Updated Date: May, 2020
Literature Review:
Gallery View, Grid View

The majority of patients have only mild symptoms; however, the percentage of patients who develop severe or critical disease is far greater than for most other respiratory viruses, including influenza. See how mild, moderate, and severe cases are defined. Assessing the percentage of patients who develop differing severities of illness is fundamentally challenging, due to the widely variable case definitions and severity definitions, as well as the lack of population-level surveillance testing to estimate asymptomatic and minimally symptomatic cases. All of these are estimates and do not apply to all populations or epidemiologic circumstances.

  • Asymptomatic Infection is present in about 20% of cases (Bi et al; Mizumuto et al; Pollan et al). One metanalysis showed asymptomatic infections account for 17% of all infections (Byambasuren et al) but this is difficult to estimate as screenings of entire populations are unavailable. As vaccines become more common, this percentage of asymptomatic infection is likely to change, as vaccinated patients are less likely to be symptomatic (see Reinfection and Breakthrough Infection).
  • Symptomatic Infection: A Chinese CDC report on approximately 72,000 symptomatic COVID cases (1% of the cases included in the study were asymptomatic), documented the following occurrence rates for mild, severe, and critical symptom presentations (Wu et al):
  • Mild Symptoms to Mild Pneumonia: approximately 81%
  • Severe Symptoms (blood oxygen saturation less than or equal to 93%, respiratory frequency greater than or equal to 30 breaths per minute, and/or lung infiltrates greater than 50% within 48 hours): approximately 14%
  • Critical Symptoms (respiratory failure, shock, multiorgan dysfunction): approximately 5%.
  • Among critically-ill patients, many receive mechanical ventilation. Median time on a ventilator ranges from 11-17 days (Chen et al; Ling et al).
  • Presentation with shock is rare, but vasopressors are eventually used in 67% of critically-ill patients.
  • Cardiomyopathy (Heart Tissue Injury) is noted in 33% of critically-ill patients (Ruan et al).

Prognostic IndicatorsCopy Link!

Updated Date: May, 2020

Demographic and Health FactorsCopy Link!

Literature Review (Comorbidities): Gallery View, Grid View
Literature Review (Sex Differences):
Gallery View, Grid View

Multiple factors have been associated with worse prognosis in people infected with SARS COV-2.

  1. Viral variant. Certain variants, such as the Delta variant, may be more severe than others. See new variants.
  2. Age: increased age is associated with more severe disease and higher rates of death (Wu et al; Chen et al; Yang et al; Qin et al).
  1. Children are less likely to have severe disease, but pediatric deaths have been reported (Bialek et al).
  2. Children appear to be as likely to contract the infection as adults, although symptomatic cases of children are more rare (Bi et al).
  1. Comorbidities and other health factors: Multiple comorbidities and/or health factors are associated with increased risk of severe COVID-19 illness. Evidence-based knowledge on this topic is continuing to develop; for ongoing updates, see the CDC’s living document. The comorbidities and other health factors associated with the strongest bases of evidence for increased risk are listed below. This list is not inclusive of all conditions which may be associated with increased risk; other common conditions which may be associated with increased risk include hypertension, moderate to severe asthma, liver disease, and others (CDC).
  1. Chronic Kidney Disease
  2. Chronic Obstructive Pulmonary Disease (COPD)
  3. Type 2 Diabetes Mellitus
  4. Pregnancy
  5. Sickle Cell Disease
  6. Smoking
  7. Cancer
  8. Down Syndrome
  9. Immunocompromised status associated with solid organ transplant
  10. Obesity (BMI of 30kg/M2 or higher)
  11. Multiple heart conditions, including heart failure, coronary artery disease, and cardiomyopathies
  1. Race: Please see Health Equity for a discussion on racial differences in COVID infection and severity.
  2. Sex: Men appear to be more severely affected by COVID-19 than women. Conclusive evidence related to sex differences is limited by methodology of existing studies (Schiffer et al).
  3. Smoking: Smoking may offer a small risk reduction for COVID infection, though it is not clear why and this finding may be subject to confounding. It does appear to be associated with worse outcomes. See Smoking for more details.

Laboratory IndicatorsCopy Link!

The most significant laboratory abnormalities associated with severe COVID-19 disease and death include the following:

Lab test

Results

Normal Ranges (For many US labs, units and values may vary)

White Blood Cell Count (WBC)

> 10 K/uL (K/uL=10^3/uL)

Male and Female- Adults: 3.4-9.6 x10^3/uL

Lymphopenia

< 1.00 K/uL (K/uL=10^3/uL)

Male and Female- Adults: 0.95-3.07 x10^3/uL

Platelets

< 150 K/uL (K/uL=10^3/uL)

Male Adults: 135-317 x 10^3/uL

Female Adults: 157-371 x10^3/uL

Creatinine

> 1.5 mg/dL

Male Adults: 0.74-1.35 mg/dL

Female Adults: 0.59-1.04 mg/dL

Albumin

< 3 g/dL

3.5-5.0 g/dL

Alanine transaminase (ALT)

> 40 U/L

Males: 7-55 U/L

Females: 7-45 U/L

Creatinine kinase (CK)

> 185 U/L

Males: 39-308 U/L

Females: 26-192 U/L

Troponin T, high-sensitivity (hs-TnT)

> ~20 ng/L

Male <23 ng/L

Female <15 ng/L

C-reactive protein (CRP)

> 125 mg/L

< or =8.0 mg/L

Lactate dehydrogenase (LDH)

> 245 U/L

Adults: 122-222 U/L

Ferritin

> 300 ug/L (Severe Disease); Ferritin > 1000 ug/L (Death)

Males: 24-336 ug/L

Females: 11-307 ug/L

Interleukin 6 (IL-6)

> 10 pg/mL

< or =1.8 pg/mL

D-Dimer

> 1000 ng/mL

< 250 ng/mL

Procalcitonin

> 0.5 ng/mL

< or =0.15 ng/mL

(Zhou et al; Huang et al; Chen et al; Wu et al; Ruan et al)

MortalityCopy Link!

Updated Date: December 16, 2020
Literature Review:
Gallery View, Grid View

Cause of DeathCopy Link!

Determining and reporting the cause of death for patients with COVID-related diseases is complex (as it is with any disease).

  • Cause of Death: This is usually the acute medical diagnosis that caused a patient to die, and often relates to a medium-term or long-term diagnosis as well. It will often include other diseases as co-morbid or contributing factors (e.g. pneumonia due to COVID-19 infection or Acute Myocardial Ischemia due to COVID-19 infection and Coronary Artery Disease).
  • Mechanism of Death: Defined as the immediate physiologic issue resulting in death (for example, hypoxemia).

A significant number of COVID-related deaths do not have clear delineation of cause of death (CEBM). The majority of people who die from COVID-19 die from respiratory failure. Because definitions of cause of death are reported differently it can be hard to determine exact numbers, but here are estimates (Ruan et al, 68 cases), (Zhang et al, 82 cases):

  • Respiratory Failure Alone: 53% - 69%
  • Circulatory Failure Alone: 7%-14.6%
  • Mixed Respiratory and Circulatory Failure, Sepsis, or Multiorgan Failure: 28-33%
  • Hemorrhage: 6.1%
  • Renal Failure: 3.1%

Tool: Improving Cause of Death Reporting
Tool: Guidance for Reporting COVID-Related Deaths

Case Fatality RateCopy Link!

Literature Review: Gallery View, Grid View

  • Case Fatality Rate (CFR) is typically the proportion of deaths from a disease relative to the number of people diagnosed with the disease in a specific period of time. Some people define a “case” as showing symptoms.
  • Infection Fatality Rate (IFR) is the proportion of deaths from a disease but relative to all infected individuals including asymptomatic people and infections that were missed. It is harder to measure, and thus most places report CFR.
  • Case Fatality Rate is variable in different countries. Range around the world seems to be between 0-16%, with most countries in the 1-3% range.

Tool: Johns Hopkins Summary of Case Fatality Ratios
Tool: Forecast Hub (Compilations of forecasts by country or state)

PathophysiologyCopy Link!

PathophysiologyCopy Link!

Updated Date: December 16, 2020
Literature Review (ACE2):
Gallery View, Grid View
Literature Review (Human Genetics)
Gallery View, Grid View

Classification: SARS-CoV-2 is a positive-stranded RNA virus with a nucleocapsid and envelope, belonging to the coronavirus family, of which seven viruses (including the original SARS-CoV in 2003 and MERS in 2013) have crossed from zoonotic origins into humans.

Cell Entry and Replication: For cell entry, the SARS-CoV-2 spike protein binds to the ACE2 receptor, expressed in nasal and bronchial epithelium, pulmonary endothelium, alveolar Type 2 cells, proximal renal tubule cells, cardiac myocytes, gastrointestinal epithelial cells, and others. Cleavage/priming by serine protease TMPRSS2 facilitates SARS-CoV2 cell entry, followed by viral replication using host cell machinery and then exocytosis (Kumar et al).

Cellular Targets and Resulting Lung Injury: The cells that express ACE2 may be the cell populations most injured by infection or targeted by the immune response. Alveolar Type 2 cells secrete surfactant, so injury may result in alveolar collapse at low opening pressures and high PEEP sensitivity, while damage to pulmonary endothelial cells may cause capillary leak and trigger an influx of monocytes and neutrophils, with formation of hyaline membranes. The highly inflamed lung parenchyma can develop microthrombi that help explain some of the thrombotic complications of COVID (Wiersinga et al).

Literature Review (Acute Lung Injury): Gallery View, Grid View

Inflammatory Cascade: Infection with the SARS-Cov-2 virus can cause apoptotic cell death, which triggers an inflammatory cascade of cytokine release, as well as the recruitment of immune cells including macrophages and dendritic cells, and later, antigen-specific T lymphocytes (Bohn et al). If the immune response is not properly checked, a state of hyperinflammation occurs, with the development of Cytokine Storm Syndrome, and sometimes multi-organ failure.

Blood type: There is evidence that A blood type is a risk factor for COVID-19 respiratory failure, and O may be protective. This was based on a genome-wide association study (GWAS) of 835 patients and 1255 control participants from Italy and 775 patients and 950 control participants from Spain. Respiratory failure was defined as a patient requiring supplemental oxygen or mechanical ventilation (Ellinghaus et al).

Literature Review (ABO): Gallery View, Grid View

Histology and AutopsyCopy Link!

Updated Date: October 1, 2021
Literature Review (Autopsy):
Gallery View, Grid View
Literature Review (Histology):
Gallery View, Grid View

  • Histology of COVID-19 associated lung disease most often shows bilateral diffuse alveolar damage with cellular fibromyxoid exudates, desquamation of pneumocytes, pulmonary edema, hyaline membrane formation, microthrombi The prevalence of microthrombi identified in the pulmonary vasculature is similar to that seen in patients with SARS-associated ARDS and higher than that seen during H1N1 influenza-associated ARDS (Hariri et al, 2021)., organizing fibrosis and superimposed pneumonia. There is evidence of direct viral injury to lung tissue, as well as inflammatory sequelae. (Xu et al, Lancet Respir Med, 2020, Hariri et al, Chest, 2021).
  • Cardiac injury and thrombotic complications are widely prevalent, including cardiac inflammatory infiltrates, epicardial edema, and pericardial effusion in some autopsies (Falasca et al; Elsoukkary et al; Geng et al).
  • Acute kidney injury, while common in hospitalized COVID patients, was found to be mild in post-mortem patients with theoretical potential for recovery (Santoriello et al).
  • Neurologic lesions in autopsy series of 43 patients (not necessarily with neurologic manifestations) showed fresh ischemic lesions in 14%, and neuroinflammatory changes with infiltration of cytotoxic T lymphocytes most pronounced in the brainstem (also cerebellum and meninges) (Matschke et al). In patients with significant neurologic decline, more severe findings have been noted including hemorrhagic lesions through the cerebral hemispheres, marked axonal injury, areas of necrosis, and pathology similar to Acute Disseminated Encephalomyelitis (ADEM). (See e.g. Reichard et al).

EpidemiologyCopy Link!

Literature Review: University of Washington Literature Report (Geographic Spread)

Literature Review: University of Washington Literature Report (Modeling and Prediction)

Tool: Outbreak.info (Epidemiology Resources)

Case Counts and PrevalenceCopy Link!

Updated Date: December 19, 2020
Tool: Worldwide case counts are published by teams at the World Health Organization, Johns Hopkins University, and others.

Prevalence estimates depend significantly on testing availability and percentage of the population that has asymptomatic infection as well as on the severity of the epidemic in a specific location. Seroprevalence studies, measuring antibodies across an entire population, can help give a better estimate of true prevalence. In one meta-analysis of 47 studies on seroprevalence covering 399,265 people from 23 countries, the SARS-CoV-2 seroprevalence in the general population varied from 0.37% to 22.1%, with a pooled estimate of 3.38% (Rostami et al). This will no doubt change over time as more people are infected.

OriginsCopy Link!

Updated date: June, 2020
Literature Review:
Gallery View, Grid View

Tool: WHO Virus Origin


COVID-19 transmission is primarily human-to-human following a suspected animal-to-human initiating event (Li et al). It is thought that it may have emerged from raccoon dogs or civets, but this is still being investigated (Mallapaty). The virus was initially recognized in December 2019 by Chinese authorities in the setting of cases of pneumonia that seemed to be clustered around a seafood market in Wuhan, Hubei Province (Wuhan Municipal Health Commission, 2019). Laboratory samples collected in December 2019 yielded evidence of a novel betacoronavirus, genetically-distinct from previously identified SARS-CoV and MERS-CoV but genetically-similar to previously-published coronavirus strains collected from bats from southwestern China (Zhu et al).

New VariantsCopy Link!

Updated Date: December 30, 2021

Literature Review (Viral Genetics) Gallery View, Grid View
Tool: Viral genomes have been published to GenBank from diverse geographies.
Tool: Reports on real-time phylogenetic tracking of the viral genome can be found at NextStrain (Hadfield et al).
Tool: CDC emerging variants.

Tool: Outbreak.info Mutation Reports (from GISAID data)

Tool: NYT Coronavirus Variants and Mutations

Tool: CDC Delta Variant.

Major New VariantsCopy Link!

Frequency of new mutations: Mutation of RNA viruses is expected and common, though less common in coronaviruses than many other RNA viruses due to “proofreading” capacity (Robson et al). Mutations started occurring in SARS-CoV-2 in the fall of 2020 (CDC), and continue to occur over time. The naming system for variants was a letter-and-number system until June 2021, when the WHO created a newer simpler naming system using greek letter names (Nature News).

Tool: Axios Variant Tracker (this outlines the major variants, their relative infectiousness and severity)

Tool: NextStrain Tracker (this gives major strain data globally)

Tool: US CDC Variant Tracker

Infectiousness and SeverityCopy Link!

The meaning of these mutations for transmission and severity depends on the exact mutation. In the Summer of 2021, the Delta variantwas identified and appeared to be more transmissible than the ancestral strain (see viral load), and also more severe. One study from the UK of over 43,000 cases showed that Delta patients had twice the risk of hospitalization compared with Alpha patients, despite overall being younger (Twohig et al). On November 26, 2021 the WHO named Omicron a new variant of concern. Omicron is unique because it has 50 new mutations not seen in combination before, with more than 30 mutations located in the spike protein, several of which are believed to make this variant even more infectious than Delta. Though some studies suggest Omicron causes less severe illness, this has not yet been definitively shown. A recent study from South Africa showed that two doses of the Pfizer-BioNTech vaccine was 70% effective against preventing hospitalizations while Omicron was the dominant variant (compared to 93% effectiveness in the period before Omicron was identified).

Infectivity and TransmissionCopy Link!

InfectivityCopy Link!

Updated Date: August 30, 2021

Viral Load, PCR Clearance, and Infectiousness TimelineCopy Link!

Literature Review (Viral Shedding): Gallery View, Grid View

Patients who are infected with SARS COV-2 and who have higher levels of virus in their respiratory tracts and oropharynx are the most infectious, regardless of their level of symptoms (Bullard et al). Symptom status does not seem to correlate predictably with viral load (Walsh et al; Lee et al; Zou L et al). Certain viral variants, like Delta, appear to cause higher viral loads (1260 higher, Li et al), and thus be more infectious.

Upper airway viral load peaks within ~5 days of symptom onset, followed by decline (Wölfel et al; Young et al). Consequently, patients appear to be most infectious in the 2-3 days before symptom onset and the 2-3 days after (Ferretti et al). PCR detection continues for a median of 20 days from time of symptom onset, with an interquartile range 17-24 days (Zhou et al). There are rare cases that remain positive up to ~60 days after infection (McKie et al).

However, viral load does not always correlate perfectly with infectiousness. It is measured by quantitative PCR, which cannot distinguish between a live viable virus or a dead or inactivated virus. The virus is very rarely culturable (our closest proxy to infectivity) after 9 days (Cevik et al). The culture data underlies the newer guidance (after November 2020) about Quarantine time. See Testing for a diagram of test positivity compared with infectivity and symptoms.

Asymptomatic PatientsCopy Link!

Literature Review (Asymptomatic): Gallery View, Grid View
Literature Review (Presymptomatic):
Gallery View, Grid View

Asymptomatic, minimally symptomatic (paucisymptomatic), and pre-symptomatic patients can all transmit the virus (Bai et al; Rothe et al; Furukawa et al), though presence of symptoms is probably associated with increased frequency of transmission. Though it is hard to estimate the prevalence of asymptomatic cases due to testing bias and few population-level studies, one metanalysis found that asymptomatic patients represented 17% or cases, and were 42% less likely to transmit than symptomatic cases (Byambasuren et al). In one study in Beijing, face masks worn by family members of pre-symptomatic COVID-19 patients were shown to be 79% effective (OR = 0.21) at reducing transmission, suggesting that presymptomatic transmission is an important mode of transmission and that masks can be effective at preventing it (Wang et al).

Recovered PatientsCopy Link!

Patients who have recovered from COVID sometimes will have fragments of viral RNA that continue to test positive by PCR. Shedding of viral RNA is longer in more severe disease, or in patients who are immunocompromised. However, recent data shows that this viral RNA does not likely represent infectious virions, but rather parts of the virus that are unable to replicate. As such, the U.S. CDC has changed its recommendations on the duration of isolation and quarantine as well as releasing patients from isolation (Cevik et al).

Vaccinated PeopleCopy Link!

We do not yet know how all available and pending vaccines will perform with respect to asymptomatic infection and transmission for all variants. This is an evolving area of research, but the data suggest that at a population-level less transmission occurs between vaccinated people. However, an individual vaccinated person who is experiencing a breakthrough infection (asymptomatic or symptomatic) can certainly transmit to others; For the ancestral strain this was thought to be less common in vaccinated people compared with unvaccinated, but for highly-contagious strains like the delta variant, transmission appears to occur at similar rates regardless of vaccination status. (CDC) Multiple studies have shown that vaccinated and unvaccinated people have similar viral loads/ infectiousness (Brown et al, Riemersma et al), at least in the first six days of infection. After six days, vaccinated people appear to have lower viral loads and be less infectious (Chia et al) Epidemiologic suggestions about what protective measures vaccinated people should take vary depending on the type of vaccine in question in that country. Follow local guidance.

Tool: Current US CDC guidance on infection prevention and safer activities for vaccinated people. (Includes helpful infographic)

TransmissionCopy Link!

Literature Review: University of Washington Literature Report (Transmission)

Basic Reproduction Number (R0)

Tool: For global estimates of R0, See Here. For the United States, state-by-state estimates of R0 are available Here (Data from The COVID Tracking Project). Please keep in mind these are merely estimates and all models are fallible.

R0 (R-naught) is a measure of transmissibility. It represents the theoretical number of secondary infections from an infectious individual. This is a property both of the infectiousness of the virus and the behaviors of humans to decrease spread.

  • An R0 > 1 is consistent with sustained outbreak.
  • An R0 < 1 means an epidemic is declining.

The R0 for COVID-19 is likely similar to, or slightly higher than, many other respiratory viruses, but because it is so highly influenced by human behavior, it can be changed. The initial R0 of COVID in Wuhan in the absence of containment measures was thought to be about 2.5 (Majumder et al). However, R0 declines with control measures (Zhao et al; Riou et al; Flaxman et al; Read et al; Shen et al). As variants of COVID develop, the R0 is likely to change.

The original ancestral strain, pre-control R0 of 2.5 is:

Aerosol, Droplet and Fomite TransmissionCopy Link!

Updated Date: August 30, 2021

Literature Review (Airborne v Droplet): Gallery View, Grid View
Literature Review (Aerosolization):
Gallery View, Grid View
Literature Review (Fomites):
Gallery View, Grid View

COVID-19 transmission primarily occurs through liquid respiratory particles (droplets, 50-100 micrometers particles) that travel through the air between people who are within a distance of about 2 meters of one another. Early in pandemic there was debate about whether transmission also occurs via aerosols (small particles under <5 micrometers), which can hang in the air for far longer and travel longer distances. Growing evidence indicates that aerosol transmission is possible, especially in poorly-ventilated spaces and with periods of exposure exceeding 30 minutes (Lancet Editorial), and the World Health Organization and US CDC both changed their guidance to include aerosol spread in spring of 2021.

Droplet Transmission: Liquid respiratory particles vary in size and are produced during breathing, talking, singing, coughing, and sneezing (CDC). Larger particles of 60-100 micrometers typically do not travel through the air farther than 2 meters (Lancet Editorial).

Airborne/Aerosol Transmission: Very small respiratory droplets, often called aerosols, remain suspended in the air and travel a distance exceeding 2 meters (Lancet Editorial). The risk of producing aerosols is heightened during coughing, sneezing, and certain medical procedures (WHO-China Joint Mission on COVID-19). See concerns about aerosolization to see a list of procedures and devices and the effect they may have on aerosols. Aerosolized particles appear to remain in the air for at least 3 hours (Van Dorelmalen et al), with some laboratory studies indicating it can be as long as 16 hours (Fears et al)

Fomite (Objects and Surfaces) Transmission: Transmission through touching contaminated objects before touching the mouth, nose, or eyes, is an inefficient mode of transmission (Kampf et al). Mathematical models suggest that the chance of an infection occurring from a contact with a contaminated surface is less than 1 in 10,000. (CDC) While viral RNA has been detectable < 24h on cardboard and < 72h on plastic or steel (Van Dorelmalen et al), attempts to culture virus from surfaces have been unsuccessful (Colaneri et al), suggesting that fomite transmission is unlikely. In cases of suspected transmission through fomites and direct contact, full exclusion of respiratory transmission as the actual mode has not been possible. Transmission through the handling of contaminated objects is presumed to be unusual (Meyerowitz et al). Adherence to standard precautions and disinfection of equipment and surfaces is still indicated (Mondelli et al).

Water and Sewage: Persistence of SARS-CoV-2 virus in drinking-water is possible; indeed, some organizations and public health departments are tracking COVID infection rates by measuring waste-water RNA (see CDC Wastewater Testing, and Larsen et al). There is no evidence to date about survival of the virus in water or sewage, but it is likely to become inactivated significantly faster than non-enveloped human enteric viruses with known waterborne transmission (such as adenoviruses, norovirus, rotavirus and hepatitis A).

Bodily FluidsCopy Link!

  1. Feces and whole blood have been shown to contain viral ribonucleic acid (RNA) on PCR studies (Wölfel et al; Young et al). Significance for transmission is unclear (Chen et al), though in one systematic review of smaller studies, replication-capable virus was found in 35% of samples (van Doorn et al), meaning that fecal transmission may be possible.
  2. Urine does not appear to contain viral ribonucleic acid (Wölfel et al).
  3. Semen and vaginal secretions: COVID-19 virus has not been detected in vaginal secretions (Qiu et al). It is detectable in semen, but transmissibility is unclear. Likelihood of transmission via respiratory secretions during sexual encounters, however, is likely (Sharun et al).
  4. Tears: a few studies have indicated presence of COVID-19 virus in tears, while others have not. Current evidence is limited, but risk of transmission through tears is thought to be low (Seah et al).
  5. Cerebrospinal Fluid: Rarely, CSF has been noted to be positive by PCR (in 2 of 578 samples in one study, but not at levels that are infectious) (Destras et al).

Household and Community TransmissionCopy Link!

Household contacts of an index case appear more likely to contract the virus than other contacts (Bi et al). Most transmission events occur within households (Luo et al). The household secondary attack rate (e.g. number of people who get infected from an index case) is very variable, thought to be about 17.2% in one meta analysis (Fung et al), though very few studies tested more than once, so many cases may not have been missed The results ranged from 10.3-32.4% when contacts were tested at least twice. One recent study that did daily testing estimated SAR at 35% excluding those who had positive tests at enrollment, 53% including cases positive on enrolment. 75% of secondary cases occurred within 5 days of the index patient’s symptom onset (Grijalva et al). However, when prevalence increases, more community (meaning with no known exposure) transmissions tend to occur, highlighting the necessity of non-pharmaceutical interventions (e.g. masks) coupled with public health strategies such as sentinel and syndromic surveillance. Having children ages 0-3 is associated with higher secondary attack rates compared with children aged 14-17 (OR 1.43), possibly due to the inability of smaller children to distance and care for themselves. (Paul et al)

Super-Spreading Events (SSEs)Copy Link!

Literature Review Gallery View, Grid View

Super-Spreading Events are when an individual directly spreads an infection to an unusually large number of others. Several cases of superspreading have occurred at choirs (Hamner et al), weddings (including a Maine wedding that led to 177 linked cases, including seven deaths), churches Daegu, South Korea, where “Patient 31” infected at least 40 others (Ryall), and even within the White House. SSEs are believed to be disproportionately responsible for COVID-19 cases globally, with several studies suggesting that ≈80% of secondary transmissions have been caused by a small fraction (≈10%) of initially infected individuals. (Althouse et al; Endo et al). SSEs are heavily dependent on sociobiological mechanisms, including individual viral load, numbers of susceptible contacts per person, residence or employment in congregate settings, and ‘opportunistic’ scenarios including temporary clustering of individuals in mass gathering events. Environmental factors also are very important with closed places, crowded places, and poor ventilation playing a significant role in SSEs. Because SSEs play such an outsized role in fueling the pandemic, they amount to a significant concern, but also serve as an opportune area for public health interventions, particularly the prevention of transmission events where over 10 people are infected (Althouse et al).

SchoolsCopy Link!

Updated Date: January 5, 2022

Schools are unique settings and are likely to contribute to COVID-19 transmission between households and within communities. However, sustained closure of in-person schooling is expected to have an adverse effect on life outcomes for children and to worsen existing inequalities.

The American Academy of Pediatrics advocates that children should be physically present in school where possible (AAP Guidance). In some places, it appears that limited reopening with some precautions has not led to significant numbers of transmission events or large outbreaks (US CDC). However, this may be very location specific: a large-scale study of over 500,000 contacts of 85,000 infected cases in India have noted that children are a significant source of spread, even despite school closures (Laxminarayan et al).

In the Fall of 2021, to minimize the amount of time students needed to quarantine and miss in-person learning, some schools began implementing a Test To Stay strategy. This involves routine serial testing paired with contract tracing, allowing school-associated close contacts to remain in school during their quarantine period.

Multiple studies have shown that mitigation measures like masks, distancing, and ventilation have a significant impact on reducing transmission in schools (Lessler et al, Doyle et al, Dawson et al, Falk et al). Schools without mask mandates are 3.5 times more likely to have COVID-19 outbreaks than schools with mask mandates based on data from early in the 2021-2022 school year in the USA (CDC). A simulation study indicates that opening windows may significantly reduce transmission, as much as 14 fold, and masks may reduce transmission as much as 8 fold (Villers et al). The exact repercussions of novel variants on these interventions has yet to be determined, but it is likely that they will continue to reduce risk.

This thorough Review of the Literature on School Transmission and Safety summarizes some of the unique challenges and recommendations (Massachusetts General Hospital COVID-19 Resource Library). Decisions on whether or not to open schools depends significantly on local policy and local epidemiology.

Tool: TH Chan School of Public Health at Harvard University Strategies to Minimize Risk
Tool: CDC Guidance on Risk Reduction and Reopening of Schools.

Tool: Rockefeller Playbook on Testing in Schools

Tool: New York Times visualization on the Impact of Opening Windows

Air TravelCopy Link!

Literature Review: Gallery View, Grid View

The risk of contracting COVID-19 on airplanes is low. 50% of the air circulated in the cabin is brought in from the outside, and the remaining 50% is filtered through HEPA filters. Air enters the cabin from overhead inlets and flows downwards toward floor-level outlets. There is relatively little airflow forward and backward between rows, making it less likely to spread respiratory particles between rows (Pombal et al). To avoid transmission, it is advised to avoid moving up and down the aisles as much as possible, and to wear a mask for the duration of the flight. A laboratory study (not real-world) designed to mimic spread within airplanes indicated that the lack of physical distancing when middle seats were permitted for occupancy may increase transmission, but this model did not account for mask wearing or vaccination (CDC).

Pets and AnimalsCopy Link!

Literature Review: Gallery View, Grid View

While transmission risk from pets is low, the United States Centers for Disease Control now recommend that social distancing rules should apply to pets as well as to humans (CDC). Dogs showed low susceptibility. Pigs, chickens, and ducks were deemed not susceptible according to early data. Evidence of viral replication was noted in inoculated ferrets and cats, with viral transmission occurring between cats (Chen et al). There is no current evidence of transmission to humans from cats and ferrets, though minks can transmit to humans (Meyerowitz et al). Virologist cited in Nature News suggests cat owners should not yet be alarmed, noting deliberate high-dose inoculation of said cats - unrepresentative of day-to-day pet/owner interactions, and that none of the infected cats developed symptoms in the aforementioned study (Mallapaty et al).

SeasonalityCopy Link!

Literature Review: Gallery View, Grid View

Experimental data suggest that the persistence of SARS-CoV-2, either on surfaces or while airborne, is somewhat sensitive to environmental conditions such as temperature, humidity, and ultraviolet radiation. Comparable environmentally-sensitive respiratory viruses often demonstrate seasonality, with greater numbers of infections during winter, and so it seems plausible that SARS-CoV-2 might demonstrate a similar pattern (Carlson et al). However, further studies suggest minimal (≈1%) reductions in SARS-CoV-2 transmission linked to environmental UV radiation (Carleton et al), and the current consensus on such environmental effects is that they are minor in real-world circumstances.

Other respiratory infections such as influenza manifest seasonal oscillations; ‘cold and flu season’ occurs when population susceptibility is high and environmental drivers such as lower temperatures, humidity, and solar radiation conspire to increase transmission, often by changing human behaviors (forcing people indoors). But current levels of immunity to SARS-CoV-2 in most countries are low enough that summer weather is not likely to be protective (Baker et al). If the virus ultimately becomes endemic, it is likely that seasonal oscillations will be observable in temperate regions, with recurrent wintertime outbreaks likely (Kissler et al).

ImmunityCopy Link!

Antibody ResponseCopy Link!

Updated Date: August 30, 2021

Rates of Antibody ResponseCopy Link!

The majority of patients with RT-PCR-confirmed COVID-19 develop antibodies against the virus within 4 weeks, with most studies ranging from 90-99% (Zhao et al; Wang et al, Arkhipova-Jenkins et al). In most patients these are neutralizing antibodies: over 90% of people seropositive for SARS-CoV-2 appear to have detectable neutralizing antibody responses (Wajnberg et al).

Types of Antibodies and SeroconversionCopy Link!

When assessing research studies, details may depend on exactly which antibodies are being assessed. Generally Seroconversion (detection of circulating antibodies) typically occurs 7-14 days after symptom onset (Deeks et al; Huang et al).

  • IgM/IgG or total antibody. Although IgM seroconversion is often thought of as occurring before seroconversion for IgG, this has not been consistently observed for SARS-CoV-2 (e.g., Qu et al; Xiang et al; Wang et al; Zhao et al). A systematic review of 66 studies showed Moderate-strength evidence that IgG levels peak 25 days after symptom onset and are often detectable still at 120 days (many did not do longer followup). IgM levels peak at approximately 20 days and then decline more rapidly. (Arkhipova-Jenkins et al)
  • Receptor Binding Domain Antibody. Antibodies directed against the receptor-binding domain (a component of the spike protein) may appear earlier than antibodies to other antigens (To et al; Okba et al).
  • IgA antibodies are important in mucosal immunity and may play an important role in the response to SARS-CoV-2 (Sterlin et al; Wang et al), but data are currently limited (Deeks et al).
  • Neutralizing Antibodies. Neutralizing antibodies prevent viral replication, usually by binding the spike glycoprotein that SARS-CoV-2 uses to enter cells. Not all antibodies are neutralizing; some bind to the virus but do not stop its activity, such as most of those that bind to the nucleocapsid. Understanding which antibodies are neutralizing is critical for Vaccine Development, Monoclonal Antibody Therapy, studying Convalescent Plasma, and determining whether seropositive individuals are Immune from Reinfection. Neutralization assays are not routinely performed clinically. They require testing the antibodies against their intended target in vitro, and are often reported by the Lethal Dose 50 (LD50) or neutralization titer (titer at which the target is inhibited) to determine if the antibody has low, medium, or high neutralizing ability.

Duration of ImmunityCopy Link!

Updated date: August 23, 2021

The duration of immunity after infection or vaccination is not conclusively known, and not consistent between individuals. Relevant host factors may include immune status, age, and severity of initial infection. Studies documenting decay of IgG antibodies or neutralizing titers may underestimate immunity, since both B and T-cell responses likely also play a significant role, and are not reflected in circulating antibody levels (Karlsson et al). Patients with mild infection lose detectable antibodies more quickly but may have an immune memory that allows them to rapidly produce antibodies on re-exposure (Stephens et al). Because of this, this section discusses the duration of antibodies, the relationship of antibodies to immunity and the duration of immunity each separately.

Duration of AntibodiesCopy Link!

The duration of circulating antibodies in the blood is variable between individuals, and likely different in those with natural vs vaccine-induced immunity. We currently have less than a year of data (8 months for vaccines) and thus do not yet know exactly how long circulating antibodies will be detectable after infection or vaccination.

  • Infection. Circulating neutralizing antibodies differ significantly in different studies, which may reflect differences in selection criteria, such as differing severity of initial disease.
  • In one study of seven month kinetics of antibodies, plasma neutralizing capacity peaked at day 80 after symptom onset and remained stable thereafter up to 250 days. (Ortega et al)
  • In a different study, neutralizing antibody titer approached baseline within a 94 day followup in one study (Seow et al.)
  • Another kinetic study showed neutralising activity above a titre of 1:40 in 50% of convalescent participants as far as 74 days (Wheatley et al)
  • Another study found neutralizing capacity in >70% of patients tested around 6 months (Wu et al)
  • Vaccination. The Moderna vaccination thus seems to show persistent antibodies through 6 month, though the meaning of these antibodies are not known (Doria-Rose et al). Pfizer reports waning antibodies after 6 months (Pfizer)

Correlation of Antibodies with ProtectionCopy Link!

Antibodies may reflect some elements of immunity, but do not necessarily reflect immune response on re-exposure (which is largely determined by T cells and memory B cells). One August 2021 study does indicate that waning antibody titers correspond with decreased protection from disease; In one antibody neutralization study of vaccine recipients, Day 57 reciprocal cID50 neutralization titers were compared with cumulative incidence of COVID for 100 days after the titer was drawn (days 57-100). They found that neutralizing titers of undetectable (<2.42), 100, or 1000 vaccine efficacy was 50.8% (−51.2, 83.0%), 90.7% (86.7, 93.6%), and 96.1% (94.0, 97.8%). Therefore, those with a negative titer still have about 50% protection, but less than those with positive neutralizing antibody titers. (Gilbert et al)

Waning Protection from InfectionCopy Link!

Note: this is a rapidly evolving area, and data may change quickly

The effectiveness of vaccine-based immunity at preventing infection may start to wane around 5-6 months, based on data from four studies in the USA which showed a declines in efficacy from 91.7% to 79.8% (Rosenberg et al), 74.7% to 53.1% (Nanduri et al), 91% to 66% (Fowlkes et al) and 86% to 76% (Moderna) 76%-42% (Pfizer) (Puranik et al) across 5-6 month followups. Slide 14 from this CDC summary shows an excellent graphical representation of these trials. The vaccines studied were the ones available in the USA (Pfizer, Moderna, J&J). The decline in efficacy also may correspond somewhat to the emergence of new viral variants like Delta, however the above CDC analysis suggests it is likely a combination of both the new variant and waning immunity. Similarly, the spike of new infections in Israel in August 2021, which has a very high vaccination rate and vaccinated most of its population around February 2021, may be a sign of waning immunity (Goldberg et al).

However, the waning in immunity appears to apply mostly to mild or moderate disease and not severe disease, hospitalization, or death. This CDC study looked at hospitalizations at 21 medical centers in the USA over 24 weeks and found no decline in vaccine effectiveness against COVID-19 hospitalization regardless of time of vaccination or “high risk” status.

Reinfection and Breakthrough InfectionCopy Link!

Updated Date: August 30, 2021
Literature Review:
Gallery View, Grid View

Reinfection Definition:

Reinfection refers to individuals who have been infected and cleared the original virus, but again show evidence of viral replication after exposure to a new SARS-CoV-2 virus (Falahi et al).

  • Prolonged symptoms from Post-COVID-19 syndrome or relapse/reactivation (also called recrudescence) of symptoms from initial infection
  • Repositivity, or residual shedding of RNA fragments or viral particles (not necessarily infectious; see Infectivity) from the initial infection
  • Breakthrough infection, or infection after vaccination in a patient who has not ever had evidence of viral replication
  • Conclusive demonstration of reinfection is sometimes difficult, since confirmation requires analysis of paired viral whole genome sequences taken during both initial and subsequent infections to be able to conclusively determine that this is a new virus (ECDC Threat Assessment Brief 2020-09-21).

Rates of Reinfection:

Reinfection is uncommon, but appears to be increasing as levels of natural immunity from prior infection wane over time, and new viral variants emerge.

  • Comparison of antibody positive and negative cohorts estimated that antibodies from natural infection conferred ~95% protection in one large study (Abu-Raddad et al) Another large study found a lower estimated protection from natural infection of ~81%, dropping to ~47% in those aged 65 years and older (Hansen et al). Dr Jetelina’s table of vaccine efficacy includes multiple studies on the efficacy of natural infection, including against viral variants.
  • New viral variants appear to cause more cases of reinfection, with the Delta variant having an Odds Ratio of 1.43 for reinfection relative to ancestral strains (Public Health England)

Clinical Course in Reinfection:

  • Symptoms in reinfection tend to be worse than initial infections, especially if the first infection was mild (Cavanaugh et al), however severe disease appears to be less likely (Qureshi et al) There is currently little data about hospitalization and mortality in reinfections.

Rates of Breakthrough Infection:

Given the vaccines are not 100% efficacious, breakthrough infections do occur. The number and severity of breakthrough infections is difficult to definitively track, though it appears rare. Breakthrough infections depend on several things: 1) the person’s immune response to the vaccine (some people are immunocompromised or have lower immune responses to the vaccine, and immunity may wane over time) 2) the variants and their ability to evade vaccination immunity and 3) frequency and nature of exposure to an infected person.

Clinical Course in Breakthrough Infection:

Early data from a study of the 10,262 reported breakthrough cases from January-April 2021 in the USA indicate that breakthrough cases carry about a 10% hospitalization and 2% mortality risk, but a full 27% of recorded cases were asymptomatic. 64% of these breakthrough infections were caused by a variant of concern (MMWR). Preliminary reports suggest that 19% people experiencing breakthrough develop some post-acute COVID (“long COVID”) symptoms (>6wks), which is higher than typical. (Bergwerk et al)

Vaccine vs Natural ImmunityCopy Link!

Updated Date: August 30, 2021

Generally vaccine-based immunity is thought to be more protective than natural immunity. In the case of the Delta variant, it may be about 2 times more protective (Cavanaugh et al). However, this is not universally the case (Gazit et al). For people who have had both the vaccine and natural infection, the natural infection seems to augment immunity similarly to how a booster shot might (Wang et al). For this reason we recommend that people who have had COVID still get fully vaccinated (see vaccination after COVID infection).

The reason for this is that natural infection produces an immune response that is unpredictable relative to vaccination. When infected with the virus, different hosts will develop antibodies to different parts of the virus, whereas with the vaccine antibodies will consistently target the spike protein. Some people with natural immunity will have high neutralizing antibody titers, and others will not. One study found that natural antibodies largely attached to only one region (E484) on the receptor-binding domain, whereas vaccine antibodies attached to many parts of the virus (Greaney et al), meaning that viral mutations may be more likely to escape natural antibodies. Further, the fact that most vaccines are given in 2 doses also likely augments immunity relative to a single infection.

VaccinesCopy Link!

Updated: June 21, 2022
Literature Review:
Gallery View, Grid View
Literature Review: University of Washington
Literature Report (Vaccines and Immunity)

Tool: Vaccine Allocation Planner (helps states and countries plan vaccine allocation)
Tool: COVID Vaccine Development Tracker
Tool: FDA COVID Vaccine FAQs
Tool: NEJM Vaccine Resources
Tool: NEJM Vaccine FAQ

MechanismsCopy Link!

Most vaccines fall into one of 4 categories. The mechanisms of the commonly available vaccines are listed below in efficacy.

  • Genetic Vaccines (typically lipid envelopes carrying SARS-CoV-2 genetic material into cells) including mRNA Vaccines
  • Code for the coronavirus “spike” protein to induce an immune response mediated by antibodies and T cells.
  • Due to the temperature-sensitivity of genetic material (Simmons-Duffin), these vaccines require very cold storage and transportation environments.
  • There is a significant amount of misinformation about mRNA technology:
  • The genetic information contained in these vaccines does not integrate with human DNA or change a recipients’ genetic code, nor are they gene therapy. The vaccines do not use nanorobotic technologies. (Reuters Fact Check)
  • mRNA does not stay in the body forever, in fact mRNA is degraded rapidly in cell cytoplasm within minutes (Chen et al).
  • There is no association with any fertility concerns. There is no effect on sperm paraters (Gonzalez et al) or any female parameters of fertility. The coronavirus’s spike protein and the placental protein syncytin-1 are completely different in structure and the vaccine does not cause immune reactions to syncytin-1 (ASRM)
  • Viral Vector Vaccines (repurposed viruses such as adenovirus carrying SARS-CoV-2 genetic material into cells) including Adenovirus-vector Vaccines.
  • Use a variety of engineered adenoviruses (common viruses that cause colds and related symptoms) as vectors to expose human cells to the SARS-CoV-2 spike. These are generally cheaper than genetic vaccines (Knoll et al), both due to ease of transport as they only require refrigeration to protect the virus vector, as well as due to less costly supply chains than those required for mRNA vaccine technology.
  • Protein-based Vaccines (delivering coronavirus proteins only)
  • Traditional “inactivated/attenuated” Coronavirus Vaccines (whole virus that is killed or weakened)

EfficacyCopy Link!

Updated date: August 30, 2021

Tool: Vaccine Table with Efficacy, including Major Variants (Compiled by Dr. Katelyn Jetelina)

Efficacy of vaccines is complex to assess, as it changes as new viral variants emerge and the immunity provided may wane. Further, often they are lumped together in studies that classify people as either vaccinated or unvaccinated and do not differentiate based on the exact vaccine studied. The above table includes data against major variants.

  1. Preventing hospitalization, critical illness, and death:
  1. All vaccines seem to offer excellent efficacy against hospitalization, critical illness, and death.
  2. A large CDC/MMWR study indicated that vaccination was associated with a 29 fold reduction in risk of hospitalization compared with no vaccine.
  3. AstraZeneca and Johnson & Johnson appear to be 95-100% effective at preventing severe disease and death.
  4. mRNA vaccines (Pfizer and Moderna) have near 100% efficacy at preventing severe disease and death, though case reports of breakthrough, critical illness, and death do occur (US CDC)
  1. Preventing all symptomatic infection:
  1. The following are the efficacies of the most commonly globally-available vaccines at preventing symptomatic infection at the time of local regulatory authorization, which typically meant with the ancestral strain (not more virulent strains like Delta).
  1. Pfizer/BioNTech (mRNA). FDA EUA cited efficacy of 95% at preventing symptomatic infection. (Pfizer EUA). Full FDA approval on August 23, 2021, cited 91% efficacy (FDA).
  2. Moderna (mRNA). FDA EUA cited 94% efficacy against symptomatic infection (Moderna EUA).
  3. Oxford/AstraZeneca (viral vector). The Oxford/AstraZeneca vaccine had an initial efficacy of 90% (Ledford; Knoll et al).
  4. SinoPharm (whole virus inactivated). 79% effective against symptomatic SARS-CoV-2 infection (WHO)
  5. Gam-COVID-Vac aka Sputnik (viral vector). The vaccine is the only vaccine that uses two different serotypes, and it appears to have 91.6% efficacy based on a phase 3 trial (Longunov). It is used in about 70 countries. However, it has yet to gain approval from the EMA or the WHO (as discussed in this Nature article).
  6. Covaxin (whole virus inactivated). 77.8% efficacy against symptomatic disease. The vaccine is approved in 15 countries but has yet to gain approval from the WHO (GAVI).
  7. Janssen/Johnson & Johnson (viral vector). FDA EUA reports an efficacy of 85% against severe disease, and around 70% for symptomatic disease (Janssen EUA).
  1. Preventing asymptomatic infection is incredibly hard to determine, as most studies do not routinely test people without symptoms. However, a few studies indicate effectiveness remains good for most vaccines. Asymptomatic infection appears to be reduced by at least 80% by both of the mRNA vaccines (Tande et al)

Mixing Different VaccinesCopy Link!

Updated date: November 12, 2021

Combining different vaccine types for different shots is an area of active research. On October, 21, 2021, the CDC announced that patients eligible for a booster can choose any of the 3 US COVID vaccines for their booster regardless of what a person received as their primary series

Two major studies have been published on this:

  1. A study of 458 individuals were sorted to get the initial full series of J&J, Moderna, or Pfizer vaccinations followed by a booster of one of the three four to six months later (Atmar et al). This study formed the basis of the ACIP recommendation to allow mixing and matching for booster shots. Notably, this study was performed using a 100ug booster for Moderna, not the 50ug booster that is currently recommended.
  1. The safety profile appears similar to boosting with the same vaccine, and includes mild reactions like fever, fatigue, and cutaneous reactions.
  2. After primary J&J series:
  1. Moderna booster gave 56.1 fold increase in IgG and 76.1 fold increase in neutralizing antibodies.
  2. Pfizer booster gave 32.8x IgG and 35x neutralizing antibody increases
  3. J&J booster gave a 4.2x igG and 4.6x neutralizing antibody increases
  1. After a primary Moderna series:
  1. Moderna booster gave 7.9 fold increase in IgG and 10.2 fold increase in neutralizing antibodies.
  2. Pfizer booster gave 9.7x IgG and 11.5x neutralizing antibody increases
  3. J&J booster gave a 4.7x igG and 6.2x neutralizing antibody increases
  1. After a primary Pfizer series:
  1. Moderna booster gave 17.3 fold increase in IgG and 31.7 fold increase in neutralizing antibodies.
  2. Pfizer booster gave 14.9x IgG and 20.1x neutralizing antibody increases
  3. J&J booster gave a 6.2x igG and 12.5x neutralizing antibody increases
  1. In a UK trial (Com-COV) one dose of AstraZeneca + one dose of Pfizer-BioNTech resulted in higher antibody levels compared with two doses of AstraZeneca, but lower antibody levels compared with 2 doses of Pfizer-BioNTech. (Shaw et al)

Efficacy on New Viral VariantsCopy Link!

Updated date: January 5, 2022

Tool: Vaccine Table with Efficacy, including Major Variants and Natural Infection (Compiled by Dr. Katelyn Jetelina)

Efficacy may change as different viral variants become more predominant, as the antibodies produced by the vaccines may have different neutralizing effects on different strains, especially if the virus mutates the area targeted by the vaccine. However, most vaccines seem to retain at least partial effect against new variants, and most of the time retain excellent protective benefit. Please see this link for a curated chart of the efficacy of six major vaccines or vaccine candidates against major variants, including links to the original literature (compiled by Dr. Katelyn Jetelina). Study estimates for effectiveness against symptomatic disease at the time the Delta variant was most prevalent were around 59% for the Astrazeneca vaccine, 67% for J&J, 66-95% for Moderna, and 39-96% for Pfizer. (Nasreen et al, Sheikh et al, Puranik et al, Pouwels et al, Elliott et al, Fowlkes et al, Sadoff et al, Israel health minister as cited in WSJ). Research on the efficacy of vaccines against the Omicron variant is still emerging but a recent study using serum samples from recipients of the Pfizer-BioNTech vaccine showed that neutralization of Omicron-infected cells was higher in recipients of 3 doses of the vaccine compared to those who had received 2 doses.

neutralization efficiency (by a factor of 100) against the omicron variant after the third dose than after the second dose; however, even with three vaccine doses, neutralization against the omicron variant was lower (by a factor of 4) than that against the delta variant. The durability of the effect of the third dose of vaccine against Covid-19 is yet to be determined.

Booster ShotsCopy Link!

Updated Date: June 21, 2022

Many countries are now recommending booster shots, due to the rise of Delta and Omicron variants as well as due to waning immunity. See Waning Protection from Infection and Breakthrough Infections for summaries of the data driving these decisions. However, even without boosters and even with the emergence of the Delta and Omicron variants, the vaccines remain very highly efficacious at preventing hospitalization and death. This has led the World Health Organization to call for a delay in rolling out booster shots in the name of global equity, as billions of people globally have not yet had the opportunity to have even a first dose (NPR), which would save many more lives.

In places where boosters are being recommended, general guidance includes:

  • Boosters for patients with normal immune systems should be lower priority than assuring patients who have not yet had any vaccines get their initial vaccination series
  • Mixing different vaccine types is permitted, and may be advantageous for some (especially giving mRNA vaccines to those who received viral vector vaccines). See mixing different vaccines.

In the United States, the CDC has made the following recommendations for booster shots:

  • Pfizer BioNTech: single booster for patients ages 5-11 and > 12 years 5 months after primary series has been completed; a second booster is recommended for patients > 50 years or >12 years who are immunocompromised 4 months after the first booster.
  • Moderna: single booster for patients > 18 years 4 months after the primary series has been completed; a second booster is recommended for patients > 50 years or >18 years who are immunocompromised.
  • Janssen: single booster 2 months after primary vaccination

Dose 3 for Immunosuppressed PatientsCopy Link!

In four recent studies, a subset of 33-50% of immunocompromised patients who did not develop an antibody response to the first two doses did develop a measurable antibody response to a third dose (CDC). A randomized control trial with Moderna found immunocompromised patients with a third dose had better protection compared to the placebo (55% vs. 18%) (Hall et al).

  • See here for more information on the timing, effect, and monitoring of vaccines in immunosuppressed patients.
  • Eligibility criteria for boosters varies by country, so please consult your local health department for guidance. In the USA current guidance includes:
  • Moderately to severely immunocompromised individuals. This includes patients with cancer on active or recent chemotherapy, solid or bone marrow transplant, primary immunodeficiencies, HIV with CD4<200, patients on certain immunosuppressive agents, and some other immunocompromised individuals. On January 3, 3022, the FDA approved a third dose of the Pfizer vaccine for children 5-11 years old who are immunocompromised. See full U.S. CDC guidance here.
  • Third dose should be >28 days after the last dose.
  • Serologic confirmation of antibodies is not yet recommended routinely

Boosters for Patients with Normal Immune SystemsCopy Link!

Countries recommending boosters are largely doing so on the basis of evidence of waning antibody levels and a handful of studies showing reduced vaccine effectiveness over time against mild to moderate disease (but not severe disease). See Waning Protection from Infection for this data.

  • Eligibility criteria for boosters varies by country, so please consult your local health department for guidance. In the USA recommendations (see full guidance here) are currently:
  • For those who received an initial Pfizer series, a booster is recommended those who are:
  1. > 5 months from second dose AND 12+ years old
  • For those who received an initial Moderna series, a booster is recommended for those who are:
  1. > 6 months from second dose AND 18+ years old
  • For those who received an initial J&J vaccine, a booster is recommended >2 months after the initial dose AND 18+ years old
  • Note, Moderna boosters are approved for a 50mcg dose, different from the 100mcg initial series dose.

Adverse Events and ReactogenicityCopy Link!

Most observed adverse events during vaccine trials were injection-related or reflected an expected immune response. Many people feel ill following vaccine administration for about 1-3 days, especially after the second dose of the vaccine This is not a sign of infection by the coronavirus.

ContraindicationsCopy Link!

The U.S. CDC considers the following contraindications: severe allergy (e.g. anaphylaxis) to a prior dose of an mRNA COVID vaccine or any of its components, immediate allergic reaction of any severity to a previous dose or any of its components (including PEG), immediate allergic reaction of any severity to polysorbate. (CDC) Reactions to non-COVID vaccines are considered a “precaution” but not a contraindication.

Routine VaccinationsCopy Link!

The CDC now states that COVID-19 vaccines and other vaccines may now be administered without regard to timing. This includes simultaneous administration of COVID-19 vaccine and other vaccines on the same day, as well as co-administration within 14 days. Other public health guidance may vary.

Vaccination Associated Cutaneous ReactionsCopy Link!

Updated Date: October 1, 2021

Cutaneous reactions to vaccination are common with COVID vaccines, as well as other vaccinations. A large red, itchy, painful, and swollen rash at the site of injection is sometimes called COVID-arm, and is a relatively common symptom of vaccination (about 1%, but variable depending on the type of vaccine). It typically occurs about a week after injection (range, 5-10 days). People with these reactions can still receive second and booster doses as they do not lead to serious sequelae (Jacobson et al). Further, many patients who had COVID-arm with a first shot will not have it on the second shot (Blumenthal et al). Pain can be managed with over the counter medications where appropriate.

Other cutaneous reactions can also occur: Amongst 405 cases of cutaneous reactions in one cross-sectional Spanish study (Català et al) of people vaccinated with Pfizer-BioNTech (40.2%), Moderna (36.3%) and AstraZeneca (23.5%), the most common cutaneous reactions were: injection-site (COVID-arm, 32.1%), urticaria (14.6%), morbilliform (8.9%), papulovesicular (6.4%), pityriasis rosea-like (4.9%) and purpuric (4%) reactions.

Vaccine-Induced Immune Thrombotic ThrombocytopeniaCopy Link!

Updated date: May 9, 2021

There have been reports of rare (tens of cases globally) of venous thrombotic disease -- and particularly cerebral venous sinus thrombosis -- in recipients of the widely deployed Oxford/AstraZeneca and Janssen/Johnson & Johnson adenovirus vector vaccines. For both vaccines, the frequency of these events appears to be far lower than the risk of severe thromboembolic complications of COVID-19 itself. As of April 15, 2021, the benefits of both Oxford/AstraZeneca and Janssen/Johnson & Johnson vaccines are thought to outweigh potential risks. From Cines et al:

  • Most of the patients are women under 50 years of age, some of whom were on estrogen-based medications.
  • Thromboses often occur at unusual sites, such as cerebral venous sinus thrombosis (CVST) or in the portal, splanchnic, or hepatic veins. Cerebral (also “central” or “dural”) venous sinus thrombosis (CVST) refers to a blood clot in the veins that drain blood flow from the brain. Obstruction of outflowing blood can lead to increased intracranial pressure and, depending on the anatomy of the clot, focal neurological symptoms that are a type of stroke.
  • At the time of diagnosis, may patients have low platelets: median platelet counts (median, 20,000 to 30,000). High levels of d-dimers and low levels of fibrinogen are common.
  • Although the mechanism of this clotting dysfunction is not certain, it appears to be a vaccine cross-reaction that causes an auto-immune thrombocytopenia.
  • If you suspect a patient has CVST or other unusual clot due to vaccination, many guidance institutions currently recommend treating that patient similarly to how you would treat a patient with heparin-induced autoimmune thrombocytopenia (HIT).
  • This generally involves (where available):
  • Close monitoring of blood counts including platelets, sending anti PF-4/heparin antibodies and serotonin release assay or heparin-induced platelet aggregation assay.
  • These patients should be treated with non-heparin containing anticoagulants such as Direct thrombin inhibitors (Argatroban, Bivalirudin, Lepirudin) or indirect FXa inhibitors (Danaparoid, fondaparinux).

MyocarditisCopy Link!

Updated Date: October 24, 2021

Receiving the COVID vaccine activates immune activity, which can cause myocarditis in a small subset of patients, particularly adolescent males. However, getting infected with COVID also causes a 16x risk of developing myocarditis (MMWR), among other risks.

  • The benefits of vaccination (specifically preventing ICU admission and death) outweigh risks in both girls and boys age 12-17 according to US CDC guidance (CDC update August 2021). See this link for a graphical representation of the CDC’s estimates of risks of myocarditis compared with COVID cases/hospitalizations/deaths for both boys and girls (as of June, 2021). (Mostly mRNA vaccines but also J&J vaccine)
  • The UK Joint Committee on Vaccination and Immunization also determined that the benefit of vaccination outweighs the risks in adolescent males, but their statement describes a more marginal difference. The reported risk of myocarditis in the UK is 3 to 17 per million for the first dose; and 12 to 34 per million for the second dose (Astrazeneca vaccine).
  • Exact data on the risks of myocarditis differentiated by vaccine type is not yet available. This is an evolving area of research.
  • Risk of myocarditis with boosters is actively being studied. Israel has administered 3.7 million boosters and to date their incidence of myocarditis with boosters is lower than with the initial series (presumably as there has been longer between doses).

Symptoms of myocarditis emerged on average 4 days after vaccination. Recovery is hoped to follow a similar course to post-MISC myocarditis patients, who tend to recover within 6 months. One study showed that 86% recovery within 35 days (the length of followup that was published) (Jain et al).

Ability to Transmit to OthersCopy Link!

It is still possible to transmit the virus to others even if vaccinated. This is especially true for highly infectious variants like Delta. It is still true even if asymptomatic. Please see Transmission.

Special PopulationsCopy Link!

Prior Infection or Antibody TherapiesCopy Link!

People who have been previously infected and/or received antibody therapies (monoclonal antibodies, convalescent plasma) can receive the vaccine. Vaccination after infection is covered here.

ObstetricsCopy Link!

Updated Date: August 23, 2021

American College of Obstetricians and Gynecologists (ACOG) and the Society for Maternal-Fetal Medicine (SMFM) strongly recommend vaccines for pregnant people. (ACOG) Pregnant women and their doctors may discuss the risks and benefits depending on the individual’s risk of acquiring COVID. Side effects, such as fever, can sometimes cause adverse pregnancy outcomes, but can be treated.

Safety:

  • Vaccines are not associated with infertility or pregnancy loss
  • In one study of 3,958 pregnant people there were no unexpected outcomes related to COVID-19 vaccination, regardless of trimester. Of the 827 people who completed pregnancy, pregnancy loss, preterm birth, babies size, congenital problems, and death were the same as background rate (Shimabukuro et al). Miscarriage rates are also the same as background rates. (Zauche et al)
  • Preliminary findings of the US Vaccine Adverse Events Reporting System (VAERS) find no specific safety concerns to the mRNA vaccines in pregnant and lactating women (Shimabukuro et al).

Efficacy: mRNA vaccines appear to produce a robust humoral immunity in pregnant and lactating women, similar to non-pregnant women, and far greater than the antibody response seen with natural infection. The antibodies appear to transfer to neonates via placenta and breast milk (Gray et al).

PediatricsCopy Link!

Updated Date: June 21, 2022

The Pfizer COVID vaccine received FDA authorization on August 23, 2021 for 16+ in the United States. In addition, the Pfizer vaccine has received Emergency Use Authorization for children ages 6 months - 4 years (3 micrograms) 5-11 (10 micrograms) as well as children ages 12-15 (30 micrograms, same dose as approved 16+ dose). All primary series are 2 doses administered 3 weeks apart. A third primary dose is recommended for patients >6 months who are immunocompromised.

The Moderna COVID vaccine has received Emergency Use Authorization for children ages 6 months - 5 years (0.2mL), 6-11 years (0.5 mL) and >12 years (0.5mL). All primary series are 2 doses 1 month apart. A third primary dose is recommended for patients >6 months who are immunocompromised.

Janssen (J&J) remains restricted to people over 18 years.

In a study of 2260 adolescents aged 12-15, the Pfizer vaccine demonstrated 100% efficacy (Pfizer). The vaccine was well-tolerated in this study, with side effects similar to those seen in people age 16-25.

  • A post-market v-safe study of >129,000 vaccinated adolescents eight months after vaccination indicated that the vaccine was very well tolerated. Amongst 8.9 million adolescents, VAERS reports were received for only one per 1,000 vaccines, and 90% were for non serious conditions. (MMWR)
  • As described in pediatrics, some immunity for neonates via breast milk likely occurs

Immunosuppressed PatientsCopy Link!

Updated date: May 26, 2021

Immunosuppressed people should get vaccinated against SARS-CoV-2, as all currently approved vaccines do not include live virus. However, vaccination may not be as efficacious as in those who are not immunosuppressed. Guidelines as to timing of vaccination and holding of certain immunosuppressive medications vary among expert panels in different specialties.

  • Vaccine Efficacy. Vaccine efficacy varies highly dependent on the type of immunosuppression, the type of vaccine, and local variant epidemiology.
  • In a study of 658 solid organ transplant recipients who received both doses of either mRNA vaccine, a month after the second dose 54% of the total cohort and 43% of those taking anti-metabolites (p<.001 for the difference in response rate) had detectable anti-spike antibodies (Boyarsky et al).
  • In a prospective study of 133 patients with chronic inflammatory diseases (rheumatologic, IBD, and neuroautoimmune—all but 9 on DMARDs or biologics), compared to 53 healthy controls, after receiving the two-dose series of either mRNA vaccine, most of those with inflammatory diseases developed a robust immune response, but with an overall 3-fold decrease in humoral response compared to the controls (p=.009). Prednisone reduced the humoral response 10-fold, with only 65% seropositivity after the second dose and no clear dose-response relationship, while B-cell depleting agents reduced the humoral response 36-fold. Antimetabolites including methotrexate reduced humoral response 2-3 fold, and JAK inhibitors showed a statistically significant reduction in antibody titers. Other therapies did not have strong impacts on humoral response, with most of these patients taking hydroxychloroquine and/or TNF inhibitors (Deepak et al).
  • In a study of IBD patients in which most were on TNF inhibitors or vedolizumab, of the 15 who were studied after receiving both doses of either mRNA vaccine, all seroconverted with robust titers (Wong et al. 2021).
  • In a study of 67 patients with hematologic malignancies, 30 of whom were receiving active therapy, 46.3% had developed no anti-spike antibody 16-31 days after the second dose of an mRNA vaccine. There was a non-statistically-significant trend toward worse response among those on active therapy, and a statistically significant worse response among those with CLL compared to other malignancies (76.9% non-response versus 38.9% for the rest of the cohort.) (Agha et al).
  • Patient Counseling
  • All patients on immunosuppression should be counseled that they potentially remain at elevated risk for SARS-CoV-2 infection compared to the rest of the vaccinated population. This is particularly true for those on glucocorticoids at any dose and/or on B-cell-depleting agents.
  • In terms of behavioral practices and masking, patients should behave as though they are unvaccinated.
  • Vaccine Timing and Immunosuppression Adjustment
  • No modifications for most drugs are suggested at present, though if starting new immunosuppression, vaccination should be completed at least two weeks prior to initiation if possible. The International Organization for the Study of Inflammatory Bowel Disease, as well as the National Psoriasis Foundation, suggest immediate vaccination for all patients currently on immunosuppression, with no alterations in timing and no holding of immunosuppressive medications (Siegel et al; National Psoriasis Foundation 2021). The American College of Rheumatology differs in opinion and makes the suggestions below:
  • For anti-CD-20 monoclonals (e.g. rituximab and ocrelizumab), vaccination should occur at the end of the dosing interval, with the second dose for 2-dose vaccines occurring at least 2-4 wks before the next infusion if possible (ACR guidelines).
  • Hold treatment for 1 week after each vaccine dose for methotrexate, cyclophosphamide, and JAK inhibitors (ACR guidelines, Feb 2021).
  • Hold subcutaneous abatacept for one week before and one week after the first vaccine dose only (ACR guidelines).
  • For intravenous abatacept, time COVID vaccination so the first shot occurs 4 weeks after infusion, with the next infusion delayed a week after the shot (ACR guidelines).
  • For bone marrow transplant, most institutions are recommending vaccination between 3 months and 12 months after transplant (expert practice).
  • Post-vaccination testing
  • Antibody testing is not necessarily a reliable indicator for predicting if there has been an immune response, because some antibody tests do not test for antibodies produced by vaccination, and because immune benefit from T cell responses is possible without having circulating antibodies.
  • Currently, recommendations do not support testing immune response after vaccination.
  • That said, in rare instances, the specialist managing the patient’s immunosuppression may opt to send a quantitative anti-spike antibody, which must be interpreted cautiously. Preliminary data (personal communication) support a strong correlation between B-cell and T-cell responses.

Tool: ACR COVID Vaccine Clinical Guidance Summary (gives recommendations for multiple clinical scenarios)

Autoimmune Conditions and History of Guillain-BarreCopy Link!

Insufficient data is available on these populations, though people with autoimmune conditions were included in trials and did not seem to have increased symptoms. To date no cases of Guillain Barre have been found with the mRNA vaccines, and it is not a contraindication to vaccination. Very rare cases have been reported in viral vector vaccines (one in the USA as of April 23, 2021).

Vaccine EquityCopy Link!

While approval of the first vaccine marked the culmination of a tremendous scientific effort, the fight against COVID-19 now faces a new challenge: a massive worldwide vaccination campaign. The same embedded structural forces driving inequities in the burden of COVID-19 must also be considered within the context of vaccine access and distribution.

Vaccine Prioritization: It is essential that COVID-19 vaccines be distributed equitably. People who should be prioritized for vaccination include (adapted from the National Academies of Sciences, 2020).

  • High-risk of COVID-related Morbidity and Mortality
  • Medical Comorbidities
  • Over the age of 65
  • High-risk of Contracting COVID-19
  • Residents of Long-term Care Facilities and Group Homes
  • Incarcerated
  • Undomiciled
  • First-responders
  • Healthcare Workers
  • Front Line Workers (e.g. Supermarkets, Factories, Schools, Agriculture, and Meat-processing Plants)

Due to generations of structural racism and socioeconomic inequalities, people of color, people with disabilities, immigrants and migrants, indigenous peoples, and the poor are all disproportionately represented in many of these groups.

Global Distribution: The distribution of vaccines among nations should follow similar principles. No country should have enough vaccines to vaccinate their entire population before another country has enough to vaccinate their high-risk populations. As of December, 2020 there is significant imbalance: Canada has ordered enough vaccines to inoculate six-times its population, the United Kingdom and the United States four-times their populations, and the European Union twice its population (New York Times).

COVAX, a global coalition including the WHO to assure vaccination, has proposed that all countries receive an adequate supply to inoculate at least 20% of their population before any nation receives additional vaccines. This will ensure that high-risk groups are vaccinated regardless of where they live. Following this initial roll-out, vaccines should be distributed based on the vulnerability of the country’s health system and the impact of COVID-19 on the country, prioritizing countries most in need (COVAX, 2020).

Vaccine hesitancy: In countries like the US, vaccine hesitancy and distrust of the medical system may further exacerbate inequity (Warren et al). This is shaped by the legacy of exploitation and oppression of marginalized groups in the name of science (for example, the Tuskegee Experiment). Meaningful community engagement and promotion of informed decision-making requires an acknowledgment that these historical and contemporary forces contribute to a rational distrust of the health system among marginalized communities (Burgess et al).

Herd ImmunityCopy Link!

Updated Date: January 24, 2021

Literature Review: Gallery View, Grid View


The minimum or critical level of population immunity - acquired either through immunization or previous infection and subsequent recovery - that is required to stymie transmission of a particular communicable disease is colloquially referred to as ‘herd immunity’. When ‘herd immunity’ is achieved, susceptible individuals are indirectly protected from infection because sufficient numbers of immune individuals serve to prevent the circulation of the pathogen to immunologically-naive individuals. The percentage of immune individuals required to achieve ‘herd immunity’ against a particular pathogen varies dramatically depending on factors such as the baseline reproduction rate of the pathogen (R0), the effective reproductive number for a given population (Rt) - which is itself influenced by the efficacy of (and societal adherence to) non-pharmaceutical interventions, population density, therapeutics, immunological factors like the length of immunity, etc.

Current estimates suggest that achieving ‘herd immunity’ against SARS-CoV-2 will not be possible without an absolute minimum of 50% of population immunity (Fonanet et al), and as high as 85% in countries with higher Rt values (On Kwok et al). Because of the significant case fatality rate of COVID, and the ancillary consequences of unnecessary cases and deaths, the WHO recommends that ‘herd immunity’ against SARS-CoV-2 be achieved through immunization campaigns and not by needlessly exposing populations to the pathogen.

Health EquityCopy Link!

What is Health Equity?Copy Link!

Updated Date: December 17, 2020

Equity focuses on the fair and just treatment of all people. By extension, addressing inequities involves eliminating avoidable, unfair or changeable differences among groups, whether these are defined socially, economically, demographically, or otherwise. Upholding equity in health allows prioritization of fair opportunities for everyone to attain their full health potential (WHO Health Systems: Equity).

The COVID-19 pandemic has disproportionately affected historically oppressed populations around the world. Due to long-standing structural inequities, people from these communities are: 1) more likely to be exposed to disease, working essential jobs and living in crowded conditions; 2) less likely have to have access to quality healthcare, including COVID-19 testing and treatment; and 3) more likely to suffer from preexisting health conditions, as a result of adverse social determinants of health, putting them at increased risk of complications and death (Warren et al).

Not all of these can be included here, but we will address several major concerns. Inclusive data collection, while important, needs to be followed by evidence-driven steps to create an inclusive pandemic response and to be the foundation for equitable public health emergency planning (Reed et al).

Providers should screen for and Address Social Determinants of Health (SDOH): SDOH are the conditions under which people are born, grow, live, work, and age (AAFP's The EveryONE Project) which act to shape the health and well-being of people in complex ways. In the context of COVID-19, living situations coupled with job insecurity increase the risk of infection and then make safe isolation and quarantine difficult. In some neighborhoods in the United States, as many as 70% of positive cases required social support to safely isolate and quarantine (Kerkhoff et al).

Resource InequityCopy Link!

Updated Date: January 20, 2021

Literature Review: Gallery View, Grid View

Wealth inequality functions as both a cause and effect of health inequity. The global imbalance of wealth among and within nations is the result of historical (and current) forces including colonialism, racism, structural readjustment, and extractive capitalism. This has left many countries with chronically underfunded health systems lacking in infrastructure, equipment, and adequate staffing.

Historically, in the face of these challenges, containment measures are often emphasized over provision of treatment and supportive care. As was seen in the Ebola epidemic, this strategy backfires by ignoring the human toll of weak treatment systems, and downplaying the impact that effective treatment has on containment: When treatment and supportive care are not available or are not high quality, it undermines confidence in public health institutions and messaging; people understandably avoid seeking care when they need it, and may not trust public education campaigns encouraging social distancing, isolation, and other precautionary measures (Farmer).

It should be noted, despite facing significant barriers to containment and treatment, a number of low- and middle-income countries have prevented COVID-19 cases and fatalities from reaching the astronomical levels seen in many wealthier nations.

Economic ConsequencesCopy Link!

The COVID pandemic has led to a global income drop for workers and exacerbated existing health gaps between rich and poor countries (AP News). Disruptions to food supplies and economies risk worsening malnutrition worldwide, and will be a severe setback to the effort to achieve the United Nations Sustainable Development Goals (Ekwebelem et al). Additionally, the pandemic will leave fragile health systems with a legacy of death and attrition in the workforce and shrinking budgets driven by unstable financial outlooks.

Racial DisparitiesCopy Link!

Updated Date: December 17, 2020
Literature Review:
Gallery View, Grid View

In the United States and United Kingdom multiple sources have demonstrated that Black and Latinx populations are disproportionately likely to be infected and/or die from COVID-19 (Garg; NYSDOH Fatalities, NYC DOH). A systematic review and meta-analysis of over 18 million patients across 50 studies from these two countries found higher COVID infection rates within Black, Latinx, and Asian communities (Sze et al). As of late November, 2020 Black and Latinx Americans have had 1.57 and 1.69-fold, respectively, as many cases as white Americans. Black deaths have been at 2.05-fold the rate of white Americans, and Latinx at 1.38-fold the white case fatality rate (Covidtracking). In the United States, rates of hospitalization among Black and Latinx COVID patients are approximately 4.7 times than among non-Hispanic white patients (Mayo Clinic; Pan et al). In terms of years of potential life lost before age 65, Black Americans are 6.7 times higher, Latinx people 5.4 times higher, Indigenous populations 4.0 times higher, and Asians 2.6 times higher compared to whites (Bassett Working Paper).

Tool: Race Statistics

Systemic health inequities affecting minority racial groups cause increased risk in the following categories:

  1. Exposure risk at work: more likely to work in healthcare, education, retail and other jobs that can’t be done from home. In the United States, Latinxs make up 21% of the essential workforce (Economic Policy Institute), but only 18% of the total population (Pew Research). In the United States, 30% of licensed practical and vocational nurses are Black. Close to 25% of the Black workforce in the United States is employed by the service industry (Mayo Clinic).
  2. Exposure risk on public transit: more likely to rely on public transport to attend work (Pew Research).
  3. Exposure risk in shared living spaces: more likely to cohabitate with others (Census).
  4. Comorbid health conditions: Black people in the United States have a significantly elevated risk of hypertension, which is well-documented, and hypertension control rates are significantly poorer in Black, Latinx, and Asian adults (with acknowledgment of heterogeneity between communities included in population groups) (Saeed et al).
  5. Access to healthcare and testing: income inequality, lower rates of health insurance, and being situated farther from health centers make it harder for many minority groups to access care.
  6. Racism in healthcare delivery: many minority patients experience consciously- and subconsciously-biased health systems and providers when they seek care. Inequitable representation among healthcare leadership and those responsible for healthcare messaging efforts may contribute to reticence from individuals and communities of color. While it is not the sole responsibility of people of color to rectify this, diversifying the types of speakers sharing public health messages may encourage communities of color to more confidently adopt evidence-based public health recommendations (Cooper et al).
  7. Chronic stress: stress and allostatic load can affect immune function.
  8. Environmental factors: risk for severe COVID has been associated with poorer air quality (Wu et al; Pozzer et al).

Indigenous CommunitiesCopy Link!

Indiginous communities are particularly affected by COVID-19. The cumulative incidence of COVID-19 among American Indian and Alaska Native persons is 3.5 times that among non-Hispanic white persons (CDC) Rates of infection often significantly exceed those in major metropolitan outbreaks (like New York City in April, 2020). As of July, 2020 in New Mexico, American Indians represented 53% of COVID deaths but only 11% of the population (Sequist et al).

Immigrants and MigrantsCopy Link!

Updated Date: December 17, 2020
Literature Review:
Gallery View, Grid View

Global migration patterns have shifted during the pandemic, decreasing in some areas but increasing in others. Job losses have resulted in a trend of workers attempting to return to countries of origin, and border closures have resulted in ~3 million people left stranded during their return journeys as of October 2020. The pandemic’s longer-term impacts on poverty and food security have yet to be revealed, but it is anticipated that migration of necessity may increase (WFP).

Noncitizens disproportionately experience the health and financial effects of pandemic, but this is often overlooked as national statistics do not always include non-citizens (for example, data is not currently collected for analysis by the USA CDC). Structural factors that shape daily life for noncitizens place these groups at greater risk of becoming infected with COVID (Langellier et al).

  • Compared with citizens, noncitizens are more likely to live in larger multi-family households where bedrooms may be shared.
  • Non-citizens are also more likely to perform work that cannot be done remotely and depend on public transit.
  • Non-citizens are not currently eligible for public financial and food assistance programs such as Social Security, TANF, and SNAP. Paradoxically, eligible documented immigrants who receive support from these public assistance programs are ineligible for citizenship based on the “public charge” test.
  • Immigration and Customs Enforcement (ICE) has detained over 50,000 undocumented immigrants in holding facilities in the United States. Detainees in such facilities are subject to all of the same infection risks as prison inmates (see People who are Incarcerated), but may be more prone to poor outcomes since ICE’s operational COVID-19 containment protocols do not consistently reflect evolving CDC recommendations (Openshaw et al; Meyer et al; Keller et al).
  • International Medical Graduates (IMGs) make up roughly 25% of the specialist workforce in America but many are serving on H-1B (temporary employment) visas that disqualify them from disability benefits if they were to get COVID at work. This also exposes family members to forcible relocation in the event of their deaths (Tiwari et al).
  • Immigrants are also at risk of being systematically overlooked or underserved in public vaccination campaigns (Foppiano et al).

Suggested policy interventions to improve health among non-citizen populations during pandemic include:

  • Elimination of citizenship barriers to public assistance programs and elimination of public assistance participation as a barrier to establishing citizenship (Langellier).
  • Ensure access to essential resources, such as food, medicine, and legal services (WFP).

Language remains one of the major barriers to quality care. Patients who cannot speak English in the United States are more likely to receive inadequate care (Ross et al). Here are strategies for communication with people with limited proficiency in the language of care providers, shared by the MGH Disparities Solutions Center:

  • Create screening and educational materials based on the languages spoken in your population.
  • Use interpreting services and tools whenever available (in-person interpreters, bilingual phones, and/or mobile screens such as iPads).
  • Use staff hotlines with people who are multilingual.
  • Target communication updates in multiple languages and through multiple platforms (posters, email, website, text messaging, etc.)
  • Create a registry with clinical staff who are multilingual and deploy them to applicable patient care sites.

People Who Are IncarceratedCopy Link!

Updated Date: November, 2020
Literature Review:
Gallery View, Grid View

People who are imprisoned are particularly vulnerable to COVID-19 infection due to overcrowding, poor ventilation, poor sanitation, lack of medical care, violence, and increased rates of chronic medical conditions (U.S. Department of Justice Special Report). Early data from the COVID-19 pandemic demonstrated up to 5 times higher rates of death among incarcerated people, despite disproportionately younger age distributions relative to nearby communities (Saloner et al). Since the start of the pandemic across all states, incarcerated persons have >3 times the per-capita number of cases as the general population (The Marshall Project). Dormitory housing has been shown to be a strong risk factor for infection (Kennedy et al).

Decarceration (release from prison) remains the most evidence-based intervention to reduce infection among incarcerated people, and by extension, the local communities that prison workers belong to (Hawks et al; Barnert et al; Okano et al). In place of full decarceration, compassionate release of low-risk offenders and elimination of cash bails that contribute to growing prison populations can also prevent infections (Nowotny et al).

When isolation and containment strategies are used in prisons, additional interventions should be supported to address the mental health burden they create for incarcerated people, especially those living with chronic mental illness (Hewson et al).

  • If available, some safe ways to support incarcerated people include waiving in-state licensure requirements for telemedicine and expanding access to virtual family visits through videoconferencing (Robinson et al).

Other solutions include mass testing of incarcerated people and prison workers, providing personal protective equipment (PPE), and improving sanitation (Akiyama et al). It is particularly critical to focus efforts on occupational health interventions that can prevent transmission of infection to nearby communities (Sears et al).

People with DisabilitiesCopy Link!

Updated Date: November, 2020
Literature Review:
Gallery View, Grid View

People with disabilities may be disproportionately marginalized by COVID-19 response efforts due to inadequate recognition of their unique needs. People with disabilities may not have equitable access to safe living situations or healthcare resources. Some disabilities do not affect severity or prognosis with COVID infection, but some disabilities may (generally due to related comorbidities such as structural heart disease). For example, if infected, individuals with Down Syndrome are five times more likely to be hospitalized and 10 times more likely to die (Wadman M).

Policies and institutional guidance should consider the needs of disabled patients.

  • Alternative support structures should be considered for patients with disabilities who are unable to participate in standard public health protocols, such as home-based COVID testing for people with autism spectrum disorder (Eshraghi et al).
  • Health policy leaders must be attentive to inequities in access to care and resources, disproportionate hardships imposed by pandemic mitigation strategies, and increased risk of harm from COVID infection in the context of pre-existing health disparities (Armitage et al).
  • Creation of equitable resource allocation protocols, especially when considering Crisis Standards of Care, should be guided by near-term survival calculations and objective measures to avoid bias against people with physical and intellectual disabilities in allocating resources (Solomon et al).

People with disabilities and their caregivers should be engaged in all stages of the outbreak response, from initial planning to implementation to assessment. During the pandemic, some strategies for providers to help patients with disabilities include:

  • If caregivers need to be moved into quarantine, plans should be made to ensure continued support for people with disabilities who need care and support.
  • Consider exceptions to Visitor Policies for patients who need support from caregivers in order to participate in care.
  • Messages should be shared in understandable ways to people with intellectual, cognitive, and psychosocial disabilities.
  • When available, masks with clear impermeable windows can improve communication for those who are deaf of hard of hearing.
  • Non-written communication (audio recordings, imaging, verbal communication) and instruction may be particularly important for this group.
  • Photographs of clinical care team members without their masks can relieve anxiety.
  • Community-based organizations and leaders in the community can be useful partners in communicating and providing MHPSS support for people with disabilities who have been separated from their families and caregivers.
  • Trauma-informed care can help build trust (CDC guide).
  • People who cannot remove a mask independently, avoid touching masks frequently, avoid excessive licking or saliva on masks, or otherwise tolerate wearing a mask should be excused from wearing one under CDC recommendations.

Tool: COVID-19 response: Considerations for Children and Adults with Disabilities, UNICEF

Tool: COVID-19 and persons with psychosocial disabilities, Pan African Network of Persons with Psychosocial Disabilities, et al

People without Secure HousingCopy Link!

Updated Date: November, 2020
Literature Review:
Gallery View, Grid View

Undomiciled (homeless) people under 65 years of age have all-cause mortality rates 5-10 higher than the general population at baseline (Baggett et al). Living conditions, higher rates of comorbidities (including substance abuse and mental illness), limited medical services, and the difficulty for public health agencies in tracing undomiciled individuals are all likely challenges during the pandemic (Tsai et al).

People Living in Congregate HousingCopy Link!

Updated Date: November, 2020
Literature Review:
Gallery View, Grid View

Skilled nursing facilities, nursing homes, and other congregate living settings often struggle with social distancing and house populations with significant medical risk factors for poor outcomes (McMichael et al).

  • In the United States, as of April 23, “there have been over 10,000 reported deaths due to COVID-19 in long-term care facilities (including residents and staff), representing 27% of deaths due to COVID-19 in those states (Kaiser Family Foundation).
  • COVID has impacted long-term care facilities around the world, with data from many countries showing 40% of COVID deaths to be connected to long-term care facilities. Rates in some higher-income countries are 80% (WHO).

Tool: Rates in Long Term Care Facilities (USA only, third chart) (Kaiser Family Foundation )

People with Substance Use Disorders (SUDS)Copy Link!

Updated Date: November, 2020
Literature Review:
Gallery View, Grid View

People with SUD are disproportionately likely to become sick with COVID and are more likely to experience severe illness and death. A team of researchers in the United States analyzing electronic health records found that 15.6% of the COVID cases were people with SUD, but people with SUD represented only 10.3% of the study population. Effects were strongest for those with opioid use disorder.

Possible explanations include higher rates of comorbid pulmonary and cardiac pathologies in people with SUD, as well as disparities in access to healthcare associated with stigma and marginalization. Black Americans with a recent diagnosis of opioid use disorder were four times more likely to become sick with COVID-19 than white peers (Wang et al).

Please see Alcohol Use Disorders and Opiate Use Disorders.

Tool: Harm Reduction Strategies For people who use substances during the COVID-19 pandemic (Harm Reduction Coalition, English/US Focus)

Intimate Partner Violence (IPV)Copy Link!

Literature Review: Gallery View, Grid View

The World Health Organization has long identified violence against women as a globally pervasive and urgent public health problem (WHO, 2013). Nearly one third of women around the world report having experienced physical and/or sexual violence perpetrated by an intimate partner (WHO). Patterns of comparable violence against men are not as well understood (Kolbe et al) but are also recognized as complex public health problem (CDC). Intimate partner violence can and does occur across all socioeconomic settings, but prevalence is affected by social determinants of health, such as economic stability, housing security, social support, and childcare access (Evans et al). It is also important to acknowledge that gender inequality is associated with IPV (McCloskey et al).

Economic dependence is a particularly salient risk factor for IPV. Job losses during the pandemic have exacerbated the economic vulnerability of women, immigrants, and workers with lower levels of education. The pandemic has also restricted the movements of people seeking alternate housing to escape IPV, and is likely affecting access to common reporting venues such as primary care delivery sites and police precincts (Evans et al). Additionally, job insecurity and economic stress are associated with a cycle of increased alcohol consumption, smoking and drug abuse (Compton et al); which in turn increases the risk of IPV (Lee et al).

Current impact data are limited, but one study comparing rates of physical IPV during the COVID-19 pandemic to rates of physical IPV during the preceding three years indicated a 1.8-fold increase in incidents, accompanied by a higher rate of severe injuries and a lower rate of reporting (Gosangi et al).

In the context of the COVID-19 pandemic, it is important to support programs that prevent IPV. Social support, cash transfer, food distribution, housing, availability and accessibility to health care, and health insurance coverage are critical to mitigating the impact of COVID-19 and preventing increasing IPV.

Tool: Identifying & Mitigating Gender-based Violence Risks within the COVID-19 Response, UNICEF, IASC

Chapter 2

COVID Testing

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

Whom to TestCopy Link!

Tool: PRIoritize_Dx, intended to help policy-makers allocate tests.
Tool: PATH COVID-19 Diagnostics Dashboard to support product selection and procurement decisions.

Tool: The Foundation for Innovative New Diagnostics (FIND), a global non-profit, conducted Independent Evaluations of test kits between April and August of 2020.

Testing Symptomatic PatientsCopy Link!

Updated Date: January 7, 2022

Prioritize testing people with symptoms suggesting acute infection (see Screening Questions and Common Symptoms).

  • The standard of care for diagnostic testing symptomatic patients is PCR-based testing, as it has the best sensitivity and specificity.
  • If testing is very limited, prioritize testing patients when it will specifically change management or isolation status/location.
  • When PCR-based testing capacity is restricted, use of the antigen test can increase testing capacity as well as offer advantages in terms of more low-cost testing with short turnaround time. The antigen test is discussed in more detail in Types of Test Section.
  • Exact testing algorithms will depend on each institution and the availability and type of testing. One potential algorithm is presented below based on whether same day testing is available or not. Same day testing has advantages.

Algorithm for Symptomatic Patients Based on Whether Same Day Testing is AvailableCopy Link!

Details for each path are discussed below:

This diagram outlines in flow-chart format content that is covered in the list below.

  1. If same-day testing is available:
  1. Fast turnaround (same-day) Nucleic Acid Amplification Test (PCR)
  1. If positive, the patient is “confirmed COVID.”
  2. If negative, categorize according to Case Definitions and clinical suspicion.
  1. If discharging home, Isolate at home and initiate contact tracing.
  2. If admitting, repeat testing in 12-24 hours.
  1. If negative on the second test:
  1. Consider alternative etiologies (influenza, malaria, other infections), and discontinue the “suspected” or “probable” Case Definition if one is found.
  2. Consider alternative testing (e.g. serology) or repeat testing (typically 72 hours from first) if clinical suspicion remains high.
  3. Consider downgrading the case to “suspected” from “probable” depending on clinical suspicion.
  1. Rapid Antigen Testing (antigen rapid diagnostic test or Ag RDT for short).
  1. If positive, admit as a “confirmed” case or isolate at home, initiate contact tracing. If in a high prevalence setting or in a symptomatic patient with likely COVID-19, confirmatory PCR testing is not needed.
  2. If negative, treat as a “suspected” or “probable” case based on Case Definitions and clinical suspicion.
  1. If discharging, isolate at home and follow-up with a call or visit, homevisit or clinic visit. Consider retesting 2 to 4 days later. Consider contact tracing if suspicion is high for COVID-19.
  2. If admitting, request PCR testing and if not available, repeat rapid antigen testing in 2-4 days (see ECDC report).
  1. If no same-day testing is available (testing is located offsite and/or turnaround times are long):
  1. Send the specimen to the facility with the fastest reliable turnaround times.
  2. Follow-up and retesting strategy:
  1. If discharging to home, isolate at home and call or arrange a visit to share results. If the test returns negative, consider retesting, especially if symptoms persist or worsen.Also, consider retesting if it is deemed important to understand whether the case is COVID-19 and doing so would lead to important contact tracing activities.
  2. If admitting, triage as “suspected” or “probable” case based on Case Definitions and clinical suspicion for disease. If the test returns negative, retest with PCR testing.
  1. If no testing at all is available:
  1. If no testing is available, use clinical judgement and risk factors to determine likelihood of COVID infection and treatment plan. Case Definitions can help. Consider other lab testing to help stratify if available, including lymphocyte count, LFTs, and C-reactive protein. Err on the side of isolation.

Testing Asymptomatic PatientsCopy Link!

Updated Date: January 18, 2022

Asymptomatic Patients with an ExposureCopy Link!

If test capacity permits, testing asymptomatic people with known exposure to COVID-19 may be helpful (ideally as a part of a contact tracing initiative).

Generally we recommend testing patients who meet criteria for an exposure as soon as possible when they become aware of the exposure and again 5-7 days after the first test. This is because initial tests are often negative early in disease. Please note that even if an initial test is negative, the patient must still quarantine until they meet criteria for release from quarantine (in selected cases, testing may be used to reduce quarantine duration). Some healthcare systems recommend against testing in these instances either because of limited testing resources, or because the concerns about false reassurance from early false negatives.

Asymptomatic Screening and Public Health SurveillanceCopy Link!

Literature Review (Not Comprehensive): Gallery View, Grid View

To understand population-level prevalence and incidence, local institutions or departments of health may perform testing (PCR or Antibody) on entire cohorts regardless of exposure or symptoms. Details on design of epidemiologic, surveillance, or infection control studies are beyond the scope of this site.

Asymptomatic people who have a high likelihood of transmission to others should they become infected may be regularly tested for COVID-19, even without a confirmed contact. This is especially important if they spend time with persons that are at risk of complications from COVID-19, the classic example is periodically testing both residents and staff of nursing homes. This is sometimes called “asymptomatic screening” or “expanded screening”.

  • Common high-risk groups that may be considered for screening:
  • Health care workers, particularly those caring for patients with COVID-19 or in high patient-flow areas
  • People living or working in congregate living settings (nursing homes, dormitories)
  • Travelers coming from high prevalence areas
  • Teachers and students
  • Other essential employees (grocery workers, sanitation workers etc).

Asymptomatic Screening Frequencies by Prevalence Indicators:

Community Spread Level

Low

Moderate

High

Highest

New Cases per 100,000 Persons in Last 7 days

< 10

10 to 50

51-100

> 100

Percentage of Tests that are Positive in Last 7 Days

< 5%

5% to 7.9%

8% – 10%

> 10.1 %

Frequency of Asymptomatic Screening

Focus on High-exposure People Weekly

Weekly

Weekly or Twice a Week

Twice a Week or More

Impact of Vaccination on TestingCopy Link!

Neither RNA- nor protein-based vaccines should have any impact on nucleic-acid based tests (NAAT) or rapid antigen tests used to diagnose COVID-19. Some antibody tests could conceivably turn positive after vaccination in rare combinations where a vaccine and test use the same viral antigens, but this is not yet verified for this purpose. Most serologic tests look for antibodies to the nucleocapsid and not spike protein, and thus do not detect vaccine-induced antibodies. As of February 2021, vaccination status should not be routinely considered in interpreting any COVID-19 test results. (CDC)

Newly symptomatic patients who have been vaccinated should still be tested, as breakthrough cases still do occur. Exposed vaccinated people are covered here (generally not needed).

Testing Previously Infected PatientsCopy Link!

Updated Date: April 23, 2021

Patients >90 days from initial illness with new symptoms of COVID-19 can be retested on a case-by-case basis. Reinfection is rare, but not impossible. Generally, patients who are <90 days from initial illness are not tested, however with the emergence of new variants this may be changing: a patient who had fully recovered and then develops new symptoms should be considered potentially re-infected with a new variant and should be re-tested.

  • If the patient tests positive, keep in mind the possibility of residual viral RNA even 90 days after initial infection, and consider alternate causes of illness (pulmonary embolism, bacterial superinfection) and (where possible) viral sequencing.
  • Recurrence of symptoms along with reemergence of positive PCR testing (particularly in patients with weakened immune systems) can occur in the absence of true reinfection.

Types of TestsCopy Link!

Updated Date: January 18, 2022

In a pandemic, clinically suspected cases should initially be isolated regardless of test status. See Screening, Case Definitions, and Isolation. All tests have both false positives and false negatives. A high index of suspicion should be used to protect staff and other patients.

Remember:

  • No currently available test fully rules out the diagnosis, especially if clinical suspicion is high. When possible, negative or positive results that are inconsistent with the clinical pictures should be discussed with someone who has expertise in diagnostic testing of COVID-19
  • None of the commercially available tests measures active, infectious virus. Whether a person who tests positive is infectious requires clinical judgment and knowledge of their disease course.
  • In all cases, please follow local public health authority guidelines in reporting all suspected, presumed, and confirmed cases of COVID-19.

Overview of COVID-19 TestsCopy Link!

Updated Date: January 18, 2022

The three most clinically relevant categories of testing for COVID-19 are:

  1. Nucleic Acid Amplification Test (NAAT): While not a perfect test, NAAT is considered the gold standard. The test uses enzymes to amplify and detect the genetic material of the virus. The most familiar is RT-qPCR (reverse transcriptase - quantitative polymerase chain reaction; related versions are often referred to as PCR or RT-PCR), but there are others. These are often collectively called “molecular tests.”
  2. Antigen Rapid Diagnostic Test (RDT): Requires less time and infrastructure to perform than NAAT tests. Uses manufactured antibodies to detect SARS-CoV-2 proteins.
  3. Antibody (IgM/IgG) RDT: Has different uses and interpretation than the nucleic acid and antigen tests. Uses manufactured antigens to detect a patient’s antibodies to SARS-CoV-2. Since this depends on the body’s immune response, it takes longer to turn positive than tests that directly detect the virus, and a negative antibody test DOES NOT rule out acute infection. The antibody test is NOT used as the sole test to diagnose active or contagious disease; it is more common in epidemiology and research. The test can be used to support the diagnosis in COVID-19 in patients that present late with symptoms (at least 8 days after the onset of symptoms) or to help assess whether a symptom or sequelae is due to a post-COVID-19 infection.

In these lists and elsewhere, tests that measure nucleic acids are often referred to as molecular assays, while antigen and antibody tests are considered immunoassays or serological assays.

General test characteristics are summarized in the table below:

RT-qPCR (or other NAAT)

Antigen (Ag) RDT

Antibody (IgM/IgG) RDT

Sample

Nasopharyngeal, oropharyngeal, saliva, lower respiratory

Nasopharyngeal, oropharyngeal, saliva, lower respiratory

Blood

False Positives

Rare, except for cases of sample contamination. However, can remain positive after virus is no longer viable.

Very low

Low to moderate, most commonly due to cross-reactivity with other coronaviruses.

False Negatives

Occasional, especially early in infection.

Moderate, not as sensitive as NAAT.

Variable. Performs poorly at the onset of the symptoms.

Turnaround time/ Laboratory Requirements

Usually hours - requires a laboratory with high technical capacity.

Under 30 min – no laboratory required.

Under 30 min – no laboratory required.

Specific data on the availability and performance of commercial COVID-19 diagnostic testing options continues to change rapidly.

Tool: PATH COVID-19 Diagnostics Dashboard to support product selection and procurement decisions. Collects data from U.S. FDA, WHO, FIND, and other lists curated by private entities. Includes information on regional or national regulatory approval.
Tool: The Foundation for Innovative New Diagnostics (FIND), a global non-profit, conducted Independent Evaluations of test kits between April and August of 2020.
Tool: The Cochrane Library, a resource by the international charitable organization Cochrane, has published independent reviews of Molecular/Antigen tests (updated 2020-08-26) and Antibody Tests (Updated 2020-06-25).

Timeframe of Test PositivityCopy Link!

Updated Date: December 19, 2020


The relative time frames of exposure, symptoms, viral markers, and antibodies are illustrated in the subsequent figure. This is an illustration of average time frames and it should be noted that information is still emerging on the timeframes of Incubation and Window Period, Infectivity, and Durable Immunity.

This diagram indicates the timeline at which different tests become positive, which is also covered in the below bullet points.

  • Infectious Period: On average, the person is most infectious 2 days prior to the onset of symptoms to about 5 days after the onset. This is referred to as the pre-symptomatic and early symptomatic time frame. In general, people are no longer infectious 10 days after symptom onset (20 days in severely ill persons, see Infectivity).
  • The Antigen Test is likely to be positive at the same period that a person is most infectious from about 2 days prior to the onset of symptoms to about 2-3 days after the onset. This is because the viral load is highest in this time period.
  • The RT-PCR test (and other NAATs) is more sensitive than antigen testing. On average, it can (but does not always) pick up cases earlier than even 3 or 4 days before the onset of symptoms or 10+ days after symptom onset.
  • The antibody test in some cases turns positive after the patient may no longer be infectious. For this reason, the antibody test is not typically used to diagnose active disease.

Nucleic Acid Amplification TestsCopy Link!

Updated Date: January 18, 2022

Literature Review (not comprehensive): Gallery View, Grid View

These tests work by amplifying minute amounts of viral RNA. PCR (technically, RT-qPCR) of a nasopharyngeal swab specimen is most widely used and should be considered the standard of care when available.

How it WorksCopy Link!

Reverse-transcriptase quantitative polymerase chain reaction (RT-qPCR) works by reverse transcribing the viral RNA genome to DNA, and then amplifying the DNA exponentially by repeatedly cycling the reaction. Samples with a small amount of virus require more cycles to reach a detection threshold than samples with a large amount of virus, allowing some RT-qPCR tests to quantify how much virus was present and approximate viral load.

Newer amplification technologies are being developed to make this process cheaper, faster, and less dependent on complex laboratory infrastructure. The term nucleic acid amplification test (NAAT) includes PCR, isothermal amplification, and CRISPR-based tests (Behera et al., Kilic et al). Xpert® Xpress SARS-CoV-2 is an example of an automated cartridge-based system on the same Xpert® machines used for diagnosis of tuberculosis that does not need a sophisticated laboratory setting. All have the common feature of detecting minute quantities of SARS-CoV-2 nucleic acids.

The main advantages to nucleic acid testing are:

  • The amplification steps allow detection of very small amounts of viral RNA, with higher specificity than serological assays.
  • Live virus is not required. Samples can be inactivated and made safe to handle.
  • Nucleic acid amplification is common in biology research. Government labs, universities, and highly-resourced clinical labs may be able to rapidly deploy new or modified tests before commercial kits are available. This may become relevant if a new strain or resistant mutation emerges.

The main disadvantages to nucleic acid testing are:

  • Most methods require significant infrastructure, usually including custom machines, reliable electricity, cold storage, a supply chain for reagents, and skilled personnel.
  • Test performance can depend heavily on how and when a sample is taken. For example, lower respiratory samples can be positive when upper respiratory samples are not.
  • Fragments of viral nucleic acids can persist in the body long after the virus has been killed. Persistently positive tests may not mean that a person is still infectious.
  • Variant strains of SARS-CoV-2 may affect the sensitivity of some nucleic acid-based tests, resulting in false negatives. Tests that use multiple genetic targets are less likely to be affected by the increasing diversity of SARS-CoV-2. As of February 2021, U.S. FDA advice is to consider negative results in clinical context and to consider repeating testing with a different test if clinical suspicion remains after an initial negative test.
  • These tests tend to take longer than antigen tests to get results, resulting in longer isolation periods for patients waiting for results. In situations where it may be difficult to reach a patient after they leave the testing site, this should be taken into consideration.

Test PerformanceCopy Link!

Sensitivity and Specificity:

On artificial samples, NAAT sensitivity and specificity approaches 100% (Giri). NAAT tests have analytical sensitivity (the lowest viral concentration where >95% tests are positive; also called limit of detection) down to 100-5000 copies of viral RNA/ml. But published real-world estimates of sensitivities for different NAATs to diagnose COVID-19 range from ~60-95%, depending in part on what reference method is used as a comparator or “gold standard.” Specificity is excellent, and false positives are rare in the absence of contamination, though there is increasing recognition that false positives due to contamination or cross-reaction with other genetic material do occur and may have significant consequences (Surkova et al)

Interpreting reported clinical sensitivities involves a range of factors:

  1. There is significant variability in how studies define a "true positive" or "gold standard":
  1. The reference standard can be "composite," including laboratory, radiographic, and clinical data. A single center study at University of Kong Kong-Shenzhen, China, compared initial RT-qPCR in 82 patients to a a retrospective diagnosis made by combining serial RT-qPCR and chest CT findings, with a resulting sensitivity of 79% and a specificity of 100% (He et al).
  2. The reference standard is often serial NAAT. since this is often the only data available for large numbers of patients. A retrospective analysis of over 20000 patients used repeat PCR within seven days (felt too short for interim infection to play a large role) and found that only 3.5% of patients initially negative by PCR subsequently tested positive. This suggested a low false negative rate, but they pointed out that this was not a true clinical sensitivity since they lacked a final confirmatory diagnosis (Long et al).
  3. The reference standard can be other previously validated NAAT’s. FindDx reports that the tests they validated agreed 92-100% when comparing to their a reference PCR assay, demonstrating small but real variability between tests.
  1. The site of sampling might not contain virus at the time of sampling. See Sample Collection.
  2. Laboratory factors (sample storage, frequency of contamination).

These same factors should be systematically considered when a clinician suspects a false negative:

  1. Exactly what other data makes me feel like the patient has COVID-19?,
  2. Do we need to repeat the sample or collect another sample type?, and
  3. Could there have been a lab error?

Typical clinical use:

If you do not know your test’s characteristics, sensitivity of ~80% may be a reasonable approximation for nasopharyngeal swabs collected at the time of patient presentation, assuming no laboratory errors.

  • If the RT-PCR is negative but suspicion for COVID-19 remains, then ongoing isolation and re-sampling several days later should be considered.
  • In practice, test results should be interpreted based on negative and positive predictive values (NPV, PPV) rather than sensitivity and specificity, since these incorporate pretest probability.

Sample CollectionCopy Link!

Literature Review (not comprehensive): Gallery View, Grid View

Upper Respiratory Tract Specimens. Most commercial kits have been evaluated on specific upper respiratory sample types. Of these, the nasopharyngeal swab is the most common and best validated; in most situations this is the best option unless a specific manufacturer recommends otherwise or there is a clinical reason to choose an alternative site.

Sites include:

  • Nasopharyngeal Swab
  • Nasopharyngeal Wash/Aspirate
  • Oropharyngeal Swabs
  • Mid-turbinate and Anterior Nasal Swabs
  • A few manufacturers allow these to be collected by the patient at-home (unsupervised) or supervised by a provider at a safe distance resulting in less risk to the HCW.
  • Saliva
  • Potential to significantly simplify sample collection cost and complexity
  • Many new platforms being developed, but still limited comparative performance data (Wyllie et al)

Tool: U.S. CDC Collection Protocol

Tool: Video Demonstration

Lower Respiratory Tract Specimens are also sometimes used, though they often require different processing and validation due to the presence of mucus. When obtaining lower respiratory tract specimens, many sampling techniques require airborne precautions for providers (see Aerosol Generating Procedures).

Sites Include:

  • Expectorated Deep Sputum (similar to sputum collected for TB testing in patients with productive cough).
  • Bronchoalveolar Lavage
  • Endotracheal Aspirates
  • Preferred in intubated patients due to higher sensitivity, though this depends on sample quality Like any respiratory sample, high quality samples are characterized by Gram stains with many polymorphonuclear cells and few epithelial cells.

The relative performance of testing different sample types, optimal timing for sample collection relative to exposure or symptoms, and the interpretation of discordant results (for example, if the nasopharynx is negative but sputum is positive), all continue to be studied.

U.S. CDC guidelines for processing of sputum specimens for SARS-CoV-2 RT-PCR recommend the use of dithiothreitol (DTT) for liquefaction of viscous mucoid/mucopurulent material prior to nucleic acid extraction

Tool: CDC Specimen Processing

Other Specimens: Viral RNA has been documented in other body sites including stool and rarely blood, but it is not known whether this represents transmissible virus (Wang et al). Testing samples from these sites requires extra laboratory expertise for sample handling and clinical expertise for interpretation. These sites should not be tested routinely.

Antigen Rapid Diagnostic TestsCopy Link!

Updated Date: January 18, 2022

Literature Review (not comprehensive): Gallery View, Grid View

How it WorksCopy Link!

This is a diagram showing the function of a lateral flow assay: sample on left, labeled antibody to a virus moves along the test to a "test line" (another antibody to the virus) and a control line (antibody to antibodies).

Rapid Diagnostic Tests (RDT’s) for viral antigens use premade, labeled antibodies to the virus to capture viral particles. The most common approach is the lateral flow test, where sample diffuses along a manufactured strip in a way that can be visually detected at the “test line” only if viral antigens are present. Note that a few manufacturers do make “rapid” NAAT assays. This discussion does not address those tests.

The main advantages to antigen RDT’s are:

  • Running a test involves adding the sample (and sometimes a single liquid reagent) and waiting for diffusion.
  • They generally do not require special trainings or machinery to run, and many are licensed to be run outside of a laboratory setting (e.g. CLIA waiver)
  • They are typically fast, cheap, and have a simple visual yes/no readout that does not require interpretation.
  • Patients can often be notified of their results while still at the testing site.
  • With more outpatient treatments becoming readily available, antigen RDT’s will be useful to implement a test and treat program (widespread screening, early detection of disease, and prompt initiation of appropriate treatment).

The main disadvantages to antigen testing are:

  • They are less sensitive than NAATs, since there is no amplification step. Negative tests may need confirmation with NAAT if clinical suspicion is high.
  • As with NAATs, performance can depend heavily on how and when a sample is taken.
  • Fragments of viral proteins can persist after the virus has been killed, though likely not as long as nucleic acids.
  • If future viral mutations change the antigen region targeted by a particular test, it will take longer to create new antigen RDT’s (which involves new manufacturing) than it would to modify most NAAT’s (which involves new reagents only).
  • Consult the manufacturer’s insert for specimen collection requirements; many are designed for nasopharyngeal swab only.

Test PerformanceCopy Link!

Clinical sensitivity for antigen RDT’s is highly variable. The average of sensitivity was 56% for four antigen RDT’s reviewed in an August 2020 Cochrane review, with 95% confidence interval of ~30-80%. The same review found much higher average specificities of 99.5% (95% confidence interval 98.1-99.9%) (Dinnes et al). The sensitivity may vary based on symptomatology, with some performing as poorly as 32% sensitive (Quidel EUA), and others as high as 79% (Alemany et al) in asymptomatic people. However, many of the cases that these tests miss may not be infectious, but this is still an area of active research.

Finding the real-world sensitivity of antigen RDT’s suffers from many of the same difficulties discussed in the NAAT section above, though these are usually compared with NAAT as a gold-standard. The minimum performance requirements for Ag-RDT set by the WHO are >80% sensitivity and >97% specificity compared to a NAAT reference assay (WHO)

Use of RDTs for Screening Symptomatic PatientsCopy Link!

Rapid Antigen RDTs are an alternative to NAAT as screening tests where testing capacity is limited and the proportion of test positivity is high (≥10%) (ECDC Recommendation). Positive and negative predictive values (PPV and NPV) of all in vitro diagnostic tests depend on disease prevalence in the target population and the test performance.

In a high prevalence setting CDC considers high prevalence to be when NAAT positivity over the last 14 days is greater than 5% or when there are greater than 20 new cases of COVID-19 per 100,000 persons within the last 14 days., rapid antigen tests will have a high PPV, meaning a positive result from a rapid antigen test is likely to indicate a true infection and may not require confirmation by RT-PCR. In contrast, any negative test result should be confirmed by RT-PCR immediately or with another rapid antigen test a few days later (where RT-PCR is very limited).

In a low prevalence setting CDC considers low prevalence to be when NAAT positivity over the last 14 days is less than 5% or when there are fewer than 20 new cases of COVID-19 per 100,000 persons within the last 14 days., rapid antigen tests will have a high NPV but a low PPV. Therefore, a negative antigen test most likely represents a true negative and may not require confirmation by NAAT, however false negatives are still possible and people being tested should be reminded that they should still take all precautions to prevent spread (e.g. masking, distancing, etc). In this situation, a negative test result may not require confirmation by NAAT, whereas a positive test will need confirmation by NAAT.

Scenarios Antigen RDT Can be Used for Screening of Asymptomatic Individuals:Copy Link!

(modified from WHO and ECDC). (For use of Antigen RDT for symptomatic individuals, including contacts, see testing symptomatic patients).

Scenarios for use of SARS-CoV-2 Ag-RDT

Populations Where RDT Can Be Used For Screening where NAAT testing is limited

Negative testing should NEVER exempt people from standard transmission prevention practices (masks, distancing, hand washing). See IPC.

Outbreak Response

To respond to suspected outbreaks of COVID-19 in remote settings, institutions and semi-closed communities

Outbreak Investigation

To support outbreak investigations (e.g. in closed or semi-closed groups including schools, care-homes, cruise ships, prisons, workplaces and dormitories, etc.)

Monitor Trends in Disease Incidence

To monitor trends in disease incidence in communities, and particularly among essential workers and health workers in regions of widespread community transmission

Community Transmission Screening for Congregate Settings

Where there is widespread community transmission, RDTs may be used for early detection and isolation of positive cases in health facilities, COVID-19 testing centers/sites, care homes, prisons, schools, front-line and health-care workers.

Testing of Asymptomatic Contacts/Contact Tracing

Testing of asymptomatic contacts of cases (either as part of outbreak investigations or household contacts) may be considered even if the Ag-RDT is not specifically authorized for this use. However, given the high pre-test probability in this population, a negative test often does not rule out infection (has a low negative predictive value) and where possible should be confirmed by NAAT or repeat RDT as described above. Even in the setting of a negative test, contacts should continue to remain in quarantine until they meet criteria to discontinue quarantine.

Antibody TestingCopy Link!

Updated Date: December 19, 2021

Literature Review (not comprehensive): Gallery View, Grid View

How it WorksCopy Link!

Antibody tests measure the host adaptive immune response, rather than the presence of the virus. This test is most often performed on circulating blood (from fingerstick or blood draw).

Adaptive immune response requires several days to make antibodies that bind to the pathogen. See Antibody Response and Durable Immunity for a more in-depth discussion of antibody patterns over time. It is not yet known what impact antibodies have on the risk of transmission to others or risk of re-infection.

The main advantages to antibody testing are:

  • Respiratory samples are not required. Antibody testing can be done on blood drawn for other reasons in clinical care. There are versions to test dried blood spots.
  • IgG may last for months to years, so it is useful in epidemiology to know who has been previously infected (even if they were asymptomatic).
  • A strategy that uses both NAAT and serology may improve sensitivity for COVID-19 over using NAAT alone. Antibody detection may identify cases with negative upper airway PCR but high clinical suspicion when timed appropriately found positive IgM in 54 of 58 probable cases without detectable nucleic acid (Guo et al).

The main disadvantages (and why antibody testing alone is not recommended to guide clinical decision making) are:

  • Antibodies take several days for the human body to develop, so antibody testing is often negative in early infection; this is known as the “Window Period.” In fact, a positive IgG argues against acute early infection.
  • False positives can occur due in patients who have been exposed to coronaviruses other than SARS-CoV-2, including some types of the common cold.
  • IgM is often less specific than IgG, so false positives may be more common for IgM results, making the test less accurate for acute infection.
  • Although antibody RDT’s using principles of lateral flow are available, the most sensitive versions require laboratory infrastructure for techniques such as ELISA.
  • The immune system simultaneously makes many different antibodies, but test manufacturers choose a single antibody to detect. This can result in increased variability between test performance from different manufacturers.
  • For these reasons, antibody testing alone is not recommended to guide clinical decision making.
  • Prior vaccination for COVID-19 is unlikely to affect the interpretation of antibody testing in most circumstances, though this is still being studied (CDC).
  • Most diagnostic tests detect antibodies without specifying whether they are neutralizing. The first test to receive a U.S. FDA EUA for specifically detecting neutralizing antibodies is the cPass SARS-CoV-2 Neutralization Antibody Detection Kit, by GenScript, USA.

Test PerformanceCopy Link!

Combined IgM/IgG testing has low sensitivity early in infection (30%) but reaches 91% by 15-21 days after onset of symptoms, in a June 2020 Cochrane Review summarizing 54 cohorts with a total of nearly 16000 patients. The same review found a high average specificity of ~98% (Deeks et al).

InterpretationCopy Link!

IgM

IgG

Interpretation

Negative

Negative

  • No serological evidence of infection with COVID-19.
  • Potentially in the “window period” before antibodies have developed
  • Also might be a weak, late or absent antibody response, particularly in older patients, those with poor nutritional status or immunodeficiency, and rarely in severe COVID-19 disease.

Positive

Negative

  • Potentially early infection, before IgG is detectable.
  • Also might be a false-positive IgM (cross-reaction to other coronaviruses).
  • IgM is often less specific than IgG, so false positives from other viruses may be more common in this case.

Negative

Positive

  • Likely either late or resolved infection.
  • Also might be a false-positive IgG (cross-reaction to other coronaviruses)..

Positive

Positive

  • Potentially active infection.
  • Also might be late or recovery phase of the disease, before IgM has declined.
  • Also possibly a false-positive resulting from cross-reaction with other coronaviruses.

Viral CultureCopy Link!

Updated Date: December 19, 2020

Viral culture is not generally used in clinical settings. Availability is very limited, since safe viral culture requires laboratories with advanced biosafety capabilities (typically BSL 3 in the USA). It is the most definitive test for the presence of viable virus, since both antigens and RNA can persist even after the virus is “killed.”

  • It is often used in research settings to tell which types of samples are potentially infectious.
  • It can be used to confirm that patient or pharmaceutical antibodies neutralize viral replication, since some antibodies might bind to the virus without inhibiting replication.

Viral SequencingCopy Link!

Updated Date: December 19, 2020
Literature Review (Novel Diagnostics):
Gallery View, Grid View

Full-genome viral sequencing is not generally useful in the acute clinical setting. When available, viral genomic sequencing from patient samples can be used for local outbreak tracing, assessing re-infection, and large-scale epidemiology. Sequencing may also be used in the future to look for mutations and decreased responsiveness to vaccines or therapeutics, though this will require significantly improved understanding of SARS-CoV-2 biology.

Several groups are developing technologies to reduce the hardware and infrastructure investment needed and finding innovative applications that may eventually impact front-line workers (Khatib et al).

Chapter 3

Infection Prevention and Control

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

Transmission PreventionCopy Link!

This section addresses transmission prevention. Please see COVID overview for the causes of Infectivity and Transmission.

Literature Review (Infection Control): Gallery View, Grid View

Wearing Face Masks and ShieldsCopy Link!

Updated Date: December 19, 2020
Literature Review:
Gallery View, Grid View

This section covers general public use of face masks and shields, for health care usage, see Personal Protective Equipment

Universal masking has been shown to reduce transmission. More data exist for medical settings, but the United States CDC and the WHO both recommend mask use in non-medical settings as well (CDC, WHO).

How do Face Masks Work?Copy Link!

  1. Face Masks provide a barrier against a high percentage of the viral particles released from a wearer’s mouth and nose (Ma et al).
  1. Wearing a medical mask has been demonstrated to result in a six-fold decrease in particle emission during breathing (Asadi et al). Systematic review of research literature on face masks shows that they reduce risk of infection by 85%, with greater effect noted in healthcare settings (MacIntyre et al). Even loose-fitting masks appear to block 51% of particles. (Brooks et al)
  1. Available evidence also indicates that face masks can protect the wearer from inhaling viral particles. Face masks with multiple layers of cloth containing higher thread counts are more effective (CDC).
  2. The effectiveness of masks may be different as new more transmissible viral variants with higher viral loads, such as Delta variant, emerge (Hetemäki et al). However, they are likely to retain some efficacy.
  3. Populations can more easily adhere to universal masking advice than stay-at-home orders in some settings. Face masks allow people to leave their homes for essential reasons with less risk to others.

Can Face Masks Harm People?Copy Link!

Face masks do not interfere with the exchange of oxygen or carbon dioxide, even in patients with severe lung impairment (Samannan et al). Depending on the face mask, it may change the rate of flow of air, which can make people feel uncomfortable, especially if they have obstructive lung disease that also impedes air flow, such as COPD or asthma. Wearing a medical mask can be uncomfortable, but will not cause oxygen deficiency or carbon dioxide intoxication. Make sure that face masks remain dry (WHO). CDC recommends face masks above age 2; WHO recommends against requiring face masks for children under the age of 5 (WHO).

Types of Face MaskCopy Link!

There are several major categories of face masks. In many places manufactured medical-grade face masks (surgical, KN95, and 95) are in short supply. If there is a shortage of medical-grade face masks, they should be reserved for healthcare workers, confirmed COVID-19 patients, patients with symptoms of COVID-19, or patients at high risk of complications (WHO). More details on different types of medical-grade masks are available in PPE Types and Uses.

Tool: Instructions on How to Make Your Own Face Mask.

Face Shields and GogglesCopy Link!

Face shields and goggles are meant to prevent droplets and sprays from entering the eyes (for example, when caring for a hospital patient or a sick family member at home).Regulatory guidance and standards on forms of eye protection are highly variable. For best protection, wear a face shield that fits snugly against the forehead and extends the full length of the face and to the point of each ear (Roberge).. There is lack of evidence to demonstrate that face shields alone are sufficient as a form of source control for protecting others (CDC). They are also not sufficient to protect the wearer when worn alone, and should generally be worn with a face mask (Roberge). When a face mask cannot be worn, a face shield can be worn instead but does not offer the same level of infection control (CDC).

When to Wear a Face MaskCopy Link!

  1. When leaving the house
  2. In quarantine/self-quarantine/isolation when contact with others is necessary
  3. In workplaces and on public transportation
  4. When entering someone else’s home to provide an essential service
  5. When indoors with people who do not belong to your household, including relatives
  6. When cleaning streets or disposing of domestic rubbish
  7. A face mask is suggested, but not absolutely necessary in some outdoor areas if a 2-meter distance can be kept from other people at all times. Consult local rules and regulations.

How to Use a Face MaskCopy Link!

  1. Wash your hands with soap and water or an alcohol-based hand sanitizer before putting on, touching, or removing a face mask (WHO). This prevents you from accidentally contaminating your face if you have coronavirus on your hands. Avoid touching the front of a face mask by touching the strings or ties instead.
  2. The face mask must be worn over both the mouth and the nose, it is not effective if used over the mouth alone. Tie securely to minimize gaps between face and mask.
  3. Avoid touching the face mask while wearing it. If you do, perform hand hygiene.
  4. When removing a face mask, undo the ties and carefully fold the face mask inside-out. Place directly in a designated area for disposal or washing, or in a plastic bag.
  5. Wash cloth face mask in soap or detergent, preferably with hot water. If hot water cannot be used, boil the mask for 1 minute after washing with detergent (WHO). Only use a cloth face mask that has been properly cleaned.
  6. If a face mask becomes damp or noticeably soiled, replace it immediately with a clean one.

Tool: WHO Infographics on How to Wear a Face Mask

Policy Interventions Around Face Mask UseCopy Link!

Adapted in large part from the South African Recommendations:

  1. Public health leaders should create media campaigns to educate the public on the use of face masks, including how to safely use them.
  2. In COVID-19 hotspots it is reasonable for policy makers to make face masks mandatory, especially in spaces where physical distancing is challenging. Educational campaigns should be prioritized over punitive measures to promote adherence.
  3. Face masks are not a substitute for other preventive measures like regular handwashing, cleaning surfaces, physical distancing and contact tracing. All must be done together whenever possible.

Physical DistancingCopy Link!

Updated Date: December 19, 2020
Literature Review:
Gallery View, Grid View

Physical DistancingCopy Link!

The World Health Organization recommends people maintain a physical distance of 1 meter between them. This is based on research on bacterial meningitis and rhinovirus spread (WHO)

In contrast the United States Centers for Disease Control (CDC) recommend a distance of 2 meters. This recommendation is based on measurements of influenza transmission, sometimes from studies in the 1930-40s (Wells).

There is no known distance cutoff that absolutely protects a person from being exposed to any droplets or aerosolized particles (see Aerosols, Droplets, and Fomites). Sneezing and coughing can create turbulent gas clouds that can spread droplets well past a distance of 2 meters (Bourouiba). However, the density of droplets seems to decline the farther away from another person you stand.

Outdoor TransmissionCopy Link!

Transmission is less likely outdoors and in other well-ventilated spaces. Systematic review of evidence indicates that COVID transmission is significantly reduced outdoors: outdoor transmission is responsible for <10 % of reported transmissions globally (Bulfone et al). In indoor spaces, low ventilation and lack of ventilation are both associated with higher transmission rates of airborne diseases (WHO). In one study of transmission in China, only one case among 7300 was associated with outdoor transmission (Qian). However, with more infectious variants such as the Delta variant there are reports of outdoor transmissions at music festivals in the USA and playgrounds in Australia, especially with shoulder-to-shoulder events. However, it is still far less likely than indoors.

Surface DecontaminationCopy Link!

Updated Date: December 19, 2020
Literature Review (Fomites):
Gallery View, Grid View

Surface decontamination can help prevent the spread of COVID-19, though transmission through transmission from surfaces and fomites is not common (see Aerosols, Droplets, and Fomites). Particular attention should be paid to cleaning high touch surfaces frequently. Instructions for making cleaning products and example cleaning schedules are found in Disinfection and Cleaning.

Hand HygieneCopy Link!

Updated Date: December 19, 2020

Effective hand washing is a proven way to remove bacteria and viruses from hands and prevent illness. The exact contribution hand washing has made to population health during the COVID pandemic is currently unknown (CDC), but it is presumed to reduce COVID transmission. Hand washing should be performed with soap and water for at least 20 seconds. Alcohol solutions with at least 70% alcohol can also be used.

When around someone with known COVID-19 infection, hand washing is always a critical protection for staff, patients, and families. Gloves should be used for all blood and body fluids.

The WHO recommends handwashing at five times:

  1. Before touching a patient
  2. Before clean/aseptic procedures
  3. After touching a patient
  4. After body fluid exposure
  5. After touching a patient’s surroundings

Exposures, Isolation and QuarantineCopy Link!

Exposure to COVIDCopy Link!

Updated Date: December 19, 2020
Literature Review:
Gallery View, Grid View

If a patient believes they have been exposed to COVID-19 it is important to assess risk in order minimize anxiety of lower-risk exposures and identify higher-risk exposures and prevent further transmission.

Definition of COVID ExposureCopy Link!

An exposure is defined differently by different groups. Some general guidance is being within 1 meter (WHO) or 2 meters (CDC) feet of an infectious COVID positive person for greater than 15 minutes or direct physical contact without PPE. It should be noted that there is no evidence for a minimum amount of time that it takes when exposed to become infected.

  • A person is considered infectious:
  • If Symptomatic: from 2 days before symptom onset until 14 days after (WHO) or until meeting criteria for discontinuing isolation (CDC) (see Releasing Patients from Isolation).
  • If Asymptomatic: from 2 days before the date of positive test until meeting criteria for discontinuing isolation.

Risk of Developing Disease After ExposedCopy Link!

Risk depends considerably on the duration and proximity of the exposure and how symptomatic the original patient was. Using a similar exposure definition to the CDC definition, investigators found 13% of exposed individuals subsequently developed COVID-19 (Boulware et al). This is higher amongst household contacts (see Household Transmission).

Prophylaxis: Casirivimab-imdevimab can be given subcutaneously for post exposure prophylaxis in select patients (those who are at high risk for progression to severe COVID-19 disease AND immunocompromised or unvaccinated) but is rarely available due to supply issues. A large trial of hydroxychloroquine as post-exposure prophylaxis demonstrated no benefit and increased risk of self-reported adverse events in the treatment arm (Boulware et al).

Testing after ExposureCopy Link!

Please see Testing Asymptomatic Patients

Quarantine and IsolationCopy Link!

Updated Date: April 23, 2021

IsolationCopy Link!

Isolation is the separation of a sick person with a contagious disease from people who are not sick. We recommend isolation for all suspected, presumptive and confirmed cases of COVID-19. Duration of isolation depends on many different factors, and this is covered in Releasing Patients from Isolation. (CDC guidelines). Facility based isolation of COVID-19 cases is discussed in Transmission Prevention in Facilities.

QuarantineCopy Link!

Quarantine is the separation of people who were exposed to a contagious disease to see if they become sick. We recommend quarantine of all persons that have been exposed to COVID-19 cases.

  • Duration of quarantine is typically 14 days from last exposure.
  • December 2, 2020 CDC guidance states quarantine can be reduced in certain circumstances. Keep in mind this may not apply everywhere, and local authorities may have longer or shorter guidelines as they consider changing evidence and resources. As the CDC states:
  • Quarantine can end after Day 10 without testing and if no symptoms have been reported during daily monitoring. With this strategy, residual post-quarantine transmission risk is estimated to be about 1% with an upper limit of about 10%.
  • Quarantine can end after Day 7 if a diagnostic specimen (collected within 48h of Day 7) tests negative and if no symptoms were reported during daily monitoring. With this strategy, the residual post-quarantine transmission risk is estimated to be about 5% with an upper limit of about 12%.
  • Please note: for COVID vaccinated patients defined as individuals two week out from final dose of COVID vaccine, the CDC states individuals may refrain from a full quarantine if they do not have symptoms of COVID-19 after contact with someone who has COVID-19. They do recommend fully vaccinated people should get tested 3-5 days after their exposure, even if they don’t have symptoms and wear a mask indoors in public for 14 days following exposure or until their test result is negative. We expect that this may change given the emergence of variants. This is liable to be different in different epidemiologic contexts, and consulting your local public health regulations is advised.

IPC for Home Quarantine or IsolationCopy Link!

Requirements for Isolation and Quarantine

Physical Distance

  • Accept no visitors from outside the home
  • Maintain a distance of >1 meter from other household members, with only one person assigned to be the caregiver to the patient (this should not be anyone at high risk of severe COVID disease.)
  • Keep patient in a well-ventilated single room, ideally on a separate floor If a fan is available, point it out of one window and keep another window open to facilitate increased air exchange.
  • Have the patient use a separate bathroom if possible; if not clean the bathroom after use
  • No visitors should come to the home during the 14 days.
  • If patient is a primary caregiver to another household member, assign someone else to take over those responsibilities
  • Know When to Seek Care

Hygiene

  • Patients and caregivers should wear masks when not physically separated. Ideally, surgical masks would be used, but cloth masks are an alternative if surgical masks are not available.
  • Caregivers should wash hands after any type of contact with the patient, before and after preparing food, and before eating.
  • All should cover their mouths when coughing or sneezing.
  • Use dedicated eating utensils for the patient. Utensils should be cleaned with soap and water
  • Use dedicated linens for the patient. Linens should be cleaned with hot water and detergent
  • Surfaces should be cleaned with soap, and high-touch” surfaces (e.g. door knobs) with a household disinfectant daily. Can use a bleach solution (1 part 5% pure bleach diluted with 9 parts water to make a 0.5% solution.)

Releasing Patients from IsolationCopy Link!

Updated Date: December 19, 2020
Literature Review (Clearance and Return to Work):
Gallery View, Grid View

Knowing when a patient has recovered from COVID infection and is no longer infectious is important to allowing them to return to contact with other individuals, including work, school, and living environments.

Time based vs testing based criteria: Most situations call for time-based criteria, because the interpretation of positive tests after infection is complicated (PCR often remains positive even in people who are no longer infectious, see Infectivity). A test-based strategy can be used for selected recovered persons for whom there is low tolerance for virus shedding and infectious risk (for example, working in healthcare facilities, residing in congregate living facilities, immunocompromised, etc). Some institutions require more stringent clearance criteria than those outlined here.

Tool: CDC When Can You Be Around Others When You’ve Had COVID-19

Tool: WHO Criteria for Releasing from Isolation
Tool: Massachusetts General Brigham Case Clearance

  1. Symptomatic patients (CDC, WHO):
  1. Time-based criteria: 10 days have passed since symptom onset
  2. 24 hours have passed since last fever without use of fever reducing medications (whichever is longer)
  3. Other COVID symptoms are improving (Note: Loss of taste and smell may persist for weeks or months after recovery and need not delay the end of isolation​)
  1. Asymptomatic patients
  1. Time-based criteria: 10 days after positive test (CDC, WHO)
  1. Patients with severe illness or patients with prolonged symptoms:
  1. Time-based criteria: The CDC recommends up to 20 days from symptom onset. WHO makes no distinction based on severity when determining duration of isolation for symptomatic patients.
  1. Currently hospitalized patients:
  1. Time based criteria: This is not universally defined. BWH uses 20 days since first positive test + 1 day after fever and symptom resolution
  2. Test based criteria: This is not universally defined. BWH uses 1 day after symptom resolution + at least 2 negative PCR swabs at least 24 hours apart
  1. Severely immunocompromised patients: Defined by the CDC as patients on chemotherapy for cancer, untreated HIV infection with CD4 T lymphocyte count < 200, combined primary immunodeficiency disorder, and receipt of prednisone >20mg/day for more than 14 days
  1. Time based criteria: 20 days after symptom onset (+ 1 days after symptom resolution). Exact determination of isolation duration in immunocompromised patients should be made in consultation with an Infectious Disease specialist (CDC)

Transmission Prevention in FacilitiesCopy Link!

Updated Date: December 20, 2020

In health care facilities, IPC is critical to reducing the spread of COVID-19.

Screening and movement: Screen all people (staff, patients, and visitors) entering clinical spaces using Screening Questions. Make modifications to patient flow to ensure patients with symptoms of or at risk for COVID-19 are appropriately classified by likelihood of disease, transported safely, and isolated in designated locations.

Physical distancing: Modify waiting and treatment areas to allow physical distancing.

  • Distances between people should be at least 1 meter (WHO recommendation), and ideally 2 meters (CDC recommendation) in all contexts.
  • Avoid gatherings of staff in confined spaces. For example, consider outdoor staff meetings or use technology to hold remote meetings. Rotate meal times to avoid crowds in dining areas and rearrange break areas to allow physical distancing so staff can eat and drink safely. Add additional work spaces to avoid congregating at nursing stations.
  • In spaces where COVID-related care is provided, the number of people (staff, patients, and visitors) should be kept to the minimum needed. Whenever possible, avoid large groupings of people.

PPE: Use appropriate Personal Protective Equipment and train staff on its use

  • Universal Masking in healthcare spaces is always necessary. Medical-grade masks should be used whenever available.
  • Wards and rooms should be clearly marked with appropriate and standardized signage indicating the category of precaution and PPE that is required to enter.

Isolation: Use appropriate facilities and protocols to isolate patients (see isolation). Positive COVID patients, PUIs, and patients without COVID symptoms should not be cohorted together. Ideally, patients should be separated as quickly as possible into separate spaces based on at least three categories (screening negative for possible COVID infection, screening low-likelihood for COVID infection, and screening high-likelihood for COVID infection). Some settings may use as many as five cohorting categories. See Likelihood Categories (Case Definitions), and Isolation.

Ventilation: Using outdoor spaces and spaces with good filtration or air turnover can decrease risk. All indoor spaces should be sufficiently ventilated and COVID care areas should be negative pressure whenever possible (see Ventilation and Filtration).

Decontamination: Clean all contact surfaces between patients for areas with frequent patient turnover (e.g. clinic rooms and triage areas) and equipment that is rotated between patients (e.g. vital sign monitors). Facilities should develop cleaning protocols for all patient and non-patient care areas.

IsolationCopy Link!

Updated Date: December 19, 2020

Isolation in Hospital Rooms and WardsCopy Link!

Never co-house a patient who screens negative in the same room or ward as confirmed positive COVID patients or PUIs. Confirmed positive patients should only ever be housed with other confirmed positives.

There is no universal set of strategic recommendations for inpatient housing arrangements. Healthcare settings vary greatly in terms of floor plan, layout, equipment, and other resources.These are some principles that can be adapted to local context.

Isolation in RoomsCopy Link!

Which patients need single rooms? In settings where most patients are kept in single or double rooms, confirmed positive COVID cases can be cohorted together in shared rooms. However, PUIs should never be roomed together, as this may result in COVID transmission from one roommate to another if one PUI is actually COVID-negative.

Requirements for rooms: The ideal room is a single private negative pressure room with transparent windows, doors that close, and continuous wireless pulse oximetry monitoring. This arrangement is often unavailable outside of critical care settings, even in the world’s best-resourced practice settings. If a room does not have adequate space and monitoring (direct patient visualization, pulse oximetry, and/or telemetry), rooming and location must balance patient safety risks and infection control needs.

Isolation in Wards and Common AreasCopy Link!

COVID-care wards should be as separated as possible (ideally in a different building) from care areas for patients who screen negative for possible COVID infection.

Separating wards by likelihood level: If single isolated rooms are not available or feasible we recommend using multiple wards or areas to separate patients by Likelihood of Disease. Wards for suspected or confirmed COVID patients should always be separate from wards for patients who screen negative for COVID symptoms. Providers should always move from low-risk patients to high-risk patients.

Whenever possible, at least three separate COVID-care wards should be established to safely cohort the following categories. For additional details, see Likelihood Categories (Case Definitions).

  1. Lower-risk PUIs, including minimally symptomatic and asymptomatic patients with known exposure.*
  2. Higher-risk PUIs, including symptomatic suspected cases and probable cases). Ideally these groups would be further subdivided into separate wards or areas based on their likelihood of having COVID (for example, separating suspected lower-likelihood cases from suspected and probable cases with a higher likelihood of disease).*
  3. Confirmed positive COVID cases.

*These first two categories require the highest level of IPC to reduce transmission, as patients in these spaces are a mix of positive and negative.

When it is not possible to separate wards by likelihood level: If separate wards for each level are impossible, PUIs patients may be cohorted within the same ward and grouped according to risk level with physical distance or Barriers between each group of patients. Since not all PUIs will have COVID, it is important to adequately distance (1-2m) PUIs from each other, arrange the ward from the least likely to the most likely patients. Strict IPC and PPE practices are imperative, and providers should try to move from low to high risk patients if possible.

PrecautionsCopy Link!

Updated Date: December 19, 2020
Literature Review (Airborne v Droplet):
Gallery View, Grid View
Literature Review (Aerosolization):
Gallery View, Grid View

Precaution Type for COVID-19Copy Link!

WHO guidance recommends standard, contact and droplet precautions when caring for suspected or confirmed COVID-19 patients. If an Aerosol Generating Procedure is being performed airborne precautions are needed (WHO).

  • Some hospitals may create their own definitions and specific policies with slightly more stringent requirements, such as Enhanced Droplet Precautions. Given concerns that coughing and sneezing may themselves cause aerosols, some hospitals may choose to put all patients on airborne precautions

Tool: CDC Guidance on Contact, Droplet and Airborne Precautions (including sample signs)

Tool: Detailed CDC Guidance Defining Different Levels of Precaution

Standard, Contact and Droplet PrecautionsCopy Link!

Standard, Contact, and Droplet precautions (drawing from CDC guidance) in the setting of COVID include the following (adapted from CDC and WHO) guidance:

  1. Use high-quality hand washing
  2. Use adequate PPE to protect against contact with the patient’s environment and droplets suspended in air (PPE is covered extensively here)
  3. Use Respiratory hygiene/ cough etiquette (cover mouths when coughing and sneezing, tissues, no-touch receptacles)
  1. Patients should wear medical masks whenever possible
  1. Use appropriate patient rooming and distancing (see “isolation” above)
  2. Use safe injection practices
  3. Use safe waste management and linen management
  4. Use designated equipment for COVID patients (or wards) and adequately sterilize equipment (stethoscope, blood pressure cuff, pulse oximeter) between each patient (e.g. with ethyl alcohol 70%).
  5. Minimize patient movement and transportation and use appropriate precautions when transport is needed (see transport below) (see “transport”)
  6. Maintain good ventilation
  1. Open doors and windows when possible, though be careful not to ventilate from COVID areas to non-COVID areas.
  1. Whenever possible, healthcare workers should move lower-risk to higher risk patients (from asymptomatic to symptomatic and then to confirmed positive patients).
  2. Some additional guidance on specific procedures, lab transport, etc is available in BWH’s ICU Strict Isolation Manual

Airborne PrecautionsCopy Link!

Use airborne precautions when there is a risk of aerosolized particles. In the hospital setting, this generally means during Aerosol Generating Procedures (AGPs). (The role of aerosols in COVID-19 transmission is discussed in Aerosols, Droplets, and Fomites). Airborne precautions should be used for all patients (not only confirmed CoVID-19 patients and PUI) for AGPs in places with high prevalence, or where testing to rule out infection prior to the procedure is not possible. In addition to gown, gloves, and eye protection, aerosol-resistant respirators (N95 masks) are needed during aerosol-generating procedures and until adequate air turnover has occurred afterward (air turnover depends on your facility, in most BWH rooms this is 47 minutes). Please see Personal Protective Equipment for guidance.

  1. Use negative pressure rooms wherever possible, or in a well - ventilated space if not (see Ventilation and Filtration).
  2. Limit the number of people in the room to the fewest necessary.
  3. There should be no other patients and no visitors present.

Aerosol Generating ProceduresCopy Link!

Updated Date: December 19, 2020
Literature Review (Airborne v Droplet):
Gallery View, Grid View
Literature Review (Aerosolization):
Gallery View, Grid View

Aerosol Generating Procedures (AGPs) must be performed with Airborne Precautions for COVID patients, and most non-COVID patients (see airborne precautions). Not all institutions use the same definition of an aerosol generating procedure. Some potential examples include:

  1. Intubation
  2. Extubation
  3. Bronchoscopy
  4. Sputum induction
  5. Cardiopulmonary resuscitation (CPR) with chest compressions
  6. Open suctioning of tracheostomy or endotracheal tube
  7. Manual ventilation (e.g. manual bag- mask ventilation before intubation)
  8. Nebulization
  9. High flow oxygen therapy and non-invasive positive pressure ventilation (e.g., CPAP, BIPAP) (though this is not universal in different institutions, and it is not clear if this increases aerosols beyond coughing, see HFNC)
  10. Oscillatory ventilation
  11. Disconnecting patient from ventilator
  12. Upper airway procedures / surgeries
  13. Upper endoscopy (including transesophageal echocardiogram) and lower endoscopy
  14. Chest physical therapy
  15. Autopsy
  16. Thoracentesis/small-bore (pigtail) chest tube placement (due to the increased risk of cough)
  17. Airway surgeries
  18. Tracheostomy changes
  19. Disconnecting patients from ventilators and ventilator circuit manipulation
  20. Upper endoscopy (including TEE)
  21. Lower endoscopy
  22. Mechanical In-Exsufflator
  23. Dental procedures
  24. Venturi mask with cool aerosol humidification (this is highly institution-dependent)

The following are NOT usually considered aerosol generating procedures:

  1. Venturi mask without humidification
  2. Nonrebreather, face mask, or face tent to 15 liters
  3. Humidified trach mask to 20L (with inline suctioning)
  4. Routine trach care
  5. In-line suctioning of endotracheal tube when ventilator circuit has a viral filter in place
  6. Labor and Cesarean section
  7. Nasopharyngeal swab
  8. Proning (unless ET tube becomes dislodged)

Patient TransportCopy Link!

Literature Review: Gallery View, Grid View

Within FacilitiesCopy Link!

Updated Date: December 19, 2020

Limit transport and movement of patients. When transport is necessary, follow guidelines outlined below.

  1. If a patient must be moved, all staff who come into contact with the patient should don clean PPE.
  2. Patients must wear face masks during transport. Generally this is a medical mask. If this is not possible, a cloth mask should be used. Surgical masks should be used over oxygen delivery devices if possible and if not, well-sealing oxygen delivery devices should be used. Some hospitals permit transport on CPAP/BIPAP or High Flow Nasal Cannula, others do not.
  3. Once a patient is in an isolation area they should not leave it unless to go to a dedicated bathroom, a specific testing or healthcare delivery location (accompanied by a healthcare worker), or upon discharge.

Interfacility TransferCopy Link!

Clinical considerations for transfer are covered in Patient Assessment under Interfacility Transfer.

IPC should be carefully considered for all pre-hospital and interfacility transport. During transport, providers and patients are frequently in close physical contact, and a patient’s COVID status may not be known. All transport systems should develop IPC guidelines adapted for their situation, including on what PPE should be used when. In addition, environmental steps to reduce transmission, such as physical barriers to separate the driver from the patient compartment and ensuring that air is not recirculated in the vehicle can be used.

Tool: CDC Recommendations for IPC for Emergency Medical Systems (EMS)

Tool: IPC Guidelines for Interfacility Transport Without Ambulance Systems (PIH)

Chapter 4

Personal Protective Equipment

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

PPE Types and UsesCopy Link!

Updated Date: April 7, 2022
Literature Review:
Gallery View, Grid View
Tool
: PPE Training Online Module

This section covers professional PPE for healthcare workers. Wearing Face Masks and Shields covers public use.

Standard recommended PPE for care of suspected, probable, and confirmed COVID-19 patients or infectious material includes gown, gloves, eye protection, and N95 respirator or medical mask. Adapted from WHO.

Item

Description

Performance Standards

Gown

  • Single-use, long sleeve, ties in back, length to middle of lower leg
  • Reusable gowns should meet performance standards before and after laundering, up to the maximum suggested number of laundry cycles
  • Some areas including the operating room and labor and delivery may require higher levels of fluid resistance.
  • EU PPE Regulation 2016/425 and EU MDD Directive 93/42/EEC
  • FDA Class I or II medical device, or equivalent
  • EN 13795 any performance level, or
  • AAMI PB70 all levels acceptable, or equivalent

Particulate Respirator

(Type N95 or greater)

Mask that covers the nose and mouth and filters particles (minimum 94-95%) without collapsing against the mouth. Some are tested for fluid resistance

  • Minimum "N95" respirator according to FDA Class II, under 21 CFR 878.4040, and CDC NIOSH
  • Minimum "FFP2 according to EN 149, EU PPE
  • Regulation 2016/425 Category III, or equivalent

Medical/Surgical Mask

Mask that covers the nose and mouth and filters minimum 98% of droplets

  • EU MDD Directive 93/42/EEC Category III or equivalent
  • EN 14683 Type II, IR, IIIR
  • ASTM F2100 minimum level 1 or equivalent

Eye Protection

(Face Shield or Goggles)

Face Shield: Made of clear plastic and completely covering the sides and length of the face. Fits snugly against the forehead with an adjustable band to tighten around the head. May be re-usable (when disinfected) or disposable

Face Shield :

  • EU PPE Regulation 2016/425
  • EN 166
  • ANSI/ISEA Z87.1

Goggles: Made of clear plastic and encloses eyes and surrounding areas; should have good seal with the skin of the face. Flexible PVC frame fits all face contours. Some have adjustable bands to secure goggles to the face. Indirect venting avoids fogging. May be re-usable (when disinfected) or disposable

Goggles :

  • EU PPE Regulation 2016/425
  • EN 166
  • ANSI/ISEA Z87.1 or equivalent

Gloves, Non-sterile/ Examination

Nitrile, powder and latex-free free single-use gloves. Ideally should have longer cuffs, reaching above the wrist so there is no gap between a gown and glove. Sizes: small, medium, large

  • EU MDD Directive 93/42/EEC Category III
  • EU PPE Regulation 2016/425 Category III
  • EN 455
  • EN 374
  • ANSI/ISEA 105,
  • ASTM D6319 or equivalent

Other Particulate Respirators

KF94 masks are similar in appearance to N95s and are able to filter 94% of particles according to standards of the South Korean government. A limited 2020 study demonstrated that KF94s provide protection from particles produced by coughing patients similar to the protection provided by N95s (Kim et al). KF94s have not been approved for healthcare worker use in the United States.

Powered Air Purifying Respirators (PAPRs)

PAPR are battery-powered respiratory protection devices that provide a higher filtration factor than N95s and other non-powered respirators. Air is blown through filter cartridges and into a breathing zone created by a tight or loose-fitting facepiece, hood, or helmet. User-friendly guidance on PAPR is available at this CDC site. International certification and regulatory standards for PAPR in healthcare settings are in slow development because PAPR are primarily certified for industrial applications (Licina et al). To be used in United States healthcare settings, PAPR must meet National Institute for Occupational Safety and Health (NIOSH) requirements. Lists of NIOSH approved PAPR are available here.

PAPR are among the most expensive forms of respiratory protection because of their battery components.There are no clinical trials available to evaluate the protective efficacy of PAPR in comparison to other forms of respiratory protection in healthcare settings, but reasonable application of the precautionary principle in consideration of their superior filtration capacities makes them attractive devices for protection against aerosols (Licina et al).

In some settings, available PAPR are reserved for healthcare workers who have failed fit seal tests or are otherwise unable to wear fitted respirators. Use of PAPR in operating rooms and other areas with sterile fields is controversial because air is not filtered upon exiting PAPR breathing zones. However, statistical differences between surgical masks and PAPR in protecting sterile fields has not been noted (Howard et al).

Seal (Fit) TestingCopy Link!

All N95 masks rely on a close seal to the face to ensure that all air is filtered through the mask. Ideally, qualitative fit testing should be performed to ensure a correct fit for each individual; this should be done annually for each type of N95. In addition, each time an N95 is used, the provider should perform a seal check, then adjust the position of the mask on their face if there is not a good seal.

Tool: Video Describing How to Perform a Seal Check

PPE During Clinical CareCopy Link!

Updated Date: April 7, 2022

Gown

Gloves

Mask

Eye protection

Droplet precautions

X

X

Medical/surgical mask

Goggles or face shield

Airborne precautions

X

X

Particulate respirator/PAPR

Goggles or face shield

General patient care (COVID not suspected)

Low COVID community transmission

Preferred: Particulate respirator/PAPR

Acceptable: Medical/surgical mask

Preferred: Goggles or face shield

High COVID community transmission

Particulate respirator/PAPR

Goggles or face shield

Aerosol generating procedures

(regardless of COVID status)

X

X

Particulate respirator/PAPR

Goggles or face shield

Low COVID community transmission defined as: ___< 50 cases per 100,000 persons____

High COVID community transmission defined as: ___>50 cases per 100,000 persons____

Note: Particulate respirator/PAPR still preferred during low transmission since testing capacity may not allow for accurate numbers

Staff Supporting Care Delivery

Administrative staff

Administrative staff should wear masks at all times; some institutions recommend eye protection for any patient interaction; plexiglass barriers can be used as an alternative

Cleaning staff

When entering a clinical area, PPE should match the PPE needed for clinical care delivery above. In addition, some cleaning supplies may require higher levels of protection from splashes or heavier gloves. When in non-patient care areas, mask is recommended

Transporters

When entering a patient care area or directly interacting with a patient, PPE should match the PPE needed for clinical care delivery above.

PPE During TestingCopy Link!

Updated Date: September 24, 2020

Recommended Personal Protective Equipment (PPE) Euring COVID-19 Testing

Test type

Sample

PPE

Antigen (Ag) RDT

Nasopharyngeal Swab or Deep sputum

N95, Gloves, Gowns, Face Shield

RT-PCR

Nasopharyngeal Swab or Deep Sputum

N95, Gloves, Gowns, Face Shield

Antibody (IgM/IgG) RDT

Whole Blood, Serum, Plasma

Masks, Gloves, Gowns. If Concern for Active (not past) Infection, Follow Local guidance for suspected COVID cases

Donning and DoffingCopy Link!

Updated Date: December 19, 2020

Putting on (donning) and taking off (doffing) PPE correctly is very important. Contamination of mucous membranes while removing PPE can expose the wearer to the virus.

Order of Donning

Order of Doffing

1. Perform hand hygiene*

2. Don gown

3. Don mask

4. Don eye protection

5. Don gloves, ensuring wrists covered

1. Remove gloves

2. Perform hand hygiene

3. Remove gown

4. Perform hand hygiene

5. Remove eye protection

6. Perform hand hygiene

7. Leave the treatment area

8. Remove mask

9. Perform hand hygiene

10. Wash hands with soap and water

*When using alcohol-based hand sanitizer, allow to dry before continuing.

Tool: WHO Infographic for Donning/Doffing PPE
Tool:
Donning Technique Video
Tool:
Doffing Technique Video
Tool
: PPE Training Online Module by Lifebox

DecontaminationCopy Link!

Updated Date: December 19, 2020

  1. Disinfecting Reusable PPE and Equipment:
  1. For most reusable items (for example, thermometers): 70% ethyl alcohol
  2. Reusable face shields can be soaked in sodium hypochlorite 0.5% for 1 hour and left in a clean, open space to dry for at least 1 hour.
  1. Decontaminating N95 Masks: Facilities may consider decontaminating N95 masks when they are in short supply. Different techniques have different levels of efficacy, and some techniques are not effective. Vaporized hydrogen peroxide, UV-C chambers and humid heat are methods that have been implemented by health care facilities. N95decon.org has an Overview of decontamination methods and In-depth Guidance on multiple methods of decontamination. Note that Alcohol and sodium hypochlorite should not be used on N95 masks as they degrade filtration efficacy.
  2. Disinfecting Surfaces: See Disinfection and Cleaning.
  3. Washing Fabric: if reusable gowns are used, they should be machine washed with warm water at 60-90° C and laundry detergent. Laundry can then be dried according to routine procedures.
  1. If machine washing is not possible:
  1. Soak linens in hot water and detergent or soap in a large drum. Use a stick to stir and avoid splashing.
  2. Empty dum and soak linens in 0.05% chlorine for approximately 30 minutes.
  3. Rinse with clean water and allow linens to dry fully in sunlight.

Conservation of PPECopy Link!

Updated Date: December 20, 2020

It is critical to conserve PPE where possible as stock remains limited globally. Local stocks and availability may vary greatly, and individual institutions or local governments may have detailed guidance for the use of PPE that differs from what is presented here. These are some general strategies that can be used to try to conserve PPE while still maximizing patient and healthcare worker safety (adapted from WHO, United States Centers for Disease Control (CDC). To ensure that global PPE shortages do not negatively impact care of any kind of patient (including TB patients and surgical patients), it is important to conserve the use of PPE in all clinical areas.

Tool: PPE Consumption Tool

Institutional PoliciesCopy Link!

  1. Decrease length of hospital stay for patients, if safe to do so
  2. Limit total personnel and visitors in treatment areas
  3. Temporarily suspend routine fit testing for N95s for employees with no COVID contact
  4. Use N95 respirators beyond the manufacturer’s shelf life for training/testing
  5. During known shortages:
  1. Develop policies for extended use of N95 respirators:
  1. Extended use refers to wearing the same respirator for repeated close contact encounters with different patients without removing the respirator between patients. CDC guidelines for extended use can be found here.
  1. Limited Reuse of N95s:
  1. Reuse refers to use of the same respirator by the same health care worker for multiple encounters and doffing it between encounters. Contact transmission may be possible with reuse. Reuse of N95s for care of tuberculosis patients is preferred over reuse of N95s for care of COVID patients because tuberculosis is not transmissible through contact.
  1. When N95 supply is limited, prioritize the use of N95s for high-risk activities, such as aerosol generating procedures.
  1. Do not stop striving to optimize PPE. Institutions remain responsible for safety even while engaging in pragmatic strategies to adapt to crises.

Care Provider ChoicesCopy Link!

  1. Minimize the number of unnecessary aerosol generating procedures. Examples:
  1. Use metered-dose inhalers (MDIs) instead of nebulizers
  2. Do not use humidification with venturi masks
  1. Plan patient care to minimize PPE use. Examples:
  1. Cluster interventions: e.g. take vital signs and give medications at the same time
  2. Time medication administrations so that interventions can be clustered (medications due at the same time)
  3. Choose medications with daily dosing or oral dosing instead of frequent IV dosing
  4. Where possible, use long-acting or scheduled protocols in lieu of protocols requiring frequent assessments and administrations for things like alcohol withdrawal and diabetic ketoacidosis
  1. Arrange patient care areas, equipment, and daily staffing assignments so that caregivers do not need to don and doff PPE as frequently. Examples:
  1. Monitors, IV pumps outside of doors
  2. Where appropriate and possible, use technology (such as phones) to communicate with patients and consultants and minimize caregiver exposure

Equivalents and AlternativesCopy Link!

  1. Due to global PPE shortages, using substitutes for N95 masks may be necessary.
  2. The CDC and the National Institute for Occupational Safety and Health provide extensive guidance on selection and use of N95 equivalents, including an updated list of approved respirators as well as counterfeit respirators.

When Recommended PPE is Not AvailableCopy Link!

Updated Date: December 19, 2020

In addition to taking all possible steps to expand PPE supply and return to normal operations, crisis strategies when recommended PPE is not available are listed below. It should be noted that none of these strategies are sufficient to protect health care workers, and these should be strategies of last resort. In addition, consider excluding healthcare workers at increased risk for SARS-COV complications from patient contact.

  1. Maintain a minimum 1 meter distance whenever possible to avoid inhalation of droplets
  2. If gloves are not available, continue vigorous hand hygiene. Wash hands frequently for more than 20 seconds each time. Avoid touching face, mucus membranes, and surfaces.
  3. When face shields or goggles are not available, use alternate eye coverings, such as glasses, to cover the eyes. If performing an aerosolizing procedure that would normally require an N95, consider:
  1. A double medical mask
  2. Remaining out of direct alignment with the patient’s nose and mouth
  3. Use ventilation and portable HEPA filtration where possible to reduce ambient virus
  4. Consider ventilated headboards for some AGPs if available

InnovationCopy Link!

While official PPE products are preferable, in instances of shortages some people may be able to create PPE alternatives by repurposing existing medical or household supplies, using 3D printers, or using innovative methods for extending the use of existing supplies. Though some have emergency use authorizations or preliminary testing data, very few of these have been officially tested and the level of protection they afford is unknown.

Tool: Frames to Enable Reuse and Improved fit of Respirators

Tool: Snorkel/Scuba Mask Face Shields with Anesthesia Filters

Tool: Elastomeric Mask Adaptations (Source 1 and Source 2)

Chapter 5

Patient Assessment

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

Screening and TriagingCopy Link!

The screening and triaging process involves three parts:

  1. A Quick Symptom and Exposure Screen to determine which patients are at risk for COVID
  2. An Acuity Triage to determine how quickly and where patients need to be seen
  3. Categorization by Likelihood (Case Definitions) to help sort patients who might have COVID by their likelihood of having it, and to help reduce transmission from likely cases to unlikely cases.

Screening QuestionsCopy Link!

Updated Date: December 20, 2020

Goal of screening: To quickly identify patients with possible COVID infections and prevent transmission of infection to other patients and healthcare workers.

Where to screen: At the point of entry. Most healthcare facilities reduce the number of available entrances and set up screening stations with trained staff at every entrance.

Whom to screen: All people entering a healthcare facility should be screened (patients, visitors, staff). Patients who are coming in for routine care should be screened prior to patient arrival if possible (typically via telephone 24 hours before the appointment) and again at the designated point of entry (whether or not the patient was already screened).

Sample Screening Questions:

  1. Do you have any of the following new symptoms?
  • Fever
  • Cough
  • Shortness of Breath
  • Muscle Aches
  • Sore Throat
  • Runny Nose
  • Loss of Smell or Taste
  1. Have you been tested for or had COVID-19 in the last 14 days?
  2. In the last 14 days have you spent at least 10 minutes within 6 feet of anyone with COVID-19 or symptoms of COVID?
  3. Are you, or a household member, currently on home isolation or quarantine, or have you traveled to a place that requires quarantine?

If the patient answers “No” to all of the above, continue routine check in. People who screen negative should be separated from those who screen positive.

If the patient answers “Yes” to any of the above, give the patient additional PPE (a surgical mask) if screening in person and go to Acuity Triage below.

Acuity TriageCopy Link!

Updated Date: December 20, 2020

Literature Review (Virtual Care): Gallery View, Grid View

Facility Acuity TriageCopy Link!

Isolation: If the patient is positive during screening, they should be treated as a possible COVID-19 case, also called a “Person Under Investigation' (PUI) and be separated from patients who screen negative.

Acuity triage: After screening positive, patients should next undergo an acuity assessment to determine how urgently they need to be seen by a medical provider. For urgent care or emergency visits, this should be done with a standardized triage system. One triage system designed for LMICs is the WHO/ICRC/MSF Interagency Triage tool (see below). Patients who are designated as higher acuity by a triage system should be seen first.. Triage should be conducted in a dedicated space with equipment to measure vital signs, and there should be clear pathways from triage to a resuscitation area for patients who are identified as critical.

Tool: WHO/ICRC/MSF Interagency Triage Tool (Pages 11-15)

Home and Virtual Acuity TriageCopy Link!

When patients screen positive over the phone prior to a visit, a provider can assess symptoms over the phone or at a home visit to determine the urgency and best location of evaluation: at home via virtual visit (telephone or video), in person (outpatient), or in an emergency unit.

Below is suggested guidance, but individualized provider assessments should always take precedent. If a provider feels that evaluation in an outpatient clinic or emergency unit is necessary,, they should ensure that the specific location recommended has appropriate IPC and PPE to safely care for PUIs as not all facilities are equipped for this purpose.

Tool: PIH Intake and Symptom Screening Tool
Tool: BWH Telephone and Video Visit Tips

Lower Risk for Complications

High Risk for Complications Age ≥ 65; residence in care facility/correctional facility/dormitory/homeless; underlying conditions: chronic lung/heart/kidney/liver disease, mod-severe asthma, immunocompromised, obesity, diabetes, immunocompromise, psychiatric or substance use disorder

Mild Symptoms (No Dyspnea or Chest Pain)

Telephone/Video/Home

Outpatient Visit

Mild Chest Tightness/Pain

Outpatient Visit

Outpatient Visit or

Emergency Unit

Moderate or Severe Chest Tightness/Pain

Emergency Unit

Emergency Unit

Mild Dyspnea Dyspnea that does not interfere with daily activities (e.g. just mild dyspnea with activities such as climbing 1-2 flights of stairs or walking briskly

Outpatient Visit

Outpatient Visit or

Emergency Unit

Moderate Dyspnea Dyspnea that limits daily activities (e.g. dyspnea that limits the ability to walk up 1 flight of stairs without needing to rest or that interferes with meal preparation or light housekeeping

Outpatient Visit or

Emergency Unit

Emergency Unit

Severe Dyspnea or Home SpO2 94%* regardless of symptoms (or significant decline from baseline) Dyspnea so severe that it renders the patient unable to speak in complete sentences and interferes with basic activities such as toileting and dressing

Emergency Unit

Emergency Unit

Danger Signs:

  • Difficulty breathing/Shortness of breath
  • Bluish lips or face
  • Gasping for air when speaking
  • Coughing up blood
  • Pain/pressure in chest (NOT associated with coughing)
  • Altered mental status or severe sleepiness
  • Inability to eat/drink or walk
  • Any other significant change in condition

Emergency Unit

Emergency Unit

*If patient has home pulse oximeter, here are Instructions. Caution on the reliability of at home pulse oximeters: Trend may be more reliable than the value itself. Dyspnea does not always correlate with oxygen saturation (Shah et al).

Likelihood Categories (Case Definitions)Copy Link!

Updated Date: December 20, 2020

During or after the acuity assessment, a clinical staff member should verify the initial screening assessment and classify patients by their risk (likelihood) of having COVID. Patients who are acutely ill or unstable should not have care delayed for this step.

Why Categorize?Copy Link!

Not all patients who screen positive on questionnaires will have COVID and it is important to try to separate patients by how likely they are to have COVID in order to avoid exposing patients who do not have COVID. Patients who have tested negative or who are not suspected to have COVID-19 should never be co-housed with COVID positive or PUI patients. Keep risk categories as separate as possible. See Levels of Isolation.

How to Categorize?Copy Link!

Someone with clinical training should categorize patients by their likelihood of having disease using standard case definitions. This process can be combined with Clinical Evaluation and can be done in multiple locations (telephone, near facility points of entry, dedicated/ prepared clinics, or COVID-ready acute patient care settings). It is important to note that:

  • Case categorization varies significantly in different hospitals and in different countries. Please follow your local guidance.
  • Clinician judgment is an important part of the decision. If the patient has an obvious alternative explanation for why they have a symptom, their risk could be downgraded. If a patient has significant exposure or classic symptoms, their risk could be upgraded even if they do not meet all criteria.
  • Testing: Test patients in these groups when possible, either before or during this evaluation.

Tool: WHO Case Definitions Handout

Adaptation of the WHO Guidelines for Case Definitions

Case type

Definition

Asymptomatic exposed

An individual who meets the definition of a COVID exposure (described under COVID Exposures but who does not have any symptoms of COVID-19. Treat as described under COVID exposure section.

Minimally symptomatic (sometimes called paucisymptomatic)

Patients not meeting suspected or probable case definitions with one or more new symptoms of fever, cough, shortness of breath, nasal congestion, sore throat, or myalgias. When testing is limited and symptoms mild, these patients may be considered lower priority for testing, but if at all possible they should be tested.

Suspected*Avoid negative terminology such as “COVID suspect”. instead say “person [or patient] with suspected COVID-19”

Anyone who meets both the following criteria:

Clinical Criteria: Acute onset of fever AND cough

OR Acute onset of three or more of fever, cough, generalized weakness and fatigue, headache, myalgia, sore throat, coryza, dyspnea, anorexia/nausea/vomiting diarrhea, altered mental status

Epidemiologic Criteria: Residing in an area with high risk of transmission of virus OR traveling to areas with community transmission OR working in any health care setting within 14 days prior to symptom onset

OR

Anyone with severe acute respiratory illness within the last 10 days who requires hospitalization

Probable*

Any of:

  1. A patient who meets clinical criteria above and has a contact with a probable or confirmed case (or known cluster)
  2. A suspected case with convincing chest imaging consistent with COVID-19 disease
  3. A person with newrecent anosmia (loss of smell) or ageusia (loss of taste) in the absence of any other known cause for these

Confirmed*

A case that has been confirmed with laboratory testing, regardless of signs and symptoms. The exact testing methodology may vary, please see Testing. Most places use RT-PCR. Many countries include positive rapid antigen tests with a contact or strong clinical history for COVID-19

*These case definitions are based on the World Health Organization classification system

Algorithm for Case Definitions

This outlines a potential flow chart for defining confirmed, probable, suspected, and minimally symptomatic potential cases.

Clinical EvaluationCopy Link!

Updated Date: December 20, 2020

HistoryCopy Link!

When assessing a patient with possible COVID-19, ask the following:

  1. Date of Symptom Onset
  1. Patients typically worsen on Day 5-10 after symptom onset and develop acute respiratory distress syndrome (ARDS) at days 7-15 (see Time Course). Patients with severe symptoms before Day 5, or with any progressive dyspnea, require close monitoring as they are more likely to decompensate.
  1. Ask about any known exposure to SARS-CoV-2 or sick contacts in the past 14 days.
  2. Ask about household members:
  1. Does anyone have increased exposure to SARS-CoV-2 (e.g. working in healthcare, schools, stores, transportation, etc.)?
  2. Is there anyone at home to help monitor the patient?
  1. Dyspnea (Difficulty Breathing)
  1. Mild: Dyspnea that does not interfere with daily activities (e.g. just mild dyspnea with activities such as climbing 1-2 flights of stairs or walking briskly)
  2. Moderate: Dyspnea that limits daily activities (e.g. dyspnea that limits the ability to walk up 1 flight of stairs without needing to rest or that interferes with meal preparation or light housekeeping)
  3. Severe: Dyspnea so severe that it renders the patient unable to speak in complete sentences and interferes with basic activities such as toileting and dressing
  1. Mental Status and Function
  1. Has there been a decline or change in alertness, memory, behavior and attention? If so, this should prompt in person evaluation
  2. Patients with recent falls or near falls should be evaluated in person and receive an assessment for traumatic injuries
  1. Chest Pain/Tightness
  1. Evaluate patients with chest pain or tightness in person. While chest pain is a feature of COVID-19 pneumonia, the high rates of cardiac and thromboembolic complications may necessitate ruling out acute coronary syndrome (ACS) or pulmonary embolism (PE).
  1. Dizziness and Hypotension
  1. Assess for orthostatic symptoms, dizziness, mental status changes, or reduced urine output as signs of possible hypotension.
  1. Age and Comorbidities
  1. See Patients with Comorbid Diseases below
  2. Geriatric patients: Older adults are at increased risk of adverse outcomes and are more likely to present with atypical symptoms such as altered mental status, decreased appetite, non-focal pain

ExamCopy Link!

In addition to standard physical exam, pay particular attention to:

  1. Vital Signs. Patients with COVID manifest significant hypoxemia without any subjective difficulty breathing (Tobin et al). See also Pulse Oximetry.
  2. Pulmonary Exam:
  1. Assess for tachypnea, cyanosis and use of accessory muscles. If present, these suggest a patient is having difficulty breathing and needs close monitoring even if oxygen saturation is normal. Dyspnea does not always correlate with oxygen saturation (Shah et al). Tachypnea can also suggest acidosis and shock.
  2. Assess lung exam: although lung exam is often NORMAL even in patients with COVID-19 pneumonia, always listen to the lungs to evaluate for wheezing or crackles that would indicate other possible or additional cause of illness (e.g. asthma/chronic obstructive pulmonary disease (COPD)/congestive heart failure exacerbation).
  1. Leg and calf swelling:
  1. COVID-19 induces a hypercoagulable state, so always assess for deep venous thrombosis (DVT). Ultrasound with Doppler is the standard modality for diagnosing DVT. D-Dimer is not validated as a tool for stratification of DVT probability in COVID-19, given elevated D-dimers in patients in the absence of thrombosis.
  2. Increased swelling in one leg should prompt consideration of deep venous thrombosis, while increased swelling in both legs more often reflects fluid overload or congestive heart failure

Differential DiagnosisCopy Link!

Keep a broad differential diagnosis, both in patients suspected of having COVID-19 and in patients with confirmed COVID-19, given the many diseases that can mimic features of COVID-19 and the risk of secondary infections or sequelae.

Mimics: Other diseases that can cause symptoms mimicking COVID-19 include tuberculosis, malaria, bacterial pneumonia, congestive heart failure, chronic obstructive pulmonary disease, urinary tract infections, and gastrointestinal illnesses. Any of these diseases can also coexist with COVID-19.

Patients should be evaluated for alternative or coexisting diagnoses based on the local burden of disease, patient risk factors, and clinical presentation. Over the course of their treatment, if a patient’s condition or symptoms change, providers should consider whether the cause is due to COVID-19 or if another process is contributing.

Coinfection: Patients with confirmed COVID-19 commonly have concurrent secondary infections. Most studies on co-infection and secondary infection are done in high-income or upper-middle income countries; it is unknown if and how co-infection patterns vary in low-income countries

  • Viral coinfection depends on local epidemiology and season
  • Bacterial coinfection is not very common (~3%), secondary bacterial infection is somewhat more common (~7%). See Bacterial Infection)
  • Malaria, dengue and other tropical diseases can co-exist with COVID

Complications:

Patients with confirmed COVID-19 can also present with or develop a number of complications:

Disease Severity and DispositionCopy Link!

Updated Date: December 20, 2020
Literature Review (Emergency Department):
Gallery View, Grid View

The decision about severity of illness and where to admit varies considerably depending on the availability of beds, the location, and the patient’s resources to monitor and care at home. This is a general set of suggestions based on BWH, PIH, and WHO criteria, and should be adapted to local needs. In some settings, patients with severe or critical COVID may need to be transferred to facilities with higher-levels of care.

Tool: WHO Classification of Disease Severity (page 13)
Tool: PIH Algorithm for Initial Patient Assessment
Tool:
MEWS (The Modified Early Warning Score for Clinical Deterioration) can offer estimates based on vital signs of the probability of ICU admission or death, and has been validated in low-income settings as well (Kruisselbrink et al).

Severity

Mild

Moderate

Severe

Critical

Location

Home

Home or Inpatient

HDU, Step-down or ICU

ICU

Oxygen saturation

(If pulse oximetry is unavailable, monitor respiratory rate)

≥ 94% on room air (ambulatory ≥ 92%)

90-94% on room air (or ambulatory < 92%)

< 90% on room air

Meeting criteria for ARDS. Or needs O2 > 6 LPM to maintain SpO2 > 92% (or rapid escalation of oxygen requirement)

Respiratory Rate

12-22

Adults: 22-30

Children:

under 2 months: ≥ 60; 2–11 months: ≥ 50; 1–5 years: ≥ 40 (WHO)

Adults >30

Children: under 2 months: ≥ 60; 2–11 months: ≥ 50; 1–5 years: ≥ 40.

Variable depending on compensation

Blood Pressure and Heart Rate

BP > 90/60 (or not significantly lower than patient’s baseline)

BP > 90/60 (or not significantly lower than patient’s baseline)

Variable (depending on HDU criteria)

Adults: SBP < 90, MAP < 65, tachycardia, other signs of shock

Children: SBP < 5th percentile or >2 SDs below norm for age, HR <90 or >160 in infants, HR < 70 or > 150 in children, capillary refill > 2 sec

Clinical appearance

Reassuring

Variable

Adults: Concerning

Children: cyanosis, fast breathing, grunting, chest indrawing, inability to drink, lethargy, or convulsions

Adults: Concerning

Children: cyanosis fast breathing, grunting, chest indrawing, inability to drink, lethargy, convulsions, mottled or cool skin

Labs

No strict criteria

ABG with pH < 7.3 or PCO2 > 50 or above patient’s baseline, Lactate > 2

Other

Home isolation requirements are met

Need for intensive nursing care

It may not be within the goals of some patients to transfer to be hospitalized, to transfer to the ICU, or to be intubated.

Vitals and MonitoringCopy Link!

Updated Date: December 20, 2020
Tool: Normal vital signs by age
Tool:
Vitals signs monitoring framework

Pulse Oximetry: Please note that pulse oximeters are less reliable in patients with darker skin tones, and accuracy is improved by trending over time or using both resting and exertional measures. See Home Pulse Oximetry for more details.

We base these recommendations on the assumption of staff and equipment availability. These frequencies may need to be adjusted based on resource availability in different settings.

Severity

Mild

Moderate

Severe

Critical

Temperature Use lower fever criteria in immunocompromised or geriatric patients: one oral temp > 37.8C or two oral temps > 37.2C (IDSA criteria)

On initial assessment

Every 4 hours

At least every 4 hours

At least every 3 hours or continuously

Oxygen Sat, Respiratory and Heart Rate

On initial assessment

Every 4 hours

Continuous or as frequent as possible

Continuous or as frequent as possible

Blood Pressure (BP)

On initial assessment

Every 4 hours

Every 2-4 hours

Continuous, or every 5-15 minutes during resuscitation (30–60 minutes once stable).

Physical exam

On initial assessment

On admission & once a shift

Once a shift minimum

Every 30–60 minutes during resuscitation.Every 2–4 hours once stable.

Mental Status

On initial assessment

Every 4 hours

Every 2-4 hours

Continuous observation or intermittent, every hour

Intake & Output

Every shift

Every 4 hours

Every 1 hour

Chest X-ray

As needed

As needed

As needed

12-lead ECG

On admission and as needed

As needed

As needed

Telemetry

As needed for clear indication

Ideally continuous for all patients, otherwise as needed for clear indication

Lab MonitoringCopy Link!

Laboratory FrequenciesCopy Link!

Updated Date: August 19, 2021

The table below provides a summary of the laboratory monitoring at a well-resourced academic tertiary institution. Monitoring labs such as IL-6 levels will not be possible in most institutions, and excellent care can still be provided without these specialized labs.

Laboratory

On Admission

Trending

De-escalating (non-ICU)

CBC with Differential

Once

Daily

Daily

BMP

Once

Daily

Daily

Magnesium

Once

Daily

Daily

LFTs

Once

QOD*

Discontinue (if stable/improving)

CPK

Once

Trend only if abnormal or clinical decline

Discontinue (if stable/improving)

Troponin

Once

Trend only if abnormal or clinical decline

Discontinue (if stable/improving)

Nt pro-BNP

Once

Trend only if abnormal or clinical decline

Discontinue (if stable/improving)

D-Dimer

Once

QOD*

Discontinue (if stable/improving)

PT/INR

Once

ICU only: QOD*

Discontinue (if stable/improving)

Fibrinogen

Once

ICU only: QOD*

Discontinue (if stable/improving)

CRP

Hospital dependent (some use to risk-stratify for baricitinib or tocilizumab)

Hospital dependent

Discontinue (if stable/improving)

IL-6

Hospital dependent (some use to risk-stratify for baricitinib or tocilizumab)

Hospital dependent

Discontinue

Ferritin

Once, or provider discretion

QOD

Discontinue (if stable/improving)

Procalcitonin

Once

Provider discretion

N/A

EKG

Once

Provider discretion

N/A

*Note: Consider discontinuation on day 8 if patient status and lab values are stable or improving

If the patient is acutely worsening

  1. Redraw all admission labs above to assess the cause of the acute change, and include any other workup that may be needed (e.g. blood cultures, urine strep pneumo and legionella, chest x-ray, EKG)
  2. Resume the regular trending lab frequency with the exception of troponin and Nt-Pro BNP which can be discontinued as soon as downtrending

When lab availability is limited, this is an alternate lab schedule:

Laboratory

On Admission

During Admission

Evaluation of Clinical Worsening

CBC/FBC with Differential

Once

Every other day or twice a week

Once

Glucose

Once

Daily

Once

Electrolytes (Na, Cl, HCO3, K)

Once

Every other day or twice a week

Once

Magnesium

Once

Once

BUN/Cr

Once

Every other day or twice a week

Once

Liver function tests

Once

Once

LDH

Once

Once

CRP

Once

Once

D-dimer

Once

Once

Common Laboratory FindingsCopy Link!

Updated Date: May, 2020

Laboratory abnormalities are more frequent and significant in patients presenting with severe disease. Many of these are associated with more severe disease or death. (Arentz; Chen; Du et al; Guan et al; Young et al; Zhang et al; Zhou et al). Some common abnormalities in COVID patients include:

Interpretation:

  • Coagulopathy:
  • Elevations in PTT and/or INR can be a sign of coagulopathy (i.e. dysfunction in the body’s clotting system which leads to an increased risk of bleeding and increased risk of clotting). Suspect disseminated intravascular coagulation when platelets drop and D-dimer, PTT, and INR increase.
  • D-Dimer:
  • An elevated D-dimer in patients with COVID-19 is not always a sign of thrombosis, though it can be. Consider other signs and symptoms and use available diagnostic methods such as ultrasound and/or CT scan to further evaluate these cases.
  • Inflammation and Cytokine Storm:
  • Inflammatory labs such as D-dimer, LDH, CRP, and ESR are often elevated in patients with severe COVID-19, so if a previously stable patient deteriorates, check these. Cytokine Storm Syndrome, an inflammatory response that can lead to shock and multi-organ failure, should be considered if the following lab parameters are met (though some patients may not meet these cut-offs):
  • CRP >50mg/L
  • And at least two of the following:
  • Ferritin >500 ng/mL
  • LDH >300 U/L
  • D-dimer >1000 ng/mL

ImagingCopy Link!

Updated Date: December 20, 2020
Literature Review (CT and Chest X-Ray):
Gallery View, Grid View
Literature Review (Ultrasound):
Gallery View, Grid View

Chest X-rayCopy Link!

Chest x-ray can help identify alternate causes of shortness of breath. Some chest x-ray findings can suggest a diagnosis of COVID-19. Normal chest x-rays do not rule out COVID: Chest X-rays may be normal in up to ~30% of COVID patients requiring hospitalization, particularly in early disease (Wong). Sensitivity 59% in one study, as compared to 86% for CT scan (Guan).

Low-risk patients with mild symptoms and confirmed PCR testing do not routinely need chest imaging. Most patients with Findings of COVID-19 Pneumonia can safely be managed at home unless clinically unstable, at high-risk of decompensation, or with pneumonia involving >50% of lung parenchyma. Where possible, portable chest X-rays are usually sufficient and require less personnel.

Consider chest x-ray in these circumstances:

  1. High clinical concern for concomitant lobar pneumonia, CHF, TB, or other etiology that could be discovered on plain film.
  2. Patients with oxygen saturation < 92% on supplemental oxygen, increased work of breathing, or new decompensation to rule out new or secondary causes
  3. High clinical suspicion but negative PCR testing (patient could have a false negative test or have been tested too early in the course).
  4. Sudden clinical change in a known COVID patient
  5. To check critical care interventions (line and endotracheal tube placement)

Tool: BWH Guide on Radiology in COVID and Guidance for Radiologists

CT ScanCopy Link!

CT scan plays no role as a screening test for patients for COVID-19, for either diagnosis or exclusion (Simpson).

CT can be used if there is a concern for other pathology. Consider CT in these circumstances:

  1. High clinical suspicion for pulmonary embolism (angiogram contrast scan)
  2. High clinical concern for concurrent abscess, empyema, loculated effusion, significant hemoptysis, pneumomediastinum, etc or if clinician feels it would substantively change management

Tool: BWH Guide on Radiology in COVID and Guidance for Radiologists

Tool: Radiopedia on COVID

UltrasoundCopy Link!

Serial ultrasound is showing promise as a low-cost method to assess disease progression. Although ultrasound findings in COVID-19 have been shown to correlate with CT scan results, the false negative rate of ultrasound is not currently known (Zani et al). A standardized approach using 12 designated zones has been proposed and is strongly recommended to allow for serial comparison (Kruisselbrink et al; Convissar et al).

Tool: POCUS 101 Complete Guide to Lung Ultrasound

Patients with Comorbid DiseasesCopy Link!

Updated Date: December 20, 2020

Patients with chronic conditions have specific risks and needs related to COVID-19 diagnosis, treatment, and social support (e.g. to allow safe isolation/quarantine if needed.) Patients with diabetes, hypertension, heart disease, and obesity have been shown to have higher rates of hospitalization and severe illness due to COVID-19. (See Prognostic Indicators)

Relevant comorbidities are covered in greater detail in different chapters, and include the following:

  • Immunosuppressed patients may have atypical presentations of COVID-19 (e.g no fever). Patients with HIV who present with respiratory symptoms should be evaluated for TB in addition to COVID-19 as clinically indicated.

Management of existing medications is an important consideration in these patients. These medications are discussed in Treatments for Comorbid Diseases and may include the following.

  • ACE inhibitors
  • Immunosuppressants
  • Nonsteroidal anti-inflammatory drugs
  • Steroids
  • Inhalers

Interfacility TransferCopy Link!

Updated Date: January 11, 2021

Reasons to transferCopy Link!

There are many potential reasons to transfer a COVID19 patient to another facility including:

When deciding whether or not to transfer, consider:

  1. Resources and specialty service availability: What resources are currently needed or will soon be needed for patient care? Are those resources available at the current facility? Are they available at the receiving facility? Consider specialized and subspecialized services such as critical care, OB/GYN, pediatrics/neonatology, and surgical specialty teams.
  2. Receiving facility capacity: Does the receiving facility have sufficient capacity to accept the patient? Receiving facilities that may normally be able to accept transfers may be overburdened as a result of the COVID-19 pandemic. Prior to transfer, the receiving facility should be contacted to discuss the transfer and verify that they have adequate resources and space to accept the transfer.
  3. Patient goals of care: What are the goals of care for the patient and family, and how does transfer fit within those goals? For example, unless there is another reason for transfer, a patient who does not want intubation and mechanical ventilation may not benefit from transfer to a facility where these services are available
  4. PPE availability: Is adequate PPE available for transfer, and at the receiving facility?
  5. Stabilization: Has the patient been stabilized as much as is reasonably possible at the current facility, or do the benefits of transfer outweigh the risks? For example, if a patient is currently at a facility without surgery capacity or the ability to perform blood transfusions, it may not be possible to fully stabilize a patient with an intra-abdominal hemorrhage and the patient may need to be transferred while still unstable. Patients should always be transferred with medications and supplies needed for ongoing treatment en route.

Stabilization Prior to TransferCopy Link!

A full discussion on stabilization for transfer is beyond the scope of this site. For the transfer of COVID19 pneumonia patients the top concern is generally the amount of oxygen required by the patient safe for transport and whether to intubate prior to transfer. This is especially true as patients considered for transfer often have a rapidly worsening trajectory and are at high risk for deterioration.

Whether to Intubate Prior to TransferCopy Link!

Intubation should not be done if it is not indicated (see Candidacy for Intubation). Intubation carries risks, especially in certain patients (e.g. patients with right heart failure or a difficult airway). The decision about whether to intubate prior to transfer should balance risks and benefits and take into consideration the following questions:

  1. Is the patient likely to require intubation en route?
  1. Consider the current clinical status of the patient (including work of breathing, vital signs, and mental status).
  2. Consider what the projected clinical course for the patient is over the time it will take for them to arrive at the receiving facility.
  1. If a patient is rapidly worsening (including a rapidly escalating oxygen requirement), intubation may be appropriate before departure regardless of transport time.
  1. If a patient is slowly worsening, but does not currently warrant intubation, transport without intubation may be appropriate if transport time is brief, while intubation prior to transfer may be needed if transport times are prolonged
  1. Is safe intubation feasible at the transferring facility? Is the transporting team able to perform a safe intubation? If neither is possible, maximize oxygen and other respiratory support (such as non-invasive ventilation, if available) for transport.
  2. Is emergent intubation possible during the transfer? Intubation during transfer may not be possible or may be higher-risk depending on provider training, vehicle space and layout, equipment available, and road conditions during transport. If emergent intubation during transfer would be difficult or impossible, intubation prior to transfer may be indicated.
  3. Are there conditions that would make emergent intubation challenging? If so and there is possibility the patient may need intubation en route, early intubation in a controlled setting prior to transfer may be preferred. This is particularly relevant if there is:
  1. Known or suspected difficult airway. Challenging airways are always difficult to manage in emergent situations, and even more so during transport when equipment is limited.
  2. Hemodynamic instability. Unstable patients are difficult to intubate under controlled circumstances and even more challenging to manage during an emergent intubation during transport.
  1. What is the capacity for monitoring, sedation and ventilation available during transport?
  1. Consider the level of training of personnel accompanying the patient, and the availability of battery-powered transport ventilators versus need for bag-valve mask ventilation during transport.
  2. In settings where transport of ventilated patients is uncommon, ensure that transport ventilators can connect to the available oxygen canisters. Ensure all ventilators have sufficient back up electrical supplies, and that providers are trained to bag patients as a back up.
  3. When transport resources are limited, it may be necessary to send trained medical staff with the patient to manage advanced equipment.
  4. In settings where monitored transport is not possible and where medical staff cannot accompany the patient, risks and benefits of intubation prior to transfer should be carefully weighted, as a dislodged endotracheal tube or an accidental disconnect of a ventilator can be fatal.
  1. Can the receiving facility manage an intubated patient? It is important that the receiving facility has the capacity and resources to manage an intubated patient. Capacity may fluctuate depending on patient volume at the receiving facility.
  2. Is the patient nearing the limits of oxygen delivery capability of the transport system? See below for specifics on air transport. Generally, mechanical ventilation for intubated patients consumes less oxygen supply than non-intubated patients on oxygen delivery devices with high oxygen flows (e.g. high flow nasal cannula or CPAP/BIPAP with a significant leak).

Calculating Transport Oxygen NeedsCopy Link!

Non-intubated patients on oxygen delivery devices with high oxygen flows (e.g. high flow nasal cannula, non-rebreather facemask, CPAP/BIPAP/NIPPV) may rapidly exhaust or exceed the available oxygen supply during transport. This can be life threatening.

  1. Calculate total oxygen demand in advance. For example, for an 8-hour transport time, a patient on a non-rebreather facemask at 15 liters per minute will require either 2 portable oxygen concentrators (may vary depending on device output) and a reliable portable power generator, or two full J cylinders (See Oxygen Cylinder Duration Calculator).
  2. Factor in a buffer in case oxygen demand increases, or the trip is longer than expected.
  3. Make sure there is at least one power backup for electrically-powered delivery devices.

Additional air transport needs: During air transport barometric pressure drops, while FiO2 stays constant. The result is less partial pressure of oxygen delivered to the alveoli and the volume expansion of any trapped gas. This can precipitate the deterioration of a patient in two ways:

  1. Worsening hypoxia at altitude. Pressurized aircraft are generally maintained at the equivalent of 5000ft (~1500m) to 8000ft (~2500m) above sea level. This is roughly the equivalent of three quarters of the oxygen delivered at sea level that is delivered in each breath. The effective altitude during transport should be accounted for when estimating oxygen needs during transport. At higher effective altitudes more oxygen will be required and less potential oxygen can be delivered than at sea level (i.e. a patient on requiring 100% FiO2 with an SpO2<100% at sea level, will desaturate when brought to altitude).
  2. Air transport can lead to expansion of gas in body cavities and can lead to pneumothorax or tension pneumothorax. Providers should be trained to recognize tension pneumothorax and perform a needle decompression if needed.

Other Factors that May Affect Transfer DecisionsCopy Link!

Factor

Recommendation

High levels of support from noninvasive ventilation with depressed level of consciousness, marginal oxygenation, tachycardia or hypotension

Consider intubation before transfer

Severe, uncorrected, electrolyte disorders

Evaluate risk of delaying transfer to correct versus starting correction and continuing to correct en route or at receiving facility

Severe obesity that cannot be accommodated in transport bed and vehicle

Consider alternate means for transfer

Unable to tolerate supine position for duration of transport

Assess if transport can be safely done in a manner tolerable to patient, including in seated position

Use of accessory muscles for spontaneously breathing patients

Consider increasing oxygen delivery or respiratory support before transfer

Receiving facility unable to provide higher level of care

Look for a different receiving facility

Receiving facility does not have available PPE or cohorting capacity for droplet and airborne transmission.

Look for a facility that has COVID care capacity

Transport team does not have adequate PPE

Transferring Facility may give PPE to transit team if possible, or alternative transport team can be selected

Pneumothorax without a chest tube

Place the chest tube prior to transit if clinically indicated and can be safely done at the transferring facility (may not be for all pneumothoraces). If the chest tube is not placed, ensure the patient transferred with trained personnel equipped for needle decompression in case of sudden worsening.

Severe hemodynamic instability

Support hemodynamics as much as possible with medical interventions; anticipate potential worsening en route and ensure transport team equipped with medications and materials as needed to address ongoing instability

Patient or family opposed to transfer

Follow guidance about patient’s Rights to Refuse Recommended Care

Futility with extremely poor short-term prognosis

Discuss goals of care with family to decide on if transfer is consistent with goals of care

Pregnancy at greater than 22 weeks gestation age without adequate obstetrics and pediatrics care available at the receiving facility

Seek a facility that has this capacity. If none is available, transfer to the location that optimizes maternal and then fetal welfare

Lack of access to a transport team capable of safe transport

Consider sending staff from the transferring facility to accompany and treat the patient en route.

Inadequate portable oxygen supply for patient’s needs

Try to obtain

Inadequate power supply for equipment

Try to obtain; assess what the minimum necessary equipment is

Tool: Tools for Interfacility Transfer and Documentation (OCC)

Tool: Interfacility Transfer Checklist

Tool: IPC Guidelines for Interfacility Transport Without Ambulance Systems (PIH)

Tool: Algorithm for COVID-19 Triage and Referral by WHO

Tool: Medical Transport Accreditation Standards, 11th Edition by Commision on Accreditation of Medical Transport Systems

Chapter 6

Home and Outpatient Management

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

Household Assessment and PreparationCopy Link!

Updated Date: December 20, 2020
Literature Review (Ambulatory Management):
Gallery View, Grid View

Whether or not a patient is clinically well enough to be managed at home is addressed in:

  1. Acuity Triage
  2. Disease Severity and Disposition

Providers should then evaluate if the patient can safely isolate at home for the full Duration of Isolation:

  1. Home Assessment and Education: Review the Requirements for Home Isolation with the patient. If possible, a healthcare or community health worker should have the patient talk them through the procedures in the patient’s particular environment/home and do a verbal walk through (or video call) of the place where they are isolating in order to ensure a safe environment.
  • For additional information on masks, social distancing, pets, and more see Transmission
  1. Communication: The patient and family should be provided with clear instructions of what to do and who to call if the condition worsens (see When to Seek Care).
  2. Caretakers, proxies, and goals of care: For all patients with COVID, it’s important to identify a single person who can be the primary caretaker at home. You should engage the patient in a goals of care conversation and have them designate someone to make decisions for them if they are unable to make decisions for themself (regardless of age, comorbidity, or severity). See Discussing Goals of Care. Caretakers should be trained in methods for masking, distancing, hand hygiene, and cleaning surfaces (see Transmission Prevention).
  3. Resources: Assess for and provide home care kits including food support, hygiene and cleaning supplies such as chlorine and soap and individual patient items such as plate, utensil, blanket and mask. Social/economic support, including food supplementation is critical and should be delivered in a way that is in line with infection prevention.
  4. Psychological support: See Psychosocial Support for information on mental health and psychological support.
  5. Close contacts: All symptomatic contacts should be tested and Quarantined. Depending on local epidemiology and practices, some places recommend that asymptomatic contacts also be tested (see COVID Exposures) and engage a contact tracing team where relevant.

Followup and MonitoringCopy Link!

Updated Date: December 2022

Follow-Up Visit ScheduleCopy Link!

Patients in outpatient isolation or post-hospitalization require followup visits based on risk level. These visits can be performed virtually if possible (by phone, or by video). In some places, community health workers may conduct these visits physically at the home if unable to connect via phone or video.

  • Individuals adversely affected by social determinants of health are less likely to be able to engage care via telemedicine and may become lost to follow-up with decreased in-person accessibility. It is critical to account for inequitable access to these resources when rolling out telehealth and virtual care models (Wood et al).

If community health workers or clinicians conduct home visits, they should have:

  • Adequate PPE. If PPE is not available, conduct the visit outside the home (rather than entering) maintaining a distance of at least 2 meters.
  • Refer patients to a different clinician or a higher level of care if needed.

Tool: PIH COVID-19 Patient Intake and Symptoms Screening Data Collection Forms
Tool:
BWH Telephone and Video Visit Tips
Literature Review (Virtual Care):
Gallery View, Grid View

Suggested Home or Phone Visit FrequencyCopy Link!

Community Health Workers:

  • For patients at low risk of complications: Visit every 2-3 days in person (or via phone or video if available).
  • For patients at high risk of complications: Visit every 1-2 days in person (or via phone or video if available). If the patient has any danger signs contact the health facility via phone or refer/accompany to the health facility.
  • After hospital discharge: Visit within 3 days of discharge (or via phone or video if available) unless the patient will return to the facility for follow up.

Clinicians:

  • For patients at low risk of complications: Day 5 of symptoms via phone or video if available (in-person if not)
  • For patients at high risk of complications: age>60, COPD, hypertension, diabetes, cardiovascular disease, chronic kidney disease, liver disease, obesity, immune deficiency or immunosuppression Days 4, 7, and 10 of symptoms by phone or video, more frequently or in-person if needed (see Acuity Triage to determine if your patient should be seen in person).
  • After hospital discharge follow-up: 2 days

Longer-Term Followup:

When to Seek CareCopy Link!

Patients should call a healthcare provider or report to a facility if any of the following danger signs develop:

COVID-19 Danger Signs

  • Difficulty in breathing/Shortness of breath
  • Bluish lips or face
  • Gasping for air when speaking
  • Coughing up blood
  • Pain/pressure in chest (NOT associated with coughing)
  • Altered mental status (e.g. confused or severe sleepiness)
  • Inability to eat/drink or walk
  • Any other significant change in condition

Pulse OximetryCopy Link!

Literature Review: Gallery View, Grid View

Why use home oximetry: If patients or community health workers have access to pulse oximetry, this can be a helpful adjunct to symptom monitoring, since hypoxemia out of proportion to respiratory effort has been seen in patients with COVID-19. Home pulse oximetry can also be used in conjunction with Awake Proning.

  • One prospective cohort study gave pulse oximeters to 77 ED or testing site patients suspected of having COVID-19 (42% with no comorbidities) and told them to measure their O2 sat three times daily and return to the ER if it dropped to <92%. This occurred for 19 patients, and of the 17 who returned to the ER, 8 returned solely because of the oxygen saturation, and 16/17 required admission. Resting home SpO2 < 92% was significantly associated with hospitalization (RR = 7.0, 95% CI = 3.4 to 14.5), ICU admission (RR = 9.8, 95% CI = 2.2 to 44.6), ARDS (RR = 8.2, 95% CI = 1.7 to 38.7), and septic shock (RR = 6.6, 95% CI = 1.3 to 32.9) (Shah et al).
  • Another randomized trial assessed a text message–based remote-monitoring program (which included twice-daily automated text messages inquiring about dyspnea and offering rapid callbacks from nurses when appropriate) supplemented with monitoring of oxygen saturation using a home pulse oximeter. Patients were randomized to the standard monitoring program in addition to home pulse oximetry versus the standard program alone. Patients in the pulse oximetry group were provided a pulse oximeter and monitored for subjective symptoms or a low or declining oxygen saturation. Among patients with test-confirmed Covid-19, there was no significant between-group difference in the number of days they were alive and out of the hospital at 30 days (mean, 29.4 days in the pulse oximetry group and 29.5 days in the standard program group; P=0.58; difference, −0.1 days; 95% confidence interval [CI], −0.4 to 0.2) (Lee et al)

How to use pulse oximeters:

  1. A caution on reliability: Pulse oximeters are less reliable in people with darker skin tones, and may read artificially high. One study showed that pulse oximetry failed to correctly identify 11.7% of black patients with Sp02 of <88% on ABG, relative to only 3.6% of white patients (Sjoding et al). They also may not work properly in patients with blood flow abnormalities in their hands such as peripheral vascular disease or Raynaud’s syndrome (Luks et al).
  1. SpO2 trending may be more reliable than a single value (i.e. baseline 100%, now at 94%) and using dynamic measures like ambulatory saturations can help identify patients who are at risk for decompensation.
  1. How to use oximeters: Have patient’s check their saturations at home at least daily and as-needed for clinical worsening. They should take one measure while sitting and one measure while ambulating (or marching in place if they are quarantined in a small area). Instruct patients to seek care if Sp02≤94% or ≤ 92% with exertion.
  1. Use the pulse oximeter on a finger without nail polish or nail abnormalities. Hands should ideally be warm and relaxed.
  2. Wait at least 20 seconds for sampling time as the Sp02 displayed is generally the average of the last 10-15 seconds
  3. If the pulse oximeter has a visible waveform (plethysmograph), the shape should show a regular rise and fall (corresponding with the pulse) when the machine is reading correctly. If the machine is not giving a reliable reading (not registering, number not steady, number very low), try it on a different finger.

Tool: How to Use Home Pulse Oximetry

Management of Mild DiseaseCopy Link!

Updated Date: February 25, 2022
Literature Review (Ambulatory Management):
Gallery View, Grid View

Medical TreatmentsCopy Link!

Treatment for COVID-19 for patients with mild disease is largely supportive. A small number of places may have antibody therapies available for outpatients.

Please see Overview of Treatment by Severity of Disease, and Treatments for more details.

  1. Patients without hypoxemia or risk factors
  1. Symptomatic Treatments:

  1. Patients without hypoxemia but risk factors (Age >60, cardiovascular disease, hypertension, diabetes, COPD, cancer, immunosuppressive medications, detectable HIV viral load or CD4 <200, TB, pregnancy, malnutrition)
  1. Symptomatic treatments
  2. COVID Treatments:

Management of Existing Medications. Medications should not be discontinued without discussing with the prescriber.

Medications we DO NOT Recommend:

Vaccinations:

  • Influenza vaccination: Recommended for all patients for whom there is not a contraindication.
  • Pneumonia vaccinations: these should be given to patients who meet criteria.
  • Please note: As SARS-CoV2 vaccines become available, present guidelines do not recommend administering them to patients with acute COVID-19 (see CDC guidance).

Non-Medical TreatmentsCopy Link!

Self-proning

  • The Awake Prone position improves dyspnea and hypoxemia in some patients with severe COVID-19 illness. Proning could be used while awaiting ambulance transfer to a health facility or as part of terminal palliation.

Psychological and social support

Referral for AdmissionCopy Link!

Patients should be referred for in-person evaluation if they develop any of the danger signs above, rapidly escalating symptoms, Sp02 below 94%, or moderate to severe dyspnea.

Where to send your patient? See Acuity Triage

Inpatient treatments to be aware of: Ambulatory providers should be aware of inpatient therapies so they can refer/admit patients appropriately.

Home Oxygen CareCopy Link!

Updated Date: January 7, 2021

Patients who require oxygen for COVID pneumonia should be cared for in a health-care facility where possible, however capacity constraints and the long course of recovery make this impossible in some circumstances. The decision to offer home oxygen therapy is complex, and should be made by a certified provider familiar with the patient’s clinical condition, resources for care at home, proximity to health care facilities, and the availability of home oxygen delivery.

This section provides a framework for providers to use when considering this treatment plan as an option for certain patients when facility-based oxygen delivery is not available. The framework below is based on published literature and an ongoing home oxygen program developed for COVID19 care in the rural United States by providers from Gallup Indian Medical Center (GIMC).

Patient SelectionCopy Link!

Patient Medical Selection Criteria:

  1. COVID-19 diagnosis established or strongly suspected with low suspicion for alternate diagnosis
  2. Hypoxia at rest or with ambulation (O2 sat < 90% on room air)
  3. Ideally, an oxygen requirement of < 2LPM NC is needed to achieve O2 saturation (SpO2) of > 92% at rest AND > 90% with exertion
  4. Stable or improving clinical trajectory. Discharging patients with hypoxia without an observation period of 48 hours is high risk, and this is better suited to stable inpatients ready for discharge.
  1. The patient’s vital signs have normalized after addition of supplemental O2 and other clinical interventions (e.g. antipyretics and fluids)
  2. 4C Mortality Score < 9
  1. Lower risk for decompensation:
  1. In outpatients: mild severity risk features/comorbidities (See: Table 1 Sardesai et al)
  2. In inpatients: mild or moderate risk features AND stability over 48 hours of observation (See: Table 1 Sardesai et al)
  1. Patients with the following risk factors may be higher risk and not ideal candidates (Age>65, BMI>40, chronic kidney disease, liver disease, immunocompromised, diabetes, hypertension, obstructive lung disease)

Patient home-care ability selection criteria:

  1. Ability to demonstrate understanding of the risks and benefits of this discharge plan. Confusion and pre-existing cognitive impairment are absolute contraindications to discharge with home oxygen. See Capacity Assessment.
  2. Ability to use and troubleshoot pulse oximeter
  3. Ability to use and troubleshoot supplemental oxygen equipment
  4. Ability to engage in telephone (or community health worker) contact follow up (i.e. reliable phone service, language services, hearing or speech impairment does not necessarily preclude communication).
  5. Ability to Isolate Safely at Home.
  1. Household members do not have high risk comorbidities and can practice safe personal protective measures
  1. Reliable power source, if discharging with home concentrator
  2. Reliable transport or plan to provide transportation resources to medical care if the patient needs to be re-evaluated (including distance, weather, or road conditions between home and hospital)

Institutional criteria:

  1. No inpatient bed is available
  2. A provider determines the home oxygen discharge plan is safe
  3. The patient has appropriate supply of oxygen (see calculator Tool and discussion below)

Tool: Determination of Eligibility for Short-Term Home O2
Resource: Home Care for Patients with Suspected or Confirmed COVID-19 by WHO
Resource: COVID19 Home Based Quality Care by HP+

Oxygen Discharge PlanCopy Link!

Patients must receive oxygen and supporting equipment as well as the instructions for use prior to discharge. They must also be educated in-person on how to use them, and counseled on return precautions using the Teach-Back technique. It is critical to confirm a working phone number for the patient.

Equipment:

  1. Oxygen supply (Cylinders or oxygen concentrators)
  1. Calculate total oxygen need, as not all sources will be able to provide continuous oxygen. Oxygen cylinders may run out too quickly to be practical for home use in patients with high needs
  2. Cylinders:
  1. If using oxygen cylinders, an oxygen conserving (i.e. pulse dose) device may be helpful
  2. Tanks must be secured to avoid potential injury, especially with small children in the household
  1. Oxygen concentrators: Most concentrators have 5 LPM max output, though some can do 10 LPM and very few can do more than 10 LPM. Patients requiring more than 2 LPM should ideally be cared for in a facility, and risks and benefits of home treatment require careful decision-making.
  1. Pulse Oximeter (confirmed working) and instructions on use
  2. Thermometer
  3. Surgical masks to wear over nasal cannula to protect contacts at home
  4. Working phone to conference with providers

Medications:

  1. Patients receiving oxygen should get Corticosteroid Therapy unless contraindicated.
  2. Patients should be discharged with other symptomatic treatments needed. See Medical Treatments for mild disease.

Instructions and education:

  1. Self Proning Handout
  2. Oxygen Self Titration Guidance
  1. Oxygen saturation readings consistently less than 92% on flow rates prescribed at initiation of home oxygen therapy (regardless of symptoms).
  1. Return Precautions:
  1. Return criteria for worsening symptoms and Danger Signs.
  2. Handout with instructions specific to the healthcare setting and available resources
  1. Oxygen management instructions
  1. Oxygen Supplier Contact
  2. Phone Number for Help Line
  3. Action plan for equipment failure, low supplies, or power failure
  1. Fire safety plan at home (Table 3 from Sardesai et al)
  2. Plan for food security

Resource: How to Use an Oxygen Concentrator

Tool: Oxygen Supply Calculator and Cylinder Duration Calculator

Tool: Tools for Home Oxygen Therapy

Follow-UpCopy Link!

Below is a sample protocol modified by one developed at GIMC.

For inpatients observed for >48 hours and then discharged on oxygen:

  1. Should be called daily for at least 2 days after discharge
  2. If remaining on 2L or less x 2 days may consider discharge from phone follow-up program, otherwise the same criteria above apply. For those on >2L, follow at least 10 days from oxygen initiation.

For patients sent home on oxygen with < 48 hours of observation (not preferred):

  1. Should be called daily for at least 6 days unless weaned off oxygen sooner
  2. After day 6, patients on 2L or less at rest x 2 days with improving trajectory may be discharged from phone follow-up
  3. After day 10, patients on stable oxygen x 2 days (even if >2L) and an improving trajectory may be considered for discharge
  4. High risk patients may remain in the program longer at provider discretion

Triggers to Consider Sending to ER

  1. Please see Home Acuity Triage. In addition to the Danger Signs listed there:
  1. O2 sat less than 90% at rest on >3L (depending on local practice)
  2. Increase in oxygen requirement of more than 2L in <24hr
  3. Inability to walk 5 steps without becoming severely short of breath
  4. Resting heart rate over 120 or resting respiratory rate >28
  5. Inability to toilet, eat, or navigate the home using bedside devices

Tool: Provider Note Template for Telephone Follow-up with Home Oxygen Patients

Test to TreatCopy Link!

Updated: December 2022

While vaccines remain the most effective way to prevent COVID infection, there are treatments available for those who have gotten infected. A test-to-treat program allows eligible patients to be tested, treated, and prescribed treatment for COVID-19 in one setting in a single visit. In this setting, a patient who tests positive for COVID-19 meets with a healthcare professional who makes a determination if the patient is eligible for oral therapy and, if eligible, provides a prescription (and, in most cases, the actual medication) to the patient. This not only allows for greater access and convenience to COVID treatment but allows for rapid initiation of treatment.

Based on efficacy data, nirmatrelvir/ritonavir (NMV/r) is the preferred agent, followed by molnupiravir.

Eligibility:Copy Link!

In general, patients who test positive and have been symptomatic for < 5 days, are at risk for complications from COVID infection but do not require a higher level of medical care are eligible for a Test to Treat program. Standard, evidence based screening and triage practices should still be practiced with the ability to stabilize and transfer to a higher-level of care if needed. Individuals with mild and moderate illness who do not require hospitalization but also are not eligible for oral antivirals (see criteria below) should be managed supportively and according to evidence-based guidelines.

Assessment:Copy Link!

  • Step 1:
  • Has the patient tested positive for COVID-19 (see Types of COVID tests), including a home test?
  • Is the patient free of signs of severe COVID-19 illness?
  • Worsening dyspnea or SpO2 < 94% on room air
  • New oxygen requirement
  • Respiratory distress
  • Altered mental status
  • Need for additional laboratory or radiologic testing
  • Is the patient less than 5 days from onset of symptoms?
  • Does the patient have high-risk factors?
  • Age ≥ 50
  • BMI ≥ 30 kg/m2
  • Pregnancy
  • Diabetes
  • Sickle cell disease
  • Neurodevelopmental disorders
  • Chronic kidney disease, stage 3b or worse
  • Cardiovascular disease, hypertension, or lung disease
  • Immunocompromising condition (e.g. HIV)
  • Tuberculosis
  • Clinician-determined medical condition, or demographic factor presumed to place the patient at high risk for disease progression

  • Step 2 (if yes to all above in Step 1):
  • Step 2A: Assess patient’s eligibility for nirmatrelvir/ritonavir (NMV/r)--If the answers are YES to each of the below, consider NMV/r for treatment of COVID-19 infection
  • AGE: Is the patient ≥18, OR ≥12yrs AND ≥40kg? (88lbs)?
  • DRUG INTERACTIONS: Confirm the patient is NOT on any drugs that interact with NMV/r and cannot be substituted?
  • Statins – hold 8 days, pitavastatin and pravastatin do not need to be held
  • DOACs—dabigatran and edoxaban likely safe, apixaban seek expert advice, avoid rivaroxaban
  • Alpha-1 blockers – hold tamsulosin and others for 8 days
  • Warfarin —monitor, INR may fall out of therapeutic range
  • Inhaled beta agonists — hold salmeterol for 8 days, formoterol/albuterol fine
  • Calcineurin inhibitors — Avoid if possible, careful monitoring and dose adjustment
  • Calcium channel blockers — monitor and consider dose decrease
  • Antipsychotics — avoid if possible, dose reduction needed
  • Opiates —consider dose decrease by 50-75% for 8 days, except methadone
  • Oral contraceptives— Barrier method recommended until next cycle
  • SSRIs — monitor, toxicity unlikely in short course
  • Triptans — hold eletriptan and zolmitriptan, sumatriptan fine
  • Benzodiazepines— monitor, consider dose reduction, don’t use triazolam
  • Chemotherapy and small molecule inhibitors— review with oncology
  • Oral corticosteroids — monitor, consider 50-75% dose reduction
  • Sildenafil/tadalafil/vardenafil — hold for 8 days
  • Rifampin — concomitant use contraindicated
  • Established ritonavir therapy — do not change established ritonavir dose
  • RENAL or HEPATIC IMPAIRMENT: Is the patient free of severe renal impairment (GFR <30) or hepatic impairment?
  • AVAILABILITY: Is NMV/r available for treatment
  • Step 2B: ineligible for MNV/r: Assess patient’s eligibility for molnupiravir. If the answers are YES to each of the below, consider molnupiravir.
  • Rule out that the patient is pregnant, trying to get pregnant, or breastfeeding?
  • Is the patient > 18 years old?

  • Step 2C: If ineligible for MNV/r or molnupiravir, consider IV antivirals if available and no contraindications.

Individuals who test positive for COVID-19 but are NOT eligible for immediate oral treatment are those with signs of severe illness including the following:

  • Worsening dyspnea or SpO2 < 94% on room air
  • New oxygen requirement
  • Respiratory distress
  • Altered mental status
  • Need for additional laboratory or radiologic testing

Dosing/Drug MonitoringCopy Link!

For pharmacology, dosing, and toxicity, please refer to: Nirmatrelvir/ritonavir and Molnupiravir

Treatment PrioritizationCopy Link!

In many places, the number of patients that are eligible for test-to-treat outweighs the supply of medications. In such situations, the following is a guide to stratify patients based on their risk of progression to severe disease.

Courtesy of USAID COVID-19 Test-To-Treat algorithm

Community Health WorkersCopy Link!

Updated Date: December 20, 2020

While Community Health Workers (CHWs) are in a unique position to help with COVID response, they are also at risk of exposure. Programs that deploy CHWs should consider how COVID will impact their work and assess risk tolerance. This is particularly applicable for CHWs who have frequent patient contact (e.g. routine home visits) and may not have access to PPE. Workflows should be evaluated, and in some instances changed, in order to adequately protect CHWs. This may include measures such as remaining outside and at a distance greater than 2 meters during routine home visits and avoiding activities requiring close physical proximity and/or contact.

Below we outline two strategies for CHW engagement in the fight against COVID. These strategies should be adapted to the local context as well as CHWs’ training, availability, funding, compensation, and access to PPE.

Strategy 1: AdvisingCopy Link!

Most CHWs should be capable of implementing this strategy which does not require them to enter homes, meet in groups, or touch patients.

Intervention

Activities

Community Education

Disseminate information, answer questions, encourage social distancing, inform when to seek care. Specific measures could include distributing fliers at houses; village communication using bullhorns; assess potential cases from a distance; distribution of paracetamol and oral rehydration solution generously (i.e. treatment and trust)

Strategy 2: AccompanimentCopy Link!

This requires sufficient funding, staffing, PPE, data systems, and integration with local health systems. Teams should map catchment areas, divide areas, and relay information systematically. Known cases should be communicated with the coordinating authorities.

Intervention

Activities

Case Finding

Screening in communities at risk. Some CHWs may also be able to do home testing (see below).

Contact Tracing

Tracing contacts and household members of known cases. Following up and assessing for symptoms. Facilitating referral to a facility when necessary.

Home Testing

Performing Rapid Tests for contacts or other community members meeting testing criteria (typically a Positive Symptom Screen) may be possible for some CHWs. This depends on training and local regulations. Some tests will be similar to rapid malaria tests (which may require little additional training for CHWs familiar with these tests), and some may be nasal swabs (this may require some additional training).

Home Based Care

Ensuring understanding of quarantine, hygiene, and distancing protocols. Conducting routine check-ins (at least twice a week) to monitor for worsening symptoms and need for referral to a facility. Facilitating transport when severe cases are identified

Chapter 7

Inpatient Management

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

MonitoringCopy Link!

Whether or not a patient needs admission is addressed in Disease Severity and Disposition.

Lab, Vitals, and ImagingCopy Link!

Lab Monitoring

Vitals and Monitoring

Chest Imaging

Cardiac Diagnostics

Medical ManagementCopy Link!

Updated Date: August 11, 2021

Although the clinical course of patients with COVID-19 is variable, there is evidence that the sickest patients do not develop severe disease until 7-14 days after their symptoms start. Because of this, clinicians should monitor inpatients closely for signs of worsening respiratory status even if a patient has been stable for several days.

MedicationsCopy Link!

Fluid ManagementCopy Link!

  • Fluid management should be conservative (small boluses of 250-500cc, with monitoring of response (urine output, hemodynamics). If possible, avoid maintenance fluids and large boluses due to risk of potentially exacerbating gas exchange and hypoxemia. As highlighted in the FACTT Trial of conservative vs. liberal fluid strategies, a conservative fluid strategy improved oxygenation, more ventilator free and ICU free days. Please see Septic Shock for more information on dynamic fluid management

Venous Thromboembolism ProphylaxisCopy Link!

  • There are multiple reports that patients with COVID-19 have a high incidence of deep vein thrombosis/venous thromboembolism (DVT/VTE). We recommend standard dose VTE prophylaxis in most patients, except in several situations. See Anticoagulation for more details.

Geriatric PatientsCopy Link!

Literature Review (Geriatrics): Gallery View, Grid View

  • Include FRAIL Frailty Screen on initial assessment, and consider a geriatrics consult if the patient has a concern for delirium, dementia, or failure-to-thrive at home.

Respiratory ComplicationsCopy Link!

Other ComplicationsCopy Link!

Oxygen CareCopy Link!

Updated Date: December 20, 2020

Oxygen Escalation PathwayCopy Link!

Management of hypoxemia in COVID-19 requires selection of an initial appropriate oxygen delivery system (e.g. nasal cannula), with escalation to a different system (e.g. simple face mask) capable of higher oxygen flow if the patient worsens or is unable to reach their target oxygen saturation (SpO2). Effective oxygen therapy is about finding a balance between delivering the lowest amount of supplemental oxygen in order to achieve normal oxygen saturations for the patient. Hypoxemia is harmful to patients, but so is giving too much oxygen (Girardis et al).

Goals of Oxygen TherapyCopy Link!

  • Initiation
  • Initiate oxygen if SpO2 is below ~94%
  • If SpO2 is not available, initiate oxygen for patients with tachypnea (RR above 22) or increased work of breathing
  • Maintenance
  • Target SpO2 92-96% on oxygen
  • 88-94% in patients with oxygen-dependent COPD
  • If SpO2 is not available, one may consider empiric escalation of oxygen therapy for tachypnea or increased work of breathing. However, these signs are weak surrogates for SpO2 when titrating oxygen therapy.

Oxygen Escalation PathwayCopy Link!

Tool: Partners In Health Oxygen Pathway

Tool: Open Critical Care Respiratory Care Pocket Reference

  1. Encourage self-proning if there are no contraindications.
  1. Proning can be used with all types of oxygen delivery systems, or for patients on no oxygen. For all oxygen systems, and non-invasive in particular, it is important to be able to monitor the patient closely enough to ensure the risks of proning do not outweigh the benefits.
  1. If patient’s SpO2 is below 94% (or if patient is tachypneic, if no pulse oximetry is available) initiate oxygen therapy
  1. Deliver by nasal cannula at 1-6 L/min
  1. If oxygen goals are not met by nasal cannula at <6 L/min then consider one of the following:
  1. Simple facemask at 6-10 L/min OR
  2. Oxymizer pendant or mustache at 6-12 L/min OR
  3. Venturi face mask at FiO2 0.4-0.6 (40%-60%)
  1. Unlike simple and non-rebreather facemasks where you set the oxygen flow rate, with Venturi masks you set the percent of oxygen (e.g. 40%). The percent of oxygen is controlled using a valve attached to either the mask or the flowmeter. First, select and attach the valve that corresponds to the correct FiO2 (or setting the percent of oxygen if the valve is adjustable). The markings on the valve will instruct you what flow rate to set. Because the valve blends pure oxygen with room air, the actual flow delivered to the patient will be higher than the flow set on the flowmeter.
  1. If oxygen goals are still not met with the above options, consider escalation to the following:
  1. Non-rebreather facemask (at 10-15L/min, do not go below 10L or carbon dioxide can be retained in the mask)
  1. For patients with severe hypoxemia, some clinicians will place a non-rebreather facemask on top of a nasal cannula. This is used when more intensive oxygen delivery systems (e.g. high flow nasal cannula, non-invasive, and intubation) are unavailable.
  1. If oxygen goals are still not met, consider one of the options in the table below. The clinical situation, availability of options at your institution, and goals of care discussions should guide selection.

Option

Ideal Candidate

Contraindications (Many are Relative)

High Flow Nasal Cannula

Patient with hypoxemia without severe work of breathing or increasing pCO2

Patient Factors:

  • Significant facial trauma or deformity
  • Unavailability or inadequate oxygen supply to complete treatment
  • Need for emergent intubation (if within goals of care)

Institutional Factors:

  • Inability to administer IPC as required by your institution

Non-Invasive Positive Pressure Ventilation (BIPAP or CPAP)

Similar criteria as for non-COVID patients, (e.g. flash pulmonary edema, heart failure, OSA, COPD flare) and those likely to only need it for a short period. Use of prolonged NIPPV in COVID19 patients remains an area of uncertainty with limited data to guide practice, potential risks to patients and HCWs and considerable practice variation.

Patient Factors:

  • Recent esophageal or gastric surgery
  • Upper gastrointestinal bleeding
  • Facial or neurological surgery, trauma, or deformity
  • Airway obstruction (eg, laryngeal mass or tracheal tumor)
  • Inability to follow commands, protect airway, or clear secretions (eg, patients at high risk or aspiration)
  • Need for emergent intubation (if within goals of care)

Institutional factors:

  • Inability to administer IPC as required by your institution
  • Limited staffing/inability to monitor

Intubation

See Candidacy. Criteria in COVID are similar to other patients (not early intubation as had been practiced earlier in the epidemic)

Not available or not within goals of care

Focus on Comfort Measures

Patients who do not want aggressive measures or escalating interventions will be unlikely to accomplish meaningful clinical benefits. The definition of “meaningful clinical benefits” will vary among people and places.

Cultural norms and legal rules vary widely.

Oxygen Weaning PathwayCopy Link!

For patients on nasal cannula attempt weaning at least once a day:

  1. Wean oxygen completely to off while monitoring at bedside with pulse oximetry, for at least 5 minutes (unless the patient rapidly desaturates)
  1. If oxygen saturation falls below SpO2 target (92% if no target specified), restart the oxygen at the lowest flow rate necessary to meet the patient’s clinical (SpO2) goal.
  2. If a patient maintains saturations above the clinical target without oxygen, oxygen therapy may be discontinued.
  1. Check oxygen saturation 30 minutes later and then again at 1 hour to ensure saturation remains adequate without oxygen therapy.

For stable patients on simple, Venturi, or non-rebreather facemasks: attempt weaning at least once a day by decreasing oxygen flow until goal oxygen saturation is met.

  1. Minimum oxygen flow rates are required for non-rebreather face masks and Venturi face masks to function properly, so do not decrease below the manufacturer recommended flow. Switch to a lower intensity oxygen delivery device once a patient is stable on the minimum flow rate for their current oxygen delivery device.
  1. Simple facemask: Minimum flow rate is often 4 to 5L/min. At this setting, the next step in oxygen weaning is to switch to nasal cannula at 5 to 6L/min.
  2. Venturi facemask: Minimum flow rate depends on the oxygen concentration setting (FiO2). In general, once a patient is stable on 40%, they are ready to attempt switching to nasal cannula at 5 to 6 L/min.
  3. Oxymizer: There is no minimum flow rate, but once a patient is stable on 4 to 5 L/min they can be switched to nasal cannula at 5 to 6 L/min.
  4. Non-rebreather Facemask: Minimum is often 10L per minute. At this setting, the next step is simple facemask at 10 L/min , or, if a simple facemask is not available, nasal cannula at 5 to 6 L/min.

Oxygen Delivery DevicesCopy Link!

Literature Review (Oxygen Delivery): Gallery View, Grid View

Concerns about AerosolizationCopy Link!

Updated Date: December 20, 2020

The degree to which different oxygen delivery devices are thought to cause aerosolization remains an area of active research, and the exact amount of aerosolization in each situation is not known. Patient factors like coughing (which produces aerosols) and viral load, as well as the dynamics of droplet particle size and dispersion, make this quite complex (Klompas et al). Meaningful distinction between “safe” and “unsafe” levels of aerosols at this point is not possible. See Aerosols, Droplets, and Fomites.

Flow rate: Lower oxygen flow rates hypothetically should reduce aerosols. However, a preprint study in healthy volunteers showed that there was no variation in aerosol level between room air, 6L/min nasal cannula, 15 L/min non-rebreather, 30L/min high-flow nasal cannula and 60 L/min high-flow nasal cannula regardless of coughing (Iwashyna et al).

This chart provides an overview, but is subject to change. Please follow your institution’s IPC Practices regarding droplet or aerosol precautions. Aerosol Generating Procedures (AGPs) are discussed here.

Device or activity

High risk of aerosolization?

More information

Coughing

Yes

There is high aerosol generation with cough: 35 fold more aerosols than are generated during extubation (Brown et al). Cough-generated aerosols rapidly spread throughout a room within 5 min (Lindsley et al). In one study, coughing was associated with 10 times greater aerosols than speaking or breathing (Hamilton et al).

Nebulizers

Yes

By design, nebulizer therapy produces aerosol particles. However, the bacterial burden in these particles appears low (O’Neil et al). It is not yet clear what the viral burden in these particles is. Jet nebulizers produce sideways aerosol dispersion between 45-80 cm (Ferioli et al). Evidence for HCW infection risk is inconsistent with 2 of 3 cohort studies found “some association” with therapy (Tran et al). Anecdotal report of COVID-19 infection associated with nebulizer therapy without use of PPE (Heinzerling et al).

Nasal Cannula

No

Simple Masks and Non-rebreathers

No

Venturi Masks

Depends on Humidification

Aerosol dispersion ranges from 30-40cm (Ferioli et al).

High Flow Nasal Cannula*

Unknown

Aerosols did not significantly increase with the use (up to 50LPM) in one small study of 10 healthy volunteers (Gaeckle et al). A different study did find that HFNO emits aerosols, but these were small (<1μm) particles generated by the machine and then passed into the patient, not coalescing with respiratory particles, and thereby unlikely to carry virus particles. (Hamilton et al)

BIPAP or CPAP*

Unknown

Aerosols did not significantly increase with the use (up to 20/10 cmH20) in one small study of 10 healthy volunteers (Gaeckle et al). In one study, CPAP (with exhalation port filter) produced less aerosols than breathing, speaking and coughing (even with large >50L/m leaks) (Hamilton et al).

By type of mask:

  • CPAP with orofacial vented mask: unable to determine smoke dispersion because it occurred equally in all directions.
  • CPAP with nasal pillows: increasing air dispersion with increasing positive pressure. At CPAP of 20 cmH2O a maximum dispersion ~25-35 cm depending upon brand of pillow interface.
  • NIPPV with full face mask: Dispersion of 60-70 cm (single limb circuit at inspiratory/expiratory pressures of 15/5 cmH2O) depending upon degree of lung injury. Dispersion of ~90 cm observed at peak pressures of 23 cmH2O.
  • NIPPV with helmet: Dispersion of 15-17 cm at inspiratory/expiratory pressures of 22/10 cmH2O. Dispersion distance of 18-27 cm observed at peak inspiratory pressure of 30 cmH2O depending upon the degree of lung injury.
  • Double circuit and tight cushion connection at the head-neck interface associated with negligible dispersion (Ferioli et al).

Choosing a Delivery DeviceCopy Link!

Tool: Oxygen Demands of Delivery Devices (if O2 supply is limited)

Tool: Open Critical Care Introduction to Oxygen Delivery Devices

Tool: ICRC-WHO Basic Emergency Care Workbook, pages 154-155
Tool: Oxygen Therapy Escalation Algorithm

It is important to know the oxygen supply capability at your facility as well as the consumption rates for different delivery devices. Depending on a facility’s oxygen supply type (liquid oxygen versus cylinders versus an oxygen generating pressure swing adsorption plant) some oxygen delivery devices may not be practical. Even relatively well-resourced facilities with liquid oxygen can exhaust supplies when ramping up use of devices like high-flow nasal cannula during a surge census. The tools above can help you determine which delivery device to use and the flow rate needed.

Estimating Fraction of Inspired Oxygen (FiO2)

Oxygen Device

O2 Flow (L/min)

FiO2

Nasal Cannula

1

0.24

2

0.28

3

0.32

4

0.36

5

0.40

Simple Facemask

6-10

0.44-0.50

Non-Rebreather Mask (reservoir must be fully inflated)

10-20

Approx 0.6-0.8

At RR ~20 and Tidal Volume ~500

20 LPM flow = ~60% FiO2

30 LPM flow = ~70% FiO2

40 LPM flow = ~80% FiO2 (Farias et al).

The values represent estimates of FiO2. Actual FiO2 delivered is dependent on multiple factors including oxygen supply quality and patients minute ventilation. One general estimation rule is using oxygen flow rate: FiO2 =0.21 + 0.03 x oxygen flow rate in L/min (Frat et al).

High-Flow Nasal Cannula (HFNC)Copy Link!

Updated Date: August 11, 2021
Literature Review:
Gallery View, Grid View

When available, high-flow nasal cannula is one option for selected patients for whom non-rebreather or Venturi mask is not adequate to maintain goal oxygenation. While standard cannulas and masks can provide flow rates of up to 15 liters per minute, an HFNC system delivers oxygen flow rates as high as 80 L/min with variable concentrations of oxygen up to 100%.

In general, HFNC has been demonstrated as an effective intervention for management of acute hypoxemic respiratory failure, improving survival (Frat et al) and reducing the need for mechanical ventilation (Ferreyro et al).

  • Several small studies show COVID-19 patients might avoid intubation using high-flow nasal cannula (HFNC) (Demoule et al). This is especially true with concomitant proning. (Tu et al; Despres et al; Xu et al).
  • In one study of 293 COVID patients in South Africa, 47% percent were weaned off of HFNC and did not require intubation (Calligaro et al).

For COVID, Indications for Use Might Include:

  • A patient who is not meeting oxygenation goals on escalating therapies (e.g. facemask, venturi mask, or non-rebreather) and does not meet criteria for intubation
  • A patient who is not meeting oxygenation goals despite maximal oxygen therapy AND intubation is either not available or not within goals of care,
  • The patient does not need significant assistance with work of breathing or hypercapnia (HFNC helps oxygenation but does not tend to help ventilation)

Contraindications:

  • Patient cannot wear or tolerate device
  • Increasing work of breathing or rising pCO2 should prompt a discussion about need for intubation
  • Inability to protect airway, or significant apnea
  • Inadequate facility oxygen supply

Infection Control Implications:

  • High flow nasal cannula is often labeled as an aerosol generating procedure (perhaps better stated as an aerosol enhancing device); However, data to support this notion or quantify risk to healthcare workers remain evolving; Nonetheless, all patients on HFNC should be required to wear a surgical mask over the cannula (Leung et al; Ferioli et al).
  • Heated and humidified oxygen must be used to avoid drying of mucous membranes and secretions to prevent ciliary damage.
  • Start with lower flow rates if possible to minimize potential aerosols
  • Transport on HFNC is often not logistically possible, so conversion to non-rebreather is recommended. In addition, non-rebreather may generate less aerosol.

Technical Use Recommendations:

  • Standard HFNC systems usually consist of a high capacity flow meter, an air-oxygen blender (typically connected to wall air and oxygen sources), tubing, cannula, and a heater-humidifier.
  • Some HFNC systems require a connection to high-pressure air in addition to high-pressure oxygen sources. There are also several systems which do not require wall air and entrain room air instead (either by Venturi effect or turbine)
  • Humidification: For optimal patient comfort and adherence, HFNC systems should deliver gas to the patient at 44 mg H2O/L or 100% relative humidity (Spoletini et al; Restrepo et al).
  • HFNC consumes significant amounts of oxygen. For example, a patient receiving HFNC at 50 L/min and 0.8 FiO2 will consume approximately 37 L/min of oxygen and 13 L/min of air. A J-type oxygen cylinder (1.45m height) contains 6800 liters of oxygen. At this rate, the cylinder would last less than 3 hours.

Tool: Open Critical Care Intro to Oxygen Delivery Devices

Tool: Open Critical Care Calculator for Duration of Oxygen Supply

Non-invasive Positive Pressure Ventilation (NIPPV)Copy Link!

Updated Date: May 16, 2021
Literature Review:
Gallery View, Grid View

When available, non-invasive positive pressure ventilation (e.g. CPAP, BiPAP) can be considered for patients with the indications for which it would normally be used (e.g. OSA, COPD flare) whether or not they have COVID.

In some institutions NIPPV is not a preferred method of delivering oxygen in worsening COVID-19-related pneumonia/ARDS, though this is an area of active research and recommendations are often changing. To see a summary of different guidance institutions recommendations, see our dashboard.

  • Some early studies indicate it may help avoid intubation, though mortality statistics remain unknown: In one study of 47 patients about a third of patients treated with CPAP were able to avoid intubation (Alviset et al). In another study of 53 patients, 83% were successfully treated with NIPPV (Brusasco et al). Careful patient selection is likely important in determining candidacy and ultimately success.
  • NIPPV may also be a way to help avoid ICU capacity overload and manage some patients on the floor (Lawton et al).
  • Patients on NIPPV need to be closely monitored as high tidal volumes or work of breathing may risk patient-induced lung injury in ARDS (Brochard).
  • Helmet NIV has been shown to be equivalent to high flow nasal cannula in moderate to severe hypoxemia. Greico et al showed no significant difference in the number of days free of respiratory support within 28 days, but did show decrease rate of endotracheal intubation and number of days free of invasive ventilation in the helmet NIV group.

For COVID, Appropriate Indications Include:

  • A patient has increased work of breathing or increasing pCO2 despite maximal oxygen therapy (including HFNC if available) AND intubation is either not available, not within goals of care, or not advised by the clinician caring for the patient.
  • Similar indications as in non-COVID-19 patients:
  • Obstructive Sleep Apnea or Tracheobronchomalacia: Patients on home nocturnal CPAP or BiPAP should continue nocturnal NIPPV.
  • Pulmonary edema
  • COPD exacerbation and other reversible hypercapnia

Contraindications:

  • NIPPV should be generally avoided in the same situations that NIPPV is avoided in COVID-19 negative patients (e.g., severe ARDS without short-term reversibility; the presence of relative contraindications such as altered mental status, aspiration risk, secretions).

Infection Control Implications:

  • Some institutions require that NIPPV be done with aerosol precautions, others do not. This is an area of active research.

Technical Use Recommendations:

  • Ensure masks/devices fit well and there is minimal air leak (which can cause significant lateral air travel (Hui et al). Full mask is preferred over nasal-only masks.

Other Considerations:

  • Prolonged use of NIPPV has been linked to malnutrition and should be monitored (Turner et al).

Prone PositioningCopy Link!

Updated Date: December 20, 2020
Literature Review (Proning):
Gallery View, Grid View
Literature Review (Self-proning):
Gallery View, Grid View
Tool:
Prone positioning protocols and checklist
Tool: Video Demonstration of Proning Technique

Benefits, Risks, and CandidacyCopy Link!

Benefits of Proning

Proning is thought to provide physiologic benefits for patients with COVID-19: It improves recruitment of alveoli in dependent areas of the lungs and may improve perfusion to ventilated areas, improving ventilation-perfusion mismatching. Typically proning is used in ventilated ICU patients, however the same benefits may be found in non-ventilated patients.

  • Intubated Proning: Proning is one of the mainstays of ARDS therapy for intubated patients, showing both 28 day and 90 day mortality benefit in the PROSEVA 2013 trial (Guerin). See also Proning of Intubated Patients.
  • Self-proning (non-intubated) in non-COVID patients: Small non-COVID-19 patient cohorts (ARDS, post-lung transplant, and post-surgery) showed association with lab, radiographic, or clinical improvement. In one observational study, (Scaravilli et al) 15 patients with pneumonia underwent a total of 43 self-proning procedures. Of the 43 procedures, 24 were performed with O2 mask, 1 with HFNC, 11 with helmet CPAP, 7 with NIPPV, with an average duration of 3 hours, range 2-8 hours. They found improvement in PaO2 and in P/F ratio: Pre-proning P/F 127 +/- 49 --> Prone 186 +/- 72 --> Post 141 +/- 64 (p < 0.05) without complications. In another limited study of 20 patients (Ding et al) with ARDS with P/F < 200 requiring HFNC or NIPPV of at least PEEP of 5 and FiO2 of 0.5 who underwent self-proning for at least 30 minutes, many fewer (45%) required intubation than would have been expected based on previous data (75%).
  • Self-Proning in COVID-19. Multiple trials have shown benefit in oxygen with proning (Coppo et al; Weatherald et al). A randomized trial of 1121 patients showed that awake proning improved outcomes: the hazard ratio for intubation was 0.75 (0.62−0.91), and the HR for mortality was 0.87 (0.68−1.11) compared with standard care. (Ehrmann et al).

Risks of Proning

  • Airway Obstruction (particularly if a patient is unconscious but not intubated)
  • Dislodged Oxygen Delivery Device
  • Facial Edema
  • Pressure Ulcerations (especially the forehead and anterior chest)
  • Pressure Neuropathies
  • Patient Intolerance
  • Intracranial Hypertension

Patient Selection

Self-proning can be used on stable patients (on room air or supplemental oxygen) and as a “rescue” for those who have escalating supplemental O2 requirements.

  • Self-proning can be done with any type of oxygen delivery system with careful consideration and ideally multidisciplinary discussion for safety. At higher levels of oxygen, the patient may require more frequent monitoring (see protocol below).
  • The patient should ideally be able to move independently and have the cognitive and physical status to supinate themselves if they become uncomfortable. This includes the ability to safely manage their supplemental oxygen, IV tubing, SpO2 monitor and other leads and attachments (within reason).
  • In certain situations, patients who are unable to position themselves may be candidates for assisted proning as a “rescue” therapy. For example, assisted proning can be considered if a patient has a low SpO2 (below 92%) on non-rebreather facemask, and HFNC, NIPPV, and intubation are all unavailable or contraindicated (see below for additional considerations).

Contraindications:

  • Absolute:
  • Inability to Supinate or Pronate Safely (see above - exception is Assisted Proning)
  • Imminent Risk of Intubation (see “when to stop self-proning”)
  • Spinal Instability
  • Facial or Pelvic Fractures
  • Open Chest or Unstable Chest Wall
  • Open Abdomen
  • Unstable Airway (Patient with oral swelling, mass, tumor or other object obstructing the airway)
  • Unresponsive Patient (May be more likely to obstruct their airway)
  • Intracranial Pressure Monitoring or Intracranial Hypertension
  • Hemodynamic Instability (Blood pressure less than 80/40 or active up-titration of vasopressors)
  • Relative Contraindications:
  • Altered Mental Status
  • Nausea or Vomiting
  • Non-invasive Positive Pressure Ventilation
  • Copious Secretions
  • Signs of Severe Respiratory Distress (Tripod position or obvious severe accessory respiratory muscle use)
  • Agitation
  • Pregnancy
  • Supporting Lines or Tubes at High Risk for Displacement (for example, a chest tube).

Awake Proning ProtocolCopy Link!

For intubated patients, please see Proning of Intubated Patients.

Awake non-intubated proning requires careful attention to a number of steps:

  1. Monitoring
  1. Oxygen monitoring: Although many patients experience improvement in oxygenation with pronation, it is possible that some patients may get worse. Therefore, it is important to have a plan for patient monitoring during pronation. However, the type and frequency of monitoring during pronation will vary by facility and patient.
  1. Continuous SpO2 monitoring is recommended if available.
  2. If continuous SpO2 monitoring is not available, we recommend checking SpO2 10 minutes after pronation to ensure stability. Although some patients will temporarily have a slight worsening in vital signs immediately after pronation, HR, BP, and SpO2 should return to close to baseline within 10 minutes. After the first 10 minutes, the interval of monitoring can be extended based on the clinical context.
  1. Telemetry: If telemetry is indicated, EKG leads can remain on anterior chest wall for continuous monitoring, avoiding pressure points.
  1. Prior to Proning
  1. Make plans in advance for toileting, contacting nurses, and cellular phone if patient has one.
  2. If possible, place the bed in reverse Trendelenburg (head above feet, 10 degrees) to help reduce intraocular pressure.
  3. Have patient empty bladder.
  4. Educate the patient.
  1. Explain the procedure and rationale of the intervention to the patient. “Lying on your stomach is what we call prone position. This position can improve your breathing, helping your lungs to expand to get oxygen to the rest of your body. It may help you feel better.”
  2. Point out any IV tubing or oxygen tubing they are connected to. Remind them this tubing should not be under them at any time.
  3. Instruct patient to roll back over and call for help if they feel worse.
  1. Arrange tubing to travel towards the top of the bed, not across the patient, to minimize risk of dislodging. Ensure support devices are well-secured to the patient. (Eg. sleeve over IV access site, position urinary catheter)
  2. Assess pressure areas to avoid skin breakdown and dress any wounds and use skin protective devices as needed.
  3. If the patient will require assistance, assess the patient’s size and weight to determine adequacy of the bed frame and the mattress in addition to the number of staff required to safely turn the patient.
  1. Prone Position
  1. The patient should lay on their abdomen (arms at sides or in “swimmer” position). Insert head supports (e.g. rolled sheets) to ensure that the head is high enough off the bed to allow for proper spinal alignment in either face down or side lying position. Position arms slightly above the head bent at the elbow. Place pillows or rolled sheets under the shins to flex the knees and allow the feet to be at a 90-degree angle. Utilize rolls to support shoulders, abdomen and pelvis where necessary. Pillows may be required to support the chest. If this is not a tolerable position, they can try laying on their side, though this may not work as well (Bentley et al).
  1. Show the patient how to choose which side to roll to so that they avoid any IV tubing and how to adjust oxygen delivery device and pillows as needed
  1. If a patient is unable to tolerate, they may rotate to lateral decubitus or partially prop to the side (in between proning and lateral decubitus) using pillows or waffle cushioning as needed. Ideally the patient should be fully proned rather than on the side as there is currently no data about whether side positioning is beneficial.
  1. Time Spent Proning
  1. Patient should try proning every 4 hrs and overnight, and stay proned as long as tolerated (ideally at least 30 minutes). Our ideal goal is 16 hrs per 24 hours (e.g. 4 times for 4 hours each session) based on common interpretations of the PROSEVA trial (Guerin). However, we realize that few (if any) patients will tolerate 16 hrs of proning per 24 hrs.
  1. Perform range of motion or repositioning of arms and legs every 2 hours
  1. When to Stop Proning
  1. We recommend continuing daily cycles of proning until the patient is on nasal cannula (<4 L/min) with SpO2>92%. The patient may choose to stop self-proning at any time
  2. Stop proning if any of the following occur:
  1. Patient intolerance. Do not administer sedation to facilitate proning.
  2. Inability to maintain SpO2 > 87% or escalating oxygen requirements concerning for potential need for intubation
  3. Development of hemodynamic instability (BP < 90/50 or HR > 140 in an adult)
  1. Assisted Proning as a Rescue Therapy
  1. Assisted/rescue proning can be used in situations where a patient is unable to position on their own and oxygen is unavailable or a patient is on the highest available level of oxygen. In these situations, the benefits of proning may outweigh the risks.
  2. The risks and benefits of assisted/rescue proning should be reassessed on a regular basis. For instance, if there is no improvement and/or close monitoring is not possible, supination may be considered in order to avoid complications (e.g. pressure ulcers). If the patient has improved but frequent monitoring is not possible, the team should discuss the risks and benefits of maintaining a prone position.
  3. Reposition the patient’s arms every two hours
  4. Perform range of motion to arms and legs every 2 hours
  5. Assess the skin frequently for areas of non-blanchable redness or breakdown, with special attention to the nose.

Triage to ICUCopy Link!

Sometimes it may be necessary to re-triage an inpatient to the ICU if they are worsening. Please see Disease Severity and Disposition for sample ICU triage criteria.

Medical DocumentationCopy Link!

Tool: Charting Tools and Templates (OCC). Contains 5 charting tools and templates that are intended to be downloaded and modified by local providers who are caring for patients with respiratory failure or critical illness in the intensive care unit (ICU) or non-ICU settings.
Tool: Adult Ventilator Protocols and Order Set Templates (OCC). Can be modified and used for ventilator management of adult patients with ARDS. It includes ARDS Net lung protective ventilation as well as orders for spontaneous breathing trials (SBTs), difficult to wean patients and cuff leak tests.

Hospital DischargeCopy Link!

Updated Date: June, 2020
Literature Review:
Gallery View, Grid View

Once a patient is breathing without oxygen and able to perform basic functions, the patient can be discharged if there is adequate ability for isolation (if still necessary) and adequate follow-up plans and social support.

Discharge CriteriaCopy Link!

Discharge requirements vary depending on the hospital. Consider discharge for patients who meet the following clinical criteria:

  • Resolution of Fever >48 hours without antipyretics
  • Oxygen Saturation ≥ 94%. In some places, patients may be discharged on oxygen (See Home Oxygen Delivery).
  • Respiratory Rate < 22.
  • Blood Pressure > 90/60.
  • No signs of increased work of breathing or respiratory distress.
  • Improvement in signs and symptoms of illness (cough, shortness of breath, and oxygen requirement)

Discharge NeedsCopy Link!

  • Confirm patient’s ability to understand and adhere to home isolation instructions
  • Confirm patient’s ability to manage daily activities with current level of support at home
  • Confirm patient has resources/social support to receive food and other necessary supplies for the duration of quarantine
  • Provide a surgical mask to all infected patients who are discharging home
  • Verify patient has a safe plan for transportation or figure out alternate transportation (infected person should wear mask in vehicle)

Discharge Against Medical AdviceCopy Link!

People are able to sign themselves out of the hospital against medical advice if they demonstrate Decision-Making Capacity. Please see Leaving Against Medical Advice for full information.

Discharge on OxygenCopy Link!

This is covered in Home Oxygen Care

FollowupCopy Link!

See Follow-up and Monitoring

Chapter 8

Critical Care Management

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

MonitoringCopy Link!

Whether or not a patient needs ICU level of care is addressed in Disease Severity and Disposition.

Lab, Vitals, and ImagingCopy Link!

Lab Monitoring

Vitals and Monitoring

Chest Imaging

Cardiac Diagnostics

Respiratory Failure in COVIDCopy Link!

Updated Date: December 20, 2020

Acute Respiratory Distress SyndromeCopy Link!

Literature Review (Acute Lung Injury): Gallery View, Grid View
Literature Review (ARDS):
Gallery View, Grid View

Tool: COVID-19 Guidelines Dashboard

Tool: Respiratory Care Quick Reference Card

Tool: Respiratory Care Protocol Templates

One of the most severe complications of COVID-19 is Acute Respiratory Distress Syndrome (ARDS). ARDS is an acute clinical syndrome associated with inflammation and damage to the lungs. In ARDS, lungs become stiff and their ability to oxygenate the blood is impaired. Worldwide, mortality rates for ARDS are estimated to be 35-46% (Bellani et al). Because ARDS has implications for the management and treatment of respiratory failure, it is important that clinicians are able to recognize and diagnose it.

Most patients with COVID-19 who require ICU care develop ARDS. See the Pathophysiology section for more information about why COVID causes ARDS. From onset of symptoms, the median time to:

COVID ARDS mortality is thought to be around 39% (95% CI 23-56%) according to a meta-analysis of studies covering more than 10,815 COVID patients with ARDS internationally (Hasan et al).

The Berlin Definition of ARDSCopy Link!

The Berlin definition of ARDS requires the following four criteria:

  1. Acute onset (within 1 week)
  2. Bilateral opacities on chest xray or CT
  3. PaO2/FiO2 ratio <300mmHg with a minimum of 5 cmH20 PEEP (or CPAP)
  4. Must not be fully explained by cardiac failure or fluid overload

The severity of ARDS is graded using this scale:

Severity

PaO2/FiO2 (on PEEP/CPAP >5)

Mortality (all cause, cohort)

Mild

200-300

27%

Moderate

100-200

32%

Severe

<100

45%

Kigali Modification of Berlin Criteria for ARDSCopy Link!

The Berlin definition of ARDS has limited applicability in settings where blood gas analyzers, chest radiographs, and/or positive pressure ventilation are not reliably available. The Kigali Modification to the Berlin criteria has been developed to address this gap (Riviello et al).

The Kigal modification requires the following four criteria to diagnose ARDS:

  1. Acute onset (within 1 week or less)
  2. Bilateral opacities on chest xray or ultrasound not fully explained by pleural effusions or masses
  3. SpO2/FiO2 <315 (no PEEP requirement; SpO2 must be <=97% for accurate estimation by this method).
  4. Must not be fully explained by cardiac failure or fluid overload

Key differences in the Kigali Modification include allowing ultrasound as a method of identifying pulmonary opacities, the use of SpO2 instead of PaO2, and the lack of requirement for PEEP. Several studies have validated the use of SpO2 in place of arterial blood gases (Chen et al; Sanz et al; Brown et al) and ultrasound in place of chest xray for diagnosing ARDS (Lichtenstein). One single center study has validated the modification in ventilated patients (Vercesi et al). However, a large validation study comparing the Kigali Modification to the Berlin Criteria is needed.

When patients are not mechanically ventilated, the FiO2 will need to be estimated.

Tool: Management of Respiratory Failure where access to arterial blood gases is limited

Tool: Respiratory Care Pocket Reference (English) (Español)

Tool: Imputed PaO2 (from SpO2) Calculator
Tool:
Respiratory Care Protocol Templates

Supplemental OxygenCopy Link!

Updated Date: August 11, 2021

See relevant sections in Inpatient Management (listed below) for options in patients that do not require intubation and mechanical ventilation. Some of these can be performed in the ICU or outside of the ICU depending on the institution:

IntubationCopy Link!

Updated Date: August 11, 2021
Literature Review:
Gallery View, Grid View

Tool: Anesthesia Pocket Guide

CandidacyCopy Link!

There are several potential indications for intubating a patient with respiratory failure. These include:

  • Persistent or rapidly worsening hypoxemia despite maximal oxygen therapies
  • Ventilatory failure (e.g. hypercapnia, fatigue, apnea or obstructive disease)
  • Severe work of breathing
  • Altered mental status that impairs either ability to protect airway or comply with oxygen therapies
  • Mechanical airway obstruction
  • Presence of a shock state
  • Presence of severe acidosis

Ultimately, the decision to intubate is based on multiple factors including the patient’s complete clinical scenario (including goals of care) as well as the availability of local resources to safely manage the airway and provide mechanical ventilation. Of note, early in the pandemic there were anecdotal reports advocating for earlier intubation of COVID patients as compared to other patients with respiratory failure from other causes. However, there are no data to support this practice. Intubation criteria for COVID patients with respiratory failure are generally the same as for non-COVID patients with respiratory failure.

Intubation ProcedureCopy Link!

Tool: Ventilator filter and humidification placement visual aid

PreparationCopy Link!

  1. Provider protections. Intubation is potentially a high risk aerosol generating procedure.
  1. Treat all intubations as a presumed COVID positive patient unless they have been fully ruled-out for COVID.
  2. Intubation should be done in a negative pressure room whenever possible (SCCM). Negative pressure rooms remove viral aerosolized particles at different rates based on the air changes/hour(ACH). OR’s are mandated to achieve at least 15 ACH’s yielding 99% airborne viral removal in 18 minutes. This is different for ICU’s and you may want to contact your facilities engineers to clarify ACH for your ICU beds. Calculate time necessary for your facility following CDC guidelines(Negative Pressure Airborne Clearance Times) and facility recommendations
  3. PPE:
  1. Intubating with the necessary PPE is often unfamiliar/difficult to many providers - consider practicing via simulation (APSF) and/or have it performed by the most experienced provider available
  2. Intubation should be done under Airborne Precautions. recommendations include disposable hair bouffant or cap, eye protection (face shield only vs face shield AND protective eyewear), either N95 or PAPR (N95 + hood for neck protection), fluid resistant gowns, double gloves, leg protection (boot covers) to below the knee These recommendations exceed the standards of the American Society of Anesthesiologists on 3/20/2020, the Society of Critical Care Medicine on 3/20/2020, and the Anesthesia Patient Safety Foundation on 2/12/2020
  1. Collect Materials: With the exception of the laryngoscope, DO NOT bring the following equipment into the room - only remove what you may need and discard or sterilize materials taken into the room after intubation even if not used. However, it is important that this equipment is easily and quickly accessible during intubation in case a difficult airway is encountered.
  1. Airway Boxes (e.g. nasopharyngeal airways, oral airway, syringes, needles, laryngeal masks, “bougie” stylet, extra endotracheal tubes (ETTs) 6.0-8.0 for adults)
  2. Medication Boxes (e.g. paralytics, vasopressors (e.g. phenylephrine, ephedrine, epinephrine, norepinephrine), lidocaine, labetalol, esmolol, propofol/etomidate, midazolam)
  3. Laryngoscope We recommend dedicated video laryngoscope if available, or the laryngoscope that is most familiar to the provider.
  1. Set up the ventilator (or bag valve mask in select circumstances):
  1. Correct placement of HEPA (bacterial/viral) filters depend on the circuit type and humidification system (See OpenCriticalCare Filter Placement FAQ). Always refer to manufacturers’ recommendations. In most circumstances a two filter setup should be used
  1. If available, place one HEPA (bacterial/viral) filter on inspiratory limb closest to the machine to protect against inadvertent backflow and device contamination
  2. Place one HEPA (bacterial/viral) filter between the endotracheal tube and the expiratory valve to avoid risk of contamination to the room. See Ventilator Filter and Humidification Placement Visual Aid.
  1. If EtCO2 monitor utilizes mainstream infrared measurement (i.e. does not sample gas into the analyzer and then exhaust into the room) then you may utilize a single HEPA (bacterial/viral) filter either between ETT and patient wye or at the expiratory limb closest to the ventilator. If EtCO2 monitor utilizes sidestream sampling of gas, then a HEPA (bacterial/viral) filter must be placed between the endotracheal tube and the EtCO2 sampling port (See OpenCriticalCare Filter and ETCO2 Placement FAQ).
  2. If no ventilator is immediately available and a bag valve mask will be used until the ventilator is available, make sure it has a HEPA (bacterial/viral) filter.
  1. Decide who will be in the room:
  1. Rapid Sequence Induction (RSI) should be performed by the most experienced airway provider, preferably with a video laryngoscope, if available (SCCM)(APSF). Always perform a difficult airway assessment to determine if RSI is appropriate, and/or what back up preparations should be taken in the case of anticipated difficulty.
  2. Limit the providers in the room to only those who are necessary. Generally this means:
  1. One person who will be intubating
  2. One assistant (often a nurse)
  3. One ventilator manager (often a respiratory therapist)
  1. Assign roles and airway plan (who will “hold/do” what)
  2. Someone should also be available immediately outside the room to access additional airway equipment as needed.
  1. Checklist prior to starting/ induction:
  1. Difficult airway assessment performed and intubation plan determined
  2. Suction available
  3. Pulse oximetry (ideally audible)
  4. Blood pressure cuff (ideally cycling q1 minute)
  5. Ventilator set up with predetermined settings entered. If using EtCO2 monitor in-line, it should be connected and ready (or color change (colorimetric) device if sidestream or mainstream capnography not available)
  6. Free-flowing IV access
  7. Post-intubation Sedation and vasopressors ready
  8. Viral filter in-line
  9. Induction medications ready
  10. Non-rebreather face mask with reservoir, connected to oxygen source with the flow turned off until ready to preoxygenate

ProcedureCopy Link!

  1. Preoxygenate the patient: Preoxygenate the patient for 3-5 minutes, and maintain preoxygenation until neuromuscular blockade (paralytic) has set in. Avoid bag valve mask ventilation if possible.
  1. If the patient is on nasal cannula, simple mask, venturi mask, or non-rebreather: tidal breathing on non-rebreather face mask at 15L/min (in general preoxygenation on nasal cannula, simple facemask or venturi mask are considered suboptimal)
  2. If the patient is on HFNC: increase FiO2 to 1.0 (100%)
  3. If the patient is on BiPAP: maintain BiPAP with tight seal until ready to intubate (turn “OFF” BiPAP flow prior to removing mask). Increase FiO2 to 1.0 (100%)
  4. If bag valve mask ventilation becomes necessary due to impending respiratory arrest or for rescue between intubation attempts:
  1. Use 2-hand technique with oral airway to create tight seal
  2. Ensure viral filter is in line
  3. Provide high frequency/low tidal volume breaths until saturation is optimized
  4. Do not remove mask for 2nd attempt intubation until end exhalation
  5. Consider use of an LMA with bacterial/viral filter to maximize airway patency
  1. Intubate the patient with an RSI technique/video laryngoscopy
  1. Use high dose neuromuscular blockade to promote rapid onset of action (practice may vary, but generally 2 mg/kg succinylcholine or 1.2-1.5 mg/kg rocuronium)
  2. Use awake intubation only when absolutely necessary as deemed by most senior clinician.
  1. After successful intubation:
  1. Inflate cuff
  2. Connect patient directly to ventilator with HEPA (bacterial/viral) filter.
  3. Endotracheal tube placement should be confirmed via quantitative in-line EtCO2 (gold standard > 3 breaths). Other methods for confirming placement include observing bilateral chest rise, hearing bilateral breath sounds, “fogging” of ETT, cuff palpation, or rising SpO2
  4. Secure ETT per hospital policy
  1. Decontaminate equipment:
  1. See Decontamination and Cleaning

Different protocols are used in different circumstances. For some specific examples, please see:

Tool: BWH Operating Room COVID Intubation Protocol

Tool: COVID19 Airway Management Checklist

Tool: South African Society of Anaesthesiologists: Recommendations for airway management for COVID-19 patients

Initial Ventilator SettingsCopy Link!

Tool: Respiratory Care Pocket Reference (Open Critical Care). Detailed pocket reference for ventilator modes and settings, including all the above tables.

  1. Obtain STAT portable chest xray: to confirm endotracheal tube location.
  1. Prioritize CXR and vent settings over procedures (such as central venous catheter placement) if possible.
  2. If portable chest x-ray is not available, confirm endotracheal tube placement with bilateral breath sounds and CO2 detection
  1. Ensure adequate sedation
  2. Set mode to volume control (AC/VC)
  1. In some settings airway pressure release ventilation (APRV)is used
  1. Set Initial tidal volume (Vt):
  1. Vt = 6 ml/kgIBW (based on ideal body weight [IBW]), see ARDSNet Table to look up tidal volume by gender and height in cm or inches.)
  1. Set initial respiratory rate
  1. Typical starting rates will be 16-24 titrated to goal minute ventilation of 6-8 L/min
  2. Consider starting rates of 24-28 titrated to goal minute ventilation of 8-12 L/min in setting of acidosis (pH < 7.25) pre-intubation. If blood gas is unavailable, higher initial minute ventilation should be targeted for patients with a pre-intubation respiratory rate above 35.
  1. Set Initial PEEP based on BMI (empirically chosen targets):
  1. BMI < 40: PEEP 5
  2. BMI ≥ 40: PEEP 10
  3. These should be readjusted after half an hour based on FiO2 and ARDSnet grid (see ARDS Oxygenation)
  1. Set Initial FiO2: 100% on intubation then rapidly wean to SpO2 92-96% (Barrot et al) (see ARDS Oxygenation)
  2. Obtain an arterial blood gas (preferred) or a venous blood gas within 30 minutes
  1. Calculate P/F ratio from initial post-intubation ABG. Adjust oxygenation as described in ARDS Oxygenation.
  2. Goal pH 7.20 to 7.45. Adjust ventilation as described in ARDS Ventilation.

Mechanical VentilationCopy Link!

Updated Date: August 11, 2021
Literature Review (Ventilator Settings):
Gallery View, Grid View

Tool: Open Critical Care Adult Ventilator Protocols and Order Set Templates. This includes sample ARDS Net lung protective ventilation as well as orders for spontaneous breathing trials (SBTs), difficult to wean patients and cuff leak tests.

This section addresses the management of mechanical ventilation for COVID ARDS specifically, not mechanical ventilation for other indications. It discusses the use of AC/VC as a ventilatory mode. Some settings may prefer APRV, which is not discussed in detail. Pressure support ventilation (PSV) mode is often used as the patient is recovering and preparing for extubation.

This section does not discuss managing COVID ARDS with concurrent obstructive lung disease (asthma, COPD), which ideally should be done by experienced clinicians only.

Improper ventilator management can permanently damage a patient’s lungs. Ventilator management requires significant infrastructure as well as training and expert guidance to call on if needed. This section assumes providers have some background knowledge about mechanical ventilation. See below for links to introductory material on mechanical ventilation.

Tool: Mechanical Ventilation Training Course (English, Spanish)

Tool: Respiratory Care Pocket Reference (English, Spanish)
Tool: COVID-19 Guidelines Dashboard
Tool:
Respiratory Care Protocol Templates

Lung Protective VentilationCopy Link!

Patients with ARDS receiving mechanical ventilation are at risk of lung damage, often referred to as ventilator induced lung injury (VILI). However, steps can be taken to reduce the risk of VILI and decrease mortality for patients with ARDS. This is referred to as lung protective ventilation (LPV).

LPV involves adjusting ventilator settings to achieve the following goals:

  • Tidal volumes (Vt) of 4-6ml/kg ideal body weight (IBW)
  • Plateau pressures (pPlat) less than 30cmH2O
  • Driving pressures less than 15cm H2O
  • Driving pressure is equal to pPlat - PEEP

ARDS Ventilation: Respiratory Rate and Tidal VolumeCopy Link!

Minute Ventilation (respiratory rate x tidal volume) helps control pH and PCO2.

  • Titrate minute ventilation to pH and not PCO2 in most circumstances!
  • Low tidal volumes are needed to protect the lungs from ventilator induced lung injury and promote lung healing. To achieve this, we tolerate hypercapnia (functionally no limitation unless clinical limits like seizure or managing increased ICP) and acidemia (pH > 7.2) (Ijland et al).

Adjusting ventilation parameters

  • First, set tidal volume
  • Follow ARDSnet ventilation where possible: Starting tidal volume of 6 cc/kgIBW.
  • Tidal volumes should always be within the 4-8 cc/kg range, ideally 4-6cc/kg based on Ideal Body Weight [IBW]. See ARDSNet table to look up tidal volume by gender and height in cm or inches.
  • Next, adjust rate to meet goal pH 7.20-7.45:
  • The respiratory rate often has to be high to accommodate low tidal volumes; typical RR is 20-35 breaths/minute. In patients with obstructive lung disease these are lower.
  • If pH > 7.45, decrease respiratory rate
  • If pH 7.10-7.25, then increase respiratory rate until pH > 7.25, or PaCO2 < 30 (maximum RR= 35 breaths/minute and check for autoPEEP (also known as intrinsic PEEP)
  • If pH < 7.10 despite maximum RR:
  • Address reversible causes of metabolic acidosis
  • Increase tidal volume up to 8cc/kg IBW or plateau pressure 30cmH20
  • Deepen sedation to RASS -3 to -5 or paralyze if needed
  • Initiation of prone ventilation (may improve V/Q matching and ventilate better)
  • Consider extracorporeal membrane oxygenation (ECMO) if available and none of the above is effective

ARDS Oxygenation: PEEP and FiO2Copy Link!

PEEP and Fi02 drive oxygenation. The goal is to deliver a partial pressure of oxygen to perfuse tissues (PaO2 ≥ 65, Sp02 ≥ 92%) while limiting lung injury from high distending pressures (with plateau pressures ≤ 30) and oxygen toxicity (with FiO2 ≤ 60%, SpO2 ≤ 96%). Extensive mammalian animal data demonstrates that hyperoxic injury occurs at an FiO2 ≥ 75% with the rate of injury increasing as FiO2 exceeds that. In multiple mammalian models, an FiO2 of 100% for 48 to 72 hours is associated with nearly 100% mortality rate. In these guidelines, we strive for FiO2 < 0.60, but wish to focus particular interest in the ARDS pathway when FiO2 >= 0.75 (i.e., increased sedation, paralysis, proning, inhaled vasodilator and ECMO consultation).

Lower limit goals for PaO2 / SpO2 are widely debated; PaO2 > 55 and SpO2 >88% are also commonly used. Our rationale relies on evidence for lack of benefit from conservative PaO2 goals in clinical trials (e.g. PaO2 > 55) and past association between lower PaO2 and cognitive impairment, although the evidence is not definitive (Barrot et al; Mikkelsen et al). Many clinicians use PaO2/FiO2 (called the P/F ratio) to guide oxygenation as it is a shorthand way of assessing the A/a gradient for the patient and seeing if their oxygenation is improving. SpO2/FiO2 ratio can be used if arterial blood gases are unavailable

  • If FiO2 >60%; patient requires ventilator optimization (ask a specialist). If persistent, see the Refractory Hypoxemia pathway.
  • It is reasonable to put a desaturating patient temporarily on 100% FiO2, but remember to wean oxygen as rapidly as possible

Adjusting Oxygenation Parameters:

  1. Typically, set PEEP and FiO2 according to the ARDSnet Tables:
  1. Within half an hour of initial ventilation settings (typically PEEP 5 for BMI <40 and PEEP 10 for BMI >40) reset PEEP and FiO2 to target oxygenation SpO2 92-96% using the following tables:

BMI < 40: ARDSnet LOW PEEP table:

FiO2

0.3

0.4

0.5

0.5

0.6

0.7

0.7

0.7

0.8

0.9

0.9

0.9

1

PEEP

5

5

8

8

10

10

12

14

14

14

16

28

18-24

BMI ≥ 40: ARDSnet HIGH PEEP table

FiO2

0.3

0.3

0.3

0.3

0.3

0.4

0.4

0.5

0.5

0.5-0.8

0.8

0.9

1

1

PEEP

5

8

10

12

14

14

16

16

18

20

22

22

22

24

Higher levels of PEEP can cause hypotension. It is important to monitor blood pressure when increasing PEEP.

  1. Readjust Frequently
  1. PEEP and Fi02 should be adjusted if:
  1. SpO2 <92% or >96% (do not use more oxygen or PEEP than is needed)
  2. PaO2 <65 or >100
  3. pPlat >30 (see this pathway)
  1. PEEP Optimization (If Needed and Familiar):
  1. In the setting of persistent hypoxemia, elevated plateau pressures, or for provider preference, PEEP optimization strategies could be considered. There is little data for how to determine optimal PEEP, and it is recommended that these be conducted by people familiar with the methods.
  1. Best PEEP trial
  2. Pressure Volume Tools
  3. Esophageal balloons. Special cases (e.g. morbid obesity, burns) may need extra diagnostics, such as esophageal balloons, which we do not recommend for routine use given limited resources and infection risk.
  4. Stress index (Video)

Mechanics: Plateau Pressure and ComplianceCopy Link!

Tool: How to Check Plateau Pressure and Compliance

Plateau Pressure:

It is important to avoid elevated plateau pressures (with goal ≤ 30) which can indicate relative lung overdistention (Slutsky et al).

  1. Check plateau pressure with every change in tidal volume, PEEP, or clinical deterioration (worsening oxygenation) but not as part of routine practice. In order to accurately measure plateau pressure, the patient must be passive (i.e. not actively breathing) on AC/VC mode with a constant flow delivery (as opposed to decelerating flow delivery).
  2. If plateau pressure is >30 cm H20, then decrease tidal volume by 1 mL/kgIBW (minimum 4 mL/kgIBW)
  3. If plateau pressure is < 25 cm H20 and tidal volume < 6 mL/kgIBW, then increase tidal volume by 1 mL/kgIBW until plateau pressure is > 25 cm H2O or tidal volume = 6 mL/kgIBW
  4. If plateau pressure is < 30 cm H20 and patient is breath stacking or dyssynchronous, then increase tidal volume in mL/kgIBW increments to 7 mL/kgIBW or 8 mL/kgIBW while plateau pressure is < 30 cm H20

Compliance:

Compliance measures can give an indication about whether a patient’s lung stiffness is improving or declining over time, and can help with prognostication and management.

Assessing Ventilator SynchronyCopy Link!

There are three main forms of asynchronous interaction between the patient and the ventilator: Asynchrony related to breath initiation and phase duration (i.e. trigger asynchrony, phase asynchrony), and mismatching related to ventilator settings of inspiratory flow and/or tidal volume. These are discussed in detail with diagnosis and treatment recommendations in Patient Ventilator Interaction.

Tool: Patient Ventilator Synchrony.

TroubleshootingCopy Link!

Resistance: Troubleshooting increased Peak Inspiratory Pressure due to high resistance: Work outside (machine) to inside (alveoli); circuit problem, ETT kink/occlusion/biting, ETT obstructed/malpositioned, large airway obstruction (mucous plug), small/ medium airway obstruction (bronchospasm); auscultation & passing a suction catheter can quickly eliminate many of these.

Compliance: Troubleshooting increased Peak Inspiratory Pressure due to reduced compliance: Work outside (patient extra-pulmonary factors) to inside (alveoli): Patient dyssynchrony requiring increased sedation (or temporary paralysis if refractory); intra-abdominal process; ET tube malpositioned (into a single tube); pneumothorax; auto-PEEP (due to incomplete exhalation, typically in setting of bronchospasm), parenchymal and alveolar process (flash pulmonary edema, pulmonary hemorrhage).

Other Modes of VentilationCopy Link!

PSVCopy Link!

This section is forthcoming

APRVCopy Link!

There exists significant practice variation around the use of bilevel ventilatory modes. APRV should only be used by providers with experience and familiarity with this mode. At this time there are no data to support the superiority of APRV in ARDS patients, including those with COVID-19. One recent small study in Australia found that APRV was associated with decreased survival (Zorbas et. al).

Tool: EMCrit APRV

Prone VentilationCopy Link!

Updated Date: December 20, 2020
Literature Review (Proning):
Gallery View, Grid View

Tool: Prone Positioning Checklist

Early proning in COVID-associated ARDS requires intensive management but can significantly improve oxygenation. Pronation is one of the only interventions shown to improve mortality in ARDS (PROSEVA) (Guérin et al). In one representative study in 62 COVID patients on ventilators who underwent prone positioning, as compared to 199 similar controls who met criteria for prone positioning but did not receive the intervention, showed a multivariate-adjusted hazards ratio for mortality of 0.57 (0.42-0.76) in the proned patients (Shelhamer et al).

For proning of non-intubated patients, please see Awake Proning.

Timing and CandidacyCopy Link!

Timing: We recommend early proning in severe ARDS (<36 hrs) and prefer to initiate proning prior to use of continuous paralytics (or inhaled pulmonary vasodilators), despite the fact that in the PROSEVA trial over 95% of patients in both the intervention and the control arm were on continuous paralytics.

  • We particularly recommend proning if a patient requires an FiO2 ≥ 60% to achieve an SpO2 ≥ 92% (or PaO2 ≥ 65) with a P:F ≤ 150 (Guérin et al).

Eligibility: The only absolute contraindications are spinal cord injury, open chest, and unstable airway. Patient size is not a contraindication. Other relative contraindications should be discussed by the clinical team .

  • For patients with a tracheostomy, we recommend that patients have their tracheostomy replaced by oral endotracheal intubation (ETT) when possible, while recognizing that decannulating a tracheostomy and placing an ETT poses an infectious risk to staff.
  • Renal replacement therapy can be performed while prone, typically via a femoral line.
  • Monitor for complications: Prone ventilation can lead to increased incidence of brachial plexopathy in the context of increased pressure to anterior portions of the arm and shoulder (Scholten; Goettler). Prone positioning for surgery has been associated with abdominal or limb compartment syndromes, or Rhabdomyolysis (Kwee).

Intubated Proning ProtocolCopy Link!

Tool: BWH MD MICU Proning Protocol

Tool: NEJM Video
Tool:
Prone Positioning Checklist

For proning of non-intubated patients, please see Awake Proning.

  1. Prepare for Proning
  1. Hold tube feeds for 1 hour prior to proning or supinating
  2. Assemble all necessary tools (pillows, props, additional persons to assist)
  1. Place Patient in Proned (“swimmer”) Position (some places have rotary beds)
  1. Have one person hold ET tube and lines to assure they are not dislodged and continuous medications are not disrupted
  1. Measure effect of proning. 1 hour post-initiation of prone ventilation:
  1. Obtain ABG. Compare pre-pronation PaO2/FiO2 to post-pronation PaO2/FiO2. Ideally there should be a 0.1 change in FiO2 (maintaining the same SpO2) or >10% change in PaO2 / FiO2. However, a lack of improvement may not be considered an absolute indication to abandon proning.
  2. Measure compliance and Plateau Pressure, PEEP, and FiO2
  3. Assess tidal volume and Adjust Ventilation Parameters
  1. If patient demonstrates improvement on proning:
  1. Prone ≥16 hrs per 24 hrs. Supine ≥ 4 hrs per 24 hrs. Repeat every day. There is no day limitation for maintaining prone ventilation and it should be repeated every day while beneficial.
  2. Discontinue neuromuscular blockade if initiated for dyssynchrony and re-assess.
  1. If patient does not improve on proning:
  1. Resupinate. Consider trying again the next day, as sometimes the recruitability of lung tissue will change over time.
  1. Consider discontinuing proning
  1. If patient has improved and meets the goals listed below after supine for >4 hrs. If patients do not meet criteria for supine ventilation then recommend ongoing prone ventilation.
  1. FiO2 < 60% to meet an SpO2 ≥ 92% (or PaO2 ≥ 65)
  2. Plateau pressure < 30
  3. pH > 7.25
  1. Return to supine position emergently, if:
  1. Unscheduled extubation
  2. Endotracheal tube obstruction
  3. Severe or significantly worsening hypoxemia, e.g. Sp02 <85% and consideration of ECMO if available
  4. Hemodynamic instability

Refractory HypoxemiaCopy Link!

Updated Date: December 20, 2020

Refractory HypoxemiaCopy Link!

If patient is hypoxic (PaO2 <75) despite PEEP optimization as above); and FiO2 > 0.6 or PaO2/FiO2 ratio < 150 then consider trying each of the following

  1. Proning: Initiate early (if not already done)
  2. PEEP: Adjust PEEP as above and request expert optimization if needed
  3. Diuresis: Assess volume status. Diurese or remove volume by renal replacement therapy if indicated.
  4. Synchrony (Paralysis): Assess Ventilator Synchrony and Sedation to achieve ventilator synchrony. If still dyssynchronous, consider Neuromuscular Blockade.
  1. Assess for improvement in oxygenation (stable oxygenation metrics while being able to reduce FiO2 by 0.1)
  2. Try stopping neuromuscular blockade daily if possible, and discontinue completely if the patient maintains PaO2>75 with FiO2<0.75 without it.
  1. Inhaled pulmonary vasodilators: consider trial of continuous Inhaled Pulmonary Vasodilators. Note however that there is no evidence of survival benefit of inhaled vasodilators in ARDS.
  2. ECMO Consultation: If available at your institution, if no improvement despite the above steps, no contraindications, and any of :
  1. Persistent PaO2 < 75 requiring FiO2 > 0.75
  2. Plateau pressure >30
  3. Refractory hypercapnia and pH < 7.2

Recruitment ManeuversCopy Link!

A recruitment maneuver is the deliberate administration of a high airway pressure for protocolized periods of time to open collapsed alveoli There are multiple protocols for performing recruitment maneuvers. Studies have shown that recruitment maneuvers are associated with a temporary increase in oxygen levels but do not impact clinical outcomes. (Brower et al; Meade et al; Oczenski et al). A more recent study of recruitment maneuvers in combination with best PEEP trials found an association with increased mortality (Calvacanti et al). Recruitment maneuvers can increase intrathoracic pressure enough to affect blood return to the heart and thus hemodynamics. We do not recommend regular use of recruitment maneuvers in the management of refractory hypoxemia.

Inhaled Pulmonary VasodilatorsCopy Link!

Literature Review: Gallery View, Grid View

There is no evidence of survival benefit of inhaled vasodilators in ARDS, and it can demand significant respiratory therapist resources (Fuller; Gebistorf et al,; Afshari et al). There is currently no evidence of the survival benefit in COVID ARDS, though data are still very limited. There is limited evidence from small studies that in about half of patients PaO2 to FiO2 ratios may be improved by >10%. In a retrospective cohort study of BWH intubated COVID-19 patients, inhaled epoprostenol did not significantly alter PaO2/FiO2. PaO2/FiO2 increased by >10% in 40% of patients (N=38), but clinical outcomes were not changed. 11 patients who failed to respond to inhaled epoprostenol were trialed on inhaled nitric oxide (iNO). On iNO, PaO2/FiO2 increased by >10% in 60% of patients (N=11). There was no change in outcome. This study was limited by a small sample size and retrospective design (DeGrado et al).

Epoprostenol Instructions:

  1. Exclude contraindications: Alveolar hemorrhage (epo has mild antiplatelet effect), LV systolic or diastolic CHF (vasodilators cause ↑ pulm blood flow → ↑ LV filling pressure → ↑ pulmonary edema & ↓ PaO2 → consider CHF if pt gets worse after starting).
  2. Measure baseline ABG for PaO2
  3. Start continuous nebulization at 0.05 mcg/kg/min based on IBW (MDcalc Online Calculator). Do not change ventilator settings, sedation, paralysis, patient position or other care that could affect oxygenation.
  4. Re-check ABG 2 hours after initiation of inhaled epoprostenol.
  • If PaO2 increased by >10% from baseline, continue inhaled epoprostenol.
  • If PaO2 not increased by >10% from baseline, discontinue inhaled epoprostenol.
  1. Weaning:
  • Attempt to wean off daily. Wean inhaled epoprostenol by decreasing 0.01mcg/kg/min every hour. Monitor SpO2 and hemodynamics.
  • Re-check ABG 2 hours after weaned off. If PaO2 worsened by >10%, restart inhaled epoprostenol.

Inhaled Nitric Oxide: Limited in vitro data notes that iNO at high doses inhibits replication of SARS-CoV, but this has not been studied in vivo (Akerstrom et al; Gebistorf et al) although clinical trials are in progress. iNO acts as a pulmonary vasodilator and can be used instead of epoprostenol. Depending on setting, continuous use of iNO can be logistically-challenging and cost-prohibitive.
Literature Review (Inhaled NO): Gallery View, Grid View

ECMO and Mechanical Cardiac SupportCopy Link!

Literature Review: Gallery View, Grid View

Respiratory failure:

In facilities where it is available, the veno-venous Extracorporeal Membrane Oxygenation (ECMO) team should be consulted for respiratory failure despite all other measures:

  • Persistent PaO2 / FiO2 ratio < 75 mmHg despite optimized ARDS management (optimized PEEP, neuromuscular blockade, proning, inhaled vasodilator).
  • Plateau pressure > 30 cm H2O on ARDSnet ventilation.
  • pH < 7.2
  • No potentially reversible causes (e.g. pulmonary edema, mucus plug, abdominal compartment syndrome)

Cardiogenic Shock:

In facilities where it is available, the veno-arterial ECMO or mechanical support team should be consulted for cardiogenic shock:

  • Dobutamine drip at 5mcg/kg/min (or unable to tolerate dobutamine due to tachyarrhythmias) and ScvO2 < 60% or CI < 2.2
  • Lactate > 4 after medical therapy

Candidacy:

The criteria for ECMO and other mechanical circulatory support varies among centers and are difficult to develop even under typical circumstances. For the purposes of general education, a hypothetical set of inclusion criteria for VA ECMO or MCS could cover:

  • Younger age (note: the inclusion of age as a criterion raises ethical issues that merit consideration by decision-makers)
  • Expected life expectancy >6 months pre-hospitalization
  • No evidence of solid or liquid malignancy
  • Able to tolerate anticoagulation
  • Platelets >50,000 or ANC > 500
  • Absence of severe peripheral arterial disease
  • No evidence of irreversible neurological injury
  • Able to perform ADLs at baseline prior to illness
  • BMI (for some devices there are BMI limitations, for others there are not)
  • No major conditions or multisystem organ failure that would preclude a reasonable chance of recovery.

Sedation and Ventilator SynchronyCopy Link!

Updated Date: December 20, 2020
Literature Review:
Gallery View, Grid View

Achieving Ventilator SynchronyCopy Link!

  1. Start analgesia and sedation immediately:
  1. Ensure analgesia/sedation infusion is at bedside prior to intubation. The sedative boluses used during intubation will wear off long before the paralytic and the patient must be sedated while paralyzed. Assume at least 60 minutes of sustained neuromuscular blockade for rocuronium (longer in renal or liver dysfunction) and cisatracurium, and 10 minutes for succinylcholine and suxamethonium.
  1. Assess patient synchrony with the ventilator:
  1. After neuromuscular blockade has worn off, assess synchrony (e.g, signs of breath-stacking, double triggering, other ventilator alarms)
  1. If synchronous, lighten sedation to the lowest level that maintains synchrony, ideally Richmond Agitation Sedation Scale (RASS) score 0 to -1.
  2. If not synchronous: First adjust ventilator settings (including flow settings, trigger settings, and modest liberalization of settings within ARDSnet criteria [Vt 4-8ml/kgIBW]). Then escalate sedation as needed to achieve synchrony
  1. If dyssynchronous despite deep sedation (RASS -4 to -5):
  1. First discuss additional ventilator changes with someone very familiar with mechanical ventilation (respiratory therapist or intensivist) if available
  2. Try Neuromuscular Blockade if unable to achieve with the above

Sedation: Pain, Agitation, Delirium ModelCopy Link!

Typical regimen in a ventilated ARDS patient: Generally ARDS patients require a period of continuous IV sedation and analgesia to establish ventilator synchrony. Assess and treat pain, agitation, and delirium in that order (sedating a patient who is in pain is typically less effective and less humane).

Detailed instructions on dosing, adjuncts, and enteral options for all three of these categories are available in the BWH Sedation Section.

  1. Pain: Low-dose opioids, in bolus or enteric form whenever possible, with adjuncts to reduce doses.
  1. First line agents are fentanyl or hydromorphone
  2. Second line agents is morphine
  1. Agitation:
  1. First line is continuous propofol in post patients
  1. Dexmedetomidine is generally reserved for patients approaching ventilation liberation; tachyphylaxis (diminishing response to the medication with repeated exposure) may occur with prolonged use
  1. Benzodiazepines can be used, but carry a high risk of delirium
  1. Delirium: If patients are agitated on daily Sedation Interruption (SAT), positive on the CAM-ICU screen, or receiving continuous sedation for >48hrs, we recommend delirium treatments.
  1. First line is nonpharmacologic mechanisms
  2. Second line is an antipsychotic (Haloperidol, Quietapine)
  3. Second line is alpha-2 agonists or mood stabilizers

Tool: BWH Detailed Sedation Recommendations including Dosing and Alternatives

Pain

Agitation

Delirium

Assess and Document

If able to self report → Numeric Rating Scale (0-10) (NRS)

If unable to Self Report → Critical Care Pain Observation Tool (CPOT) (0-8)

Richmond Agitation Sedation Scale (RASS) (-5 to +4)

Bispectral Index (BIS) in patients receiving Neuromuscular Blockade

CAM ICU-modified (+ or -)

Frequency

At least every 8 hours on all patients

If receiving intermittent or continuous analgesia or sedation, every 2 hours

At least every 8 hours on all patients

If receiving intermittent or continuous analgesia or sedation, every 2 hours

At least every 8 hours on all patients

Interpretation

Patient is in significant pain if:

  • NRS > 4
  • CPOT > 3

Sedation/agitation depth defined according to RASS scale

Usual goal RASS is 0 to -1

Delirium present if: CAM-modified is positive

Treatment

First line: Fentanyl or Hydromorphone

Second line: Morphine

First line: Propofol

Second line: Dexmedetomidine or benzodiazepines

First line: non-pharmacologic

Second line: antipsychotics

Third line: Alpha-2 agonists and mood stabilizers

Strategies to Minimize Medication ShortagesCopy Link!

  1. Boluses of benzodiazepines and opioids are preferred to continuous infusions. If continuous infusions are used, boluses should be administered prior to starting the infusion as well as when infusions are up-titrated. Bolus doses are typically 50-100% of the hourly infusion dose.
  2. Use the lowest dose that can achieve the desired effect
  3. Change IV medications to enteral medications if appropriate, especially as patients are weaning.

Strategies to Avoid Prolonged SedationCopy Link!

  1. Daily SAT:
  1. We recommend a daily spontaneous awakening trial/sedation interruption unless contraindicated.
  1. Wean quickly, while monitoring for withdrawal:
  1. Patients who have been on continuous sedation for less than 7 days can be weaned rapidly with minimal concern for withdrawal.
  2. Otherwise, wean sedation and analgesics by at least 20% per day
  1. Faster weaning is frequently possible due to drug accumulation in tissues
  2. Consult a pharmacist (if available) if concerns for withdrawal
  1. Use adjuncts:
  1. Adjuncts including alpha-2 blockers, antipsychotics, enteral agents, and non-pharmacologic delirium prevention can facilitate weaning
  2. Try ventilator adjustments to facilitate Ventilator Synchrony.

Tool: SAT and SBT Algorithm and Orderset Template

Neuromuscular BlockadeCopy Link!

When to use: Neuromuscular blockade (NMB) is often used as a last measure to achieve ventilator synchrony in patients on Assist Control or Mandatory ventilation modes to help reduce lung injury from ventilator dyssynchrony. It should be used after alternative approaches to achieving ventilator synchrony have been pursued (see Ventilator Synchrony). It has also been used as part of standard therapy for moderate-severe ARDS, although recent data have brought this practice into question (NHLBI).

Neuromuscular blockade should always be administered with adequate sedation and for the shortest possible duration.

Use when Proning: NMB is usually not required for proning, as most patients can be proned with deep sedation alone, however, it can be used in boluses prior to proning or supination.

Safety and Monitoring:

  1. Always use analgesia and sedation: Neuromuscular blockers have no sedative or analgesic properties. Patients receiving neuromuscular blockade must be on medications for both analgesia (e.g. an opioid) and sedation (e.g. propofol or a benzodiazepine) to avoid having a patient who is paralyzed but wakefl. This must be initiated prior to starting NMB.
  1. Monitoring Sedation: Ideally you should use RASS to measure sedation before initiating neuromuscular blockade and a BIS monitor after. RASS cannot be used to assess sedation after initiating neuromuscular blockade.
  1. Before Initiating NMB: Target sedation scale to RASS - 4 to -5
  1. Where BIS monitoring is used to assess level of sedation, it may not be reliable before neuromuscular blockade is initiated due to facial muscle activity. Some institutions do not routinely use BIS.
  1. After initiating NMB: Target BIS of 40-60 (RASS cannot be used in paralyzed patients). If BIS is not available, continue deep sedation with the dose of sedative and analgesic that had been required to achieve RASS -5 prior to starting NMB.
  1. Closely monitor heart rate and blood pressure. Unexplained high heart rates and/or hypertension may be a sign that the patient is under-sedated. Patients require higher doses of opioids or sedatives over time to achieve the same level of sedation, so assess this daily.
  1. Even if BIS is available, monitoring is imperfect, and can be falsely low in the setting of edema or hypotension or falsely high with ketamine administration.
  1. After stopping NMB: Washout time for paralytics should be allowed (and, if using, “Train of Four” with a peripheral nerve stimulator should be 4/4) before sedation weaning (see tool for using “Train of Four” below)
  1. Use the lowest effective dose for the shortest possible time: Prolonged neuromuscular blockade may contribute to weakness, prolonged weaning, and delayed recovery. Corticosteroids may increase risk of severe myopathy.
  1. Monitoring Paralysis: Use the minimum dose needed for intended effect
  1. Ventilator synchrony should be the primary indicator of when a patient is adequately paralyzed. Consider bolus, not continuous, dosing for intermittent dyssynchrony.
  2. Some institutions use a peripheral nerve stimulator (“Train of Four”) to assess paralytic effect and minimize doses. The goal is still vent synchrony not a number of twitches. Use the minimal amount of paralytic necessary for synchrony. (See tool for using “Train of Four” below)
  1. Stopping NMB: Try stopping neuromuscular blockade after 48 hours (earlier if synchrony can be achieved by other means) and daily thereafter unless the patient is too unstable.
  1. The clinical context and goals of mechanical ventilation should guide decisions regarding continuation of neuromuscular blockade. For example, if synchrony with lung protective ventilation settings is clinically indicated and cannot be achieved with sedation alone, then it is reasonable to continue neuromuscular blockade.
  2. Assess response in oxygenation by assessing the SpO2/FiO2 or PaO2/FiO2 ratio. Consider discontinuing if the patient maintains PaO2>75 with FIO2<0.75 and is synchronous.

Tool: Train of Four Monitoring (Winnipeg Regional Health Authority)

Dosing Strategy:

  1. Try bolus dosing before continuous dosing:
  1. Use boluses to facilitate pronation/supination or for intermittent ventilator dyssynchrony in adequately sedated patients. Practice patterns vary on how much asynchrony to tolerate, but there is some evidence that even minor asynchrony can have mortality implications (Blanch et al).
  2. Convert to a drip if there is persistent ventilator dyssynchrony requiring >3 bolus doses in 2 hours, with re-evaluation every 24-48 hours.
  1. First Line Agents:
  1. Cisatracurium (Preferred in renal or hepatic dysfunction, though supplies globally are limited)
  1. Dosing - Intermittent Bolus: 0.1-0.2 mg/kg Infusion: 0-5 mcg/kg/min
  1. If Cisatracurium is not available, Rocuronium is often used.
  1. Dosing - Intermittent Bolus: 0.6-1.2 mg/kg Infusion: 0-20 mcg/kg/min Start infusion at 3-5 mcg/kg/min
  1. If concerns for tachyphylaxis (decreasing effect with prolonged exposure), consider rotating to an alternative agent (vecuronium or atracurium)

Cisatracurium

Atracurium

Vecuronium

Rocuronium

Duration/Recovery (min)

80-180

20-40

30-60

20-30

Renal excretion (%)

Hoffman Elimination

Hoffman Elimination

50

20-30

Effect of renal failure

No change

No change

Increased, especially metabolites

Minimal

Hepatic excretion (%)

Hoffman Elimination

Hoffman Elimination

35-50

< 75

Effect hepatic failure

No change

No change

Variable, mild

Moderate

Histamine release

No

Dose dependent

No

No

Liberation from the VentilatorCopy Link!

Updated Date: December 20, 2020
Literature Review:
Gallery View, Grid View

Spontaneous Awakening and Breathing Trials (SAT/SBT)Copy Link!

Tool: SCCM Guide on Spontaneous Awakening and Breathing Trials (SAT/SBT) (See page 5 for visual algorithm)

Tool: SAT and SBT Algorithm and Orderset Template (OCC). This includes sample ARDS Net lung protective ventilation as well as orders for spontaneous breathing trials (SBTs), difficult to wean patients and cuff leak tests.

Tool: SBT Protocol

Aim to liberate the patient from mechanical ventilation as soon as safe and feasible. Prolonged intubation is associated with ventilator-associated pneumonia (VAP) with median-time to VAP onset of 8 days in a retrospective study of 191 COVID patients in Wuhan (Zhou).

All patients with improving or stable respiratory disease should have FIO2 and PEEP titrated at least daily and should have a Spontaneous Awakening trial (see below) to ensure that their neurologic status remains intact, even if they are not yet a candidate for extubation.

Daily Spontaneous Awakening Trial (SAT): all patients on mechanical ventilation should be assessed daily for whether they meet criteria for SAT:

  1. Is the patient on high ventilator settings, proned, paralyzed within the last 6 hours, hemodynamically unstable typically HR > 120 or unstable arrhythmia or, MAP < 65, or vasopressor requirement equivalent of norepinephrine > 10 mcg/min, though none of these is absolute? Have they had recent myocardial ischemia, elevated intracranial pressure, or are they sedated for non-intubation medical reasons (e.g. seizure)? If so, do not attempt an SAT
  2. Otherwise, stop sedatives (and analgesics not needed for pain) until a RASS of 0 is achieved
  3. Ask the patient to do the following (ideally they should do 3 of 4, though this is not absolute): If the patient becomes highly agitated they may not be able to follow commands but could still be able to be extubated if they are able to take large breaths. This is common in young people.
  1. Open their eyes
  2. Look at their caregiver,
  3. Squeeze the hand
  4. Put out their tongue
  1. Check the patient’s hemodynamics for 5 minutes. The patient should have none of the following issues:
  1. RR >35?
  2. No spontaneous breaths initiated in 5 minutes
  3. SpO2 <88% for 5 minutes
  4. Acute arrhythmia?
  5. Marked work of breathing or agitation.
  1. If the patient passes part b and c, then move on to SBT
  1. If a patient fails, sedatives are started at half the prior dosage and titrated up as needed. Often patients will need a longer washout time, or better Management of Delirium.

Spontaneous breathing trial (SBT): all patients on mechanical ventilation and passing SAT should be assessed daily for SBT (adapted from AHRQ).

  1. Does the patient meet these criteria?:
  1. FiO2 ≤ 50%, PEEP ≤ 10cmH2O for BMI ≤ 40 (or ≤ 16 cmH2O for BMI > 40 at provider discretion) with SpO2 ≥ 92% in the supine position
  2. Hemodynamically stable (defined as HR < 120, MAP > 65, and vasopressor requirement of norepinephrine < 10 mcg/min and no unstable tachyarrhythmias)
  3. No other medical contraindications to increased respiratory effort or decreased sedation
  1. If so, change their ventilator settings:
  1. SBT consists of Pressure Support ventilation mode with a pressure support = 5cmH2Oand PEEP = 5cmH2O (consider PEEP of 10cmH2O for BMI > 40)
  2. SBT is discontinued (patient is re-sedated and ventilator settings changed) if the patient develops:
  1. Evidence of increased work of breathing with RR > 30
  2. Hypoxia (SpO2 < 92%)
  3. Hemodynamic instability
  4. Rapid shallow breathing index (RSBI) = respiratory rate/tidal volume > 105
  1. If not terminated for the above reasons, terminate all SBTs after 30 minutes.
  1. If the patient does well and the medical team deems them ready to be extubated (see below), proceed with extubation.
  2. If the patient does not do well or is not ready to be extubated, they can be returned to their prior mode of ventilation or to a new mode. Typically we choose:
  1. AC/VC if ARDS is ongoing and the clinicians anticipates >1 day of need for ongoing intubation
  2. PSV (with higher pressure support) if the clinicians believe the patient may be able to be extubated within a day or deems it appropriate

ExtubationCopy Link!

Extubation should be considered if patient:

  1. Passes SAT and SBT
  2. Is able to follow commands (with RASS ideally 0 to -1)
  3. Coughs on deep suctioning and has a gag reflex
  4. Does not require deep suctioning more than every 2 hours
  5. Does not need any other medical interventions prior to extubation
  6. The patient has enteric access if needed.
  1. We recommend placing an NG or Dobhoff tube (with bridle if possible) prior to extubation for patients intubated for >48h given the frequency of swallowing issues post-extubation in these patients and the challenges in obtaining swallowing evaluations in ICU extubation
  1. In patients with risk factors for laryngeal edema (e.g. traumatic intubation, prone positioning, prolonged intubation >6 days, or anasarca) , it may be wise to check a cuff leak (ability of air to move around the ET tube) prior to extubation. No cuff leak increases the likelihood of reintubation (LR: 4) and a cuff leak reduces the likelihood of reintubation (LR: 0.5) (Girard et al).
  1. In patients without a cuff leak, but are otherwise ready for extubation, systemic steroids can be administered to reduce the risk of post-extubation stridor.

Tool: Endotracheal Cuff Leak Protocol

Extubation Procedure:

  1. Clarify goals of care if the patient fails extubation and whether reintubation should be attempted.
  1. Make sure adequate supplies for Reintubation and Airway Management are available and nearby if needed.
  1. Use airborne precautions, don appropriate PPE, minimize staff
  2. Place the patient on 1.0 FiO2 on the ventilator
  3. Ensure the selected supplemental oxygen device that the patient will use after extubation is at bedside. Optimal selection may help reduce the risk of reintubation. The selection of devices varies widely depending on practice patterns. One suggestion is that patients who have hypercapnia be extubated to NIPPV. Other patients at high risk for reintubation may get NIPPV or HFNC where available. Patients with prolonged intubation should be extubated to HFNC where possible. Other oxygen delivery devices are often adequate, but prepare for the potential to increase oxygen delivery rapidly if needed (Hernández et al; Oulette et al; Hernández et al).
  4. Place absorbent pad or towel on patient chest. Consider placing a drape on top of the patient to prevent exposure to any coughing that may occur.
  5. Secure the feeding tube to nose.
  6. Suction mouth and loosen tape securing ETT to patient.
  7. Turn all gas flows to “OFF” (may still have some O2 flow as a safety mechanism for most machines)
  8. Deflate the ET tube cuff, and extubate the patient.
  9. Immediately place the oxygen delivery device on the patient (typically at a high flow or FiO2 rate).
  10. Immediately discard ETT, absorbent pad or towel, and drape
  11. Ensure the patient is adequately oxygenating and ventilating (Sp02, respiratory rate)
  1. Consider blood gas half an hour after extubation
  1. Doff PPE and ensure adequate air turnover in the room before taking off airborne precautions. Assuming an air changeover of 6 times an hour, this is 47 minutes.

Barriers to LiberationCopy Link!

Weaning can fail in the setting of the following conditions (address appropriately) (Boles et al)

  1. Respiratory factors:
  1. Ongoing pneumonia or pulmonary inflammation
  2. Bronchoconstriction
  3. Glottic and airway edema, sputum production, impaired cough
  1. Cardiac factors:
  1. Cardiac dysfunction or shock
  1. Neuromuscular factors
  1. Weakness and prolonged immobility
  2. Effects of steroids or neuromuscular blockade
  1. Neuropsychological factors
  1. Delirium
  2. Sedating medications
  1. Metabolic factors
  1. Malnutrition
  2. Electrolyte disturbances (hypophosphatemia, etc)

Tracheostomy ManagementCopy Link!

Literature Review (Tracheostomy Management): Gallery View, Grid View

  1. For tracheostomy procedure, see BWH guide on Tracheostomy
  2. This section is in development

ShockCopy Link!

Updated Date: December 20, 2020

Undifferentiated ShockCopy Link!

Definition: Acute onset of new and sustained hypotension (MAP < 65 or SBP < 90) with signs of hypoperfusion requiring intravenous fluids or vasopressors to maintain adequate blood pressure

Time course: Patients rarely present in shock on admission. Natural history seems to favor the development of shock after multiple days of critical illness.

Etiology: The range of reasons for shock in COVID is broad, and includes

  1. Myocardial Dysfunction
  2. Secondary Bacterial Infection
  3. Cytokine Storm Syndrome
  4. Sedation Effects in Intubated Patients

Workup:

  1. Assess for severity of end organ damage:
  1. Urine output, mental status, lactate, BUN/creatinine, electrolytes, liver function tests
  1. Obtain a FULL infectious/ septic workup, which includes all of the following:
  1. Labs: CBC (FBC) with differential. Note that most COVID patients are lymphopenic (83%). However, new leukocytosis can occur and left-shift can be used as a part of clinical picture (Guan et al). Two sets of blood cultures, liver function tests (for cholangitis/acalculous cholecystitis), urinalysis (with reflex to culture), sputum culture (if it can be safely obtained), procalcitonin at 0 and 48h if available (do not withhold early antibiotics on the basis of procalcitonin alone), urine Strep pneumo and legionella antigens if available
  2. Portable chest x ray (avoid CT unless absolutely necessary)
  3. Full skin exam
  1. Assess for cardiogenic shock
  1. Assess extremities: warm or cool on exam
  2. Assess patient volume status: JVP, CVP, edema, CXR
  3. Assess pulse pressure: If < 25% of the SBP, correlates highly with a reduction in cardiac index to less than 2.2 with a sensitivity of 91% and a specificity of 83% (Stevenson et al).
  4. Perform point of care ultrasound, if available, to assess for gross LV/RV dysfunction
  1. For transthoracic echocardiography (TTE) protocols see Advanced Cardiac Diagnostics.
  1. Labs: Obtain a central venous O2 sat or mixed venous O2 sat if the patient has central access, troponin x2, NT proBNP, A1c, lipid profile, TSH
  2. EKG (and telemetry)
  3. Calculate estimated Fick Cardiac Output
  1. MDcalc online calculators: Fick CO, BSA
  1. Consult cardiologist if available if any suspicion of cardiogenic shock
  1. Assess for other causes of shock:
  1. Vasoplegia:
  1. Run medication list for recent cardiosuppressive medications, vasodilatory agents, antihypertensives
  1. Adrenal insufficiency:
  1. Unless high pretest probability of adrenal insufficiency, we recommend against routine cortisone stimulation testing
  1. Obstruction:
  1. PE (given the elevated risk of thrombosis)
  2. Tamponade (given elevated risk of pericarditis)
  3. Obstruction from PEEP
  1. Cytokine Storm Syndrome
  2. Allergic reactions to recent medications
  3. Neurogenic shock is uncommon in this context
  4. Hypovolemia:
  1. Bleeding
  2. Insensible losses from fever
  3. Diarrhea/vomiting

Differentiating ShockCopy Link!

Tool: Differentiating Shock

Type of Shock

Cardiac Output

SVR

CVP/Wedge

ScvO2, MvO2

Other features

Cardiogenic

Low

High

High

Low

Distributive (sepsis,cytokine, anaphylaxis)

High

Low

Low

High

Obstructive

Low

High

High

Low

Hypovolemic

Low

High

Low

Low

Neurogenic

Low

Low

Low or normal

Low

Decreased HR

Septic ShockCopy Link!

Literature Review: Gallery View, Grid View

The rates of sepsis and septic shock are not reported consistently in currently available case series. One meta-analysis of 21 studies (47,344) found that 4.7 % of patients developed shock (95CI 0.9–8.6 %), though this was pooled septic and other forms (Hu et al). Sepsis can be caused by the coronavirus itself (viral sepsis), multisystem organ failure, or by Secondary Bacterial Infection.

We recommend instituting early empiric antibiotics for suspected septic shock and following a conservative fluid management strategy.

AntibiosisCopy Link!

  • Early empiric antibiotics should be initiated within 1 hour (see Antibiotics)
  • Choice of agent varies widely depending on local bacterial epidemiology and the availability of antibiotics. For septic shock it should include broad gram positive and gram negative coverage.

Pressors and Fluid Management:Copy Link!

Pressors. Goal MAP > 65mmHg. While there is emerging data that lower MAP thresholds may be beneficial, we recommend following this threshold for now.

  1. Norepinephrine is the preferred initial vasopressor for undifferentiated shock and septic shock.
  1. If norepinephrine is not available, we recommend epinephrine (Myburgh et al).
  1. Dopamine should only be used as an initial pressor if other pressors are unavailable
  1. When norepinephrine requirement is above 10, we recommend adding a second agent (typically vasopressin if available).
  1. If vasopressin is unavailable, epinephrine should be used as a second pressor
  1. Sometimes phenylephrine is also needed, though this should be used with caution if there is concern for cardiogenic shock.
  2. Corticosteroids. Consider increasing from COVID-dosing Corticosteroids to stress-dose hydrocortisone at 50mg IV every 6 hours in patients on > 2 pressors. If hydrocortisone is not available, equivalent steroids with both

Conservative Fluid Management:

Literature Review (Intravenous Fluids): Gallery View, Grid View

Do not give conventional 30cc/kg resuscitation. COVID-19 clinical reports indicate the majority of patients present with respiratory failure without shock. ARDS is mediated in part by pulmonary capillary leak, and randomized controlled trials of ARDS indicate that a conservative fluid strategy is protective in this setting (Grissom et al; Famous et al; Silversides et al). Conservative fluid management is also part of the most WHO guidelines.

If vasopressors are unavailable or limited, a conservative fluid strategy may not be possible for patients with shock. In these situations, fluid management should be guided by focusing on either shock (i.e. fluid resuscitation) or respiratory failure (i.e. conservative fluid strategy), depending on which is the most immediately life-threatening problem. Seeking expert opinion is also recommended.

  1. Give 250-500cc IVF and assess in 15-30 minutes for response:
  1. Increase > 2 in CVP
  2. Increase in MAP or decrease in pressor requirement
  1. Use isotonic crystalloids; Lactated Ringer’s solution is preferred where possible. Avoid hypotonic fluids, starches, or colloids
  1. Repeat 250-500cc IVF boluses; Use dynamic measures of fluid responsiveness
  1. Pulse Pressure Variation: can be calculated in mechanically ventilated patients without arrhythmia; PPV >12% is sensitive and specific for volume responsiveness
  2. Straight Leg Raise: raise legs to 45° w/ supine torso for at least one minute. A change in pulse pressure of > 12% has sensitivity of 60% & specificity of 85% for fluid responsiveness in mechanically ventilated patients; less accurate if spontaneously breathing
  3. Ultrasound evaluation of IVC collapsibility should only be undertaken by trained personnel to avoid contamination of ultrasound

Tool: Conservative Fluid Management protocols are available from from FACCT Lite trial (Grissom et al).

Cardiogenic ShockCopy Link!

Cardiogenic shock occurs in hospitalized COVID-19 patients, can occur late in the course, and portends a poorer outcome. The mechanisms are still being researched but potentially include direct viral toxicity, acute coronary syndrome, stress or inflammatory cardiomyopathy. Please see here for more information on Acute Cardiac Injury, Acute Coronary Syndromes, and Myocarditis.

Time course: Cardiogenic shock may present late in the course of illness even after improvement of respiratory symptoms.

WorkupCopy Link!

  1. Significant concern for cardiogenic shock if any of the following are present with evidence of hypoperfusion (e.g. elevated lactate):
  1. Elevated NT-proBNP OR
  2. ScvO2 < 60% (PvO2 < 35 mm Hg) OR
  3. Point of care ultrasound or echocardiogram with depressed LV and/or RV function
  1. Rule out acute coronary syndrome and complete the initial work up as described in Acute Coronary Syndromes.
  2. Ongoing monitoring:
  1. Labs: ScvO2 (central venous O2 sat obtained by upper body central line) or ScvO2 (mixed venous O2 sat) every 8-12 hours or with clinical change, lactate every 4-6 hours, liver function tests daily (for hepatic congestion)
  2. Daily EKGs or as needed with clinical deterioration
  3. Trend troponin to peak
  1. All cardiogenic shock cases require cardiology consult if available.
  1. PA catheters may be placed bedside by experienced providers, with preference for use only in mixed shock or complex cases with cardiology guidance

ManagementCopy Link!

Close collaboration with a cardiologist is recommended if possible.

Goals:

  • Mean Arterial Pressure 65-75, Central Venous Pressure 6-14, SCvO2 > 60%
  • If invasive monitoring is used: PCWP 12-18, PAD 20-25, SVR 800-1000, CI > 2.2
  • Note: Achieving MAP goal is first priority, then optimize other parameters

How to Achieve Goals:

  1. Continue titration of norepinephrine infusion for goal MAP 65-75. If norepinephrine is not available, then epinephrine (adrenaline) can be used.
  2. Initiate diuretic therapy for CVP > 14, PCWP >18, PAD > 25
  3. Initiate inotropic support:
  1. Dobutamine drip for SCvO2 < 60%, CI < 2.2 and MAP > 65. Start at 2mcg/kg/min. Increase by 1-2mcg/kg/min every 30-60 minutes for goal parameters. Alternative strategies should be considered once dose exceeds 5mcg/kg/min. Maximum dose is 10mcg/kg/min.
  1. Ensure negative inotropes such as beta blockers, calcium channel blockers and antihypertensives are discontinued.
  2. If the patient is failing to meet goals despite the above, consider Mechanical Support if it is available.

Cytokine Storm SyndromeCopy Link!

Updated Date: November, 2020
Literature Review:
Gallery View, Grid View

MechanismCopy Link!

Also called cytokine release syndrome, CSS is an umbrella term used for many different cytokine-driven illnesses that share certain aspects of pathophysiology but differ in serum cytokine patterns, timing, and other factors (Henderson et al). Viral infections, especially EBV and influenza, are a known cause of cytokine storm, and it has been implicated in SARS and MERS-associated ARDS as well (Schulert et al, Kim et al).

Cytokine Storm Syndrome in COVID: A subgroup of patients with severe COVID-19 have an immune hyperactivation that resembles CSS (Mehta et al, Henderson et al).

  • Evidence of cytokine storm syndrome in COVID-19 includes correlation of elevated D-dimer, ferritin (a marker of macrophage activation), and soluble IL-2 receptor (a marker of T lymphocyte activation) with severe disease course (Zhou).

Mechanisms: Cytokine storm reflects impaired control of immune responses, leading to leukocyte activation and release of cytokines such as IL-1, IL-6, and IFN-gamma (Mangalmurti et al; Vabret et al; Henderson et al; Tay et al). CSS usually originates from dysfunctional interactions between the innate immune system and the adaptive immune system as follows: (See illustration in Subbarao et al). The adaptive immune system takes 5-7 days to respond to a new antigen, which may explain why CSS usually occurs around/after this timepoint in the course of disease:

  1. The adaptive immune system fails to kill activated innate immune cells.
  2. If the innate immune cells are not shut down, both the innate cells (especially monocytes and macrophages) and adaptive cells continue to produce pro-inflammatory cytokines, activating positive feedback loops
  3. The immune response fails to move toward the resolution phase and instead causes amplification of the immune response, especially systemic inflammatory cytokine production.
  4. These inflammatory cytokines cause upregulation of complement proteins, clotting factors, and other substances that can cause target cell damage. This in turn leads to further inflammation.

Consequences: Cytokine storm syndrome has a number of downstream clinical consequences, including:

  • Secondary to hypotension/AKI/ATN, microthrombi, or other mechanisms

ManagementCopy Link!

Diagnosis: Suspect cytokine storm in patients with the following lab and clinical findings.

  1. Clinical findings of severe COVID-19:
  1. Escalating supplemental oxygen requirement or work of breathing
  2. Shock/septic physiology
  3. Unexplained myocardial dysfunction
  4. ICU admission
  1. Labs suggestive of possible cytokine storm:
  1. General markers: neutrophilia, lymphopenia, elevated hepatic transaminases, elevated LDH
  2. Disseminated Intravascular Coagulation markers: elevated D-dimer, thrombocytopenia, falling fibrinogen, prolonged PT / PTT.
  1. Fibrinogen is also an acute phase reactant, so it may be elevated in CSS. If fibrinogen levels fall rapidly from baseline, or fall below the normal range, consider active DIC.
  1. General inflammation markers: Elevated C-reactive protein (CRP), ESR, ferritin (all of these markers are non-specific)
  1. Ferritin even in severe CSS in COVID-19 is only moderately elevated (typically no higher than low 1000s), in contrast to other types of CSS.
  1. Targeted immune cell activation markers: soluble IL-2 receptor (sCD25), IL-6
  1. These tests may not be available, or may take several days to result and should not delay clinical care.
  1. Keep in mind that procalcitonin is downstream of IL-6 and IL-1, so it is not a specific marker of infection in the setting of cytokine storm

Screening: All hospitalized patients with COVID-19 should receive laboratory screening for CSS. Please see Lab Monitoring for recommendations

  1. CSS may show up in the labs before it appears clinically, and suggestive lab findings merit early consideration of immunomodulators given higher risk of progression to ARDS, shock, and multiorgan failure (Chen).
  2. CSS should be considered if the following lab parameters are met (though some patients may not meet these cut-offs):
  1. CRP >50mg/L
  2. At least two of the following:
  1. Ferritin >500 ng/mL
  2. LDH >300 U/L
  3. D-dimer >1000 ng/mL

Monitoring: Patients with suspected or confirmed CSS should receive the following monitoring labs:

  1. CRP and fibrinogen are dynamic and should be checked daily, along with daily basic labs (CBC with diff, BMP)
  2. D-dimer, ferritin, LDH, LFTs tend to change more slowly and can therefore be checked every 2 days
  3. sIL2R and IL-6 can be monitored 1-2 times per week
  1. Keep in mind that serum IL-6 levels often go up after tocilizumab and sarilumab, likely because the cytokine is displaced or blocked from the receptor by the IL-6 receptor-blocking antibody. Therefore, monitoring IL-6 after tocilizumab or sarilumab is not clinically useful.

Management: The management of CSS in COVID-19 is actively evolving. If there is suspicion of developing or ongoing cytokine storm, the patient should be started on Corticosteroids (if they are not already on them). If they are worsening, they should be discussed with infectious disease, rheumatology, and/or pulmonary/critical care specialists before initiating other immunomodulatory agents (IL-1 and IL-6 agents typically).

Cardiac ArrestCopy Link!

Updated Date: November, 2020
Literature Review:
Gallery View, Grid View

In-Hospital Cardiac Arrest ManagementCopy Link!

Out-of-hospital cardiac arrest (OHCA) is not covered in this guide, only in-hospital (IHCA). Because lay people and emergency responders often initiate chest compressions, risk to bystanders is a concern. Follow local guidance.

Tool: BWH Hospital Cardiac Arrest Code Quick Guide

Cardiac Arrest OutcomesCopy Link!

In a study of 5019 critically ill patients across 68 U.S. hospitals, 14% had a cardiac arrest, and 57% of those received CPR. The most common rhythms were PEA (49.8%) and asystole (23.8%). Only 12% of the patients who underwent CPR survived to hospital discharge (Hayek et al).

Early Goals of Care ConversationsCopy Link!

To avoid unnecessary codes in patients with an irreversible underlying condition leading to cardiac arrest, patients should all have goals of care discussions on admission. Those who are at high-risk for acute decompensation should be identified early and code status confirmed with the patient and family. In some places and circumstances it may be appropriate to not offer resuscitation if there is no reasonable chance of recovery or if there are no critical care resources to care for the patient if ROSC is achieved. Rules, laws, regulations, as well as cultural and religious values about code status vary widely and local laws must be obeyed.

Minimizing Healthcare Worker ExposureCopy Link!

Code Responses to COVID-19 patients are high-risk events for healthcare worker exposure due to aerosolization with chest compressions and intubation

  • Use PPE:
  • CDC guidelines recommend N95 respirator, face shield, gown and gloves be used by all code responders during code events (CDC Guidelines, 2020).
  • Minimize personnel:
  • Use an automated compression device where available to minimize personnel.
  • Cover the patient’s face:
  • If it does not interfere with oxygen equipment, place a surgical facemask and/or a blanket over the patient’s face prior to chest compressions while awaiting a definitive airway.
  • Prepare code equipment:
  • To limit transmission of virus while passing meds/supplies into the patient’s room from the code cart, consider creating Code Bags inside the Code Cart pre-packed with necessary code meds (Epinephrine, Bicarbonate, Calcium etc.) and IV/lab supplies.

Cardiopulmonary ResuscitationCopy Link!

Compressions:

  1. Shock early: If the patient is on monitoring and has shockable rhythm (VF/VT), defibrillate as soon as possible (even if this means pausing compressions before 2 minutes is complete)
  2. Proned patients: If a patient has been proned for ventilatory purposes and they develop cardiac arrest, a decision should be made by the medical and nursing team whether to de-prone the patient.
  1. If the patient is able to be safely de-proned in an efficient manner, the medical team should do so
  2. If the patient is not able to be de-proned due to limited staff, or concerns about extubation or line/tubing entanglements, the team should proceed with reverse precordial compressions, also called Reverse CPR (Brown et al).
  1. Reverse precordial compressions are performed by placing a clenched fist beneath the sternum while administering compressions to the midthoracic spine between the inferior scapulae (Sun et al). This is optimally performed with one person administering compressions and one person holding counter-pressure beneath the sternum.

Airway:

  1. Avoid bag valve mask or rescue breathing: Until a definitive airway is obtained, compression-only CPR with passive oxygen delivery should be performed. Multiple studies have shown that compression-only CPR is non-inferior to standard CPR (Svensson et al).
  1. If the patient is already on high-flow nasal cannula or non-invasive ventilation (CPAP, BiPAP) these can be continued
  1. Proceed with Rapid Sequence Intubation as early as possible if the patient does not have a shockable rhythm,
  1. To maximize the success rate for intubation, airway interventions should be carried out by experienced individuals and chest compressions should be stopped briefly during intubation (Cheung). Pausing compressions is a deviation from usual cardiac arrest care, however this is acceptable to maintain the safety of code responders and minimize attempts at intubation. See Intubation. Chest compressions should resume once the endotracheal tube (ETT) cuff is inflated.
  2. If the pause in chest compressions is excessive and endotracheal intubation does not seem likely, consider a laryngeal mask or other extraglottic airway device.
  1. Initial Ventilator Settings
  1. Patients should be placed on the following settings, consistent with AHA ACLS guidelines (Edelson et al) unless the patient was already on the ventilator (in which case they should not be disconnected) or clinical information suggests different ventilator settings be used:
  1. Vt 500 cc, RR 10, PEEP 5cm H20, FiO2 100%
  2. Post-return of spontaneous circulation patients should be placed on Lung Protective Ventilation

Reversibility:

  1. It is important to attempt to identify and treat reversible causes before stopping the code. Hypoxemia, hypo/hyperkalemia, hypoglycemia, hypovolemia, acidosis, hypothermia, pulmonary embolus,acute coronary syndrome, tension pneumothorax, cardiac tamponade, toxin
  2. Cause of Death in COVID patients is largely respiratory failure (see Cause of Death)
  3. Terminating Resuscitative Efforts
  1. Legal rules on the termination of resuscitative efforts vary by location and should always be observed. Within these parameters, avoid prolonged resuscitation if there is no easily reversible etiology identified. Medically, it is reasonable to stop resuscitation efforts if return of spontaneous circulation (ROSC) has not been achieved within 30 minutes as the chance of meaningful recovery is low (Goto)
  2. In intubated patients, failure to achieve an ETCO2 of greater than 10 mm Hg by waveform capnography after 20 minutes of CPR should be considered as one component of a multimodal approach to decide when to end resuscitative efforts (Mancini et al).
  1. Post-Resuscitation Care
  1. Dispose of, or Clean all equipment used and any work surfaces.
  2. Doff PPE.
  3. If ROSC is achieved, provide usual post-resuscitation care consistent with current recommended guidelines including Targeted Temperature Management where possible (Donnino et al).

Brain Death and Targeted Temperature ManagementCopy Link!

Please see BWH guidelines for more information about Diagnosing Brain Death and Targeted Temperature Management after cardiac arrest.

Prevention of ComplicationsCopy Link!

Updated Date: September 24, 2020

Best Practice ChecklistsCopy Link!

(As adapted from WHO interim SARI guidance)

Goal

Intervention

Reduce days of invasive mechanical ventilation

Reduce incidence of ventilator associated pneumonia

  • Oral intubation is preferable to nasal intubation in adolescents and adults
  • Keep patient in semi-recumbent position (head of bed elevation 30-45 degrees)
  • Use a closed suctioning system, periodically drain and discard condensate in tubing
  • Use a new ventilator circuit for each patient, once patient is ventilated, change circuit if it is soiled or damaged but not routinely
  • Change heat moisture exchanger when it malfunctions, when soiled, or every 5-7 days

Reduce incidence of venous thromboembolism

Reduce incidence of catheter-related bloodstream infection

  • Use checklist of steps for sterile insertion, verified by an observer in real time
  • Use a daily reminder to remove catheter if no longer needed

Reduce incidence of pressure ulcers

  • Turn patient every 2 hours

Reduce incidence of stress ulcers and gastrointestinal (GI) bleeding

  • Give early enteral nutrition (within 24-48 hours of admission).
  • If available, also give histamine-2 receptor blockers (e.g. famotidine 20mg IV BID) or a proton pump inhibitor (e.g. pantoprazole 20-40mg IV daily) if history of GERD or GI bleed

Reduce incidence of ICU - related weakness

  • Actively mobilize patient early in the course of illness when safe to do so
  • Assure Nutrition and give a multivitamin with minerals, Thiamine 100mg, and Folate 1mg daily

Tool: BWH COVID-19 ICU Bundle
Tool: OCC ICU Daily Rounding Checklist

Critical Illness Neuropathy and MyopathyCopy Link!

ICU-acquired weakness has been observed in 25-46% of ICU patients. Duration of ventilation, corticosteroid administration, multi-organ dysfunction, sepsis, hyperglycemia, and renal replacement therapy have all been correlated with ICU-acquired weakness (De Jonghe). Data are mixed regarding correlation of steroids and NMBA administration with ICU-acquired weakness (Doughty).

Tool: BWH General Guidelines on Critical Illness Neuropathy and Myopathy

Nutrition in ICU PatientsCopy Link!

Updated Date: June, 2020
Literature Review:
Gallery View, Grid View

Nutrition (ICU)Copy Link!

Enteral nutrition is recommended for intubated patients. It is important to maintain muscular strength and reduce stress ulcers.

  1. In most patients:
  1. Standard 1.5 calorie tube feeds (e.g. Osmolite 1.5 @10mL/hr., advance by 20mL Q6h to goal 50mL/hr)
  1. If renal failure and high K or phosphorus:
  1. Renally formulated tube feeds (e.g. Nepro @ 10mL/hr, advance by 10mL Q6h to goal 40mL/hr)
  1. If on pressor support:
  1. Hold tube feeds due to the risk of intestinal ischemia if on:
  1. Any two pressors
  2. Epinephrine > 5 mcg/min
  3. Norepinephrine > 10 mcg/min
  4. Phenylephrine >60 mcg/min
  5. Vasopressin >0.04 units/min
  1. If unable to tolerate enteral nutrition support given escalating or multiple vasopressors TPN should be considered.
  1. If paralyzed:
  1. It is safe to feed while patients are on paralytic agents such as cisatracurium
  1. If prone:
  1. Patients requiring proning may continue to receive tube feeding.
  2. The tube feeds should be held for one hour prior to turning the patient.
  3. Prokinetic agents may be beneficial during proning to enhance gastric emptying and decrease risk of vomiting.

Glucose Management (ICU)Copy Link!

Glucose Management and DKA:

  1. Goal glucose range is typically 140-180, though some places prefer tighter control
  2. Management of DKA in COVID is challenging given the frequent need for blood sugar checks and the PPE/Donning/Doffing involved. In some instances, subcutaneous insulin might be used instead of a drip.

Tool: BWH Guidelines on ICU Management of DKA for COVID Patients

Procedures and LinesCopy Link!

Updated Date: August, 2020

Arterial and Venous CathetersCopy Link!

Literature Review (Central Line): Gallery View, Grid View
Literature Review (Arterial Line):
Gallery View, Grid View

Placing and removing arterial and central venous catheters is the same in COVID patients as it is in others. Given the duration of time and proximity to the patient’s face, some institutions may want to consider treating these as aerosol generating procedures.

Provider should make sure to don and doff PPE properly, take only needed supplies in the room, and clean all durable equipment thoroughly on departing.

Tool: NEJM video on Arterial Lines
Tool:
NEJM video on Central Venous Catheters

Arterial Line HeparinCopy Link!

In the COVID-ICU we have seen frequent arterial line thrombus formation. A possible means to prevent this is the use of heparinized saline in the arterial line pressure bag.

Patient selection:

  • Requiring more than 1 arterial line placements (or re-wiring) due to thrombus
  • Clinical team discretion based on patient specific factors (e.g. line access issues)
  • Contraindications: history of heparin-induced thrombocytopenia and/or currently on systemic anticoagulation

Product and dosing:

  • Heparin infusion in normal saline 2 units/mL 500 mL bag. Not all pharmacies will have the correct formulation of heparin for this aim.
  • Typical dose: 5mL/hr (10 units/hr) continuous via the arterial line

Nasogastric Tubes and ThoracentesesCopy Link!

Placement is standard, however given the proximity to the patient’s oropharynx and tendency to cough, treat these as aerosol generating procedures.

Tool: Tulane video on NG tubes

Tool: NEJM video on Thoracentesis

BronchoscopyCopy Link!

Literature Review (Bronchoscopy): Gallery View, Grid View
Tool:
BWH COVID-Specific Bronchoscopy Guidelines.

Chapter 9

Psychosocial Support

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

Psychological Impact of PandemicCopy Link!

Updated Date: September 24, 2020
Literature Review:
Gallery View, Grid View

The COVID-19 pandemic has introduced a number of new stressors and challenges to individuals and communities. Necessary infection control actions (such as physical distancing, quarantine, and isolation) can be stressful and psychologically impactful. Mitigation of harm in the context of pandemic response requires attention to the psychological needs of populations, families, and individuals (CDC).

Particular attention to the stressors of quarantine is warranted at a time when a very large portion of the global population is under quarantine. Quarantine of 10 days or longer is considered long-duration and is particularly stressful (adapted from Brooks et al).

Stressors associated with quarantine can include:

  • Frustration, boredom, and loneliness
  • Inadequate supplies and access to regular medical care
  • Insufficient information
  • Fears about becoming infected and/or infecting others
  • Financial loss and socioeconomic distress
  • Stigmatization and rejection from family, neighbors, co-workers or friends
  • Challenges of resuming one’s “normal” routine

Anxiety and DepressionCopy Link!

Updated Date: August, 2020
Literature Review (Anxiety and Depression):
Gallery View, Grid View
Literature Review (Trauma and Stress- Related Disorders):
Gallery View, Grid View
Literature Review (Psychosis):
Gallery View, Grid View
Literature Review (Suicide):
Gallery View, Grid View

Anxiety and Post-Traumatic Stress Disorder (PTSD)Copy Link!

Anxiety and PTSD are significant risks for patients with COVID infection. Meta-analyis of psychological studies on survivors of Middle-East Respiratory Syndrome (MERS) and previous SARS outbreaks has indicated a PTSD prevalence of over 30% (Rogers et al). As of November 2020, Lanzhou University researchers have established a living systematic review for accurate ongoing study on prevalence of PTSD and other mental health disorders in COVID patients (Shi et al).

See treatments for Nonpharmacologic and Pharmacologic Management of acute anxiety

DepressionCopy Link!

Updated Date: August, 2020

In the United States, prevalence of self-reported depression in adults was four times higher at the end of June, 2020, than it was in the second quarter of 2019 (Czeisler et al). Symptoms of depression can include a dysphoric (unhappy) mood, difficulty concentrating, social withdrawal, disrupted sleep, decreased appetite, fatigue, tearfulness, and a sense of worthlessness, hopelessness, and helplessness.

Non-Pharmacologic interventions for Depression: see Coping and Support Strategies.

Tool: Supportive Resources (Hotlines, therapy resources, and mindfulness/CBT Training)

Pharmacologic interventions for Depression: Continue home medication regimen if one is in place. Starting a Selective Serotonin Reuptake Inhibitor (SSRI) may help, but will likely take six weeks to take effect. For depression with sleep disruption and low appetite, mirtazapine 7.5 milligrams is a good choice.

AddictionCopy Link!

Updated Date: August, 2020
Literature Review:
Gallery View, Grid View

Alcohol Use DisordersCopy Link!

Clinicians have anecdotally noted significant changes in patients’ drinking habits during the pandemic in association with the stresses of isolation, with increased risk of relapse and dysfunctional drinking. Survey data in the United States indicated a 14% general increase in frequency of alcohol use above baseline levels, with a 41% increase in days of heavy drinking specifically in women (Pollard et al).

Alcohol Withdrawal: For unsupervised mild to moderate withdrawal, consider use of anticonvulsant medications such as gabapentin, topiramate or carbamazepine (SAMHSA Guidelines) or a short course of benzodiazepines.

If clinically appropriate for a COVID-19 positive inpatient, a scheduled benzodiazepine or phenobarbital taper may be preferable to a protocol requiring frequent assessment and administration of benzodiazepines (which is often more labor-intensive for the nursing team, requiring more PPE use and close patient contact for appropriate monitoring)

  • Instead of scoring-based systems (e.g CIWA), consider less-frequent as needed or scheduled benzodiazepine tapers
  • Consider phenobarbital load/taper if familiar, as this requires less frequent monitoring in most cases.

Tool: EM Crit Review of Literature on Phenobarbital (contains several example dosing regimens)
Tool: Addiction Resources for Patients and Families

Opioid Use DisordersCopy Link!

Updated Date: August, 2020

If opioid replacement therapy is available for addiction treatment in your practice location, make all efforts in accordance with local guidelines to prevent the pandemic from disrupting patient access to these medications.

The Harm Reduction Coalition has created a tip sheet for adapting services which can be viewed here. Recommendations include equipping program participants with extra supplies, if available (such as the overdose rescue medication Naloxone), and considering 1-month prescriptions for patients who are taking buprenorphine. In the USA the United States Drug Enforcement Administration (DEA) has changed guidelines during the pandemic to help reduce interruptions in care (Covid-19 Info Page).

Buprenorphine in the USA: Practitioners who prescribe buprenorphine are now able to prescribe it to new patients with opioid use for maintenance treatment or detoxification treatment following an evaluation via telephone voice calls, without first performing an in-person or telemedicine evaluation, if deemed clinically appropriate and safe.

Tool: Decision Tree for Prescribing Controlled Substances During COVID-19 (USA specific)
Tool: Addiction Resources for Patients and Families

Addiction Resources for Patients/FamiliesCopy Link!

(US-centric, but some are more widely available)

  1. Online Alcoholics Anonymous (AA) Groups
  2. Dharma Online Meetings
  3. In the Rooms Online Support - A global recovery online community
  4. Al-Anon Online Meetings - Al-Anon is an organization for anyone who is affected by alcoholism in a family member or friend. Utilizes Twelve Step principles.
  5. Virtual Narcotics Anonymous (NA) Groups: (Online) or (Phone)
  6. Marijuana Anonymous Online Meetings
  7. Smart Recovery Online Meetings
  8. Tools for Smoking Cessation
  9. Herren Project - Meetings 7 nights per week at 7:30 PM EDT via Zoom platform for a peer support based live online recovery meeting. Each meeting begins with a speaker, followed by fellowship and sharing.
  10. Connections - Free smartphone app for sobriety - features include sobriety tracking, access to e-therapy, messaging with trained counselors and peers, journaling, resource library including videos and testimonials

Palliative and Symptom ManagementCopy Link!

Updated Date: September, 2020
Literature Review (Palliative Care):
Gallery View, Grid View

Discussing Goals of CareCopy Link!

Literature Review: Gallery View, Grid View
Tool: The REMAP Framework. Reframe, Expect Emotion, Map Values, Align, Propose Plan (Childers et al)
Tool: Hospital Medicine #1: Goals of Care & Code Status (goals consistent with intubation)
Tool: Hospital Medicine #2: Goals of Care & Code Status (goals not consistent with intubation)
Tool: ED #1: Goals of Care & Code Status (goals not c/w intubation)
Tool: Experts at VitalTalk have created a COVID-19 Communication Guide. See also: Suggested Language for COVID-19 scenarios

Providing Palliative CareCopy Link!

Tool: Essential Package for Palliative Care (Medications, Equipment, Locations, Personnel).

Palliative care is ethically imperative care focused on prevention and relief of suffering of adult and pediatric patients and their families facing life-threatening illnesses, including COVD-19. Palliative care, including relief of symptoms and provision of psychosocial support, should be available to all patients with COVID-19 at all stages of illness.

Caretakers and family members should be given access to adequate training in caregiving and infection control and to appropriate personal protective equipment. They should have access to the same psychological, social and spiritual care as patients, and also bereavement support.

Managing SymptomsCopy Link!

Literature Review: Gallery View, Grid View

Symptom

Non-pharmacologic Treatment

Pharmacologic Treatment

Dyspnea

Oxygen and/or a fan on the patient’s face can decrease sensation of dyspnea

If pulmonary edema also may be present, consider furosemide. If the patient also may have COPD or asthma, use bronchodilators and consider steroids.

Pharmacologic and Opioid Treatment to Reduce Dyspnea.

Respiratory Secretions at End of Life

Communicate with families to expect sounds. Reassure them that although the “rattling” sound is distressing to hear, the patient is not experiencing difficulty breathing or having to clear phlegm from their throat.

See here for guidance on respiratory secretions that are troubling for the patient

Pain

For mild pain: paracetamol/acetaminophen or other non-opioid agents

For moderate or severe pain: Opioid Agents

Nausea / Vomiting

Consider and treat other causes such as gastritis, constipation or anxiety

If pharmacologic treatment is needed, there are many options.

Haloperidol can be used if other drugs are not available or ineffective

Constipation

Can be caused by slow transit due to opioid, anticholinergic medicines, immobility, volume depletion. Treat these first.

Some options include bisacodyl 5 – 10mg orally QD – BID or Lactulose 15 – 30ml orally QD - BID

Anxiety

Most common cause in patients with COVID-19 is dyspnea. Anxiety usually resolves when dyspnea resolves or is adequately relieved (see above)

If treating dyspnea is not adequate, there are many other options.

Depression

See Psychosocial Support

See above

Delirium & Agitation

Use the Confusion Assessment Method (CAM) to help diagnose. Consider placing patient in a quiet location, frequent re-orientation, promoting normal sleep-wake cycles.

Pharmacologic management is covered in Delirium (including terminal delirium). Avoid benzodiazepines in most circumstances.

Tool: Pallicovid.app for one-page guides, pocket cards, nursing resources, and related information.

Caring for an Imminently Dying PatientCopy Link!

Signs and symptoms of imminent death include:

  • Somnolence
  • Warmth, and later cooling and mottling of extremities
  • Change in respiratory pattern, intermittent apnea, Cheyne-Stokes pattern
  • Gurgling sounds from oropharynx (often more distressing to family than patient)

Symptom management:

  • Should follow the guidelines provided in sections above

Communication

Compassionate Cessation of Ventilator SupportCopy Link!

In some settings there is a legal and social framework for cessation of ventilator support if families prefer it or if it is no longer in the patient’s best interest. If this is pursued, for staff and visitor safety, we do not recommend physical extubation, but rather patient should be weaned down to PSV 0/0 with FiO2 0.21 to maintain a closed circuit

Tool: Ventilator withdrawal protocol: Von Gunten and Weissman, Palliative Care Fast Fact 33
Tool:
Palliative sedation: Salacz and Weissman, Palliative Care Fast Fact 106

Communicating EffectivelyCopy Link!

Psychological Skills:

Tool: ASC Guidance on Basic Psychosocial Skills: A Guide for COVID-19 Responders
Tool:
NURSE Skills for Responding to Emotion (Name, Understand, Respect, Support, Explore) (Back et al)

Assess Understanding & Delivering Information

Tool: ASK-TELL-ASK (Back et al).

Managing Uncertainty

Key Skill: Pairing hope and worry (Jackson et al).
E.g. “I hope you will improve AND I am also worried because your oxygen level is getting worse.”

Rights to Refuse CareCopy Link!

Updated Date: December 20, 2020

Capacity AssessmentCopy Link!

Literature Review: Gallery View, Grid View

Capacity is an essential part of consent and the process of decision-making. The evaluation of capacity is decision-specific and time-specific, and involves a need to balance autonomy and beneficence. A capacity assessment can be completed by any physician. There are four decision-making abilities that patients must demonstrate in order to have decision-making capacity:

  1. Ability to understand relevant information
  1. Patient must be able to understand basic information about their current condition, possible options, and risks/benefits associated with these options
  1. Ability to understand the situation and its consequences
  1. This is the ability to recognize how the information provided by medical professionals is related to one’s own situation
  1. Ability to reason
  1. Ask the patient to describe how they reached their decision, and which factors influenced this decision-making process
  1. Ability to communicate and express a consistent choice

“Competency” is a legal status based on a global assessment of a patient’s ability to perform actions important to their health and survival. “Capacity” refers specifically to the ability to make decisions, and is based on functional assessment by a clinician. Capacity can change over time and in relation to different decision-making scenarios. Competency may be defined differently depending on the legal system.

Tool: Several structured tools for capacity assessment exist including the Aid to Capacity Evaluation (ACE)

Declining Medical InterventionsCopy Link!

  1. Assess urgency of intervention and consequences of refusal. The clinician needs to distinguish between tolerable (acceptable) risks and intolerable risks. Only intolerable risks require Assessment of Capacity. Intolerable risks might include behavior that is new and unprecedented (not consistent with past behavior as best as can be determined by obtaining collateral from the patient’s chart or associates) and behavior that is causing significant harm to self or others.
  1. Assess different possible causes for refusal: delirium, anxiety, agitation, volitional (lack of motivation, for example in the context of depression), and psychosocial stressors
  1. Patients who refuse testing or COVID-19 isolation precautions will likely require a Capacity Assessment. Given that symptomatic patients seem to transmit the virus to others, the patient will need to demonstrate that they understand the risk to themselves and to the people around them.
  1. If the patient has capacity and the decision harms only themself (for example, refusing testing), this is within their rights. Note that if a patient is living in a congregate setting, this may interfere with their living situation.
  2. If the patient has capacity and the decision is endangering others (for example leaving their isolation room without precautions and exposing other patients), try first to reason with them. Many hospitals have policies around rearranging or declining care to a patient who is harming others.
  3. If a patient lacks capacity and is harming others, safe use of restraints is allowed in most hospital settings.

Leaving Against Medical Advice (AMA)Copy Link!

  1. Identify psychosocial stressors and address them as thoroughly as possible. For patients with unstable housing, do your best to call any available shelters. Local departments of health, departments of housing, and non-governmental organizations may have specific isolation options for undomiciled patients.
  2. Patients with COVID who request to leave AMA will likely require a Capacity Assessment.
  3. If a patient with COVID is found to have capacity to leave AMA but poses significant infection risk to others (is unable or unwilling to self-isolate, lives with high-risk individuals), discuss this with the legal and/or risk team at your hospital if you have one. In many (but not all) places the Department of Public Health will have to be alerted, and they will pursue potential contact tracing or notification of susceptible contacts. Facilities should put in place plans to notify the local or state health department, ideally with an established system and not a case-by-case basis.
  1. Note that in most places it is illegal to share medical information about the patient with his or her contacts without permission, though some places make exceptions to this rule if the contact is imminent danger (e.g. if the person has made a credible threat of bodily harm), and many local laws (like HIPAA) has exceptions for communicable disease. If you suspect that a specific susceptible person may be placed at grave risk by your patient, it may be appropriate to call and report (WHERE?)
  1. If a patient is tested but leaves AMA before receiving their results, every attempt should be made to inform them of their results status, and the information needed to prevent transmission.

Tool: CDC If You are Sick or Caring for Someone

Supporting Vulnerable GroupsCopy Link!

Updated Date: September, 2020

Defining Vulnerable GroupsCopy Link!

Vulnerable groups are those needing additional psychological, social, and material support in order to adapt to the pandemic. Groups in need of support are identified by conditions which render individuals potentially more vulnerable to the effects of pandemics (such as poverty or pre-existing mental health conditions). They can also be identified on the basis of quarantine/hospitalization status or occupation. This section suggests some ways in which vulnerable groups may be assisted by health providers, local governments, service organizations and/or community leaders and members.

Vulnerable populations include (IASC with PIH adaptation):

  • Older adults, especially those with cognitive decline or dementia;
  • People living with disabilities with pre-existing health conditions; people living with disabilities, including psychosocial disabilities;
  • People living with disabilities in crowded living conditions (e.g. prisoners, people in detention, refugees in camps and informal settlements, older adults in nursing homes, people in psychiatric hospitals, inpatient units or other institutions);
  • Homeless individuals or individuals with unstable housing
  • People living with disabilities at particular risk of discrimination or violence, such as those at risk due to COVID-19-related stigma (e.g. specific ethnic groups, health workers)
  • People exposed to gender-based violence, including sexual violence;
  • Pregnant, postpartum or in post-abortion, and lactating women;
  • Children, adolescents and their caregivers;
  • People with difficulties in accessing services (e.g. migrants).

Supporting Vulnerable GroupsCopy Link!

Older adults, People with Disabilities, and people with severe mental illness may be particularly vulnerable and in need of support. For example, older adults with cognitive deficits, decline, and/or dementia may become more anxious, agitated, and withdrawn during the outbreak and while in quarantine. People with severe mental illness may need additional support from organizations, communities, and family members to enhance treatment success, stop the spread of the virus, and maximize prevention.

  1. Assist people with accessing information. Messages should be shared in ways that are understandable to the individual.
  2. If caregivers need to be moved into quarantine, plans should be made to ensure continued support for those who need it.
  3. Engage families and other support networks in providing information and promoting infection prevention measures (handwashing, universal face covering, etc.)
  4. Assist people in continuing to access necessary medical care.
  5. People requiring caregiving and their caregivers should be included in all stages of the outbreak response.

Tool: See This Document From WHO for more recommendations for supporting both general and specifically vulnerable populations.

Coping and Support StrategiesCopy Link!

Updated Date: December 20, 2020
Literature Review (Health Care Worker Mental Health):
Gallery View, Grid View

Psychological First AidCopy Link!

Psychological First Aid (PFA) OverviewCopy Link!

Psychological first aid (PFA) is a structured way to have conversations that allows anyone (citizens, mental health care providers, community health workers, etc) to provide humane support to a fellow human being. PFA involves helping people to feel safe, connected to others, calm, and hopeful; have access to social, physical, and emotional support; and feel able to help themselves, as individuals and communities. These efforts must respect the safety, dignity, and rights of participants (WHO Outline). PFA does not need to be done by a professional, and it is not professional counseling.

Psychological First Aid TrainingCopy Link!

The recommended length for a PFA training is 3 hours and topics include: principles of PFA, special considerations for vulnerable populations, and referral pathways for additional psychological care (as required). PFA training materials and activities should be adapted to the context.

  • Anyone, mental health care providers, community health workers, general healthcare workers, support staff, etc., can be trained in and provide PFA.
  • PFA training can be standalone or adapted to be a part of existing COVID-19 related trainings. Additional trainings can be conducted as needed.

Tool: WHO PFA Manual (available in multiple languages).

Tool: PFA and MHPSS training materials developed by Partners In Health are available upon request. Please click here to complete the material request form.

Supporting PatientsCopy Link!

The following is adapted from WHO; Banerjee, and PIH

All emotions and stress responses are “normal”. The following strategies can be used by healthcare workers to support distressed people who have been recently exposed to a serious crisis event.

  1. Be attentive to safety and basic needs: offer immediate assistance by looking for ways to make people feel safe and comfortable (blankets, water, somewhere to sit, etc). If directing quarantine or isolation, keep it as short as possible and restricted to what is scientifically indicated.
  2. Be attentive to how you communicate: provide clear, practical communication about the nature of disease, reason for quarantine and treatment, and other essential information. Information will often need to be repeated and must be developmentally, culturally, and language appropriate.
  • Do not pressure the person to tell you what they have been through or how they are feeling.
  • Allow for sharing silence. When possible, allow people to talk as little or as much as they want to.
  • Validate (show acceptance towards) feelings and thoughts. Avoid temptation to judge the rightness or wrongness of people’s reactions to experiences.
  1. Help people feel calm: keep your tone of voice calm and soft; make eye contact; remind them you are there to help and they are safe (if it is true). If feeling disconnected from their surroundings, it may help them to make contact with the environment and themselves.
  • Guide them to place and feel their feet on the floor.
  • Ask them to tap fingers or hands on their lap.
  • Guide them to notice some non-distressing things in the environment (such as things they can see, hear, or feel).
  • Focus together on breathing, and breathing slowly.
  1. Encourage healthy coping habits: for a list of these, see the bottom of this section.
  2. Connect people with other supportive individuals: connect them to available therapy resources, spiritual practitioners, and family members. Provide telephone and video resources, as available, to facilitate connections.
  3. Support emotional re-adjustment after the crisis, by encouraging: acceptance of the event and the losses; identification, labeling, and expression of emotions; and recovery of a sense of control and mastery over our lives.

Supporting Family MembersCopy Link!

The following is adapted from the Center for the Study of Traumatic Stress

  1. Manage uncertainty and elevated distress: remind family that their loved one is being cared for, and that the majority of hospitalized individuals are successfully treated and are able to return home.
  2. Support connection and communication: create opportunities for the family to communicate safely with the hospitalized family member. Set expectations for frequency of updates from healthcare providers.
  3. Support advance planning: recommend obtaining ready access to the patient’s medical, legal, and financial documents during hospitalization should the need arise. If health status deteriorates, encourage important conversations prior to the need for intubation to understand last wishes and provide opportunities to say goodbye.

Supporting ChildrenCopy Link!

Tool: My Hero is You, Storybook for Children on COVID-19, IASC
Tool:
Responding to the Mental Health and Psychosocial Impact of COVID-19 on Children and Families, UNICEF
Tool: Early Childhood Focused COVID-19 Resources

Tool: Resource for Discussing COVID-19 with Children

Supporting Healthcare WorkersCopy Link!

Updated Date: April 23, 2021

Frontline workers, including nurses, doctors, ambulance drivers, case identifiers, community health workers, and others are experiencing new occupational stressors during the pandemic. Identifying and reducing sources of stress and hardship is a shared responsibility of organizations and individuals.

  1. Reduce stress by taking every possible measure to reduce risk of physical and psychological harm to healthcare workers and their families.
  2. Include frontline workers in designing interventions to reduce workplace stress and moral distress.
  3. Show empathy and be available: understand that everyone is likely to be feeling overwhelmed.
  4. Ensure good quality communication and accurate information updates to all staff.
  5. Provide opportunities for additional education so that healthcare workers can adapt to new patient needs. See psychological first aid section.
  6. Rotate workers from higher-stress to lower-stress duties.
  7. Partner inexperienced workers with their more experienced colleagues, especially when doing community outreach.
  8. Initiate, encourage, and monitor work breaks.
  9. Protect staff from financial harm when needing to quarantine or care for sick family members (provide paid time off whenever possible).
  10. Implement a flexible work schedule/time for workers. Staff may need extra time to attend to personal matters.
  11. If you are a team leader or manager in a healthcare facility, facilitate access to mental health and psychosocial support services.
  12. Managers must be able to role-model self-care strategies to mitigate stress

Tool: Mental Health and Psychosocial Considerations for Volunteers in COVID-19, IFRC

Self-Care ResourcesCopy Link!

Specific suggestions are available in other sections for:

Therapy Resources and Mindfulness ToolsCopy Link!

Tool: Supportive Resources (Hotlines, therapy resources, and mindfulness/CBT Training)

Self-Care StrategiesCopy Link!

  1. It is normal to feel sad, distressed, worried, confused, or angry. Talk to people you trust about your feelings. Social distancing does not mean emotional distancing; use technology to connect with loved ones.
  2. Maintain a consistent routine and a healthy lifestyle as much as possible, including eating, drinking, sleep, bathing, and exercise.
  3. Keep routines consistent but flexible: don’t allow anxiety to dictate an overly rigid schedule for children and others.
  4. Maintain a positive tone within households to manage tensions and provide a sense of safety, power, and responsibility to children, elders, and others.
  5. Seek help from someone you trust if you or anyone in your home is in danger of experiencing violence or abuse.
  6. Avoid using tobacco, alcohol, or other drugs to cope with negative emotions.
  7. Consume television and internet news from reliable sources and only once or twice per day. After checking news, engage in another activity or focus on something you enjoy.
  8. Engage in relaxation activities and/or spiritual exercises, such as mindful breathing, meditation, or religious practice.
  9. Find safe ways to help others in the crisis and get involved in community activities.
  10. If you feel overwhelmed, talk to a healthcare worker, social worker, or another trusted person in your community (such as a religious leader or community elder) by phone or video. Make a plan about where you would go to seek help for physical, mental, or psychosocial healthcare if needed.

Integrating Mental Health into Health SystemsCopy Link!

Updated Date: April 23, 2021

The following guidelines, drawn from a collaborative effort of the Inter-Agency Standing Committee (IASC), Partners In Health (PIH), and 55 other humanitarian agencies part of the IASC Reference Group, help teams to mount a mental health and psychosocial response to the COVID-19 pandemic.

Tool: IASC Guidance on Operational considerations for Multisectoral Mental Health and Psychosocial Support Programmes during the COVID-19 Pandemic. (Resources available in several languages)

Tool: PIH Cross-Site Mental Health guidelines, training materials, tools

The IASC Guidelines recommend that multiple levels of interventions be integrated within outbreak response activities. During the first acute emergency stage, the focus of mental health and psychosocial support response should be to work closely with the broader health and public health system to strengthen efforts of Ministries of Health on infection risk management and treatment, including: 1) prevention of COVID-19 transmission; 2) support of surveillance, case triage and contact tracing; 3) support of persons testing positive and those in quarantine/isolation, as well as their family members; and 4) support for health care workers

Recommended Initial Actions:

  1. Assess available resources for COVID-19 response (human, financial, and other).
  2. Facilitate collaboration between government agencies, non-profit organizations, and community organizations to ensure a coordinated response.
  3. Develop a plan to assist existing mental health and psychosocial services in adapting to pandemic conditions in order to meet new and ongoing population needs.
  4. Establish a strategy for supporting specific groups affected by the pandemic, including individuals testing positive for COVID, people in quarantine/isolation, the elderly, people with disabilities, and children.
  5. Ensure Community Health Workers and other front-line workers have the most up to date information about infection control and prevention and locally available resources.
  6. Train all front-line workers on essential psychosocial care principles, including communication techniques, and referral pathways.
  7. Develop a monitoring and evaluation system for mental health and psychosocial service activities

Maintaining and Adapting Ongoing Services to Meet Psychosocial NeedsCopy Link!

Community level:

  • Ensure community health workers and traditional healers have adequate PPE for home visits.
  • Create a list of the most vulnerable patients in care and determine an essential social support package.
  • Create a list of high-risk patients to ensure remote care, safety plans, and/or sufficient medication supply is provided.
  • Maintain at least a 2 meter distance between the patient and caregivers, except when medically necessary. Explain to patients and caregivers the need for this prevention measure.

Health facility level

  • Limit the need for patients to come to the health facility by shifting to remote services, particularly for those at high risk for contracting COVID-19 and home visits to the most vulnerable patients. See additional guidance below on remote services.
  • Ensure seating areas in the waiting room are adequately spread out.
  • Limit the number of people accompanying a patient to the health facility.
  • Work with the pharmacy team to obtain extended medication supply for stable patients.

Remote services and supervision

  • Determine the best, most accessible platform for calls with patients considering costs and network reliability.
  • Ensure a private and safe space is available for both the provider and the patient.
  • Utilize safe, secure, and appropriate channels of communication.
  • Establish referral pathways with supervisors for those with severe psychological distress.
  • Allocate resources for talk time to ensure staff and patients connect via phone and video call remotely.
  • Define clear times when providers will be available for sessions.
  • Schedule times for regular clinical supervision with staff.

Chapter 10

Treatments

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

COVID-19 Guidelines DashboardCopy Link!

Please see our Dashboard for color-coded evidence based summary of guidance from multiple institutions on all major treatments. New updates weekly!

Overview by SeverityCopy Link!

Updated Date: November 13, 2022
Literature Review (Therapeutics):
Gallery View, Grid View
Tool:
Medication Interactions with COVID-19 Treatments; COVID-19 (Therapeutics) Guidelines Dashboard

Clinical Severity

Treatment Considerations

COVID-19 Without Hypoxemia

AND

Without Risk Factors

1. Symptomatic Treatment

COVID-19 Without Hypoxemia or Radiographic Evidence of Disease

BUT

With Risk Factors: Age >60, cardiovascular disease, hypertension, diabetes, COPD, cancer, immunosuppressive medications, detectable HIV viral load or CD4 <200, TB, pregnancy, malnutrition (BMI <18 in adults, yellow MUAC for children < 5 years old)

See example outpatient treatment algorithm here (USAID) and here (MGB)

1. Symptomatic Treatment

2. Nirmatrelvir/ritonavir if indicated and available

3. Remdesivir if indicated and available

4. Molnupiravir if indicated and available when the above two options are unavailable

5. Monoclonal antibodies can be considered if circulating variants are susceptible. Currently, the circulating variants are resistant to all available monoclonal antibody products

6. Closer Monitoring and advance to other therapies (see below) if clinical condition worsens

COVID-19 Diagnosis with Hypoxemia

1. Symptomatic Treatment

2. Corticosteroids

3. Consider Remdesivir (if available and recommended by your institution)

4. Consider Tocilizumab, Baricitinib, or Anakinra

5. Consider availability of clinical trials

COVID-19 with Critical Illness or ARDS

1. Symptomatic Treatment

2. Empiric antibiotics initially (commonly Ceftriaxone and Azithromycin or Doxycycline for community-acquired pneumonia), with adjustment at 24-48 hours based on workup (see Bacterial Infections)

3. Corticosteroids

4. Consider Tocilizumab, Baricitinib, or Anakinra if within 24 hours of admission to the intensive care unit

5. Consider availability of clinical trials

Note: Remdesivir is NOT recommended if requiring intubation

Oral Antiviral Outpatient Test-to-Treat AlgorithmCopy Link!

Full downloadable algorithm (that is available for re-use) can be found here

A second example of an outpatient treatment algorithm, from Mass General Brigham, can be found here

CorticosteroidsCopy Link!

Updated Date: April 3, 2021
Literature Review:
Gallery View, Grid View

Tool: COVID-19 (Therapeutics) Guidelines Dashboard

RecommendationsCopy Link!

  1. Low-dose systemic corticosteroids are recommended for COVID-19 positive patients who require supplemental oxygen or are critically ill
  2. Clinical considerations in patients when initiating steroids:
  1. Monitor glucose, WBC, mental status, blood pressure, risk of myopathy (especially in those who are paralyzed for > 48 hours)
  2. Assess for risk of Strongyloides and test and/or empirically treat as needed.
  3. If patient has other risk factors requiring initiation of stress ulcer prophylaxis, initiate famotidine or a proton pump inhibitor as indicated
  4. Contraindications:
  1. Hypersensitivity to steroids
  2. Relative contraindication: invasive fungal infection

EvidenceCopy Link!

  1. The Randomized Evaluation of COVID-19 Therapy (RECOVERY) trial found that dexamethasone dosed at 6 mg daily for up to 10 days (n=2104) had lower rates of 28-day mortality compared to usual care (n=4321) (22.9% vs. 25.7%; age-adjusted RR 0.83, 95% CI 0.75-0.93, p<0.001). Dexamethasone reduced deaths in mechanically ventilated patients (29.3% vs. 41.4%, RR 0.64, 95% CI 0.51-0.81) and patients receiving supplemental oxygen (23.3% vs. 26.2%, RR 0.82, 95% CI 0.72-0.94), but not among patients who did not require respiratory support (17.8% vs. 14%, RR 1.19, 95% CI 0.91-1.55) (Horby et al).
  2. The WHO Rapid Evidence Appraisal for COVID-19 Therapies (REACT) Working Group conducted a prospective meta-analysis of 7 randomized trials in which 647 of 1703 COVID-19 patients died (38%). 28-day all-cause mortality was lower among patients who received corticosteroids compared to those who received usual care or placebo (OR 0.66, 95% CI 0.53-0.82, p<0.001). Of note, this meta-analysis included the RECOVERY trial results (Sterne et al).
  3. Additional data on corticosteroids for COVID-19 is evolving
  1. Additional randomized controlled trials: Jeronimo et al; Angus et al; Dequin et al; Tomazini et al; Edalatifard et al; Ghanei et al
  2. Non-randomized cohorts: Fadel et al; Fernández-Cruz et al; Keller et al; Wu et al; Nelson et al; Lu et al; Wang et al; Bani-Sadr et al; Sanz-Herrero et al; Yuan et al; Wu et al; Bartoletti et al; Monreal et al; Salton et al; Li et al; Liu et al; Fatima et al.
  3. Additional meta-analyses: Yang et al; Cano et al; Pasin et al.
  4. Previous studies have shown negative effects of corticosteroids on similar viruses, albeit without the hyperinflammatory response frequently seen in COVID-19. There is no clinical evidence of net benefit from steroids in SARS-CoV, MERS-CoV or influenza infection. Observational data show increased mortality, more secondary infections, impaired viral clearance and more adverse effects in survivors (e.g., psychosis, diabetes, avascular necrosis) with steroid use at varying doses compared to usual care (Lee et al; Stockman et al; Lansbury et al; Arabi et al).
  1. The WHO makes a strong recommendation for corticosteroids in patients with severe and critical COVID-19 and a conditional recommendation not to use corticosteroids in patients with nonsevere COVID-19 (WHO COVID-19 Living Guidance, September 2020). The National Institutes of Health, Infectious Diseases Society of America, American Thoracic Society, and Society of Critical Care Medicine all suggest the use of steroids in patients requiring supplemental oxygen or on mechanical ventilation (NIH Treatment Guidelines, February 2021; IDSA Treatment Guidelines, September 2020; ATS COVID-19 Updated Guidance, July 2020, SCCM COVID-19 Guidelines, March 2021).

DosingCopy Link!

Dosing regimens to consider include:

Corticosteroid

Dose

Duration

Dexamethasone (preferred if available)

6mg IV or PO daily

10 days

Hydrocortisone

50mg IV Q8h

10 days

Methylprednisolone

15mg IV BID

10 days

Prednisone/Prednisolone

40mg PO daily

10 days

  1. If also treating shock, hydrocortisone 50mg IV Q6h is recommended until improvement in shock, followed by consideration of steroid dosing as above to complete 10 days of total treatment. Indications for steroids in shock include:
  1. Any shock in a patient with chronic steroid use >10mg prednisone daily
  2. Multipressor (>2 pressor) shock without history of chronic steroid use
  1. Some patients with adrenal suppression may need higher doses of supplemental corticosteroids

AntiviralsCopy Link!

RemdesivirCopy Link!

Updated Date: November 13, 2022
Literature Review:
Gallery View, Grid View

Tool: COVID-19 (Therapeutics) Guidelines Dashboard

RecommendationsCopy Link!

  1. Remdesivir, if available, is recommended for outpatients within 7 days of symptom onset and at high risk for progression to severe disease (see an example of an outpatient treatment algorithm here), or hospitalized patients with COVID-19 disease, but not yet requiring intubation (NIH Treatment Guidelines, August 2022; IDSA Treatment Guidelines, November 2022; WHO Treatment Guidelines, September 2022).

PharmacologyCopy Link!

Remdesivir is a nucleotide prodrug metabolized to an analog of adenosine triphosphate, which inhibits viral RNA-dependent RNA polymerase, causing premature termination of RNA transcription

EvidenceCopy Link!

  1. Key inpatient randomized controlled trials: Beigel et al; Pan et al; Spinner et al; Ader et al
  2. Key outpatient randomized controlled trial: Gottlieb et al
  3. Additional randomized controlled trials: Goldman et al; Wang et al; Barrat-Due et al
  4. Key meta-analyses: Rochwerg et al
  5. Non-randomized cohorts: Mozaffari et al; Garcia-Vidal et al; Holshue et al; Grein et al; Antinori et al; Pasquini et al; Olender et al; Kalligeros et al; Garibaldi et al; Lapadula et al
  6. Pharmacology reviews: Aleissa et al; Jorgensen et al

DosingCopy Link!

  1. 200 mg IV loading dose, followed by 100 mg IV daily for 4-9 days for a total 5 to 10-day duration The package insert notes an infusion time of 30-120 minutes. If the patient is able to tolerate it, shorter infusion times (30-60 minutes) are preferred as remdesivir's active metabolite (GS-443902) is active intracellularly and achieves higher intracellular AUCs if infused over 30 minutes compared to 120 minutes (Humeniuk et al).
  1. 5-day duration is preferred in the majority of patients
  2. For outpatients or patients hospitalized for non-COVID-19 indications incidentally found to be COVID positive and mildly symptomatic, but without an oxygen requirement, a 3-day course of remdesivir should be sufficient (Gottlieb et al)

Monitoring and ToxicityCopy Link!

  1. Elevated transaminases (AST, ALT), acute kidney injury, phlebitis, constipation, headache, and nausea
  2. Remdesivir is co-formulated with sulfobutyl ether β-cyclodextrin (SBECD), so there is a theoretical risk of accumulation in renal failure promoting further renal injury, similar to intravenous voriconazole. If remdesivir is being considered in patients with renal impairment, the expected benefits of treatment should outweigh the potential risks prior to initiation (Adamsick et al). If remdesivir is used in renal impairment (eGFR <30 mL/min), the powder formulation is preferred as it has less SBECD content/vial than the liquid formulation (3 grams vs. 6 grams of SBECD)

Nirmatrelvir/ritonavirCopy Link!

Updated Date: March 2, 2022
Literature Review:
Gallery View, Grid View

Tool: COVID-19 (Therapeutics) Guidelines Dashboard

RecommendationsCopy Link!

  1. If available, nirmatrelvir/ritonavir is recommended for the treatment of mild-to-moderate COVID-19 in outpatients ≥ 12 years old who are at high risk for progression and within 5 days of symptom onset
  2. More information on the FDA’s nirmatrelvir/ritonavir EUA can be found here
  3. An example of an outpatient treatment algorithm can be found here

PharmacologyCopy Link!

  1. Nirmatrelvir is a SARS-CoV-2 main protease (Mpro) inhibitor. Inhibition of Mpro renders SARS-CoV-2 incapable of processing polyprotein precursors, preventing viral replication.
  2. Co-packaged with ritonavir to inhibit CYP3A4 metabolism of nirmatrelvir, which is required for the protease inhibitor to achieve therapeutic levels

EvidenceCopy Link!

  1. Key randomized controlled trial: Hammond et al
  2. Non-randomized cohorts: Arbel et al; Najjar-Debbiny et al; Malden et al

DosingCopy Link!

  1. Nirmatrelvir 300 mg (two 150 mg tablets) with ritonavir 100 mg by mouth twice daily for 5 days, initiated within 5 days of symptoms onset
  1. Dose reduction for moderate renal impairment (eGFR ≥ 30 to < 60 mL/min): Nirmatrelvir 150 mg (one 150 mg tablet) with ritonavir 100 mg by mouth twice daily for 5 days
  2. Not recommended in patients with severe renal impairment (eGFR <30 mL/min) or severe hepatic impairment (Child-Pugh class C)

Monitoring and ToxicityCopy Link!

  1. Ritonavir, as a strong CYP3A4 inhibitor, interacts with many medications. Please refer to the University of Liverpool drug-drug interaction checker to assess potential drug interactions. An example one-pager of drug interactions from Mass General Brigham can be found here
  2. Adverse reactions occurring in more than 1% of patients in clinical trials included dysgeusia, diarrhea, hypertension, and myalgias
  3. Hepatic transaminase elevations, clinical hepatitis, and jaundice have occurred in patients receiving ritonavir
  4. While highly unlikely, nirmatrelivir/ritonavir use may lead to a risk of HIV-1 developing resistance to HIV protease inhibitors in individuals with uncontrolled or undiagnosed HIV-1

MolnupiravirCopy Link!

Updated Date: February 15, 2022
Literature Review:
Gallery View, Grid View

Tool: COVID-19 (Therapeutics) Guidelines Dashboard

RecommendationsCopy Link!

  1. If available, molnupiravir is recommended for the treatment of mild-to-moderate COVID-19 in adult outpatients who are at high risk for progression and within 5 days of symptom onset only if other outpatient treatments (COVID monoclonal antibodies, nirmatrelvir/ritonavir, remdesivir) are not accessible or clinically appropriate
  2. Not recommended during pregnancy
  1. Women of child-bearing potential should use contraception during treatment and for 4 days after course completion
  2. Men of reproductive potential sexually active with females of child-bearing potential should use contraception during treatment and for at least 3 months after course completion
  1. More information on the FDA’s molnupiravir EUA can be found here
  2. An example of an outpatient treatment algorithm can be found here

PharmacologyCopy Link!

  1. Molnupiravir is an oral ribonucleotide prodrug that is metabolized into N4-hydroxycytidine (NHC), which is then phosphorylated to form the active ribonucleoside triphosphate NHC-TP. NHC-TP inhibits SARS-CoV-2 replication by inducing RNA mutagenesis

EvidenceCopy Link!

  1. Key randomized controlled trial: Bernal et al
  2. Other randomized controlled trials: Fischer et al; Arribas et al; Caraco et al

DosingCopy Link!

  1. Molnupiravir 800 mg (four 200 mg capsules) by mouth every 12 hours for 5 days, initiated within 5 days of symptom onset

Monitoring and ToxicityCopy Link!

  1. Molnupiravir is not recommended for use during pregnancy due to potential embryo-fetal toxicity
  2. Molnupiravir is not authorized for patients < 18 years old as it may affect bone and cartilage growth
  3. Adverse reactions occurring in more than 1% of patients in clinical trials included diarrhea, nausea, and dizziness

Other AntiviralsCopy Link!

FavipiravirCopy Link!

Literature Review: Gallery View, Grid View

Not recommended for routine use

See BWH Summary

Umifenovir (Arbidol)Copy Link!

Literature Review: Gallery View, Grid View

Not recommended for routine use

See BWH Summary

Lopinavir/RitonavirCopy Link!

Literature Review: Gallery View, Grid View

Not recommended for routine use

For patients on Antiretrovirals for HIV, we do not recommend changing existing ART regimens for the purposes of prophylaxis or treatment of COVID-19

See BWH Summary

InterferonsCopy Link!

Literature Review: Gallery View, Grid View

Not recommended for routine use

See BWH Summary

AntibodiesCopy Link!

Monoclonal AntibodiesCopy Link!

Updated Date: November 13, 2022
Literature Review:
Gallery View, Grid View

Tool: COVID-19 (Therapeutics) Guidelines Dashboard

RecommendationsCopy Link!

  1. The United States Food and Drug Administration (FDA) has issued emergency use authorizations (EUA) for five investigational monoclonal antibody therapies, bamlanivimab-etesevimab, casirivimab-imdevimab, sotrovimab, tixagevimab-cilgavimab, and bebtelovimab. However, no monoclonal antibody products are currently active against the circulating SARS-CoV-2 variants and thus are not recommended at this time.
  2. Bamlanivimab-etesevimab, casirivimab-imdevimab, sotrovimab, and bebtelovimab are authorized for the treatment of outpatients with mild to moderate COVID-19 disease who are at high risk for progressing to severe COVID-19 and/or hospitalization. More information on the EUAs can be found here for bamlanivimab-etesevimab, here for casirivimab-imdevimab, here for sotrovimab, and here for bebtelovimab.
  3. Currently, no monoclonal antibody products are available for post-exposure prophylaxis as casirivimab-imdevimab and bamlanivimab-etesevimab are not active against the Omicron SARS-CoV-2 variant Casirivimab-imdevimab and bamlanivimab-etesevimab were authorized for post-exposure prophylaxis in patients who have a recent confirmed SARS-CoV-2 exposure, are at high risk for progression to severe COVID-19 disease, AND are immunocompromised or unvaccinated
  4. Tixagevimab-cilgavimab is authorized for pre-exposure prophylaxis in patients who:
  1. Are not currently infected with SARS-CoV-2 and who have not had a known recent exposure to an individual infected with SARS-CoV-2, and
  1. Have moderate to severe immune compromise due to a medical condition or receipt of immunosuppressive medications and may not mount an adequate immune response to vaccination, or
  2. For whom vaccination with any available COVID-19 vaccine is not recommended due to a history of severe adverse reaction
  1. More information on the tixagevimab-cilgavimab EUA can be found here
  1. Monoclonal antibody products have varying activity depending on the most prevalent SARS-CoV-2 variant in your local area. If located in the United States, please refer to the HHS/ASPR website for the most up-to-date information.
  2. An example of an outpatient treatment algorithm can be found here

PharmacologyCopy Link!

  1. Tixagevimab and cilgavimab (AZD7442, Evusheld) are recombinant human IgG1κ monoclonal antibodies that bind to non-overlapping regions of the receptor binding domain of SARS-CoV-2 spike protein, blocking viral entry into host cells
  2. Bebtelovimab (LY-CoV1404) is a recombinant human IgG1κ monoclonal antibody to the spike protein of SARS-CoV-2, blocking viral entry into host cells
  3. Other monoclonal antibody products are not currently authorized by the FDA due to the current circulating variants
  1. Bamlanivimab-etesevimab (LY-CoV555 and LY-CoV016) are two recombinant neutralizing human IgG1κ monoclonal antibodies to the spike protein of SARS-CoV-2, blocking viral entry into host cells
  2. Casirivimab-imdevimab (REGN10933-REGN10987) are two recombinant human monoclonal antibodies to the spike protein of SARS-CoV-2 (IgG1κ and IgG1λ, respectively), blocking viral entry into host cells
  3. Sotrovimab (VIR-7831) is a recombinant human IgG1κ monoclonal antibody to the spike protein receptor binding domain of SARS-CoV-2, blocking viral entry into host cells

EvidenceCopy Link!

Outpatient Treatment

  1. Key randomized controlled trials: Weinreich et al; Dougan et al; Weinreich et al; Gupta et al; O’Brien et al
  2. Other randomized controlled trials: Chen et al; Gottlieb et al

Inpatient Treatment

  1. Key randomized controlled trials: RECOVERY; Lundgren et al; Self et al; Lundgren et al

Post-exposure Prophylaxis

  1. Key randomized controlled trials: Cohen et al; O’Brien et al

Pre-exposure Prophylaxis

  1. Levin et al

DosingCopy Link!

  1. Tixagevimab 300 mg and cilgavimab 300 mg IM once as two separate injections.
  1. For patients who previously received 150-150 mg dosing, an additional 150 mg of tixagevimab and 150 mg of cilgavimab administered as two separate IM injections may be given
  2. Redosing with 300-300 mg is recommended every 6 months, timed from the most recent tixagevimab-cilgavimab dose
  1. Bebtelovimab 175 mg IV once over at least 30 seconds as soon as possible after a positive SARS-CoV-2 test and within 7 days of symptom onset
  2. Other monoclonal antibody products are not currently authorized by the FDA due to the current circulating variants
  1. Bamlanivimab 700 mg and etesevimab 1400 mg IV once over at least 21 minutes administered as soon as possible after a positive SARS-CoV-2 test and within 10 days of symptom onset While the 2800-mg dose was the only dose to show a significant reduction in viral load at day 11 in BLAZE-1, the FDA’s EUA authorized the 700-mg dose for use due to issues with limited drug supply
  2. Casirivimab 600 mg and imdevimab 600 mg IV (or subcutaneous, but IV is strongly preferred) once over at least 20 minutes as soon as possible after a positive SARS-CoV-2 test and within 10 days of symptom onset
  3. Sotrovimab 500 mg once over 30 minutes as soon as possible after a positive SARS-CoV-2 test and within 10 days of symptom onset

Monitoring and ToxicityCopy Link!

  1. Patients must be monitored during administration and for at least 60 minutes following the infusion or injection of any of the monoclonal antibody cocktails to assess for signs of hypersensitivity
  2. The most common adverse events reported in clinical trials have been nausea, diarrhea, dizziness, headache, pruritus, and vomiting
  3. Understanding the local incidence of SARS-CoV-2 variants can help guide which monoclonal antibody product may be most appropriate for use.
  1. The NIH has an OpenData Portal with further information if looking for more information than the below table
  2. Monoclonal antibody products have varying activity depending on the most prevalent SARS-CoV-2 variant in your local area. If located in the United States, please refer to the HHS/ASPR website for the most up-to-date information and CDC’s Nowcast for current variant estimates in your region

COVID Variants of Concern

Fold Reduction in Susceptibility

WHO label

Variant

Key Substitutions Tested

Bamlanivimab-Etesevimab

Casirivimab- Imdevimab

Sotrovimab

Bebtelovimab

Tixagevimab-Cilgavimab

Alpha

B.1.1.7

N501Y

No change

No change

No change

No change

No change

Beta

B.1.351

E484K, K417N, N501Y

>325

No change

No change

No change

No change

Gamma

P.1

E484K, K417T, N501Y

252*

No change

No change

No change

No change

Delta

B.1.617.2

L452R, T478K

No change

No change

No change

No change

No change

Epsilon

B.1.427/ B.1.429

L452R

11

No change*

No change*

No change

No change

Iota

B.1.526

E484K

11

No change*

No change*

No change

No change

Kappa

B.1.617.1

L452R, E484Q

6*

No change

No change

No change*

No change

Lambda

C.37

L452Q, F490S

No change*

No change*

No change*

No change*

No change*

Mu

B.1.621

R346K, E484K, N501Y

116*

No change*

No change*

5.3*

7.5*

Omicron

BA.1

Numerous

>2,938*

>1,013*

No change*

No change

12-30

Omicron

BA.1.1

BA.1 + R346K

N/A

N/A

No change*

No change

176

Omicron

BA.2

Numerous

N/A

N/A

15.7 (EC50)

25.3-48.1 (EC90)

No change

5.4

Omicron

BA.2.12.1

BA.2 + L452Q

N/A

N/A

N/A

No change

5*

Omicron

BA.2.75

BA.2 + D339H, G446S, N460K, R493Q (reversion)

N/A

N/A

N/A

No change

2.4-15*

Omicron

BA.4

Numerous

N/A

N/A

N/A

No change

33-65*

Omicron

BA.5

Numerous

N/A

N/A

N/A

No change

2.8-16

Omicron

BA.4.6

BA.4 + R346T

N/A

N/A

N/A

No change

>1000*

Omicron

BF.7

BA.5 + R346T

N/A

N/A

N/A

No change*

Not promising

Omicron

BQ.1

BA.5 + K444T, N460K

N/A

N/A

N/A

>672*

Not promising

Omicron

BQ.1.1

BA.5 + R346T, K444T, N460K

N/A

N/A

N/A

>672*

Not promising

No change: < 5-fold reduction in susceptibility for all except < 2-fold reduction for casirivimab-imdevimab

* Performed in pseudotyped virus-like particles rather than authentic SARS-CoV-2 virus

Convalescent PlasmaCopy Link!

Updated Date: September 13, 2021
Literature Review:
Gallery View, Grid View

Tool: COVID-19 (Therapeutics) Guidelines Dashboard

RecommendationsCopy Link!

  1. The FDA has issued an Emergency Use Authorization (EUA) for convalescent plasma for the treatment of hospitalized COVID-19 patients.
  2. Despite the EUA, convalescent plasma is not routinely recommended for COVID-19 patients given the lack of conclusive evidence. This stance is also supported by both the National Institutes of Health (NIH) Guidelines and Infectious Diseases Society of America Guidelines.
  3. Because of the feasibility of local production, convalescent plasma may be a particularly appealing therapeutic option in low and middle-income countries. However, given the unproven benefits and potential risks, the International Society of Blood Transfusion recommends that convalescent plasma use be limited to the context of clinical research studies (Smid et al).

PathophysiologyCopy Link!

  1. Convalescent plasma originates from patients who have previously recovered from a viral infection and are now able to donate their immunoglobulin-containing blood
  2. The presumed mechanism of action is that antibodies present in convalescent plasma may suppress viremia

EvidenceCopy Link!

  1. The first multicenter randomized clinical trial was published on June 3, 2020, in which 103 COVID-19 patients were randomized to receive convalescent plasma (n=52) or standard of care (n=51). In this open label trial, clinical improvement occurred within 28 days in 51.9% of the convalescent plasma patients compared to 43.1% of the standard of care patients (HR 1.4, 95% CI 0.79-2.49). There was also no significant difference in mortality between groups, with 15.7% mortality in the convalescent plasma group compared to 24% with standard of care (OR 0.65, 95% CI 0.29-1.46). The findings of this study are limited however due to early termination of the trial. To provide 80% power, 200 patients were required in the analysis, but only half of that number were enrolled. Further studies are still warranted (Li et al).
  2. In a 160-patient, randomized, double-blind, placebo-controlled trial among older patients (≥65 years with comorbidities or ≥75 years without comorbidities), early receipt of high-titer convalescent plasma (<72 hours of symptoms) reduced the incidence of severe COVID-19 (RR 0.52, 95% CI 0.29-0.94) (Libster et al).
  3. PlasmAr randomized 333 hospitalized COVID-19 patients to receive convalescent plasma (with a minimum SARS-CoV-2 antibody titer of 1:400, n=228) or placebo (n=105). At day 30, there were no significant differences between groups in clinical status (OR 0.83, 95% CI 0.52 to 1.35) or overall mortality (10.96% in convalescent plasma group vs. 11.43% in placebo group, -0.46 percentage points, 95% CI -7.8 to 6.8) (Simonovich et al).
  4. PLACID randomized 464 hospitalized COVID-19 patients to receive convalescent plasma in addition to standard of care (n=235) or standard of care alone (n=229). Progression to severe disease or death by day 28 occurred in 19% of patients in the convalescent plasma arm compared to 18% in the control arm (RR 1.04, 95% CI 0.71 to 1.54) (Agarwal et al)
  5. CONCOR-1 randomized hospitalized patients 2:1 to receive convalescent plasma (n=625) or standard of care (n=313). Intubation or death occurred in 32.4% of patients in the convalescent plasma arm compared to 28% patients in the standard of care arm (RR 1.16, 95% CI 0.94-1.43, p=0.18). Patients receiving convalescent plasma had more serious adverse events (33.4% versus 26.4%; RR 1.27, 95% CI 1.02-1.57, p=0.034) (Bégin et al)
  6. The Mayo Clinic has published on their expanded access program, which to-date has analyzed 20,000 hospitalized COVID-19 patients who received convalescent plasma. In the cohort, the risk of serious adverse events was low and the seven-day mortality rate was 8.6% (8.2 to 9%). Noted side effects included cardiac events (0.37%), sustained hypotension (0.27%), thrombotic/thromboembolic complications (0.16%), transfusion-associated circulatory overload (TACO) (0.18%), transfusion-related acute lung injury (TRALI) (0.1%), and allergic transfusion reactions (0.13%), (Joyner et al; previous report with 5,000 patients: Joyner et al).
  1. A subgroup of 3,082 patients had anti-SARS-CoV-2 IgG antibody levels determined prior to transfusion. In the high-titer group (defined as a signal-to-cutoff ratio of >18.45), 22.3% of patients died within 30 days, compared to 27.4% in the medium-titer group and 29.6% in the low-titer group. These retrospective results hint that high-titer convalescent plasma may be more effective than plasma with lower antibody levels (Joyner et al).
  1. Numerous smaller case series and cohort studies have been published to-date without reaching definitive conclusions on the use of convalescent plasma in COVID-19 (Shen et al; Duan et al; Zhang et al; Salazar et al; Salazar et al; Rogers et al; Liu et al; Tworek et al)

DosingCopy Link!

  1. Optimal therapeutic dosing is not yet known. Most ongoing studies are assessing one infusion of 1-2 units (200-500 mL)

Monitoring and ToxicityCopy Link!

  1. Plasma transfusions in general are safe and well-tolerated in most patients. Potential side effects however include:
  1. Mild fever
  2. Allergic reactions, including serum sickness on rare occasions
  3. Transfusion-associated circulatory overload (TACO)
  4. Transfusion-related acute lung injury (TRALI) (Gajic et al)
  5. Potential risk of another infectious disease from donor, although risk is incredibly low with modern blood bank techniques
  1. There is a theoretical concern that convalescent plasma may lower a patient’s INR if on warfarin, similar to (but to a lesser degree than) fresh frozen plasma.

ImmunomodulatorsCopy Link!

Anti-IL-6 Agents (e.g. Tocilizumab)Copy Link!

Literature Review (Tocilizumab):Gallery View, Grid View
Literature Review (Sarilumab):
Gallery View, Grid View
Literature Review (Siltuximab):
Gallery View, Grid View

Tool: See BWH Summary

Tool: COVID-19 (Therapeutics) Guidelines Dashboard

Primarily based on the RECOVERY and REMAP-CAP trials, tocilizumab may be considered in patients with severe COVID-19 in addition to corticosteroids if early in the patient’s hospital admission and with no contraindications to its use (Horby et al, Gordon et al). The WHO now makes a strong recommendation in favor of using tocilizumab in severe or critical COVID-19 based on the results of their meta-analysis, which found that 28-day all-cause mortality was lower among those receiving IL-6 inhibitors (21.8%) compared to usual care or placebo (25.8%) (OR 0.86, 95% CI 0.79-0.95) (Shankar-Hari et al). BWH’s example algorithm for use of tocilizumab or baricitinib in addition to corticosteroids can be found here.

JAK Inhibitors (e.g. Baricitinib)Copy Link!

Literature Review (Baricitinib):Gallery View, Grid View

Literature Review (Tofacitinib):Gallery View, Grid View

Literature Review (Ruxolitinib):Gallery View, Grid View

Tool: See BWH Summary

Tool: COVID-19 (Therapeutics) Guidelines Dashboard

Based on the ACTT-2 and COV-BARRIER trials, baricitinib may be considered in patients with severe COVID-19 in addition to corticosteroids on a case-by-case basis if early in the patient’s hospital admission and with no contraindications to its use (Kalil et al, Marconi et al, Ely et al). BWH’s example algorithm for use of tocilizumab or baricitinib in addition to corticosteroids can be found here.

Anti-IL-1 Agents (e.g. Anakinra)Copy Link!

Literature Review (Anakinra): Gallery View, Grid View
Literature Review (Canakinumab): Gallery View, Grid View
Literature Review (Rilonacept): Gallery View, Grid View

Tool: See BWH summary

Based on the SAVE-MORE trial, anakinra may be considered in patients with severe COVID-19 in addition to corticosteroids on a case-by-case basis if early in the patient’s hospital admission and with no contraindications to its use (Kyriazopoulou et al). BWH’s example algorithm for use of tocilizumab or baricitinib in addition to corticosteroids (in which anakinra is now a part of) can be found here.

Symptomatic TreatmentsCopy Link!

CoughCopy Link!

Updated Date: December 7, 2020

Non-Pharmacologic Therapy

  1. Drink plenty of fluids, preferably warm if possible
  2. A teaspoon of honey may help ease coughing symptoms
  3. Cough drops or hard candy may also be used

Pharmacologic Therapy

  1. Wet cough, difficulty clearing thick sputum: cough expectorant such as guaifenesin
  2. Dry cough: cough suppressant such as dextromethorphan
  3. Severe cough that disrupts sleep or results in pain, dyspnea, or vomiting: opioid such as morphine at same doses as used for refractory dyspnea.

DyspneaCopy Link!

Updated Date: December 7, 2020

Dyspnea is a common physical symptom of severe COVID-19. Dyspnea may be severe. Dyspnea from COVID-19 should first be treated with oxygen and/or medications as discussed in other sections. Other underlying causes (such as severe anemia, pleural effusion, pneumothorax, or acidemia) should be ruled out and/or treated.

Non-Opioid ManagementCopy Link!

  1. Non-Pharmacologic Therapy for Dyspnea
  1. Positioning: sitting patient up in bed, if possible.
  2. Bedside fan to blow air onto face.
  3. Relaxation techniques (see section on Anxiety).
  1. Pharmacologic Therapy
  1. NSAIDs and/or acetaminophen may be used.
  2. Lorazepam can be used to ease the anxiety associated with dyspnea, but would avoid in patients who have had a previous paradoxical reaction (i.e. worsened agitation).

Opioid ManagementCopy Link!

Tool: Partners In Health Decision Tree for Opioid Treatment of Severe Dyspnea

Opioids are effective for relief of dyspnea that does not respond to treatment of underlying causes (e.g. severe anemia, severe anemia, pleural effusion, pneumothorax, or acidemia). Opioid therapy is an important component of the Essential Package for Palliative Care, which can be accessed here

  1. Candidates for Opioid Treatment of Dyspnea
  1. Opioids should be used to treat dyspnea in patients for whom survival is unlikely and treatment is focused solely on comfort and control of symptoms.
  2. Other patients with significant refractory dyspnea despite maximal treatment but expected to survive can receive opioids to treat dyspnea, although this should be done carefully in order to minimize the side effect of respiratory suppression.
  1. General Principles:
  1. Always use as needed (PRN) boluses to address acute, uncontrolled symptoms. PRN bolus dosing should be 10-20% of the 24-hour opioid dose
  1. For Opioid Naive Patients:

Normal GFR

Abnormal GFR (<50)

(Not absolute contraindication to morphine, but caution should be taken due to drug stacking)

No COPD

  • Morphine 5-10mg PO q3h PRN (use the 20mg/ml concentrate)

  • Morphine 2-4mg IV q2h PRN
  • Hydromorphone 1-2mg PO q3h PRN

  • Hydromorphone 0.1-0.2mg IV q2h PRN

COPD

  • Morphine 2-5mg PO q4h PRN (use the 20 mg/ml concentrate)

  • Morphine 1-2 mg IV q2h PRN
  • Hydromorphone 2-4mg PO q4h PRN

  • Hydromorphone 0.2-0.4mg IV q2h PRN

  1. If frequent doses are needed, schedule an effective morphine dose Q4H and add a rescue dose as needed at 10% of the total daily dose
  2. If patient is not well-managed with the above, add opioid infusion:
  1. Consider drip If > 3 bolus doses in 8 hours
  2. Calculate initial dose with total mg used/8 hours
  1. e.g. 1+2+2+2= 7 mg; begin drip at 7mg/8 hr = 1 mg/h
  2. Depending on symptoms and goals of care, consider reducing hourly rate by 30-50%. If patient is at end of life, would use 100% of hourly rate.
  1. Continue PRN dosing at current dose (if effective) or titrate as per above
  1. For Opioid tolerant patients:
  1. If able to take PO:
  1. Continue current long-acting doses if renal and hepatic function tolerate
  2. Continue current oral PRN dose if effective q4h prn
  1. If ineffective, increase dose by 50% and order range of up to 3 x basal dose
  1. e.g. 5 mg PO MS q3h prn; increase to 7.5 mg; 7.5-22 mg PO q3h PRN
  1. If unable to take PO, severe or rapidly escalating symptoms:
  1. Convert as-needed PO doses to IV pushes as needed
  1. Use the IV Conversion chart (see chart below)
  2. Decrease PRN dose by ⅓ for incomplete cross-tolerance when switching between opioid classes
  1. e.g. to convert 20 mg of oxycodone to IV hydromorphone: 20 mg oxy = 1.5 mg IV hydromorphone; 1.5 mg x ⅔ =1 mg IV
  1. Convert PO long-acting/ sustained release opioids to an infusion:
  1. Calculate 24-hour dose of PO sustained release (SR) morphine
  1. Divide by 3 for the total 24h mg IV (Morphine PO/IV = 3:1)
  1. Divide the 24h mg IV total by 24h for the hourly drip rate (mg)
  1. e.g. 30 mg SR PO morphine q8 hr= 90 mg PO in 24 h; 90 mg /3 = 30 mg IV dose; 30 mg / 24 h ~1 mg/hr IV morphine infusion
  1. Continue PRN dosing. PRN dose should be 100-200% of opioid drip rate
  1. e.g. 1 mg/hr IV morphine infusion; PRN dose is 1-2 mg IV q2h

Abbreviated Opioid Equianalgesic Table (for complete table and an example conversion see DFCI Pink Book)

Drug

PO/PR (mg)

Subcut/IV (mg)

Morphine

30

10

Oxycodone

20

n/a

Hydromorphone

7.5

1.5

Fentanyl

(See table below for transdermal conversions)

n/a

0.1 (100 mcg)

PainCopy Link!

Pharmacologic:Copy Link!

Opioid management of pain should be managed similarly to Opioid Management for Dyspnea.

AnxietyCopy Link!

Updated Date: December 7, 2020
Literature Review (Anxiety & Depression):
Gallery View, Grid View

NonpharmacologicCopy Link!

Feelings of uncertainty and fear can fuel anxiety

  1. Important to first acknowledge and normalize distress reactions
  2. Correct misinformation. Provide accurate information (regarding patient’s current medical condition and next steps, regarding hospital protocols and measures being taken for safety)
  3. Offer counseling (Spiritual, Psychocological, Social Work)
  4. Offer mindfulness strategies
  5. Strategies for reducing distress
  1. Restful sleep, eating regular meals, exercising
  2. Talking to loved ones (by telephone or video chat)
  3. Diaphragmatic breathing (breathing to inflate the abdomen)
  4. Muscle relaxation

PharmacologicCopy Link!

  1. Continue home psychotropic medication regimen if possible
  2. For patients with evidence of delirium
  1. Quetiapine 12.5-25mg TID PRN or Haloperidol 1-2.5 mg orally or IV Q4H as needed (can also be scheduled Q6 – 8 H)
  1. For patients without evidence of delirium
  1. Quetiapine 12.5-25mg TID PRN
  2. Lorazepam 0.5-2 mg PO/SL TID PRN; 0.5-2 mg IV TID PRN or Diazepam 2.5-5mg every 6 to 24 hours
  1. For patients with risk of respiratory depression or history of respiratory illness
  1. Buspirone 5-15mg PO TID
  1. For moderate or severe anxiety in a patient expected to survive
  1. Fluoxetine 20mg orally daily. Increase dose as needed every 7 days to achieve good effect, maximum 80mg per day. Other selective serotonin uptake inhibitors (SSRIs) that can be used instead of fluoxetine include sertraline (50mg orally QD, increase weekly as needed to a maximum of 200mg QD) and citalopram. Beware of QTc prolongation with some SSRIs.

Anxiety Related to Dyspnea or End of LifeCopy Link!

  1. Benzodiazepines (if patient is not delirious; can use in either intubated or non-intubated pts — use with caution in older patients)
  1. Lorazepam (longer half-life) 0.5-2 mg PO/SL q4-6h PRN; 0.5-2 mg IV q2h PRN
  2. Midazolam (shorter half-life) 0.2-0.5 mg IV slowly q 15 min PRN or 0.1-0.3 mg/hr IV infusion
  3. Diazepam 2.5-5mg every 6 to 24 hours
  1. SSRI/SNRI: Continue home dose if possible. If NPO, replace with prn benzodiazepine

Respiratory SecretionsCopy Link!

Updated Date: December 7, 2020
Literature Review (Airway Clearance):
Gallery View, Grid View
Literature Review (Dornase):
Gallery View, Grid View

  1. Patients can develop thick secretions from Covid-19 itself or secondary bacterial pneumonia
  2. Nebulized treatments may help with airway secretion management, but published evidence is not available
  3. Options include:
  1. Normal (0.9%) saline nebulizer BID
  2. N-acetylcysteine (“Mucomyst”) nebulizer BID or TID
  1. N-acetylcysteine can cause bronchoconstriction
  2. Pre-treat with inhaled albuterol just prior to delivery
  1. Nebulized hypertonic (3-7%) saline once daily
  1. Hypertonic saline can cause bronchoconstriction
  2. If using, start with 3% saline to assess response and bronchoconstriction.
  3. Pre-treat with inhaled albuterol just prior to delivery
  1. Dornase alfa 2.5mg nebulizer once daily
  2. Dornase can cause bronchoconstriction, mucosal bleeding, and can clog the HEPA filter, requiring intermittent replacement by RT
  3. Avoid in the setting of bloody secretions
  4. Pre-treat with inhaled albuterol just prior to delivery
  5. It would be reasonable to consider other agents, including N-acetylcysteine, first given the need to change HEPA filters. In addition, a RCT for dornase nebulizer versus saline will begin shortly at BWH. However, if persistent secretions, it is reasonable to try dornase nebulizer
  1. Although avoided if possible since it is an aerosol generating procedure, bronchoscopy for pulmonary clearance can be performed if needed on COVID-19 confirmed or PUI patients.

Secretions at the End of LifeCopy Link!

Pharmacologic management (not to be used with secretions with significant mucus). Avoid using > 2 of these at the same time; if more than one is required, monitor for development of anticholinergic crisis

  1. Glycopyrrolate 0.2 – 0.4mg IV q2hrs prn secretions, rattling sound
  2. Hyoscyamine sulfate 0.125-0.25mg PO q4hrs prn secretions, rattling sound
  3. Scopolamine 1.5mg TD q72hrs if patient not awake and no apparent delirium or history of delirium. Note that the patch will take ~ 12 hours to take effect
  4. Hyoscine butylbromide (alternative formulation of scopolamine) 20mg orally/IV/SC Q6H PRN or scheduled.

Nausea and VomitingCopy Link!

Updated Date: December 7, 2020

  1. Consider reversible etiologies such as gastritis, constipation, anxiety.
  2. Match treatment to etiology of nausea:
  1. Chemoreceptor Trigger Zone (blood brain barrier breakdown)
  1. haloperidol, metoclopramide, ondansetron, olanzapine, aprepitant
  1. Gastrointestinal:
  1. ondansetron, metoclopramide, dexamethasone (if malignant obstruction)
  1. CNS cortical centers:
  1. lorazepam for anticipatory nausea, dexamethasone (tumor burden causing ICP)
  1. Vestibular:
  1. meclizine, scopolamine, diphenhydramine
  1. Additional information can be found at the DFCI Green Book (page 11 for more dosing recommendations):
  1. Ondansetron 8-24mg/day IV/PO (usually on a q6h PRN schedule, max single dose 16mg) *causes constipation* Beware QTc prolongation.
  2. Haloperidol 0.5-2 mg IV/PO q 4-8 hours *extra-pyramidal effects unlikely at these low doses*
  3. Metoclopramide 10-40 mg IV/PO TID-QID *pro-motility*
  4. Olanzapine 2.5-10 mg PO/dissolvable daily *off label, effective for concurrent anxiety, will not exacerbate constipation*
  5. Prochlorperazine 10 mg PO TID-QID (max 40 mg/day) 25 mg PR BID *very sedating, overlaps with haloperidol, metoclopramide, perphenazine*
  6. Meclizine 25-50 mg PO daily

DeliriumCopy Link!

Updated Date: December 20, 2020
Literature Review:
Gallery View, Grid View

  1. Non-Pharmacologic:
  1. Frequent reorientation when appropriate
  2. Early mobilization (getting out of bed)
  3. Promotion of sleep-wake cycles via use of room lighting and stimulation and quiet location
  4. Timely removal of unnecessary restraints, catheters, lines, and other devices
  5. Ensuring use of glasses/hearing aids once patient is sufficiently alert
  6. Reverse contributing medical conditions as able
  7. Consult psychiatry services if available

  1. Pharmacologic
  1. Avoid delirium-causing medications (anticholinergics, benzodiazepines, opioids) whenever possible
  2. Treat comorbid symptoms and underlying medical illness
  3. For agitation/aggression:
  1. Antipsychotics
  1. Haloperidol: Mild agitation: 0.5-1.0 milligrams intravenously, or 1- 2 milligrams by mouth every 6 hours and 1-2 milligrams every 2 hours as needed; Moderate agitation: 2-4 milligrams intravenously; Severe agitation: 4-10 milligrams; Maximum dose: 20 milligrams / 24 hours
  2. If unresponsive to treatment, olanzapine (Zyprexa), 2.5 to 5 milligrams (by mouth, sublingual, or intravenously) every 12 hours. Based on onset (6h), PO/SL olanzapine should not be used PRN for agitated delirium Maximum dose: 30 milligrams / 24 hours. **do not combine with benzodiazepines given by other routes, due to increased risk of respiratory depression**
  3. If haloperidol/olanzapine not effective or contraindicated, can try:
  1. Quetiapine (Seroquel) 12.5-50 milligrams every night at bedtime, can increase to every 6-12 hours. Titrate up to effect by 50 mg – 100 mg/day. Max dose: 600-800 mg/day
  2. Aripiprazole (Abilify) 5 milligrams by mouth daily; maximum dose 30 milligrams daily
  1. Alpha 2 Agonists - helpful for patients for ventilator weaning; also good option if prolonged QTc
  1. Dexmedetomidine (Precedex) intravenously - easy to adjust dosing given short half-life
  2. Consider use of clonidine 0.1 milligrams twice daily (can uptitrate) - available as a transdermal patch as well.
  1. Mood Stabilizers
  1. Valproic Acid (good option if prolonged QTc): Start at 125-250 milligrams intravenously every 8 hours three times daily, however, COVID patients are seeming to need escalations in doses (up to anti-manic dosing of 15-25 milligrams/kilogram) in combination with antipsychotics (such as haloperidol or olanzapine, as above).
  1. Others
  1. For regulation of sleep/wake cycle: Mirtazapine (Remeron): 7.5 milligrams (can increase, but it is more sedating at lower doses)
  1. Considerations for Geriatrics Patients
  1. High risk for delirium given restrictive visitor policy, disorienting effect of PPE use by staff, difficulty hearing/identifying caregivers through masks
  2. Avoid delirium-causing medications such as anticholinergics and benzodiazepines (See here for a comprehensive Beers Criteria List)
  3. If acutely agitated, not redirectable by non-pharmacologic means, trial 12.5 milligrams trazodone x 1 as needed, repeat dose at 30 min if no effect
  4. Use antipsychotics (such as haloperidol, olanzapine, quetiapine) as last resort only, and only if QTc is < 500. Dose reductions should be used (suggestions: Haloperidol, Mild agitation 0.25 -0.5 mg IV or 1 to 2 mg PO q6h and 1 mg q2h PRN; Moderate agitation: 1-2 mg IV; Severe agitation: 2 mg IV Maximum dose: 20 mg / 24 hours)

AnticoagulationCopy Link!

Prophylactic DosingCopy Link!

Updated Date: October 31, 2021
Literature Review (Prophylaxis):
Gallery View, Grid View

Tool: COVID-19 (Therapeutics) Guidelines Dashboard

For Patients Who are Not Critically IllCopy Link!

  1. Background:
  1. For non-critically ill inpatients, VTE rates appear to be similar to general ward patients amongst those receiving standard prophylactic anticoagulation (Hill et al).
  2. The REMAP-CAP, ACTIV-4a, and ATTACC multi-platform response-adaptive randomized controlled trials evaluated therapeutic versus standard dose heparin prophylaxis in COVID-19 patients, separating by moderate vs critically ill (see below for a discussion of these results in critically ill patients) (REMAP-CAP, ACTIV-4a, and ATTACC (Non-critically ill). The primary outcomes was a composite of organ support-free days and number of days free of cardiovascular or respiratory organ support Respiratory organ support included non-invasive ventilation and high flow nasal cannula up to 21 days among patients who survived to hospital discharge.
  1. For moderately ill hospitalized patients, the therapeutic heparin arm appears to have an beneficial effect with an OR of 1.27 (95% credible interval 1.03 - 1.58). Major bleeding events to be 1.9% in the therapeutic arm compared to 0.9% in the standard dose arm. The difference in treatment effect was 4% favoring therapeutic dose, with a 0.7% difference in major bleeding, resulting in an approximately 3% difference in outcome.
  2. These results are controversial for several reasons:
  1. First, the proportions of participants assigned to each trial arm were recalculated based on a single interim analysis performed on December 15, 2020 to favor randomization to the therapeutic dose arm, which can introduce bias. This approach is called response-adaptive randomization. While this approach theoretically provides therapeutic advantages to study participants and could be more ethical, it can also introduce selection bias, counteract the benefits of initial randomization, actually reduce total absolute numbers of individuals assigned to the favored arm, and can lead to challenges in interpreting the final results (Proschan, et al. Clin Inf Dis 2020, Park, et al., Clin Epidemiol 2018) The chance for bias is greater when based on fewer or a single interim analysis, as was done in this trial.
  2. Second, it is challenging for a clinician to gauge whether an individual patient would have qualified for the trial. For example, participants who were thought not to need hospitalization for more than 72 hours were excluded, and some platforms only enrolled patients within 72 hours of hospital admission, while others enrolled patients within 14 days.
  3. Third, the composite endpoint does not elucidate individual patient-specific risks and benefits between clotting and bleeding, especially since major bleeding events were higher in the therapeutic dose arm.
  1. Recommendations:
  1. For mildly-ill outpatients, we do not recommend prophylaxis
  2. For moderately-ill inpatients, we recommend standard dose prophylaxis. (American Society of Hematology) This is due to the difficulty in determining which individual patients might benefit from higher doses, the flaws of the study as above, and the bleeding risk. However, clinicians should weigh individual risk factors for VTE and bleeding.
Standard Dosing VTE ProphylaxisCopy Link!

VTE Dosing Weight Adjustment

CrCl ≥ 30mL/min

CrCl < 30mL/min

(or enoxaparin unavailable)

Standard

Enoxaparin 40mg daily

Heparin 5000 units Q8H

Obese (≥120kg or BMI ≥ 35)

Enoxaparin 40mg BID or 0.5mg/kg Daily

(max dose 100mg daily)

Heparin 7500 units Q8H

Low Body Weight (< 50kg*)

Enoxaparin 30mg daily

Heparin 5000 units BID-TID

*LBW does not have a universal definition for LMWH dosing, we define it differently in the non-ICU (<50kg) and ICU (<60kg) populations to help achieve our targeted anticoagulant effect, though this remains an active area of research

For Critically Ill PatientsCopy Link!

  1. Background:
  1. Experts are divided as to whether standard, intermediate, or full dose anticoagulation provides the optimal balance of benefit of anticoagulation with risk of bleeding for COVID patients (Bikdeli et al). ICU patients are at additionally elevated risk for VTE events even on standard prophylaxis (Klok et al.; Middeldorp et al.; Klok et al.; Llitjos et al; Nahum et al.; Moll et al). Therefore, several observational studies and randomized clinical trials have addressed the use of intermediate and therapeutic dose heparin prophylaxis.
  1. Intermediate dosing: The INSPIRATION randomized controlled trial (INSPIRATION Investigators) compared intermediate to standard dose heparin prophylaxis in ICU patients and reported no difference in venous or arterial thromboses, need for ECMO, or 30-day mortality. The intermediate dose group had more thrombocytopenia, but no significant differences in major bleeding.
  2. Therapeutic dosing: The REMAP-CAP, ACTIV-4a, and ATTACC multi-platform response-adaptive randomized controlled trials evaluated therapeutic versus standard dose heparin prophylaxis in COVID-19 patients (REMAP-CAP, ACTIV-4a, and ATTACC (Critically-ill). The primary outcomes was a composite of organ support-free days and number of days free of cardiovascular or respiratory organ support Respiratory organ support included non-invasive ventilation and high flow nasal cannula up to 21 days among patients who survived to hospital discharge. For ICU patients, the trial was stopped early for futility, though for non-critically ill hospitalized patients there is a possible benefit (see above).
  1. Recommendations:
  1. Recommendations between different societies are split on anticoagulation in ICU patients.
  1. Standard dosing: As of October, 2021 the American Society of Hematology recommends standard dosing. Further data on therapeutic dosing is forthcoming.
  2. Intermediate dosing: As of October, 2021 BWH uses intermediate dosing for critically ill patients and post-critically ill patients. Their randomized controlled trial data do not suggest that intermediate dose heparin reduces the risk of progression of COVID-19 or death, but does decrease VTE death, and thus far does not suggest an increase in bleeding rates.
  3. Full dosing: As of October, 2021 UCSF has started using full dose anticoagulation for critically ill patients requiring oxygen support.

Intermediate Dosing VTE ProphylaxisCopy Link!

  1. Inclusion (BWH recommendations):
  1. COVID-19 confirmed and PUI patients with critical illness at any point during hospitalization
  2. Platelets >25,000
  1. Exclusion:
  1. If Platelets <25,000 or bleeding, hold prophylaxis and start thromboembolic deterrent stockings and sequential compression devices

VTE Dosing Weight Adjustment

CrCl ≥ 30mL/min

CrCl < 30mL/min

(or enoxaparin unavailable)

Standard

Enoxaparin 40mg BID

Heparin 7,500 units Q8H

Obese (≥120kg or BMI ≥ 35)

Enoxaparin 0.5mg/kg BID

(max dose 100mg BID)

Heparin 10,000units Q8H

Low Body Weight (< 60kg)*

Enoxaparin 30mg BID

Heparin 7,500 units Q8H

*LBW does not have a universal definition for LMWH dosing, we define it differently in the non-ICU (<50kg) and ICU (<60kg) populations to help achieve our targeted anticoagulant effect, though this remains an active area of research

VTE Prophylaxis in LMICsCopy Link!

Given limited evidence and resource constraints in LMICs, we recommend standard dosing of VTE prophylaxis as opposed to a tiered dosing approach, in accordance with published guidelines (Ahmed et al).

Therapeutic DosingCopy Link!

Updated Date: October 1, 2021
Literature Review (Anticoagulation):
Gallery View, Grid View
Literature Review (TPA):
Gallery View, Grid View

Tool: COVID-19 (Therapeutics) Guidelines Dashboard

  1. Recommendations for therapeutic anticoagulation of patients with known DVT or PE remain the same as prior to COVID-19.
  1. While some institutions are considering full dose anticoagulation in severe COVID-19 disease without known VTE, our interpretation of the data is that the risks outweigh the benefits at this time, unless documented DVT or PE. Preliminary data from Wuhan suggest that prophylactic LMWH or heparin may be of benefit in those patients with severe COVID-19 and D-dimer levels > 6 times the upper limit of normal (Tang et al).
  2. A propensity score-matched cohort study of 3,772 participants compared COVID-19 patients receiving anticoagulation/antithrombotic therapy prior to diagnosis to patients without prior anticoagulation/antithrombotic therapy. No statistically significant difference in survival or time to mechanical ventilation was observed (Tremblay et al.)
  3. In settings where diagnostic testing is limited, initiating empiric therapeutic anticoagulation for hospitalized COVID-19 patients with high suspicion of DVT/VTE may be done in accordance with local clinical practice guidelines.
  4. Fixed dose subcutaneous heparin may be used for therapeutic anticoagulation in settings where IV heparin or low-molecular weight heparin are unavailable or impractical.
  1. Therapeutic subcutaneous unfractionated heparin dosing: 333 units/kg then 250 units/kg SC every 12 hours
  1. If the patient is on direct oral anticoagulants (DOACs) or Warfarin for Afib or VTE, assess on an individual basis whether to switch to a parenteral anticoagulant with a shorter half-life (LMWH or heparin) based on clinical status.
  1. Consider the same clinical criteria used for non-COVID-19 patients. For example:
  1. Consider LMWH or heparin in COVID-19 patients with AKI, procedures that require time off therapeutic anticoagulation or clinical instability (e.g., patients requiring critical care).
  2. Continue home anticoagulation regimen in clinically stable COVID-19 patients without other contra-indications, with close monitoring of factors that could influence pharmacokinetics (e.g., antibiotics that could increase the effect of Warfarin; renal function for DOACs).
  1. DOACs can be continued in patients on steroids and remdesivir. The benefits likely outweigh the risk of potential interactions between medications (e.g., by the induction of CYP3A4 or the multidrug efflux pump P-glycoprotein by dexamethasone).
  1. Speculative use of therapeutic anticoagulation or tissue plasminogen activator (TPA)
  1. While therapeutic anticoagulation has been used empirically in some severe COVID-19 patients in Wuhan given the possible microthrombi in pulmonary vasculature, our interpretation of the data is that the risks outweigh the benefits at this time, unless documented DVT or PE (Hardaway et al).
  1. Similarly, TPA has been proposed as a possible therapeutic. We recommend against TPA for ARDS

AspirinCopy Link!

Updated Date: December 7, 2020

Aspirin can continue to be used in patients in whom it is indicated (i.e. cardiovascular disease prevention), but at this point in time, there is not enough evidence to support its use strictly for COVID-19 prevention and/or treatment

  • A small retrospective study showed a possible improvement with aspirin started before or early during admission. This has yet to change clinical practice (Chow et al).

AntibioticsCopy Link!

Choice of AntibioticsCopy Link!

A discussion of the risks/benefits of empiric antibiosis and suggested initial regimens is found under Bacterial Infections, whether or not to give empiric antibiosis, and choice of agent. A common initial regimen for community acquired pneumonia is ceftriaxone and azithromycin or doxycycline.

Treatments for Comorbid DiseasesCopy Link!

RAAS InhibitorsCopy Link!

Angiotensin Converting Enzyme Inhibitors (ACEi) and Angiotensin II Receptor Blockers (ARB)

Examples: Lisinopril, Enalapril, and Captopril
Updated Date: May 10, 2020
Literature Review:
Gallery View, Grid View

Tool: COVID-19 (Therapeutics) Guidelines Dashboard

RecommendationsCopy Link!

  1. For outpatients, ACEi/ARBs should not be discontinued
  2. For inpatients, ACEi/ARBs should not be discontinued unless otherwise indicated (e.g., acute kidney injury, hypotension, shock)

Pathophysiology and EvidenceCopy Link!

  1. Data so far indicate that use of ACE inhibitors or angiotensin-receptor blockers (ARBs) is not associated with worse outcomes in patients with COVID-19 (Mancia et al, Reynolds et al).
  1. The REPLACE COVID and BRACE Corona Trials showed indifferent outcomes regarding hospitalization duration, death, AKI requiring RRT etc.) (Cohen et al; Lopes et al).
  1. The evidence that currently exists favors continuing these medications unless otherwise indicated to stop them because their abrupt discontinuation, particularly in those who have heart failure or have had a myocardial infarction, may lead to clinical instability and adverse outcomes (Vaduganathan et al). The American College of Cardiology, American Heart Association and Heart Failure Society of America joint statement recommends against discontinuing ACEi and ARBs in patients with COVID-19 (Bozkurt et al, HFSA/ACC/AHA Statement Addresses Concerns Re: Using RAAS Antagonists in COVID-19, 2020)
  2. Background: SARS-CoV-2, the virus that causes COVID-19, enters via the same cell-entry receptor as SARS-CoV, namely angiotensin-converting enzyme II (ACE2) (Paules et al). SARS-CoV-2 is thought to have a higher affinity for ACE2 than SARS-CoV. ACE2 is expressed in the heart, lungs, vasculature, and kidneys. ACE-inhibitors (ACEi) and angiotensin-receptor blockers (ARBs) in animal models increase the expression of ACE2 (Zheng et al), though this has not been confirmed in human studies. This has led to the hypothesis that ACEi and ARBs might worsen myocarditis or precipitate ACS. It has also been hypothesized that the upregulation of ACE2 is therapeutic in COVID-19 and that ARBs might be protective during infection (Gurwitz).

StatinsCopy Link!

Examples: Simvastatin, Rosuvastatin, Pravastatin
Updated Date: December 7, 2020

  1. Statins can continue to be used in patients in whom they are indicated, but at this point in time, there is not enough evidence to support their use strictly for COVID-19 prevention and/or treatment

Calcium Channel BlockersCopy Link!

Examples: Amlodipine, Nifedipine, Diltiazem, Verapamil

Updated Date: January 7, 2021

  1. Calcium channel blockers can continue to be used in patients in whom they are indicated, but there is not enough evidence to support their use for COVID-19 prevention and/or treatment

Non-Steroidal Anti-Inflammatory Drugs (NSAIDs)Copy Link!

Updated Date: May 10, 2020
Literature Review:
Gallery View, Grid View
Tool:
COVID-19 Guidelines Dashboard

RecommendationsCopy Link!

  1. Concern has been raised that NSAIDs may worsen COVID-19 disease. This has not been proven clinically to-date, so we cannot make a recommendation for or against their use at this time

PharmacologyCopy Link!

  1. SARS-CoV-2 binds to cells via ACE2. ACE2 is upregulated by ibuprofen in animal models, and this might contribute to increased pathology (see “Angiotensin Converting Enzyme Inhibitors (ACE-I) and Angiotensin II Receptor Blockers (ARB)” section of this chapter).

EvidenceCopy Link!

  1. Reports from France indicate possible increase in mortality with ibuprofen in COVID-19 infection, but these reports have not been corroborated (Fang et al; Day). WHO clarified on March 20, 2020 that it does not recommend avoiding NSAIDs as initially stated March 18th (WHO, COVID-19 Interim guidance, March 2020)

BronchodilatorsCopy Link!

Updated Date: December 7, 2020
Literature Review:
Gallery View, Grid View

Bronchodilators are not indicated for COVID alone in the absence of other indications, such as asthma or COPD. Some patients may have bronchoconstrictive responses to infection and so they may help some patients, but should not be used as a default treatment for all patients. When using, try to use meter-dose inhalers (MDIs) with a spacer instead of nebulizers where possible to decrease aerosols

NebulizersCopy Link!

Literature Review: Gallery View, Grid View

Nebulizers should be used very sparingly as they pose a risk to staff due to aerosolized virus. An inhaler with a spacer will provide similar benefit in most patients. Limit nebulizers to patients with severe wheezing who do not respond to inhalers. Any nebulizers should be done on airborne precautions (e.g. N95 mask use for all staff and private room for the patient, with negative pressure if possible). Airborne precautions should be continued for at least 1-3 hours after the treatment finishes.

Tool: Instructions on How to Make a Spacer With a Water Bottle: WHO-ICRC Basic Emergency Care Course (p.158)

Meter-Dose InhalersCopy Link!

Meter-Dose Inhalers (MDIs) can still be used and normal, and should be used with a spacer for efficacy.

Tool: For simple instructions on how to make a spacer with a water bottle, see page 158 of the WHO-ICRC basic Emergency Care Course.

Inhaled CorticosteroidsCopy Link!

Literature Review: Gallery View, Grid View

To treat COVID:

One small study with industry funding suggested a decrease in requiring urgent medical care amongst outpatients with COVID and no underlying lung condition who were treated with Budesonide. (Ramakrishnan et al) Larger studies are ongoing, and this remains experimental.

  • At this time is no convincing data to support ICS as a treatment for COVID in patients without underlying asthma or COPD, and negative effects on viral clearance remain unknown. We do not recommend using inhaled corticosteroids routinely unless as part of a clinical trial.

To treat asthma or COPD:

Whether ICSs are harmful or protective against COVID-19 is debated. In theory, ICS use might reduce local lung immunity and increase susceptibility to disease. However, ICS use reduces the frequency of exacerbations of COPD and asthma, and might even and might even reduce replication of the SARS-CoV-2 virus (Jeon et al). In a large retrospective cohort study of over 140,000 COPD and Asthma patients, use of ICS had no bearing on Asthma-related COVID-19 mortality, and may be associated with increased mortality in COPD patients (though this is confounded by COPD severity). Regardless, use does not appear to be associated with any significant benefit (Schultze et al). The Asthma Section discusses ICS use in the setting of asthma and COVID.

  • If ICS is indicated for treatment of asthma or COPD, these should be continued (or initiated as needed). We do not recommend changing management related to COVID risk
  • If a patient on ICS develops COVID at this time we do not recommend changing withdrawing the ICS

Blood ProductsCopy Link!

Updated Date: May 1, 2020
Literature Review:
Gallery View, Grid View

  1. In general, treat bleeding rather than numbers.
  2. We recommend a restrictive transfusion strategy (Hct > 21, Hgb > 7). Randomized controlled trials of ICU patients have shown that a conservative transfusion strategy (Hgb > 7) is associated with less pulmonary edema, fewer cardiac events and no evidence of harm compared to a liberal transfusion strategy (Hébert et al; Holst et al; Gajic et al).
  3. If hemodynamically stable, transfuse 1 unit at a time and reassess needs.
  1. A conservative approach to transfusions is encouraged given risks associated with blood product transfusions, limited supply (blood drives are limited by social distancing), and volume overload being of particular concern in COVID patients.
  1. Fresh frozen plasma (FFP) or 4 factor-PCC (lower volume) should be given for active bleeding in the setting of known or suspected coagulation abnormalities.
  2. For warfarin reversal, use 4 factor-PCC given longer effect and lower volume.
  1. If PCC is unavailable, FFP and vitamin K (10mg IV administered over 60 minutes) should be given
  2. If FFP is unavailable, vitamin K should still be given, although it can take hours to have an effect
  1. Massive transfusion protocol, as a very limited resource, will need to be activated only by a senior clinician.
  2. Tranexamic acid: only for ongoing oozing/bleeding with over DIC and hyperfibrinolysis.
  3. Procedures: If the patient is at high bleeding risk, the most experienced operator should perform the procedure to minimize complications.
  1. We recommend avoiding subclavian lines when placing central venous catheters in coagulopathic patients.

Patient

DVT ppx

Transfusion Thresholds

Transfuse 1 unit at a time

RBC

Platelets

Cryo

FFP

No bleeding,

Plts > 30k

LMWH daily or

SC UFH TID

Hgb < 7, If ACS,** Hgb > 10

n/a

Fibrinogen < 100

INR > 10

No bleeding, but patient requires anticoagulation

Heparin gtt

PTT goal depends on indication

Hgb < 7, If ACS,** Hgb > 10

Plts < 30k

Fibrinogen < 100

INR > 10

No bleeding,

Plts < 30k

SCDs*

Hold pharmacologic

Hgb < 7, If ACS,** Hgb > 10

Plts < 10k

Fibrinogen < 100

INR > 10

Minor Procedures

(a-lines, CVCs)

Continue pharmacologic ppx in most patients

SCDs* if not using pharmacologic

Hgb < 7, If ACS,** Hgb > 10

Plts < 10k

Fibrinogen < 100

INR > 3

Mild Bleeding or Rigors (increases risk of ICH in thrombocytopenia)

Continue pharmacologic ppx in most patients

SCDs* if not using pharmacologic

Hgb < 7, If ACS,** Hgb > 10

Plts < 20k

Fibrinogen < 100

INR > 3

Intracranial Hemorrhage

+ SCDs*

Hold pharmacologic if able

Hgb < 7, If ACS,** Hgb > 10

Plts < 75k

Fibrinogen < 100

INR > 1.7

Serious Bleeding#, Trauma or Major Procedure

(includes LP)

+ SCDs*

Hold pharmacologic if able

Transfuse for active bleeding

Plts < 50k or higher

Fibrinogen < 100

INR > 2

(INR > 1.4 for LP)

* SCDs = sequential compression devices = “pneumoboots”

** ACS = Acute Coronary Syndrome

# Intracranial hemorrhage and massive bleeding are not included here.

ImmunosuppressantsCopy Link!

See Baseline Immunosuppression

Other Miscellaneous AgentsCopy Link!

HydroxychloroquineCopy Link!

Updated Date: November 15, 2020
Literature Review:
Gallery View, Grid View

Tool: COVID-19 (Therapeutics) Guidelines Dashboard

RecommendationsCopy Link!

  1. Hydroxychloroquine and chloroquine are not recommended in the treatment of COVID-19 outside of clinical trials
  2. Despite an initial EUA, the FDA concluded that it is unlikely that chloroquine or hydroxychloroquine will be effective in treating COVID-19 and that the benefits do not outweigh the risks for use in COVID-19, thereby revoking EUA 039 originally authorized on March 28, 2020 (FDA EUA Revocation Letter June 15, 2020). The World Health Organization, National Institutes of Health and Infectious Diseases Society of America also recommend against its use (WHO Treatment Guidelines, May 2020; NIH Treatment Guidelines, October 2020; IDSA Treatment Guidelines, August 2020)

PharmacologyCopy Link!

Hydroxychloroquine (HCQ) is an anti-malarial 4-aminoquinoline shown to have in vitro activity against diverse RNA viruses, including SARS-CoV-1 (Touret et al).

EvidenceCopy Link!

  1. The first randomized control trial for COVID-19 post-exposure prophylaxis was published on June 3, 2020. Asymptomatic patients who had household or occupational exposures to others with COVID-19 for more than 10 minutes within 4 days of exposure were randomized to receive either placebo (n=407) or hydroxychloroquine 800 mg once, 600 mg in 6-8 hours, then 600 mg daily for 4 additional days (n=414). The incidence of new illness compatible with COVID-19 was 11.8% in the hydroxychloroquine arm and 14.3% in the placebo arm (absolute difference -2.4%, 95% CI -7 to 2.2%, p=0.35). Side effects were more common in the hydroxychloroquine arm (40.1% vs. 16.8%), but no serious adverse reactions were reported (Boulware et al).
  2. The Randomized Evaluation of COVID-19 Therapy (RECOVERY) trial in the United Kingdom has enrolled over 11,500 patients to-date into multiple treatment arms, two of which are hydroxychloroquine and standard of care. While it hasn’t yet been published, the hydroxychloroquine arm of the study ceased enrollment on June 4, 2020 due to lack of benefit. The independent Data Monitoring Committee found that the hydroxychloroquine arm (n=1542) had similar outcomes in terms of 28-day mortality compared to the standard of care arm (n=3132) (25.7% vs. 23.5%, HR 1.11, 95% CI 0.98-1.26, p=0.10) (RECOVERY statement June 5, 2020).
  3. The WHO-initiated SOLIDARITY trial was randomized across 30 countries and >11,000 hospitalized COVID-19 patients comparing 5 potential COVID-19 treatment regimens (remdesivir, hydroxychloroquine, lopinavir/ritonavir, interferon, and combination lopinavir/r with interferon) against placebo. In the hydroxychloroquine portion of the trial, hydroxychloroquine (n=947) did not lead to a significant difference in 28-day mortality compared to placebo (n=906), 11.0% (104 deaths) vs. 9.3% (84 deaths) respectively (RR=1.19, 95% CI 0.89-1.59, p=0.23) (Pan et al).
  4. The NIH’s ORCHID trial has also stopped enrolling patients after the data and safety monitoring board (DSMB) determined that while hydroxychloroquine did not cause additional harm, it also was very unlikely to provide benefit to hospitalized patients with COVID-19 (NIH press release June 20, 2020).
  5. A number of other studies have also shown no positive impact with the addition of hydroxychloroquine in COVID-19 patients (Tang et al; Geleris et al; Mahevas et al; Borba et al; Magagnoli et al).

AzithromycinCopy Link!

Literature Review: Gallery View, Grid View

Tool: COVID-19 (Therapeutics) Guidelines Dashboard

There is not sufficient supporting evidence to use azithromycin for COVID-19 disease outside of clinical trials, unless concomitant community-acquired pneumonia is suspected and atypical coverage is needed. Numerous studies have raised concerns about the deleterious effects of hydroxychloroquine and azithromycin combination therapy (Mercuro et al; Bessière et al; Chorin et al).

For more information on the pharmacology and evidence, please see BWH’s Protocols.

IvermectinCopy Link!

Updated Date: January 2, 2021

Literature review: Gallery View, Grid View

Tool: COVID-19 (Therapeutics) Guidelines Dashboard (New Updates Weekly!)

RecommendationsCopy Link!

  1. A small number of low-quality studies have published data on the use of ivermectin as a therapy for COVID-19. At this time, it is not possible to make any conclusions regarding the efficacy of ivermectin therapy for the treatment of COVID-19. We do not recommend ivermectin use for the treatment of COVID-19 at this time. The United States FDA has issued a warning against using ivermectin intended for animals for the treatment of COVID-19
  2. Patients should be assessed for risk of concurrent Strongyloides Infection. Ivermectin may be indicated for empiric treatment of strongyloides in patients with COVID-19 to prevent complications from corticosteroid therapy.

PharmacologyCopy Link!

  1. The antiviral activity of ivermectin is not entirely clear, but it is postulated that ivermectin may inhibit importin ɑ/β1 receptor, which transmits viral proteins into the host cell nucleus (Caly et al).

EvidenceCopy Link!

  1. In vitro, Caly and colleagues infected cells with SARS-CoV-2 and exposed them to 5 μM of ivermectin over 72 hours. At 24 hours, there was a 93% reduction in viral RNA and at 48 hours, the effect increased to loss of essentially all viral material (~5000-fold decrease). The ivermectin concentration resulting in 50% inhibition (IC50) was estimated to be ~2 μM (Caly et al).
  1. Multiple subsequent studies have shown that ivermectin dosing would need to be much higher than the current maximum approved dosing in order to reach the needed concentration in vivo (Schmith et al; Jermain et al; Momekov et al).
  2. Two letters to the editor have also challenged the initial in vitro study (Bray et al).
  1. A small randomized control trial compared ivermectin 12 mg daily for 5 days (n=22) or ivermectin 12 mg x1 and doxycycline daily for 5 days (n=23) against placebo (n=23). Ivermectin monotherapy showed a reduction in time for viral clearance with a mean duration 9.7 days vs. 12.7 days for placebo (p=0.02), but ivermectin + doxycycline did not show a reduction (11.5 days). Further studies are required and conclusions can't be made from this study (Ahmed et al).
  2. A small prospective controlled (non-randomized) trial compared 2 to 3 doses of ivermectin in combination with 5 to 10 days of doxycycline (n=70) with standard care (n=70). Ivermectin/doxycycline therapy was associated with reduced time to recovery of 10.6 days compared to 17.9 days for placebo (p<0.0001). These results are not peer-reviewed and further studies are needed in order to make any conclusions regarding ivermectin therapy and COVID-19 (Hashim et al).
  3. A retrospective cohort reviewed the impact of ivermectin use in 280 COVID-19 patients in four Florida hospitals. Mortality was less in patients who received ivermectin (n=173, 15% mortality) compared to those who received standard of care (n=107, 25.2% mortality). After adjusting, the mortality difference between groups remained significant (aOR 0.27, 95% CI 0.09-0.80, p=0.03). These findings however require randomized controlled trials for confirmation (Rajter et al).
  4. Additional case series have been published on the use of ivermectin in COVID-19 (Camprubi et al).

DosingCopy Link!

  1. The dose needed to obtain therapeutics concentrations is likely not feasible (see Evidence above). For parasitic diseases, ivermectin dosing ranges from 150 to 400 μg/kg. Published reports thus far have utilized doses of 200 μg/kg once or 12 mg once daily for 5 days
  2. Nausea and vomiting, rash, CNS effects (dizziness, drowsiness, ataxia), itching, eosinophilia, tachycardia, hypersensitivity reactions. Toxicities with higher-than-approved doses are not yet fully understood (Navarro et al).

NitazoxanideCopy Link!

Literature Review: Gallery View, Grid View

Nitazoxanide should not be used outside of clinical trials as overall clinical evidence is lacking and optimal dosing is not known.

For more information on the pharmacology and evidence, please see BWH’s Protocols.

FamotidineCopy Link!

Literature Review: Gallery View, Grid View

Tool: COVID-19 (Therapeutics) Guidelines Dashboard

Famotidine can be used in patients in whom it is indicated (GERD, stress ulcer prophylaxis), but there is not enough evidence to support its use for COVID-19 prevention and/or treatment at this time

Vitamins & MineralsCopy Link!

ZincCopy Link!

Literature Review: Gallery View, Grid View
We do not recommend routine use of zinc for the treatment or prevention of COVID-19, except as part of a clinical trial. For more information on the pharmacology and evidence, please see BWH’s Protocols.

Vitamin CCopy Link!

Literature Review: Gallery View, Grid View
While this idea has been popular on social media, there is currently no evidence to support low- or high-dose vitamin C in COVID-19 patients. For more information on the pharmacology and evidence, please see BWH’s Protocols.

Clinical TrialsCopy Link!

Literature Review: Gallery View, Grid View
Clinical trials are essential for advancing the management of COVID-19, but are challenging in the setting of a new and evolving virus with an ever-changing standard of care. When able, patients are encouraged to enroll in active clinical trials.

  1. National Institute of Allergy and Infectious Diseases (NIAID)-supported COVID-19 studies
  2. Mass General Brigham COVID-19 studies
  3. World Health Organization COVID-19 studies
  4. Clinicaltrials.gov COVID-19 studies
  5. Many large adaptive trials are underway, including RECOVERY, SOLIDARITY, DisCoVeRy, REMAP-CAP, and ACTIV

Baseline ImmunosuppressionCopy Link!

Updated Date: May, 2021

COVID-19 Risk by MedicationCopy Link!

The list below summarizes existing data about the risks of severe COVID for patients on baseline immunosuppressive medications used as disease modifying therapies.

**PLEASE NOTE:

  • This section applies to patients who were already on these agents when they contracted coronavirus. The use of immunosuppressive medications as therapeutics for COVID-19 specifically (e.g. colchicine, glucocorticoids, tocilizumab/sarilumab, and tofacitinib/baricitinib, are covered in corticosteroids and immunomodulators)
  • This review does not include agents for which there is no evidence of true immunosuppression or increased infection risk (such as drugs like interferon beta, glatiramer, or hydroxychloroquine).
  • Most of this data is preliminary and rapidly evolving, and much of it is derived from smal@l and/or uncontrolled studies. It is also very challenging to keep up to date, and thus may not include the most recent studies for all drugs. If you

Class and/or Mechanism

Example Agents

Risk

Data

Anti-B cell Agents

(CD20, CD19, or BAFF)

Rituximab (CD20), Ocrelizumab (CD20), Ofatumumab (CD20), Obinutuzumab (CD20), Inebilizumab (CD19), Belimumab (BAFF)

Increased risk of severe COVID and death.

Concern for potential poor response to SARS-CoV-2 vaccination in patients with B cell depletion from anti-CD20 therapies (Baker et al).

In a review of 1540 MS patients, ocrelizumab and rituximab were associated with hospital admission (aPRs=1.19 and 1.58), ICU admission (aPRs=3.53 & 4.12), and mechanical ventilation (aPRs=3.17 & 7.27) compared to dimethyl fumarate, with similar associations when comparing a combined anti-CD20 cohort with either all other DMTs or with natalizumab (MS Virtual 2020). In a multivariate analysis of 3729 patients in the Global Rheumatology Alliance database, rituximab use was associated with a 4.04 (2.32-7.03) odds ratio of death compared to methotrexate monotherapy. (Strangfeld et al)

Alkylating Agents

Cyclophosphamide

Insufficient data but thought likely higher risk given degree of immunosuppression

Anti IL-1 Agents (Prior to infection. See Anti-IL1 agents as a therapeutic for COVID)

Anakinra, Canakinumab, Rilonacept

Insufficient data

Anti IL-4 Agents

Dupilumab

Insufficient data

One paper suggesting that mechanistically, it is not expected that dupilumab should worsen COVID-19 (Patruno et al)

Anti IL-5 or IgE Agents

Mepolizumab, Reslizumab, Benralizumab, Omalizumab

Insufficient data exists, but some suggestion that there is no increased risk of severe COVID or death.

Only case reports on mepolizumab, with favorable outcomes (e.g. Azim et al.) A series from the Belgian Severe Asthma registry identified no deaths or ICU admissions in patients on anti IL-5 or IgE agents; however, only 8 patients on these agents had a positive SARS-CoV2 IgG and 6 had a positive PCR for diagnosis (Hanon et al.)

Anti IL-6 Agents (Prior to infection. See Anti-IL6 agents as a therapeutic for COVID)

Tocilizumab, Sarilumab, Siltuximab

Insufficient data

Anti IL-17 Agents

Secukinumab, Ixekizumab, Brodalumab

Registry data (Mahil et al) and case reports (e.g. Gisondi et al.) in the dermatological literature suggest favorable outcomes with these agents, though there is insufficient data to stratify by individual agents.

In a registry analysis of 374 psoriasis patients, use of a nonbiologic therapy was associated with an increased risk of hospitalization compared with a biologic, odds ratio 2.84 (1.31-6.18), as was use of no systemic therapy, odds ratio 2.35 (0.82-6.72). This was true regardless of class of biologic (comparing TNF-alpha inhibitors to IL-23 inhibitors to IL-17 inhibitors) (Mahil et al)

Anti IL-23 and IL-12 Agents

Risankizumab, Tildrakizumab, Guselkumab (IL-23),

Ustekinumab (IL-23 and IL-12)

Insufficient data

Anti-T Cell Agents IgG1

Abatacept, Basiliximab

Insufficient data

Antimetabolites

Cladribine

No increased risk of severe COVID-19 or death (Cladribine)

A review of 700 cases in the literature of MS patients with COVID-19 showed no increase in severe COVID-19 or death for patients on cladribine but only 3% of the patients in the study were on cladribine (Mohn et al). In a report of 46 cladribine-treated COVID patients in the Merck Global Patient Safety Database, only 4 cases were classified as “serious”, none required mechanical ventilation, and none died (MMWR).

Antimetabolite (inosine monophosphate dehydrogenase inhibitor)

Mycophenolate

Data is conflicting but suggests possible increased risk of severe COVID or death.

In a multivariate analysis of 3729 patients, immunosuppressant use was associated with a 2.22 (1.43-3.46) odds ratio of death compared to methotrexate monotherapy; of 296 patients on immunosuppressants in the study, more than half were on MMF (68 on monotherapy, 81 on combination therapy.) (Strangfeld et al.) In a French cohort of 694 patients, mycophenolate use was associated with an increased odds ratio of severe COVID in multivariate analysis 6.6 (1.47-29.62). (Filière des Maladies Autoimmunes et Autoinflammatoires Rares) In a study from the Johns Hopkins CROWN registry, of the 108 patients on on immunosuppressive medications there was no significant difference in need for mechanical ventilation, mortality, or length of stay. Outcomes were not stratified by agent, but a substantial number (15) of these patients were on mycophenolate (Andersen et al).

Antimetabolite (Antifolate)

Methotrexate

Insufficient data, however appears possibly lower than mycophenylate (see above)

Antimetabolite (Pyrimidine synthesis inhibitor)

Leflunomide, Teriflunomide

No increased risk of severe COVID-19 or death.

A review of 700 cases in the literature of MS patients with COVID-19 showed no increase in severe COVID-19 or death for patients on leflunomide/teriflunomide (Mohn et al)

Antimetabolite (Thiopurine)

Azathioprine, Mercaptopurine

Increased risk of severe COVID.

Analysis of 1439 cases in the SECURE-IBD registry showed an association between any thiopurine use and severe COVID, adjusted OR compared with TNF-alpha inhibitor monotherapy 4.08 (1.73-9.61), adjusted OR for combination thiopurine + TNF-alpha inhibitor therapy versus TNF-alpha inhibitor monotherapy 4.01 (1.65-9.78). (Ungaro et al). In a multivariate analysis of 3729 patients in the Global Rheumatology Alliance database, immunosuppressant use was associated with a 2.22 (1.43-3.46) odds ratio of death compared to methotrexate monotherapy; of 296 patients on immunosuppressants in the study, 63 were on azathioprine monotherapy and 51 on azathioprine combination therapy. (Strangfeld et al.)

Aminosalicylates

Sulfasalazine, mesalamine

Increased risk of severe COVID with aminosalicylates.

Analysis of 1439 cases in the SECURE-IBD registry showed an association between mesalamine/sulfasalazine use and severe COVID, adjusted odds ratio 1.70 (1.26-2.29) but unmeasured confounders may have affected the data (Ungaro et al). Analysis of 525 IBD pts internationally in SECURE-IBD registry showed an increase in the primary combined endpoint of ICU admission/ventilation/death for patients on sulfasalazine/5-ASA (adjusted OR, 3.1; 95% CI, 1.3–7.7) (Brenner et al). In a multivariate analysis of 3729 patients in the Global Rheumatology Alliance database, sulfasalazine use was associated with a 3.6 (1.66-7.78) odds ratio of death compared to methotrexate monotherapy (Strangfeld et al.)

Calcineurin Inhibitors

Tacrolimus, Cyclosporine

Data is not suggestive of an increase in mechanical ventilation, in-hospital mortality, or length of stay for tacrolimus

In a study from the Johns Hopkins CROWN registry of 2121 patients consecutively admitted with COVID, 108 were on immunosuppressive medications, and there was no significant difference in need for mechanical ventilation, mortality, or length of stay compared to patients not on immunosuppression. Outcomes were not stratified by agent, but a substantial number (32) of these patients were on tacrolimus (Andersen et al).

Anti-Complement Pathway Inhibitors

Eculizumab, Ravulizumab

Likely no increased risk of severe COVID (expert consensus) (Korsukewitz et al).

Corticosteroid: Oral, GI selective

Budesonide (oral)

Unlikely to increase likelihood of severe COVID-19 given high first-pass metabolism/low systemic bioavailability.

Corticosteroids: Systemic (see corticosteroids for use as a therapeutic for COVID)

Prednisone

Dexamethasone

Dose-dependent increased risk of severe COVID and death.

Analysis of 525 IBD pts internationally in SECURE-IBD registry showed adjusted odds ratio of severe COVID of 6.9 (2.3-20.5) for chronic corticosteroid use (Brenner et al). A case series of 600 patients from the Global Rheumatology Alliance showed increased rates of hospitalization for patients on ≥10 mg of prednisone daily (adjusted OR 2.05, 1.06-3.96, p=0.03) (Gianfresco et al). In a registry-based study of 636 patients with multiple sclerosis, glucocorticoid use in the past two months carried an adjusted odds ratio of 2.62 (1.33-5.17) for hospitalization, 1.57 (0.49-4.97) for ICU admission, 4.17 (1.13-15.4) for death (Salter et al).

JAK Inhibitors

Tofacitinib

Baricitinib

Ruxolitinib

Conflicting data in small studies.

Analysis of 37 patients on tofacitinib among 2326 on at least one IBD medication showed no difference in hospitalization, severe COVID, or death despite the fact that the patients on tofacitinib were less likely to be in disease remission (Agrawal et al) A prospective case series of 126 rheumatology patients in New York who developed proven or suspected COVID while on biologic or immunomodulatory therapy found a significant association of JAK inhibitors with increased hospitalization only in the group with spondyloarthropathy (OR 17.6 [95% CI 1.04–299.69, P = 0.047), but not with RA (OR 2.50 [95% CI 0.43–14.52, P = 0.31). The study may be subject to significant confounding (Haberman et al)

Lymphocyte-depleting agents

Alemtuzumab (CD52), Blinatumomab (CD19/CD3), Daratumumab (CD38), Elotuzumab (SLAMF7)

No increased risk of severe COVID-19 or death.

A review of 700 cases in the literature of MS patients with COVID-19 showed no increase in severe COVID-19 or death for patients on alemtuzumab, but only 1% of the patients in the study were on alemtuzumab (Mohn et al).

mTOR inhibitors

Sirolimus

Everolimus

Insufficient data

NrF2 Activators

Dimethyl Fumarate

No increased risk of severe COVID-19 or death.

A review of 700 cases in the literature of MS patients with COVID-19 showed no increase in severe COVID-19 or death for patients on DMF, though selection bias may affect results (Mohn et al).

Other/Complex

Colchicine

Insufficient data

PI3K Inhibitors

Idelalisib, Copanlisib

Insufficient data

Selective Adhesion Molecule Inhibitors (integrin)

Natalizumab,

Vedolizumab

No increased risk of severe COVID-19 or death. (Natalizumab)

Insufficient data (Vedolizumab)

A review of 700 cases in the literature of MS patients with COVID-19 showed no increase in severe COVID-19 or death for patients on natalizumab (Mohn et al).

Sphingosine-1-receptor modulator

Fingolimod

No increased risk of severe COVID-19 or death.

A review of 700 cases in the literature of MS patients with COVID-19 showed no increase in severe COVID-19 or death for patients on fingolimod (Mohn et al).

TNF-alpha inhibitors

Etanercept, Infliximab, Adalimumab,

Certolizumab, Golimumab

No increased risk of severe COVID or death; may decrease risk of poor outcomes.

Analysis of 525 IBD pts internationally in SECURE-IBD registry showed no increased risk of severe COVID with TNF-alpha inhibitors (Brenner et al).

A case series of 600 patients from the Global Rheumatology Alliance showed decreased rates of hospitalization for patients on TNF-alpha inhibitors compared to the rest of the patients in the series (adjusted OR=0.40, 0.19-0.81; p=0.01) (Gianfresco et al).

Management of Baseline ImmunosuppressionCopy Link!

These recommendations mostly derive from expert opinion, vary internationally, and are constantly evolving. Please verify with your institution and local guidance.

In general:

  • In COVID-negative patients, continuation of baseline immunosuppression is recommended, though corticosteroids may be tapered, but these decisions should be made with the relevant specialist(s).
  • In COVID-positive patients, ,anagement of baseline immunosuppressants depends on the type and severity of their chronic illness, the specific immunosuppressants they take, and the severity of their SARS-CoV-2 infection.

DermatologyCopy Link!

Particularly relevant diseases include psoriasis, atopic dermatitis, and hidradenitis suppurativa. This table synthesizes guidelines from the following societies:

Consider entering psoriasis patients with COVID in the following registry: PsoPROTECT.

Patient SARS-CoV-2

Status

Immunosuppressive Initiation

Immunosuppressive Continuation

Negative

Consider risks/benefits, consider postponing rituximab/considering alternative therapy.

Continue all therapies, but consider reducing corticosteroid dose, or discontinuing if high risk. (European Guidelines)

Asymptomatic/Mild Disease

Do not initiate.

Consider risks/benefits.

Moderate-Severe Disease

Do not initiate.

Consider discontinuation of all therapies.

GastroenterologyCopy Link!

Particularly relevant diseases include Crohn’s disease and ulcerative colitis.

This table synthesizes guidelines from the following societies:

Consider entering IBD patients with COVID in the following registry: SECURE-IBD Registry

Patient SARS-CoV-2

Status

Immunosuppressive Initiation

Immunosuppressive Continuation

Negative

For systemic agents: If considering biologic plus another immunosuppressant, consider risk/benefit on a case by case basis. Consider initiation with TNF-alpha inhibitor monotherapy as an alternative; might consider adalimumab given lower risk of immunogenicity compared to infliximab.

Taper steroids where possible. For UC patients with flare, consider budesonide MMX or beclomethasone to minimize systemic steroid exposure. For patients with active small bowel or ileocecal Crohn’s, consider budesonide. In uncontrolled UC patients, optimize oral 5-ASA dose with or without topical rectal 5-ASA. Consider treatment discontinuation in high risk patients.

Asymptomatic/Mild Disease

No specific guidance given.

Consider a transition to budesonide. Stop thiopurines, methotrexate, tofacitinib. Per British guidelines: Hold all biologics for 2 weeks. Per IOIBD guidelines: Uncertainty surrounding whether to stop TNF-alpha inhibitors, ustekinumab, or vedolizumab.

Moderate-Severe Disease

No specific guidance given.

Taper/stop prednisone (if steroid not indicated for COVID) as appropriate or transition to budesonide.

Stop thiopurines, methotrexate, TNF-alpha-inhibitors, tofacitinib. Per British guidelines: Hold all biologics until after COVID-19 resolution. Per IOIBD guidelines: Uncertainty surrounding whether to stop ustekinumab, vedolizumab.

NeurologyCopy Link!

Particularly relevant disease include multiple sclerosis, myasthenia gravis, Lambert Eaton myasthenic syndrome, and other immune-mediated diseases such as neuromyelitis optica, autoimmune encephalitis, certain myopathies, or neuropathies such as CIDP.

This table synthesizes guidelines from the following groups:

Consider entering MS patients with COVID in the following registry: COVID-19 Infections in MS and Related Disease

Patient SARS-CoV-2

Status

Immunosuppressive Initiation

Immunosuppressive Continuation

Negative

Consider on a case-by-case basis any cell-depleting therapies like rituximab, deploy pulse corticosteroids only with very clear indication. Consider delaying immunosuppressive initiation if disease activity permits. Per British Guidelines: Particular caution for ocrelizumab, alemtuzumab and cladribine. Limited evidence suggests that for MS, natalizumab has the lowest risk among high-efficacy treatments.

Generally continue the current regimen. Consider delaying retreatment courses if disease activity permits. Same medication guidance as initiation.

Asymptomatic/Mild Disease

Do not initiate immunosuppression.

Pause immunosuppression (except chronic steroids), particularly cell-depleting therapies, except in cases with risk of sudden/life-threatening relapse

Moderate-Severe Disease

Do not initiate immunosuppression.

Pause immunosuppression (except chronic steroids), particularly cell-depleting therapies, except in cases with risk of sudden/life-threatening relapse

RheumatologyCopy Link!

This table synthesizes guidelines from the following societies:

Consider entering patients with rheumatologic diseases who contract COVID into the EULAR registry if in Europe or the Global Provider-Entered Registry if elsewhere, both available through the following link: COVID-19 Global Rheumatology Alliance.

Patient SARS-CoV-2

Status

Immunosuppressive Initiation

Immunosuppressive Continuation

Negative

Per American guidelines: glucocorticoids should be used at the lowest possible dose.

Initiate medications, including biologics, when conventional synthetic DMARDs fail to control disease activity adequately. Uncertainty surrounding the safety of JAK inhibitors. Per British guidelines: Same as continuation.

Per American guidelines: glucocorticoids should be used at the lowest possible dose. Continue current DMARDs. Per British: Avoid pulse methylprednisolone except for major organ flares, switch IV biologics to SQ options where possible. Assess whether rituximab can be given as a single pulse or time between treatments extended.

Exposure to SARS-CoV-2

No specific guidance given.

Per American guidelines: Continue sulfasalazine. Stop hydroxychloroquine, conventional immunosuppressants (tacrolimus, cyclosporine, MMF, azathioprine, etc.), non-IL-6 biologics, and JAK inhibitors pending 2 weeks symptom free. May consider continuation of IL-6 biologics. Uncertainty surrounding continuation of methotrexate and leflunomide. Per European guidelines: No agreement on this point.

Documented or Presumed SARS-CoV-2 Infection

No specific guidance given.

Per American guidelines: Stop sulfasalazine, methotrexate, leflunomide, hydroxychloroquine, conventional immunosuppressants (tacrolimus, cyclosporine, MMF, azathioprine, etc), non-IL-6 biologics, and JAK inhibitors pending 2 weeks symptom free. May consider continuation of IL-6 biologics. Per British guidelines: Continue sulfasalazine and hydroxychloroquine. Temporarily stop all biologic and synthetic DMARDs.Per European guidelines: In patients with COVID-19, address DMARD management on a case-by-case basis with rheumatologist input.

Solid Organ TransplantCopy Link!

This table synthesizes guidelines from the following societies:

Consider entering solid transplant patients with COVID in the following registry: COVID-19 Transplant Registry. See also the Transplant Library for an up-to-date compendium of papers surrounding transplant and COVID-19.

Patient SARS-CoV-2

Status

Immunosuppressive Initiation

Immunosuppressive Continuation

Negative

Do not alter immunosuppression.

Do not alter immunosuppression.

Asymptomatic/Mild Disease

No specific guidance given.

Depends on the organ in question, with great variance between societal recommendations.

Please see the above linked guidelines for more specifics.

Moderate-Severe Disease

No specific guidance given.

Per British Guidelines: Stop MMF/azathioprine. Consider stopping or reducing calcineurin inhibitor—should dramatically reduce or stop in critically ill patients. Steroids as per treatment for COVID.

Per ISHLT (heart/lung transplant): Consider holding mycophenolate, azathioprine, and calcineurin inhibitors.

Per ERA-EDTA (renal transplant): Discontinue all immunosuppressant drugs except steroids. Increase/start prednisone at 15-25 mg/day. Carefully consider continuing low dose calcineurin inhibitor in patients with higher risk of rejection.

Chapter 11

Obstetrics

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

Literature Review (Obstetrics): Gallery View, Grid View
Tool
: Information for Pregnant Patients About COVID-19 in 7 Different Languages (Pregistry)
Tool: PIH Flow Chart of Pregnancy Care During the COVID-19 Pandemic, From Screening and Triage Through Labor and Delivery to Postpartum Care.

Clinical Course in PregnancyCopy Link!

Clinical PresentationCopy Link!

Updated Date: February 2, 2021

Signs and SymptomsCopy Link!

In general, pregnant women present with signs and symptoms similar to the non-pregnant population (Zaigham; Wu et al; Liu et al; Afshar). In a study of 23,000 pregnant and 386,000 non-pregnant women with symptomatic COVID-19, the most frequently reported symptoms among pregnant women were cough (50%), headache (43%), muscle aches (37%), fever (32%), sore throat (28%), shortness of breath (26%), chills (24%), loss of taste or smell (22%), nausea or vomiting (20%), diarrhea (14%), runny nose (13%), fatigue (13%) and abdominal pain (8%). (Zambrano et al).

Asymptomatic and Presymptomatic InfectionCopy Link!

Rates of asymptomatic and pre-symptomatic infection amongst pregnant women vary in different studies. In one systematic review, 75% of positive mothers in universal screening were asymptomatic, and only 25% were symptomatic (Allotey et al). Depending on local epidemiology, prevalence in asymptomatic pregnant women may be high enough to warrant universal screening. See Screening. A series of 215 pregnant patients from New York demonstrated a 13.7% rate of positive nasopharyngeal swab testing for SARS-CoV2 amongst asymptomatic women (Sutton et al).

Differential DiagnosisCopy Link!

The clinical presentation for COVID-19 can mirror other diseases in pregnancy and other diseases can mimic COVID-19. Keep a broad differential.

  • If a patient is presenting with COVID-like symptoms, particularly a fever, a high clinical suspicion for alternative or comorbid processes such as chorioamnionitis, pyelonephritis, malaria, or influenza is needed.
  • Laboratory abnormalities may overlap with obstetric diagnoses (e.g. transaminitis and pulmonary findings can confound a diagnosis of preeclampsia or HELLP syndrome).
  • If a patient develops new signs / symptoms during hospitalization or labor, COVID-19 needs to be considered regardless of the likelihood of other common infectious processes (such as epidural fever or chorioamnionitis). In a study from two New York hospitals of 14 asymptomatic patients with positive SARS-CoV2 nasopharyngeal swabs admitted to labor and delivery for obstetric indications, 8 developed fever while inpatient as their only COVID symptom, while 2 developed severe disease requiring ICU care (Breslin et al).

Maternal OutcomesCopy Link!

Updated Date: February 16, 2022
Literature Review:
Gallery View, Grid View

Severe disease and ICU admission:

Pregnant women are at increased risk of severe disease compared to the general population (ACOG COVID–19 FAQs). That said, most pregnant women present with mild disease, and critical disease is uncommon. In a study of 23,000 pregnant and 386,000 non-pregnant women with symptomatic COVID-19, the pregnant patients were more likely to be admitted to the ICU (10.5 versus 3.9 per 1000 cases, RR 2.9), end up on a ventilator (2.9 versus 1.1 per 1000 cases, aRR 2.9), or die (1.5 versus 1.2 cases per 1000, aRR 1.25) (Zambrano et al). A metanalysis of 192 studies comprising over 67,000 patients likewise showed ICU admission OR of 2.13, Invasive ventilation OR of 2.59, and need for extracorporeal membrane oxygenation OR of 2.02, and maternal death OR 2.85 (Allotey et al.) Another systematic review of 77 studies including over 11,000 pregnant and postpartum women also showed that those with COVID-19 disease had higher relative risk of ICU admission (1.62, 1.33 to 1.96) and need for invasive ventilation (1.88, 1.36 to 2.60) (Allotey et al).

Traditional risk factors for severe disease as delineated by the CDC may also apply to pregnant women.(https://www.cdc.gov/coronavirus/2019-ncov/need-extra-precautions/people-with-medical-conditions.html)

Obstetric OutcomesCopy Link!

Updated Date: February 16,2022
Data on first and second trimester infections with COVID-19 are limited. Most of the current data are on women infected in the third trimester and most studies were conducted in high-resource settings.

Pre-eclampsia:

In one metanalysis of 42 studies comprising over 438,000 pregnant people, COVID was associated with an OR of 1.33 for pre-eclampsia. Compared with mild disease, severe disease was strongly associated with preeclampsia (OR 4.16). (Wei et al)

Preterm birth and cesarean sections: Current data suggests that maternal SARS-CoV2 infection increases the risk of spontaneous preterm birth, iatrogenic delivery, and Cesarean delivery.

  • A metanalysis of 42 studies comprising over 438,000 pregnant people found that COVID was associated with an OR of 1.82 for preterm birth. Compared with mild disease, severe disease was strongly associated with preterm birth (OR 4.29). (Wei et al)
  • A meta-analysis of 17 studies from the US, Europe, China, and Brazil showed that approximately one fifth of the infants of mothers with SARS-CoV2 infection were born preterm (mostly iatrogenic preterm delivery, not spontaneous), and nearly half were born by Cesarean (Khalil et al). In a U.S. study of 3900 infants born to mothers with SARS-CoV2 infection, more of the infants were born preterm than the national average (12.9 versus 10.2 percent.) (Woodworth et al).
  • In contrast, among 263 infants initially enrolled in the PRIORITY study, no difference in adverse outcomes, including preterm birth, NICU admission, and respiratory disease, were found (though Black and Latina women were underrepresented in this study, Flaherman, 2020). A more diverse cohort of 252 SARS-CoV2-infected pregnant women and 3122 negative pregnant in Dallas also suggested no differences in a composite outcome of preterm birth (RR 1.02, (95% CI 0.70-1.48), preeclampsia with severe features, or cesarean for abnormal fetal heart rate (Adhikari et al).

Effect of disease severity: Obstetric outcomes may be affected by severity of maternal disease. In a cohort of women with severe/critical disease, at least 50% delivered preterm (amongst the subset with critical disease this was > 80%), most of these via Cesarean section (Pierce-Williams et al).

Fetal OutcomesCopy Link!

Updated Date: February 16,2022
Literature Review: Gallery View, Grid View

Miscarriage (Spontaneous Abortion) and StillbirthCopy Link!

In a metanalysis of 42 studies comprising over 400,000 people who were pregnant, COVID infection was associated with an Odds Ratio of 2.11 for stillbirth. (Wei et al). A similar association was also suggested by a recent CDC study which showed a larger number of stillbirths among pregnancies with a COVID 19 diagnosis between March 2020 and September 2021 (DeSisto et al). This association can have a plausible mechanism explained by placental pathological findings due to SARS Co V2 infections.

There is limited data about spontaneous abortion in the first and second trimester. One small case control study suggests no association, but is very limited by sample size (Cosma et al). A larger prospective cohort through the PRIORITY study also suggests no increased risk of pregnancy loss in infected women without severe disease in the first trimester (Jacoby et al, 2021).

Placental Perfusion and IUGRCopy Link!

Placental changes from maternal SARS-CoV2 infection could possibly increase the risk of intrauterine growth restriction (and other adverse pregnancy outcomes). In a case series including 16 placentas of patients with COVID-19 disease, decidual arteriopathy secondary to maternal vascular malperfusion was more likely to be identified in placentas from COVID patients compared to historical controls (Shanes et al). However, in a study that included 252 infants of infected mothers, placental pathologic abnormalities were not associated with disease severity and there was no difference in the rate of small for gestational age (SGA) (Adhikari et al). Other studies have also found increased maternal vascular malperfusion, fetal vascular malperfusion and acute and chronic placental inflammation (DiGirolamo et al)

PrematurityCopy Link!

See Obstetric Outcomes

Neonatal Outcomes and NICU AdmissionCopy Link!

Data on neonatal outcomes, as with prematurity, has been conflicting and may be severity-related. However, in one metanalysis of 192 studies comprising 67,000 pregnancies, it appears that COVID infection is associated with higher rates of NICU admission (OR 4.89). (Allotey et al).

Vertical Transmission and Neonatal OutcomesCopy Link!

See Vertical Transmission (Pediatrics)

Antenatal CareCopy Link!

Updated Date: February 16, 2022
Literature Review (Prenatal Care): Gallery View, Grid View

Screening and TestingCopy Link!

ScreeningCopy Link!

All pregnant patients should be screened for symptoms of COVID-19 at, and ideally before, each visit.

Why screen?: Symptom screening and selected testing of asymptomatic and symptomatic pregnant women is recommended to:

  1. Identify cases early and establish a plan to monitor for symptom progression and how to seek care if needed
  2. Decrease transmission to other patients and healthcare providers during in-person visits, and to determine suitability for in-person versus virtual care
  3. Plan for the personnel, equipment, and rooms needed if there is a chance the patient will be COVID-positive during labor and delivery
  4. Educate patients about decreasing transmission to family living in the same space (including the new neonate, where relevant)

Symptom Screening: Symptom screening can be conducted over the phone, ideally 24 hours before the patient presents for care, and should occur again upon arrival at the facility. See Symptom Screening.

Tool: PIH Obstetric and COVID-19 Screening & Triage Algorithm (not applicable in the US where this is individualized by institution)

TestingCopy Link!

Asymptomatic patient testing: Testing criteria vary by institutional protocol and resource availability. Currently the BWH model is to test the following asymptomatic patients:

  1. All pregnant patients on admission to Labor and Delivery (we repeat testing every 3 days if pt is still admitted)).
  1. Currently accompanying support person is not tested
  1. All planned admissions for scheduled induction of labor or cesarean delivery
  2. Patients with exposure to a person with respiratory illness or known COVID (see COVID exposures).
  1. If the exposure is not confirmed to be COVID and there is a significant possibility that it could be influenza, consider influenza testing and prophylaxis.

Symptomatic testing: All obstetric patients with symptoms of COVID are

  1. Patients who test positive and have mild/moderate illness not requiring admission (by official criteria) are managed at home with frequent check ins from clinic staff. If there is any evidence of worsening symptoms, they are brought in and evaluated in the hospital. Currently, therapies can be offered to outpatients without severe disease (including monoclonal antibodies, IV remdesivir, and nirmatrelvir-ritonavir [Paxlovid]). Per ACOG and SMFM, pregnant women should not be excluded from these options. At our institution, pregnant women can be referred for monoclonal antibody treatment. Given the limited supply, IV remdesivir is an option in case of limited antibody supply. SMFM supports the use of nirmatrelvir-ritonavir in pregnant patients with COVID.Patients with comorbidities chronic lung/heart/kidney/liver disease, mod-severe asthma, immunocompromised, obesity, diabetes, psychiatric or substance use disorder or any concerning signs for moderate or severe disease, dyspnea or chest pain, Sp02 <94%, RR >30 may need to be admitted
  1. See Severity Assessment

Routine Antenatal CareCopy Link!

Visit FrequencyCopy Link!

Due to the risk of asymptomatic transmission of COVID-19, some institutions are changing routine outpatient care to protect women and their families from facility-based transmission. The WHO Focused Antenatal Care Model recommends a minimum of 4 antenatal visits which cluster risk assessments, screenings, distribution of essential medications, and essential patient education about obstetric danger signs. Although not officially endorsed by the American College of Obstetricians and Gynecologists (ACOG), there are published guidelines for routine antenatal care during the SARS-CoV2 pandemic (Boelig et al), and the Society for Maternal Fetal Medicine has published guidelines for modified ultrasound frequency (SMFM Ultrasound Practice Suggestions). Exact practice patterns will depend on available resources, patient medical complexity, local epidemiology, and provider discretion.

It is also important to note that prenatal care can be provided safely in-person when appropriate precautions and safety measures are used. Women should be counseled that it is still essential to obtain care throughout pregnancy to ensure the health of the patient and her fetus.

Patients with Low Risk of Obstetric ComplicationsCopy Link!

Gestational age

Visit type

FIGO Recommendations

MFM Guidelines (Boelig et al.)

<11 weeks

Telephone

OB Intake (History-taking)

~12 weeks

In Person

History-taking

Assessment for risk-factors and comorbidities relevant to COVID-19

Routine labs

Genetic screen if available

Ultrasound if available for dating and nuchal translucency

Initial OB labs

Ultrasound, if available, for dating +/- nuchal translucency

16 weeks

Telephone

~20 weeks

In Person

Routine care

Anatomy scan, if available (at 19-20 wks, favor 20 wks in high BMI patients)

Routine care

Anatomy ultrasound

~24 weeks

Telephone or as needed

Consider checking BP at home or ambulatory setting if possible

Oral glucose tolerance test

~28 weeks

In person

Routine care

Rhogam, if applicable

TDaP if applicable

Routine care

Labs/vaccines

30 wks

Telephone

Consider checking BP at home or ambulatory setting if possible

32 weeks

In person

Ultrasound for placental location if low-lying placenta or placenta previa on prior ultrasound/ growth scan as per local practice

Repeat ultrasound as indicated

34 weeks

Telephone or PRN

Consider checking BP at home or ambulatory setting if possible

36 weeks

In person

Routine care

GBS swab as indicated

Repeat ultrasound as indicated

GBS swab, repeat HIV screen

37-41 weeks

Telehealth

Recommended in-person at 38 wks in FIGO schedule

Routine care and kick counts weekly

Postpartum

Telephone or PRN

High risk may require in-person visit

See Poon et al (algorithm #1) and FIGO Global Interim Guidance: Safe Motherhood and COVID-19

Patients with Intermediate Risk of ComplicationsCopy Link!

Hypertension not on medication, gestational diabetes or pregestational diabetes not on medication, advanced maternal age > 40 years old, BMI > 35, uncomplicated dichorionic diamniotic twin pregnancies, history of intrauterine growth restriction (IUGR), or preeclampsia in a prior pregnancy are considered intermediate risk. We generally recommend a similar approach to first and second trimester screening as low-risk patients plus the following for specific conditions;

  • History of IUGR or pre-eclampsia in a prior pregnancy: Ultrasound for estimated fetal weight at 30-32 weeks. Repeat ultrasound every 6 weeks. (Identification of IUGR places patient in high risk category, see below)
  • Dichorionic Twins: Ultrasound every 4 weeks for fetal growth beginning at 28 weeks.
  • Diet-Controlled Pregestational Diabetes or Gestational Diabetes: Third trimester ultrasound for assessment of fetal weight at discretion of care provider
  • Fetal Anomalies: Growth surveillance tailored to fetal anomaly with more frequent surveillance in the face of hydrops, polyhydramnios, or concern for genetic syndrome
  • Short Cervix but > 2.5 cm: Monitor cervical length every 2 weeks until 28 weeks if cervical length remains stable.
Patients with Higher Risk of ComplicationsCopy Link!

Hypertension on medications, gestational hypertension, pregestational or gestational diabetes on medications, monochorionic twins and higher-order multiples, IUGR in the current pregnancy, preeclampsia, maternal renal disease, other complex maternal or fetal comorbidities are considered higher risk for complications. Patients with other prior medical conditions, or who develop complications during pregnancy, may also be included in this category per provider discretion. These patients require additional surveillance during pregnancy and may develop additional morbidity if care is rationed or deferred. The frequency of antenatal visits and fetal surveillance during pregnancy depends on specific conditions and available resources, and not all the following may be available in every setting.

In addition to standard antenatal care, in settings where antenatal testing is available, consider the following:

  • Hypertension or diabetes: Growth ultrasound every 6 weeks with weekly non-stress tests (NSTs) for interval fetal testing.
  • Monochorionic twins: Ultrasounds every 2 weeks to monitor for evidence of twin-to-twin transfusion syndrome.
  • IUGR: Weekly NSTs with biophysical profile (BPP) and umbilical artery Doppler every 4 weeks in place of NST. Increase NSTs to twice weekly in the setting of abnormal Dopplers.
  • Short Cervix < 2.5 cm before 25 weeks: Weekly ultrasound for cervical length at the discretion of the OB care provider until 25 weeks. A final cervical length ultrasound can be considered after cerclage placement.

Obstetrical ReferralCopy Link!

Referral to obstetrical care for patients cared for by Community Health Workers, midwives, and other practitioners should continue based on the usual indications. Community Health Workers (CHWs) are important for the identification of cases needing referral in the community. A framework for establishing roles of CHWs during COVID-19 is discussed here. Patients with symptomatic COVID-19 may not be appropriate for delivery in all facilities. Institutions without both critical care and obstetrical care services and the necessary PPE and oxygen support should prepare for transferring severe cases of COVID-19 in pregnant women.

Social Screening and Patient EducationCopy Link!

Social screenings remain an essential service, perhaps more than ever given the frequency of economic and food insecurity during pandemic. Providers should provide referrals to food and economic assistance where available. In addition, economic instability and home quarantine/isolation may contribute to increased rates of intimate partner violence and/or child abuse, as well as unplanned pregnancy. Clinical providers should continue to screen patients for these conditions and provide referral when appropriate.

Patient education should include the following:

  • Anticipatory guidance about pregnancy/childbirth.
  • Coronavirus-related changes being made to visit schedules or care protocols (including infection prevention measures)
  • Ways to protect themselves from contracting coronavirus at home and in the community (see Transmission Prevention)
  • Information on how to contact a maternity provider (their own obstetrician if they are followed by a single doctor, or a provider at their antenatal clinic or maternity ward) with questions or symptoms
  • Review of obstetric and COVID danger signs with advice on when to seek care
Danger signsCopy Link!

COVID-19 Danger Signs

Obstetric Danger Signs

Difficulty breathing/shortness of breath

Bluish lips or extremities

Gasping for air when speaking

Coughing up blood

Pain/pressure in chest when NOT coughing

Dizziness when standing

Altered mental status or severe sleepiness

Inability to eat/drink or walk

Labor pains

Rupture of membranes

Vaginal bleeding

Decreased fetal movement

Severe headache

Visual changes

Convulsions

Persistent fever

Unexplained abdominal pain

COVID VaccinationCopy Link!

Updated: February 16, 2022

The COVID-19 vaccines currently available have not been tested in pregnancy and thus there is no current safety data specific to pregnancy. There is also no data that indicates the vaccine should be contraindicated in pregnancy. Over the past few months, data from prospective studies support safety and immunogenicity of COVID vaccines (Gray et al). As such, the American College of Obstetricians and Gynecologists (ACOG) recommends that all eligible persons aged 12 years and older, including pregnant and lactating individuals, receive a COVID-19 vaccine or vaccine series. The mRNA COVID-19 vaccines are preferred over the J&J/Janssen COVID-19 vaccine for all vaccine-eligible individuals, including pregnant and lactating individuals, for primary series, primary additional doses (for immunocompromised persons), and booster vaccination.

For more information and full recommendations on boosters, see vaccines.

Treating COVID in PregnancyCopy Link!

Updated Date: February 16, 2022
Literature Review: Gallery View, Grid View

Both ACOG and SMFM have provided guidance for the management of pregnant women with COVID-19 disease. However institutions have individualized their practice based on their resources and community prevalence of the disease.

Antenatal MonitoringCopy Link!

Patients with COVID-19 infection (at any point in their pregnancy) should receive increased fetal surveillance as we do not yet know the full impact of infection on fetal development and there is evidence of placental effects of SARS Co V2 infection. There is not yet standard guidance on this, though BWH will often suggest:

  1. Growth ultrasound surveillance starting when the pregnancy is in the third trimesterweeks, and continuing every 3-4 weeks until delivery
  2. Weekly fetal nonstress test beginning when the pregnancy is at 32-34 weeks until delivery. (Can use biophysical profile at the times of growth surveillance ultrasounds in lieu of nonstress tests to reduce unnecessary testing.)

Clinical Severity AssessmentCopy Link!

See Acuity Triage and Disease Severity and Disposition for advice on the appropriate care location for patients in general. The initial assessment of pregnant women should include assessment of gestational age and fetal well-being. For telephonic triage, this includes verbal assessment of fetal movement (if appropriate by gestational age); for in-person assessment, fetal heart rate monitoring should be performed as appropriate for gestational age.

Pregnant women, due to both younger age and physiologic adaptation, can compensate for medical illness in a way that masks its severity until their physiologic reserves are suddenly exhausted. Classically, they will appear quite well until they rapidly deteriorate. Suggested criteria for observation or admission are as follows. The appropriate admission or observation location (e.g. medical ward, labor and delivery, or intensive care unit) should be made in conjunction with the obstetric care team and depending on local policies:

Triage assessments that may be possible at home or in healthcare settings:

  1. COVID-19 danger signs and Obstetric danger signs
  1. Even patients with mild COVID may warrant admission for obstetric indications
  1. SpO2 < 94% or <92% with exertion on room air if a pulse oximeter is available.
  2. Blood pressure abnormalities if a BP monitor is available
  1. Hypotension (typically systolic < 90 or diastolic < 50, unless baseline blood pressure is below these criteria in the prior antenatal record)
  2. Hypertension (typically systolic > 140 or diastolic pressure > 90 on two separate measurements four hours apart in the absence of chronic hypertension)
  1. Persistent fever > 38°C (101°F) despite antipyretic
  2. Respiratory rate > 30
  3. Maternal tachycardia > 120 (not improved with fluids if in healthcare setting)

Triage assessment in healthcare settings:

  1. Lab abnormalities including transaminitis, elevated creatinine, or new thrombocytopenia < 150K
  2. Category 2 fetal heart rate on tracing (or, if unavailable, on intermittent auscultation) despite adequate maternal resuscitation.
  1. For tracings: see criteria from Bailey et al
  2. For intermittent auscultation: See criteria from American College of Nurse-Midwives
  1. The presence of maternal or fetal comorbidities:
  1. Maternal examples may include pulmonary disease, heart disease, diabetes, HIV infection, or hypertension including preeclampsia.
  2. Fetal examples include intrauterine growth restriction, monochorionic twin pregnancy, or other markers of placental insufficiency.

Intermittent Auscultation of Fetal Heart RateCopy Link!

This technique can be used if continuous fetal monitoring is unavailable (American College of Nurse-Midwives).

  1. First auscultate FHR between contractions and when the fetus is not moving. At the same time, assess the mother's radial pulse to ensure that the FHR is auscultated and not the mother’s.
  2. After establishing the baseline rate, auscultate the FHR for 15 to 60 seconds at various intervals depending on the stage of labor.
  1. If not in labor or in latent phase, every 30 to 60minutes.
  2. In active labor, every 15 to 30 minutes
  3. In second stage, every 5 to 15 minutes)
  1. Non-reassuring findings include the presence of any of the following:
  1. Irregular rhythm
  2. Presence of FHR decreases or decelerations from the
  3. baseline
  4. Tachycardia (sustained FHR above 160bpm for >10 minutes in duration)
  5. Bradycardia (sustained FHR below 110bpm for >10 minutes in duration)

Mild Illness (Outpatient)Copy Link!

Updated Date: February 16, 2022

Care for pregnant COVID-19 patients with mild disease largely mirrors that of the general population. See also Home and Outpatient Management.

  • Symptomatic management is appropriate for mild cases of confirmed or suspected COVID-19.
  • Educate patients about Isolation, as well as COVID and obstetric warning signs.
  • Maintain adequate oral hydration
  • Patients should ideally receive follow-up telephone contact by a provider within 48 hours after initial diagnosis, and then on a regular schedule based on risk thereafter.
  • Therapeutics specific to COVID-19 disease in pregnancy are discussed below. Currently monoclonal antibodies, IV remdesivir, and nirmatrelvir-ritonavir (Paxlovid)are options.

Moderate or Severe Illness (Inpatient)Copy Link!

Updated Date: February 2, 2021

Compared to other patients, there are some minor modifications in the care of pregnant patients with moderate or severe COVID-19, including a higher target oxygen saturation and different considerations for proning. See also Inpatient Management for general adult management guidelines.

  • All women on admission to the maternity ward should identify someone to make healthcare decisions in the event that they are unable to (healthcare proxy) and communicate goals of care.
  • For patients with moderate or severe disease, routine care, including imaging (with shielding where possible) should not be withheld due to pregnancy.
  • If antibiotic treatment is administered, antibiotics contraindicated in pregnancy (e.g. tetracyclines, fluoroquinolones) should be avoided.
  • In cases of maternal hypoxia, given the benefits of proning, a supported prone position or Lateral Sims can be used, with cushioning used to avoid direct pressure on the abdomen (Halscott et al).
  • Target oxygen saturation for pregnant women is 95% or greater.
  • Discharge planning should involve follow-up with a maternity care provider within 48 hours, and then on a regular schedule based on risk thereafter, and increased antenatal monitoring.

Critical IllnessCopy Link!

Updated Date: February 2, 2021

ManagementCopy Link!

Most management of the critically-ill pregnant patient is unchanged from that of the general population (see Critical Care Management), but differences exist.

  1. Team: Given the need for fetal monitoring and potentially for urgent delivery, care should ideally include a multidisciplinary critical care and obstetrics team in the location that makes the most sense based on the patient’s anticipated clinical needs. Neonatology if available should be notified of any pregnant patient admitted to the hospital at or beyond 22 weeks.
  2. Physiologic targets: Modifications from standard care should reflect the physiologic changes of pregnancy including:
  1. Increased cardiac output (heart rate and stroke volume)
  2. Decreased systemic vascular resistance
  3. Increased minute ventilation (driven by respiratory rate)
  4. Physiologic compensated respiratory alkalosis
  5. Decreased pulmonary functional residual capacity
  6. Increased GFR and volume of distribution
  7. Expanded plasma volume
  8. Alterations in clotting cascade to promote coagulation
  1. Intubation should be discussed earlier than for an equivalent non-pregnant patient given increased airway edema and aspiration risk in pregnancy as well as limited functional residual capacity.
  1. Ventilation should target the respiratory alkalosis of pregnancy maintaining a pCO2 < 45 and a pH > 7.35. Permissive hypercapnia may be acceptable but inability to maintain these targets alongside other standards of care for lung-protective ventilation should prompt a discussion with the obstetric care team. Depending on the gestational age and clinical status, this may prompt more liberal parameters, additional fetal monitoring, or consideration of delivery.
  1. Prone positioning in pregnancy has been reported and should be considered for standard indications (Dennis et al). See Proning. A supported prone position or Lateral Sims can be used, with cushioning used to avoid direct pressure on the abdomen (Halscott et al). Early discussion of the feasibility of this strategy and gestational-age dependent patient positioning considerations should take place with the obstetric team.
  2. Steroids: Require balancing fetal and maternal needs, see Therapeutics in Pregnancy.
  3. Fetal monitoring: A plan for fetal monitoring should be established by the obstetric team. Fetal status may be evaluated twice daily (via non-stress test, if available) with increased frequency if alterations in fetal status are identified. Spontaneous preterm birth rates in critically-ill populations are very high. The obstetrics team should share anticipated outcomes at a given gestational age with the critical care team to inform their management.

Tool: BWH Quick Tips for Intensivists Caring for Critically Ill Obstetric Patients
Tool: Prone Positioning for Pregnant Women With Hypoxemia Due to COVID-19 (Tolcher et al)

Delivery in Critically Ill PatientsCopy Link!

  1. When to call obstetrics: Fetal distress or unexplained maternal tachycardia, hypertension, increasing sedation requirements, or tachypnea require obstetric evaluation for possible preterm labor. See also Tocolysis.
  1. Contraindications to labor (e.g. placenta previa, prior uterine surgery, in some settings nonvertex presentation) should be noted prominently in the patient’s chart and birth plan
  1. Equipment: A vaginal delivery kit, cesarean delivery kit, and neonatal warmer and resuscitation kit should be at the bedside in anticipation of spontaneous delivery or maternal cardiac arrest for all patients.
  2. Planning for emergent delivery: The multidisciplinary team should all be aware of the pathway for emergent delivery for emergent maternal or fetal indications based on gestational age and maternal stability.
  3. Planning for urgent or scheduled delivery: Delivery may become necessary in select circumstances where oxygenation or ventilation are thought to be impaired by pregnancy.
  1. Anticipate the autotransfusion after delivery as blood returns from the uterus into the circulation. Diuresis and PEEP optimization may be needed.
  2. Vaginal delivery is the preferred mode of delivery for patients in the absence of contraindication to labor. The decision for vaginal versus Cesarean delivery in the critically-ill patient should be individualized taking into account parity, gestational age, monitoring needs, and likelihood of a successful vaginal birth.

Cardiac ArrestCopy Link!

Maternal cardiac arrest at or beyond 20 weeks, or when the uterus is at the level of the umbilicus, includes specific modifications to account for the impact of the gravid uterus (Jeejeebhoy et al).

  1. Provide immediate left uterine displacement by either pushing or pulling the uterus off the IVC.
  1. This is a dedicated role for a member of the code team.
  1. Ensure the IV used for code medications is above the level of the diaphragm to ensure no interference with return to circulation.
  2. Remove and detach all fetal monitors.
  3. Prepare to begin a resuscitative hysterotomy (perimortem Cesarean delivery) if 4 minutes elapse without return of spontaneous circulation (ROSC) with goal for delivery of the fetus by 5 minutes after the cardiac arrest.

Labor and Delivery for COVID PatientsCopy Link!

Updated Date: February 16, 2022
Literature Review: Gallery View, Grid View

Care of COVID-19 patients during labor generally follows routine protocols, with minor modifications, but should include increased maternal and fetal monitoring. See Infection Control in Obstetric Settings for details on infection control measures.

Guidance on management of COVID disease is provided in the United States by ACOG and SMFM with the individualization of guidelines done at the institution level.

LaborCopy Link!

Support PersonCopy Link!

WHO recommends women have a companion of their choice with them during childbirth as part of supporting women’s rights to a “safe and positive childbirth experience” (World Health Organization). We recommend that women should be allowed a screened, asymptomatic support person. The support person must wear a mask and refrain from walking around the ward.

Maternal AssessmentCopy Link!

Assess severity of COVID-19 symptoms at minimum every 4 hours. In case of deterioration, make an individualized assessment regarding the risks and benefits of continuing labor vs proceeding with an emergency Cesarean.

Fetal Monitoring and AmniotomyCopy Link!

Consider continuous electronic fetal monitoring, where available

  • Internal monitors (fetal scalp electrodes, intrauterine pressure catheters) may be used for usual indications, given that current data thus suggest only maternal-to-fetal placental transmission of SARS-CoV2 and the virus has not been reliably isolated in amniotic fluid (see placental transmission).
  • Amniotomy may still be utilized for labor management as clinically indicated (SMFM and SOAP Labor and Delivery Considerations)

Labor AnalgesiaCopy Link!

  • Nitrous oxide: Some institutions avoid nitrous oxide in COVID+ patients due to risk of aerosolized infectious droplets and risk of viral contamination of the breathing system and equipment. There is variability in practice and if used, adequate PPE and a bacterial viral filter should be used.
  • Remifentanil PCA: There is no data currently to guide practice in COVID19, but should be used with caution due to risk of respiratory depression, especially in women with respiratory symptoms or need for supplemental oxygen.
  • Neuraxial analgesia: Recommended for standard indications to laboring women. May confer additional benefit in women with COVID-19 as the ability to rapidly convert the epidural to surgical anaesthesia in the event of operative delivery being required would potentially avoid the need for general anesthesia, which is a risk to the patient and also an aerosol generating procedure.
  • Other practices are generally the same as for non-COVID patients (Bampoe et al).

TocolysisCopy Link!

If antenatal corticosteroids are administered, tocolysis can be administered until completion. Tocolysis is not contraindicated in SARS-CoV2 infection, but the choice is highly individualized, weighing the fetal benefit of slowing spontaneous preterm labor (more acute at certain gestational ages) against the potential maternal side effects of the individual agents. In preterm labor, intraamniotic infection may need to be ruled out before using tocolysis. Ruptured membranes are a contraindication.

  • Indomethacin can be used in most cases until 32 weeks.
  • Nifedipine is a commonly used tocolytic from 32-34 weeks. Caution that using magnesium sulfate and a calcium channel blocker concomitantly can lead to decreased muscle contractility and can worsen respiratory depression. Caution must be used in pregnant patients requiring oxygen as nifedipine is a potent systemic and pulmonary vasodilator that can worsen hypotension or V/Q mismatch. Nifedipine is contraindicated for women with cardiac manifestations of COVID.
  • Magnesium Sulfate is not the tocolytic of choice for SARS-CoV2 infected patients who do not have an additional indication (e.g. fetal neuroprotection, seizure prophylaxis).

Magnesium SulfateCopy Link!

Magnesium sulfate is routinely used for both fetal neuroprotection and preeclampsia/seizure prophylaxis. In SARS-CoV2 infected patients without respiratory symptoms, it should be used for the usual obstetric indications. For patients with respiratory symptoms, the benefits of therapy should be weighed against potential risks of maternal respiratory depression (less relevant if the patient is intubated). If renal function is impaired, doses should be adjusted accordingly.

  • Respiratory depression typically occurs at serum concentrations of 10-13 mg/dL and will be preceded by loss of deep tendon reflexes often occurring at levels of 7-10 mg/dL.
  • A single 4-gram bolus of magnesium sulfate without subsequent infusion may serve as an alternative to usual dosing in the setting of mild respiratory distress.

DeliveryCopy Link!

Updated Date: February 16, 2022

Timing of DeliveryCopy Link!

  • Timing and mode of delivery should be individualized based on clinical status of the patient, gestational age, and fetal condition.
  • Timing of delivery should not be dictated by maternal COVID-19 infection in most cases. According to ACOG, for women with COVID infection early in pregnancy who recover, no alteration in the usual timing of delivery is indicated. For women with COVID infection in the third trimester who recover, it is reasonable to defer delivery untl quarantine status is lifted (to avoid transmission to the neonate)
  • Medically indicated deliveries should not be delayed in an individual who is COVID-19 positive.
  • If a woman requires ventilatory support, it may be reasonable to expedite delivery via induction or Cesarean delivery as it may be more difficult to ventilate with a gravid uterus.

Mode of DeliveryCopy Link!

  • Vaginal delivery is not contraindicated. There is no evidence that suggests women with suspected or confirmed COVID-19 cannot give birth vaginally or would be safer having a Cesarean section or operative vaginal delivery.
  • Operative Delivery: Data on perinatal transmission at this time does not preclude the use of forceps or vacuum.
  • Cesarean Delivery:
  • Cord Clamping: Delayed cord clamping has known neonatal benefits. Given the lack of evidence of significant vertical transmission of SARS-CoV2, delayed cord clamping is not contraindicated.
  • Placental tissue and miscarried embryos/fetuses should be treated as infectious tissues and disposed of appropriately.

Neonatal ResuscitationCopy Link!

Respiratory distress in a neonate can have a variety of etiologies, including perinatal COVID-19 transmission, prematurity, sepsis, and asphyxia. Treatment therefore should be based on clinical presentation. COVID-19 testing is not routinely performed in infants. If performed, testing should be delayed to 24-48 hours of life as maternal secretions can cause false positivity; a follow-up test should be performed at 48-72 hour intervals. Throat and nasopharyngeal swabs are generally used (Puopolo et al).

Further management of COVID-19 exposed infants is reviewed in the Pediatric section.

Postpartum CareCopy Link!

Updated Date: February 2, 2021

BreastfeedingCopy Link!

See Breastfeeding (Pediatrics)

Skin-to-Skin ContactCopy Link!

There are significant benefits to mother/newborn skin-to-skin contact including mother-infant bonding, increased success with breastfeeding, and stabilization of infant glucose levels and body temperature. However, neonates may be at increased risk for severe infection (see pediatric severity). Shared decision-making is recommended. It is reasonable to counsel a mother to practice skin-to-skin contact with her baby, while wearing a mask and after handwashing (World Health Organization). See also breastfeeding and distancing for infected caregivers.

ContraceptionCopy Link!

Given decreased in-person contact with the healthcare system during COVID-19, offer postpartum contraception (especially long-active reversible contraceptives and progesterone-only methods) and initiate prior to discharge if the patient desires.

Pain ControlCopy Link!

Pain control is per provider preference. Non-steroidal anti-inflammatory drugs (NSAIDs) may be used and are safe in breastfeeding for most patients. Currently there is no evidence of severe adverse effects in patients with COVID-19 as a result of the use of NSAIDs.

Hospital DischargeCopy Link!

  • Timing of discharge depends on the clinical status of mother and newborn, the mode of delivery, and the ability to isolate safely.
  • Teaching: In addition to routine counseling, patients should be taught about handwashing/physical distancing, postpartum depression, and COVID-19 signs/symptoms, and should be screened for the need for social or economic support.
  • Follow-up:
  • For mild disease, full visit (by phone/video if possible) within a week after discharge.
  • For moderate or severe disease, check-up call from a healthcare provider within 48 hours after discharge and a full visit (by phone/video if possible) a week after discharge. These patients should also be offered home community health worker or nursing services for monitoring of symptoms and vital signs.

Therapeutics in PregnancyCopy Link!

Symptomatic TreatmentsCopy Link!

Updated Date: February 2, 2021

  • Acetaminophen/paracetamol: Fever may be associated with an increased rate of birth defects, especially neural tube defects, We recommend acetaminophen for pregnant women with a temperature ≥ 100.40F, up to 1000mg in a single dose, not to exceed 3000mg in 24 hours. Acetaminophen is also the preferred antipyretic for influenza in pregnancy (ACOG Guidance on the Assessment and Treatment of Pregnant Women with Suspected or Confirmed Influenza; SMFM, Am J Obstet Gynecol). Nonsteroidal anti-inflammatories should be avoided during pregnancy (though not for COVID reasons).
  • Cough suppressants: Guaifenesin and dextromethorphan are safe in pregnancy.

Thrombosis ProphylaxisCopy Link!

Updated Date: February 16, 2022

COVID-19 infection and pregnancy are both independent risk factors for thrombosis and there is a theoretical additive risk of thrombosis. Thrombosis in pregnancy is especially common in the third trimester and postpartum and can contribute to severe morbidity and mortality.

  • Outpatients: There are no universal practices on VTE prophylaxis for outpatients. Non-pregnant outpatients typically are not treated with any thromboprophylaxis. Low-dose aspirin is safely used in pregnancy for several indications, most commonly in preventing preeclampsia (ACOG Guidance on Low-Dose Aspirin Use During Pregnancy). At BWH, we discuss the option of low dose ASA for pregnant women who have been diagnosed with COVID disease and are managed in the outpatient setting.
  • Inpatients: Different institutions adopt standard or intermediate-dosing thromboprophylaxis. Generally pregnant women are treated with the same regimens as non-pregnant patients at most institutions. The NIH treatment guidelines recommend that pregnant women hospitalized with severe COVID disease receive prophylactic dose anticoagulation unless contraindicated. At BWH, patients with resolved COVID disease are offered low dose ASA to continue in the outpatient setting
  • Postpartum patients: Some post-partum patients may merit prophylactic anticoagulation after delivery. This is very practice- and patient- specific, depending on risk factors, mode of delivery (surgical or vaginal), and patient mobility. Expert guidance is mixed or absent. The Royal College of Obstetricians and Gynecologists recommends 10 days of thromboprophylaxis post-discharge for pregnant patients or postpartum patients within 6 wks of discharge who have been admitted for COVID-19 (RCOG)

Systemic CorticosteroidsCopy Link!

Updated Date: February 2, 2021

Corticosteroid treatment in pregnancy must balance maternal COVID recovery and fetal lung maturation with the desire to minimize unnecessary fetal exposure. Certain corticosteroids (methylprednisolone, prednisone, prednisolone, and hydrocortisone) have limited placental transfer and can be used to treat the mother but limit fetal exposure.

Corticosteroids for COVID: A 10-day course of corticosteroids is recommended for adults with an oxygen requirement due to COVID-19 infection.

Corticosteroids for Fetal Lung Maturity: A 48-hour course of corticosteroids is recommended for fetal lung maturity between 24+0 and 33+6 weeks of gestation in patients at high risk of preterm birth within seven days.

Recommendations:

  1. If a patient requires corticosteroids for both fetal lung maturity and COVID (generally in weeks 24-33 6/7)
  1. Dexamethasone 6mg IM q12h x 4 doses. (This high dose is for fetal lung maturation.)
  2. After the initial 48 hours of dexamethasone, many patients can be switched to hydrocortisone, methylprednisolone, or prednisone to reduce fetal exposure.. See Corticosteroids for dosing. The RECOVERY trial in the U.K. (the initial study proving the utility of dexamethasone in COVID-19 patients with an oxygen requirement) used prednisolone 40 mg administered by mouth daily or hydrocortisone 80 mg IV twice daily for pregnant patients. Recovery Collab, NEJM 2020.
  1. If a patient meets criteria for corticosteroids because of COVID but does not require corticosteroids for fetal lung maturity (generally < week 24 or > week 34)
  1. Recommend hydrocortisone, methylprednisolone, or prednisone to minimize passage to the fetus.
  1. If a patient meets criteria for corticosteroids for fetal lung maturity, but not for COVID, recommendations remain the same as for non-COVID patients
  1. Betamethasone 12 mg IM Q24 hours for two doses
  2. Dexamethasone 6mg IM q12h for four doses.
  1. Only betamethasone or dexamethasone should be used as other options do not adequately transfer across the placenta
  1. If a non-breastfeeding postpartum patient meets criteria for corticosteroids for COVID
  1. Recommendations remain the same as for a non-pregnant COVID patient.
  1. If a breastfeeding postpartum patient meet meets criteria for corticosteroids for COVID
  1. Recommend methylprednisolone, prednisone, prednisolone, or hydrocortisone. See Corticosteroids for dosing.

Other Therapeutics in PregnancyCopy Link!

Updated Date: February 16, 2022

  • Remdesivir: Data is limited in pregnancy. Pregnancy was an exclusion criterion for participation in the initial remdesivir trials for COVID-19. A retrospective review of 86 pregnant and postpartum women who did receive remdesivir recorded serious adverse events in 16%, generally Grade 3 transaminase elevations, generally thought to be related to underlying disease, (though remdesivir can cause transaminitis.) One mother died of underlying disease; there were no neonatal deaths (Burwick R et al). There is no known fetal toxicity with remdesivir. SMFM and ACOG recommend this be offered to pregnant patients who meet criteria for treatment of COVID disease
  • Immunomodulators: Anti-IL6 Agents (Tocilizumab, Siltuximab, Sarilumab). Tocilizumab does cross the placenta. Post-marketing data analysis of pregnancy outcomes in 288 evaluable women out of 399 who were exposed to tocilizumab shortly before or during pregnancy revealed no substantial increase in adverse pregnancy outcomes. However, this series was too small and diverse to demonstrate the safety of this agent in pregnancy (Hoeltzenbein et al).
  • Convalescent plasma and monoclonal antibodies: Pregnancy is not a contraindication to receiving convalescent plasma or current monoclonal antibodies, if otherwise meeting criteria.

Chapter 12

Pediatrics

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

Epidemiology in ChildrenCopy Link!

Literature Review (Pediatrics): Gallery View, Grid View
Updated Date: February 1, 2021

There are many challenges associated with quantifying the true number of children infected with SARS-CoV-2 and other relevant epidemiologic factors:

  1. The lack of population-level testing, low levels of access to testing for children in particular, and the focus on testing only patients presenting with symptoms.
  2. The heterogeneity across data sources with regard to the chronological definition of a “child” and surveillance methods used (PCR vs. rapid antigen tests vs. clinical suspicion).
  3. The lack of disaggregation by age in pediatric data sources. This makes it challenging to tease out epidemiologically significant differences that may exist between neonates, infants, young school-aged children, and adolescents (Cruz et al).

Susceptibility: Data from a systematic review of contact tracing and population-based studies suggests that children are less susceptible to infection than adults.

  • One analysis of 32 studies comprising 41,640 children and adolescents and 268,945 adults showed that the pooled odds ratio of being an infected contact (PCR or serology confirmed) in children (relative to adults) was 0.56 (Viner et al).
  • However, there is a significant amount of variability between studies. One seroprevalence study of household secondary infections reported that children and adults appear equally likely to become infected while quarantined in a household with another infected individual (Brotons et al).
  • Lower susceptibility may be less true for adolescents: The above analysis found lower seroprevalence in children compared with adults, although seroprevalence in adolescents appeared similar to adults (Viner et al).

Viral variants change susceptibility. Delta variant appears to affect children more than prior variants have, with fully 18% of new cases reported in children in the USA (AAP, a higher fraction than before), which could represent increased susceptibility or social factors (e.g. fewer precautions). The delta variant also could increase severity, with 1.9% of pediatric patients ending in hospitalization as of August, 2021 (CDC Data Tracker).

Prevalence: The prevalence of SARS-CoV-2 is lower in children compared to adults (MMWR, 2021). In addition, children account for a smaller percent of total cases than their population prevalence would suggest, which appears to be due to lower susceptibility rather than asymptomatic carriage. A large cohort study from Italy reported that children accounted for 1.8% of total SARS-CoV-2 infections (Bellino et al). As mentioned above, this may change with new variants.

Clinical CourseCopy Link!

SymptomsCopy Link!

Updated Date: February 5, 2021

Common SymptomsCopy Link!

Many children present with minimal or no symptoms (see asymptomatic). Children who are infected tend to exhibit milder symptoms compared to adults (Wald et al).

Those who do exhibit symptoms tend to report:

In infants <3 months old a wide range of fever, respiratory, gastrointestinal, cardiac and neurologic findings have been reported. A high index of suspicion for SARS-CoV-2 is needed in young infants presenting with generalized symptoms such as fever, decreased feeding or lethargy even in the absence of respiratory symptoms (Mithal et al).

Uncommon ManifestationsCopy Link!

Since SARS-CoV-2 symptoms vary widely in children, when community infection rates are high, it is important to maintain a high level of suspicion for children with unusual presentations. Case reports have described previously healthy children presenting with:

Asymptomatic InfectionCopy Link!

Many children infected with SARS-CoV-2 are asymptomatic. While a study from Milan concluded that the asymptomatic carrier rate is not higher in children compared to adults (Milani et al), studies differ widely on estimated asymptomatic carrier rates in children. For example:

  • Studies from China and Korea reported that approximately one in four PCR+ children were asymptomatic (Qiu et al, Han et al).
  • A small study from France found that 45% of PCR+ children were asymptomatic (Poline et al).
  • A study from India reported that 73% of PCR+ children were asymptomatic (Fakiri et al).
  • A study from Barcelona found that almost all (99.9%) PCR+ children living in a household with an infected adult were asymptomatic or very mildly symptomatic (Brotons et al).

Another large study from 28 children’s hospitals in the US that conducted PCR testing on asymptomatic children presenting for medical or surgical care found that positivity rates among asymptomatic children across 25 regions varied from 0% to 2.2% with a pooled prevalence of 0.65%; positive rates among asymptomatic children were significantly associated with the incidence in the general population during the period when testing occurred (Sola et al).

Asymptomatic children can still transmit to others, including to vulnerable older adults:

  • Viral loads and duration of viral shedding may be similar in asymptomatic and symptomatic children. In a study from France, asymptomatic, PCR+ children exhibited similar viral loads to those with symptoms suggesting a comparable potential to spread infection (Poline et al).
  • The duration of viral RNA detection is only slightly shorter in asymptomatic children (mean of 14.1 days after initial positive test result) compared to symptomatic children, though the importance of duration of PCR positivity in infectiousness remains unknown (see infectiousness timeline), (Han et al).

SeverityCopy Link!

Severe manifestations: For those with a more severe course, manifestations may include (Derespina et al, Nepal et al):

Lab indicators of severity: Similar to adults, children with severe disease had significantly higher levels of CRP and procalcitonin at admission and significantly higher peak IL-6, ferritin and D-dimer levels during hospitalization (Zachariah et al). Other variables associated with increased disease severity include: lymphopenia, elevated WBC count, elevated platelet count, elevated creatine kinase MB, and elevated proBNP (Qiu et al; Kainth et al; Chao et al).

Age:

  • Adolescents: Adolescents and those with comorbidities are at higher risk for developing severe disease (Derespina et al; Shekerdemian et al). A study comparing characteristics of infected children requiring hospitalization and intensive care found that adolescents >15 years old were overrepresented in the hospitalized cohort. Adolescents >15 years old were also overrepresented in the critically ill cohort (DeBiasi et al).
  • Infants and Neonates: Some studies show that neonates are also more likely to be hospitalized (DeBiasi et al) and have severe infection (Bellino et al). In a systematic review of 52 studies on outcomes of pregnant women with SARS-CoV-2 infection and their newborns showed only one study was deemed to be “at low risk of bias.” Results were also highly variable across studies, with neonatal death rates ranging from 0-11.7%, suggesting that the evidence base on infant and neonatal severity remains poor (Vergara-Merino et al). However, most cases are still mild to moderate and improve with supportive care (Mark et al; Leibowitz et al; Nathan et al; Paret et al; McLaren et al; Zhang et al). Rarely, neonatal infection can be associated with life-threatening pulmonary disease (Coronado et al). Cases of silent hypoxemia requiring respiratory support have also been reported, highlighting the importance of training parents to recognize signs of possible hypoxemia, such as poor breastfeeding or changes in skin color (Sinelli et al).

Literature Review (Neonatal Care): Gallery View, Grid View

Hospitalization, ICU rates, and death: Rates of hospitalization, ICU level-of-care, and mortality tend to be lower in children than adults, but are highly variable depending on testing patterns, case definitions and resources.

  • According to data from the US Department of Health, as of September 2020, children represented 1.7% of total SARS-CoV-2 hospitalizations and 0.07% of total deaths (Sisk et al).
  • These rates may vary by age and geographic location:
  • One large retrospective cohort study that included people <25 years of age in the US found that 7% of SARS-CoV-2-infected children required hospitalization, and of those hospitalized, 28% required ICU-level care and 9% required mechanical ventilation. Case fatality rate was 0.2% (Bailey et al).
  • Another that included 51 children <13 years of age who were hospitalized with SARS-CoV-2 symptoms in South Africa reported that 21.6% required ICU-level care and 2% died (Van der Zalm et al).

TimecourseCopy Link!

Updated Date: February 1, 2021


Incubation and window period: (see Adult Incubation and Window Period)

  • In one study, children were found to be pre-symptomatic for a median of 2.5 days (i.e. symptoms developed approximately 2.5 days after viral levels were detected) (Han et al). This pre-symptomatic period is slightly shorter than is estimated for adults (5 days, see Incubation).
  • Testing during this phase may be unreliable (see Window Period).

Duration of symptoms

  • In one large cohort study, symptoms lasted for a median of 11 days (Han et al). This period also appears to be slightly shorter than is estimated for adults (see Timecourse)

PCR positivity timeline: As with adults, testing positive via PCR does not necessarily correlate with infectivity, as the test may detect non-infectious viral particles (see Infectiousness Timeline).

  • A large cohort study of children who underwent PCR and antibody testing for SARS-CoV-2 between March and June 2020 found that the median duration of PCR positivity was 19.5 days (Bahar et al). A study from Korea reported a mean duration of 17.6 days(Han et al).
  • This period of positivity may be slightly shorter in infants: In one cohort of infants <1 year of age, time between symptom onset to negative detection of SARS-CoV-2 RNA was 13 days (Liu et al).

ComorbiditiesCopy Link!

Updated Date: February 1, 2021

AsthmaCopy Link!

While asthma is the most frequent underlying diagnosis in SARS-CoV-2 infected children, patients with asthma do not seem to be overrepresented among those requiring hospitalization (DeBiasi et al). It is unclear whether there is an increased risk of SARS-CoV-2 susceptibility among children with asthma or whether infection increases risk of asthma exacerbation, but (similar to adults) mild asthma does not appear to be linked with higher morbidity (see Asthma). Treatment of concurrent asthma and SARS-CoV-2 is the same as treatment of each individually, including oral corticosteroids if required. Meter-dosed inhalers are preferred to nebulizers in health facilities where possible due to increased risk of viral transmission (Abrams et al). See Nebulizers, Bronchodilators, Inhaled Corticosteroids.

Congenital Heart DiseaseCopy Link!

Congenital heart disease may be a risk factor for severe disease, though data remains limited. A case series of 7 children with congenital heart disease who contracted SARS-CoV-2 reported that all 7 developed acute decompensation (Simpson et al).

DiabetesCopy Link!

As with adults, children with diabetes are at risk for severe complications of SARS-CoV-2 infection. The main strategy for decreasing risk is to optimize glycemic control. Patients with T1D and SARS-CoV-2 should adhere to standard sick-day guidelines with increased frequency of monitoring of blood glucose and ketones. Frequent changes in dosage and correction in insulin boluses may be required to maintain normoglycemia. In patients with T2D and SARS-CoV-2, dosage of drugs such as Metformin should be adjusted or held to decrease the risk of lactic acidosis (Ho et al).

Sickle Cell Disease or ThalassemiaCopy Link!

Differentiating between standard SARS-CoV-2 infection and Acute Chest Syndrome (ACS) is important for patients with Sickle Cell Disease (SCD). Testing and radiographic appearance (diffuse vs. subpleural changes) may help with differentiation. A close watch for pulmonary hypertension is necessary. Aggressive blood transfusion or exchange transfusion is recommended in those who develop or are at high risk for developing ACS. This commentary describes topics specific to patients with SCD and Thalassemia including hydroxyurea, medication concerns, thrombosis prophylaxis, and recommended blood tests (Taher et al).

Trisomy 21Copy Link!

Children with Trisomy 21 and associated comorbidities such as congenital heart disease, pulmonary hypertension, or obstructive sleep apnea who become infected with SARS-CoV-2 should be considered high risk and monitored closely (Newman et al; Krishnan et al).

Immune DeficienciesCopy Link!

The clinical course of immunocompromised children (including HIV, malnutrition and cancer) is generally not more severe than other children (Kainth et al; Marais; Wald et al). In a cohort of 50 children and adolescents hospitalized with SARS-CoV-2 infection, infants and immunocompromised patients were not at increased risk for severe disease requiring mechanical ventilation. However, obesity and hypoxia on admission were associated with severe disease (Zachariah et al; Fernandes et al).

Vertical TransmissionCopy Link!

Updated Date: February 1, 2021
Literature Review (Vertical Transmission):
Gallery View, Grid View

This section addresses transmission to neonates and breastfeeding infants. While studies differ on how transmission rates compare between children, between children and adults, and between adults (Zhu et al; Brotons et al, Viner et al; Szablewski et al; D’Agostino et al) the mode of transmission in these scenarios is likely the same. Please also see Transmission Prevention.

Placental TransmissionCopy Link!

Definitive evidence that SARS-CoV-2 crosses the placenta and infects the fetus is lacking, though the possibility is not ruled out (Lamouroux et al). Expression of the main receptor (ACE2) used for entry of SARS-CoV-2 is rare on the surface of the placenta and could explain the low rates of placental infection.

A few cases where placental tissues have been positive for SARS-CoV-2 have been described, as have a few cases of possible in utero transmission during the third trimester, some in premature infants (Gupta et al). Many neonatal transmissions are likely due to respiratory transmission from mother to neonate after birth. While it can be hard to definitively prove that transmission occurred intrauterine or intrapartum, testing the umbilical cord, blood and amniotic fluid in the first hour after birth may provide more definitive answers to the source of infection (Lo Vecchio).

  • In a systematic review of studies including 936 pregnant women with SARS-CoV-2 infection, viral RNA was found in 3.2% of neonatal nasopharyngeal swabs, 2.9% of cord blood samples, 7.7% of placenta samples, and 0% of amniotic fluid and infant urine samples (Kotlyar et al). Another review of approximately 800 patients reported similar findings (Kyle et al).
  • Other cohort studies of neonates born to mothers infected with SARS-CoV-2 reported no evidence of vertical transmission (Dumitriu et al; Verma et al).

However, neonates of infected mothers may have an inflammatory response without direct infection. Another case report described a systemic inflammatory response in an infant born to a mother with SARS-CoV-2; though the infant tested negative for SARS-CoV-2, the systemic inflammation likely occurred in response to the virus in the absence of vertical transmission (McCarty et al).

BreastfeedingCopy Link!

Updated Date: February 1, 2021
Literature Review:
Gallery View, Grid View

Virus in BreastmilkCopy Link!

Breast milk isolated from SARS-CoV-2 positive mothers has only rarely been found to contain SARS-CoV-2 viral RNA, and it is unclear if the viral RNA detected is capable of infecting an infant (Groß R et al). If there is any transmission through breast milk, it is likely very uncommon (Centeno-Tablante et al). In situations where there is concern, pasteurization can eliminate the infectivity (Conzelmann). Many preliminary laboratory and clinical reports support the safety of breastfeeding regardless of mother’s infection status as breast milk is unlikely to be a source of infection for infants (Furman et al; Chan et al; Lugli et al). However, transmission by respiratory droplets from mother to infant during the process of breastfeeding is possible due to the necessary proximity.

Breastfeeding RecommendationsCopy Link!

While breast milk itself is unlikely to transmit the virus, there is still uncertainty around neonatal risk from breastfeeding that has led to wide variations in care practices for newborns. Breast milk helps protect newborns against many illnesses and has been shown to contain neutralizing antibodies to SARS CoV-2 in mothers who are immune (Demers-Matheiu et al; Fox et al; Pace et al); it is also known to be the best source of nutrition for most infants. Despite the inherent proximity in direct breastfeeding, healthy neonates do not often become infected: In a study of 62 neonates (95% breastfed) who roomed in with mothers with SARS-CoV-2 infection, only one infant was diagnosed with the virus, and that infant developed only transient mild dyspnea (Ronchi et al). In a cohort of 188 infants delivered at a large county medical center, neonatal infection occurred in 3% of infants, predominantly among infants born to asymptomatic or mildly symptomatic women (Adhikari et al).

For healthy neonates, recommendations on breastfeeding differ:

  • The American Academy of Pediatrics prefers separation of healthy newborns from infected mothers and feeding initially with expressed breast milk (AAP).
  • The World Health Organization prefers rooming together and breastfeeding (WHO).
  • The Center for Disease Control and Prevention (CDC) recommends shared decision-making (Gupta et al).

For neonates requiring intensive care, isolation from SARS-CoV-2 positive individuals is recommended. However, feeding with expressed breast milk may still be permitted.

Instructions for mothers with SARS-CoV-2 infection who wish to breastfeed or express breast milk include:

  1. Wash hands (and, if possible, breasts) with soap and water before and after touching the infant or any pumps or bottle parts
  2. Avoid using a pump shared by others
  3. Wear a mask or cloth face covering during breastfeeding or pumping
  4. Follow manufacturer instructions for proper pump cleaning after each use, cleaning all parts that come into contact with skin or breast milk
  5. If the child is not being breastfed directly, pumped breast milk should be fed to the infant by a healthy caregiver to minimize exposure (Sullivan et al)

Distancing for Infected CaregiversCopy Link!

The decision about whether an infected caretaker should be physically distanced from an infant to minimize respiratory transmission is personal and recommendations vary widely. Providers and families should engage in shared decision-making, balancing the risks to the neonate, the availability of non-infected caretakers, the desire to breastfeed, Higher rates of breastfeeding are noted in unseparated dyads both in the hospital and at home (Popofsky et al) and the psychological harms to parents and neonate that can result from early separation. Provided that mothers adhere to proper respiratory and skin hygiene measures including hand washing and masking, many studies suggest that the benefits of rooming together after birth may outweigh the risks if mothers are able to care for their newborns (Ronchi et al; Kaufman et al; Williams et al; Boscia). However, given the lack of definitive answers surrounding neonatal risk, some studies and institutions still recommend that infants be separated from infected mothers (use barriers, distance 1-2 meters) and fed initially with either formula or expressed breast milk if there is a well caregiver who can attend to the infant until the mother meets criteria to discontinue isolation (Zeng et al; ACOG).

Clinical AssessmentCopy Link!

Initial EvaluationCopy Link!

Updated Date: January 24, 2021

Infection Control: Children presenting in person with possible signs and symptoms of SARS-CoV-2 infection should be evaluated in separate clinical spaces by providers wearing appropriate PPE per IPC guidance.

Acuity TriageCopy Link!

Any child with confirmed or suspected SARS-CoV-2 infection should be triaged based on Disease Severity using definitions of mild, moderate, severe, and critical to determine appropriate treatment location and disposition. Healthy children and adolescents with mild symptoms may be candidates for phone-based triage and monitoring (see mild disease). The following patients should be evaluated at a healthcare facility:

  • Any child with altered mental status, respiratory distress, or signs of organ dysfunction such as decreased urine output should be immediately triaged to an emergency unit.
  • Any febrile infants <3 months should be evaluated in person due to their increased risk of severe disease and bacterial infection.
  • Infants and young children with >3 days of fever and any other symptoms should be seen in person so a trained clinical can rule-out bacterial supra-infection and assess hydration and respiratory status.

Tool: Risk Categorization Algorithm for Children with Suspected COVID Infection
This algorithm helps classify children with suspected infection into severity categories and suggests an appropriate location of care and management approach. Note that we do not recommend routine use of Azithromycin except with suspected secondary bacterial infection (Feketea et al).

School EvaluationCopy Link!

Recommendations about school closures, epidemiology in schools, and minimizing transmission in schools are available here.

Tool: Algorithms for Parents and Schools for Symptomatic or COVID-Exposed Children These tools can help parents, students, administrators, and school nurses approach students with suspected SARS-CoV-2 symptoms or exposure (Orscheln et al).

Differential Diagnosis: It is important to consider a broad differential including bacterial co-infection (Mithal et al) when working up symptomatic infants and children. The Differential Diagnosis for children with suspected SARS-CoV-2 infection is similar to adults. Other respiratory viruses to consider include: influenza, adenovirus, RSV, parainfluenza, and metapneumovirus; atypical pneumonias such as mycoplasma or chlamydia pneumoniae can also present similarly. Severe disease, especially MIS-C may be confused with Toxic Shock Syndrome (TSS), Kawasaki Disease (KD), or severe sepsis.

SARS-CoV-2 TestingCopy Link!

Updated Date: February 1, 2021

Nucleic Acid Amplification and Rapid Testing: Indications for testing children for SARS-CoV-2 vary by local epidemiology and practice. The timing and type of testing for children is similar to adults (CDC), but the sample collection technique may be different as children may not tolerate nasopharyngeal swabs. Many places use mid-turbinate swabs (or back-of-throat swabs) instead. These less invasive swabbing techniques are likely 5-10% less sensitive (Boston Children’s Hospital).

Serology: The timeframe to antibody response (seropositivity) is likely similar to adults as well. In one study, the median time from PCR positivity to seropositivity was 18 days, but median time to reach adequate levels of neutralizing antibodies was 36 days (Bahar et al).

Laboratory TestingCopy Link!

Common laboratory findings in children infected with SARS-CoV-2 are similar to those of adults and include: leukocytosis, lymphopenia, and elevated inflammatory markers, D-dimer and troponin (Gonzalez-Dambrauskas). We recommend the same laboratory monitoring schedule recommended for adults.

ImagingCopy Link!

Common radiologic findings for children infected with SARS-CoV-2 are similar to those of adults and include bronchial thickening and ground-glass opacities in a basilar and peripheral distribution. Peribronchial patterns and bronchial wall thickening appear to be more common in pediatrics (Chen et al). These findings were noted in both symptomatic and asymptomatic children (Castagnoli et al).

Ultrasound: A preliminary report from Italy noted a high concordance between radiologic and lung ultrasound findings in children infected with SARS-CoV-2, suggesting that lung ultrasound may be a reasonable method to detect lung abnormalities in children (Denina et al).

CT Scan: Routine use of CT scanning is not recommended. In pediatric patients with SARS-CoV-2 infection but minor or no upper airway symptoms, a chest CT may be normal in up to 50% of cases, creating a false sense of security while exposing children to ionizing radiation. Thus, CT scanning of the chest is not a suitable screening tool to rule out infection in children (Merkus).

Management by SeverityCopy Link!

Updated Date: Jan 22, 2021
Tool: ID COVID-19 Treatment Guidelines
Tool:
NIH COVID-19 Treatment Guidelines

Routine supportive care is the mainstay of therapy for children with SARS-CoV-2 and should be provided to all pediatric patients, even those with severe or critical disease (Chiotos et al; Larson et al).

Severity Classification and Vitals MonitoringCopy Link!

To classify disease by severity, see severity and disposition (which includes pediatric vitals).

For suggested monitoring frequency by severity, see vitals and monitoring.

Mild DiseaseCopy Link!

Most patients with mild disease and no significant risk factors can be managed at home.

Medications: Most cases can be managed in an outpatient setting with symptomatic treatment.

  1. In otherwise healthy patients without comorbidities, currently we do not recommend disease-targeted therapies
  1. Certain patients with comorbidities may be candidates for outpatient monoclonal antibodies on a case-by-case basis
  1. We do not recommend VTE prophylaxis apart from ambulation
  2. Existing medications generally should be continued:
  1. Immunosuppressants: case-by-case
  2. Nonsteroidal Anti-Inflammatory Drugs: can continue in most patients
  3. Inhalers: can continue; avoid nebulizers if possible due to increased risk of transmission
  1. Comorbidities may need extra attention and monitoring (e.g. T1D or adrenal insufficiency)

Household and Caregivers: Children should have a designated caregiver (ideally someone who has already been exposed), and should isolate within the household as much as possible (see household assessment and preparation).

Followup: As with adults, disease may progress in severity in the second week, so patients with new symptoms should be re-evaluated to exclude evolution to ARDS, severe neutropenia and possible secondary infection (Venturini et al). We recommend a followup schedule similar to the one recommended for adults.

When to seek care: Patients should seek care if they have worsening symptoms or danger signs. Infants and young children with >3 days of fever should be seen in person so a trained clinician can rule-out bacterial supra-infection and assess hydration and respiratory status. In some patients, home pulse oximetry may be helpful.

Moderate or Severe DiseaseCopy Link!

MedicationsCopy Link!

  1. Corticosteroids should be considered in patients with oxygen requirements or severe disease
  2. Convalescent Plasma may be beneficial in some hospitalized patients
  3. Remdesivir is generally not recommended, but further data in pediatric patients is forthcoming
  4. VTE Prophylaxis is indicated in some, but not all patients
  5. Symptomatic Treatments are discussed below
  6. As with adults, most existing medications (“home” medications, or medications taken regularly prior to hospitalization) can be continued unless cessation is otherwise indicated (e.g. for renal failure). Immunosuppressants should be discussed on a case-by-case basis.

Fluid ManagementCopy Link!

  1. A conservative approach to intravenous fluid administration should be used in children with SARS-CoV-2 infection in the absence of shock and dehydration. This is especially true in settings where mechanical ventilation is limited, as aggressive fluid resuscitation may worsen oxygenation and pulmonary compliance (Schultz; WHO). If the patient does need resuscitation, initial fluid management should be conservative (around 10mg/kg boluses while monitoring hemodynamics and urine output). Avoid large boluses (20mg/kg or more) which may compromise gas exchange and exacerbate hypoxemia (Maitland et al).
  2. Maintenance fluids are only indicated if the patient is dehydrated and unable to take oral fluids.

Oxygen TherapyCopy Link!

Oxygen delivery and escalation is covered in Oxygen Therapy.

Critical CareCopy Link!

Respiratory Failure and ARDSCopy Link!

Updated Date: January 29, 2021

While most children with acute SARS-CoV-2 infection have mild symptoms, children, especially with comorbidities, can develop severe pneumonia, respiratory failure, and ARDS (Liguoro et al). Severe pneumonia is defined by the WHO clinical signs of pneumonia (cough or difficulty breathing) and at least one of the following:

  • Central cyanosis or SpO2 < 90%; severe respiratory distress (i.e. fast breathing, grunting, severe chest indrawing); general danger signs (i.e. inability to breastfeed or drink, lethargy or unconsciousness, or convulsions)
  • Fast breathing (in breaths/min): < 2 months: ≥ 60; 2–11 months: ≥ 50; 1–5 years: ≥ 40 (WHO)

Oxygen TherapyCopy Link!

  1. Goals of Therapy: SpO2 >90% and manageable work of breathing (i.e. age-appropriate respiratory rate and no subjective or objective signs of labored breathing such as retractions and nasal flaring)
  2. Begin supplemental oxygen therapy by nasal cannulaNasal cannula or prongs tend to be well tolerated in young children. when SpO2 < 90%
  1. If emergency signs such as obstructed or absent breathing, severe respiratory distress, central cyanosis, shock, coma, or convulsions are present, use a higher initiation target of < 94% and consider proceeding directly to intubation and mechanical ventilation
  1. Titrate oxygen to target saturation SpO2 > 90% (> 94% if emergency signs are present) and manageable work of breathing. If goals of therapy are not met:
  1. Escalate support by delivering oxygen via face mask with a reservoir bag. If this is insufficient, evaluate the patient’s risk for developing ARDS including age, timing, origin, and imaging criteria as ARDS and oxygenation (Pediatric Acute Lung Injury Consensus Conference Group):

Oxygen Delivery Device

Patient Considered At Risk for ARDS if:

Noninvasive ventilation

(Nasal mask, CPAP or BiPAP)

FiO2 > 40% to maintain SpO2 88-97%

Noninvasive ventilation

(Oxygen via mask, nasal cannula or high flow)

SpO2 88-97% with oxygen supplementation at minimum flow:

<1 year: 2 L/min

1-5 years: 4 L/min

5-10 years: 6 L/min

>10 years: 8 L/min

Mechanical ventilation

Oxygen supplementation to maintain SpO2 >88% but oxygenation index <4 or oxygen saturation index <5 (OSI; [FIO2 × mean airway pressure × 100]/SpO2)

  1. If the patient is not at risk for developing ARDS:
  1. Escalate to high flow nasal cannula (HFNC) if available. If goals of therapy are still not met or HFNC is not available:
  1. Escalate to noninvasive positive pressure ventilation (NIPPV), bubble continuous positive airway pressure (bCPAP) or bilevel positive airway pressure (BiPAP) if available.
  1. If the patient is at risk for ARDS:
  1. NIPPV should be considered in early disease to improve gas exchange and work of breathing (The Pediatric Acute Lung Injury Consensus Conference Group). Some locations will also use HFNC for this indication.
  1. If the patient is at risk for ARDS and has severe hypoxia or work of breathing: proceed to intubation with low tidal volume ventilation unless unavailable or otherwise contraindicated.
  1. A child with respiratory distress and hypoxia should be closely monitored for signs of clinical deterioration as children can rapidly progress to respiratory failure and shock. If the child worsens to moderate or severe ARDS, the child should be intubated and mechanically ventilated by a trained healthcare worker using an appropriately sized cuffed endotracheal tube (WHO; Kache et al).

Acute Respiratory Distress SyndromeCopy Link!

Children with SARS-CoV-2 infection can progress to ARDS. Pediatric ARDS has similarities to adult ARDS, but both the definition and management take into account pediatric-specific conditions (e.g., cyanotic heart disease) and physiology. Pediatric ARDS is defined by the Pediatric Acute Lung Injury Consensus Conference Group as (Kemani et al):

  1. Age: Exclude patients with perinatal related lung disease
  2. Timing: Within 7 days of a known clinical insult
  3. Origin: Not fully explained by cardiac failure or fluid overload. For special populations (cyanotic heart disease, chronic lung disease, and left ventricular dysfunction), the acute deterioration in oxygenation should not be explained by the underlying disease.
  4. Imaging: New infiltrate(s) consistent with acute pulmonary parenchymal disease
  5. Oxygenation:

Noninvasive ventilation (PARDS, no severity stratification)

PF ratio <300 or SF ratio <264

Mechanical ventilation (Mild PARDS)

OI 4-8, OSI 5-7.5

Mechanical ventilation (Moderate PARDS)

OI 8-16, OSI 7.5-12.3

Mechanical ventilation (Severe PARDS)

OI >16, OSI >12.3

OI: oxygenation index; OSI: oxygen saturation index. Use PaO2-based metrics when available. If PaO2 is not available, wean FIO2 to maintain SpO2 ≤ 97% to calculate oxygen saturation index (OSI; [FIO2 × mean airway pressure × 100]/SpO2) or SpO2:FIO2 (SF) ratio.

Mechanical VentilationCopy Link!

For children with SARS-CoV-2--associated ARDS requiring mechanical ventilation, target:

  • Initial tidal volumes of 6 ml/kg per ideal body weight (PBW). Tidal volume should then be adapted to disease severity: 3–6 mL/kg PBW if poor respiratory compliance, and 5–8 mL/kg PBW with preserved compliance.
  • Plateau pressures <28 cmH2O
  • pH 7.15–7.30
  • Optimized positive end expiratory pressure (PEEP): PEEP titration should be individualized to the patient and the ARDS phase
  • Note: These targets are different from those used in adult patients! More detailed instructions on mechanical ventilation parameters and adjustment, synchrony, and weaning is available in the adult mechanical ventilation section.

Proning: Consider Prone Positioning in children with ARDS and severe hypoxemia. Methodology is similar to proning in adults.

Sedation and Neuromuscular Blockade: Consider intermittent or continuous neuromuscular blockade in cases of significant ventilator dyssynchrony despite adequate sedation, refractory hypoxemia, or refractory hypercapnia (WHO). Information on adult sedation and neuromuscular blockade is available for reference, though pediatric practice is different.

Extracorporeal Membrane Oxygenation (ECMO): If resources are available, ECMO should be considered in pediatric patients to manage ARDS and/or cardiac failure (myocarditis, arrhythmias, PE) (Kache et al). Information on adult ECMO is available here. Criteria for children are often different depending on the institution.

ShockCopy Link!

Updated Date: March 5, 2021

Septic ShockCopy Link!

While sepsis and septic shock are rare manifestations of SARS-CoV-2 infection in children (Liguoro et al), they can occur, and should be promptly recognized and treated. Sepsis is a dysregulated immune and inflammatory response to an infection that can cause life-threatening organ dysfunction (Singer et al). In children, it is defined as a suspected or proven infection with ≥ 2 age-based systemic inflammatory response syndrome (SIRS) criteria, one of which must be abnormal temperature or WBC count:

  • Abnormal temperature (>38.5°C or <36°C)
  • Tachycardia for age or bradycardia for age if <1 year
  • Tachypnea for age or need for mechanical ventilation
  • Abnormal WBC count for age or > 10% bands (Goldstein et al)

Signs of septic shock in children may include any combination of the following:

  • Altered mental status
  • Bradycardia or tachycardia (HR < 90 bpm or > 160 bpm in infants and heart rate < 70 bpm or > 150 bpm in children)
  • Peripheral vasodilation and bounding pulses (warm shock), or prolonged capillary refill (> 2 sec) and weak pulses (cold shock)
  • Fast breathing
  • Mottled or cool skin, or petechial or purpuric rash
  • Elevated blood lactate
  • Reduced urine output
  • Hyperthermia or hypothermia
  • Hypotension (SBP < 5th percentile or > 2 SD below normal for age) as a late sign (Davis et al; WHO).

The immediate goals when managing pediatric septic shock are to maintain perfusion to the organs and treat the underlying infection. Below is a brief overview of pediatric septic shock management (Weiss et al; WHO; Kache et al):

  1. Antimicrobial therapy: Within 1 hour of recognition, initiate empiric, broad-spectrum antimicrobial therapy.
  2. Monitoring and Targets: Perfusion targets include age-appropriate MAP, urine output (1 mL/kg/hr), and improvement of skin mottling and extremity perfusion, capillary refill, heart rate, level of consciousness, and lactate. Monitor frequently to guide fluid resuscitation and vasoactive medications.
  1. In settings where accurate MAPs cannot be easily obtained, systolic blood pressure is an acceptable alternative.
  2. Given the concurrence of cardiogenic and septic shock in some patients (particularly those with MIS-C), we recommend performing a thorough cardiac evaluation including ECG, echocardiography and cardiac biomarkers (troponin, CK and CK MB, mixed venous oxygen saturation if central line is present) on all patients who present in shock to rule out cardiac involvement and mixed shock, even if presenting with a distributive picture.
  1. Fluid resuscitation: Within 1 hour of recognition, initiate fluid resuscitation:
  1. Use balanced/buffered crystalloids for fluid resuscitation rather than albumin or normal saline. Do not use hypotonic fluids, starches or gelatin.
  2. Fluid resuscitation may lead to volume overload and capillary leak, exacerbating respiratory failure. It is important to discontinue fluid administration if the patient is not responding or benefiting, or if risks of precipitating respiratory failure outweigh marginal benefits of ongoing fluids. In these cases, vasoactive medications may be preferable.
  1. In healthcare systems with intensive care capacity including ventilatory support, administer bolus fluids, 10–20 mL/kg per bolus, up to 40–60 mL/kg, over the first hour, titrated to clinical markers of perfusion Improved tachycardia, improved blood pressure, capillary refill time, level of consciousness, and urine output.
  1. Discontinue if signs of fluid overload develop or respiratory consequences outweigh benefits.Jugular venous distension, pulmonary edema, or new or worsening hepatomegaly
  1. In healthcare systems without intensive care capacity:
  1. If the child has a normal blood pressure for age, initiate maintenance fluids and do not administer fluid boluses
  2. If the child has hypotension, administer bolus fluids, 10–20 mL/kg per bolus, up to 40 mL/kg over the first hour with titration as above
  1. Vasoactive medications: Administer vasoactive medications if signs of fluid overload are apparent, or signs of shock (listed above) persist after two fluid boluses. Monitor blood pressure frequently and titrate the vasoactive medication to the minimum dose necessary to maintain perfusion and prevent side-effects.
  1. Initiate either epinephrine or norepinephrine, ideally through a central venous catheter. Diluted vasoactive solutions can be initiated through a peripheral intravenous catheter if central access is not available.
  1. Dopamine can be used in places where epinephrine and norepinephrine are not available.
  2. For children requiring high doses of catecholamines (definitions vary between institutions), consider adding vasopressin if available
  3. Inodilators (milrinone, dobutamine or levosimendan) should not be used routinely, and are typically not used for septic shock in the absence of evidence for cardiac dysfunction. They can worsen peripheral blood pressure and thus should only be considered in cases of refractory hypoperfusion and evidence of cardiac dysfunction by practitioners familiar with their use.
  1. Corticosteroids: There is insufficient evidence to recommend for or against glucocorticoids to treat refractory shock in children with SARS-CoV-2. However, corticosteroids may be indicated for the treatment of SARS-CoV-2 infection.

Tool: See Surviving Sepsis Campaign International Guidelines for more detailed recommendations. Surviving Sepsis Campaign: Initial Resuscitation Algorithm for Children (Bundle)

Cardiogenic ShockCopy Link!

Cardiogenic shock can occur with SARS-CoV-2 infection in the setting of sepsis or MIS-C. Cardiogenic shock is characterized by circulatory failure due to impaired of cardiac contractility.

Signs and symptoms of cardiogenic shock in children may include any combination of the following:

  • Shortness of breath
  • Jugular venous distention
  • Hepatomegaly
  • Peripheral edema
  • Pulmonary crackles
  • Altered mental status (e.g., lethargy, confusion)
  • Decreased perfusion
  • Tachycardia
  • Decreased urine output
  • Cardiac murmur
  • Gallop
  • Decreased peripheral pulses
  • Arrhythmia
  • Hypotension
  • Cardiomegaly and/or pulmonary edema on chest x-ray

The immediate goal when managing any type of pediatric shock, including cardiogenic shock, is to maintain perfusion and restore oxygen delivery to the organs. The recommendations below pertain to pure cardiogenic shock in the absence of congenital heart disease. Additionally, undifferentiated shock can occur (e.g., septic shock with myocardial dysfunction) and clinical judgement and frequent reassessment should be used to determine appropriate therapy.

  1. Optimize cardiac output
  1. Fluid resuscitation with balanced/buffered crystalloids is indicated in patients with cardiogenic shock only after clinical assessment demonstrating preload insufficiency, ideally by echocardiogram. If indicated, use small volume boluses (5-10ml/kg) over 30-60 min and assess frequently for clinical response and signs of fluid overload and respiratory compromise.
  2. In children with fluid overload, ventricular dysfunction and adequate blood pressure (with or without the use of vasoactives), consider the use of diuretics (e.g., furosemide infusion) to achieve euvolemia.
  3. In patients without hypotension, dobutamine or milrinone can be administered to decrease afterload and improve cardiac output.
  4. In patients with hypotension, epinephrine can be administered to improve inotropy. Be aware that epinephrine doses > 0.05 µg/kg/min can increase afterload.
  1. Treat reversible causes such as electrolyte abnormalities, arrhythmias, pulmonary embolism, pneumothorax, tamponade, sepsis.
  1. If sepsis is suspected, initiate empiric, broad-spectrum antimicrobial therapy.
  1. Optimize ventilation/gas exchange:
  1. Provide oxygen therapy as above with a goal SpO2 >94%
  2. If either noninvasive or invasive ventilation are clinically indicated, be aware of the cardio-pulmonary interactions and prepare for the possibility of cardiac arrest on intubation.
  1. Monitoring and Targets: Early goal-directed therapy should be based on clinical (volume status, urine output, blood pressure, mental status) and laboratory (end organ function [blood urea, creatinine, transaminases], blood pH, lactate levels, BNP/NT-proBNP, troponin) measurements as well as echocardiogram.

Other ICU ManagementCopy Link!

  1. Renal Replacement Therapy: For children with fluid overload or renal dysfunction who are unresponsive to diuretic therapy, consider renal replacement therapy.
  2. Nutrition: After adequate resuscitation (e.g., no longer requiring escalating doses or in the process of weaning vasoactive medications), initiate enteral nutrition for children with no contraindications. Parenteral nutrition need not be initiated in the first 7 days of admission.
  3. Transfusions: Do not routinely transfuse hemodynamically stable children with a blood hemoglobin concentration ≥ 7 g/dL.

Cardiac ArrestCopy Link!

Updated Date: February 1, 2021

The American Heart Association (AHA), in collaboration with the American Academy of Pediatrics (AAP) among other groups, compiled interim guidance to help rescuers treat pediatric and neonatal victims of cardiac arrest with suspected or confirmed SARS-CoV-2 infection (Topjian et al). Some guidance specifically related to children and neonates include:

  • For out-of-hospital cardiac arrest: lay rescuers should perform chest compressions and consider mouth-to-mouth ventilation, if willing and able, given the higher incidence of respiratory arrest in children. A face mask or cloth covering the mouth and nose of the rescuer and/or patient may reduce the risk of transmission if the rescuer is unwilling to perform direct mouth-to-mouth ventilation.
  • For an in-hospital cardiac arrest in a patient who is intubated: consider adjusting respiratory rate to 10 breaths/min for pediatric patients and 30 breaths/min for neonates.

Neonatal ResuscitationCopy Link!

For neonates born to mothers with confirmed or suspected SARS-CoV-2 infection, suction of the airway after delivery should not be performed for clear or meconium-stained amniotic fluid since suctioning is an aerosol-generating procedure (AGP). Since endotracheal installation of medications such as epinephrine is also aerosol-generating, intravenous delivery via a low-lying umbilical venous catheter is the preferred route of administration during neonatal resuscitation.

Multisystem Inflammatory Syndrome in ChildrenCopy Link!

Updated Date: January 24, 2021
Literature Review:
Gallery View, Grid View

Multisystem Inflammatory Syndrome in Children (MIS-C), also known as Pediatric Inflammatory Multi-System Syndrome Temporally Associated with SARS-CoV-2 (PIMS-TS or PIMS), is a rare manifestation of SARS-CoV-2 that has been described in children and young adults in multiple case series (Panupattanapong et al, Riphagen et al, Verdoni et al, Toubiana et al, Pouletty et al, Jones et al).

PathophysiologyCopy Link!

The pathophysiology of MIS-C is not yet well understood, but is likely related to immune dysregulation.

Innate immune mechanisms:

  1. One possible contributor is an innate immune response called neutrophil extracellular traps (NETs), which are webs of cell-free DNA, histones, and neutrophil granule content (Jiang et al). An overabundance of NETs, or NETosis, can cause an exaggerated systemic inflammatory response (Mozzini et al) and promote thrombosis (Martinod et al). NETs have been shown to be elevated in the plasma of patients infected with SARS-CoV-2, and higher concentrations are seen in patients with respiratory failure (Zuo et al).
  2. A dysregulated innate immune response and a subsequent cytokine storm (Cytokine Storm Syndrome) with endothelial damage may also contribute to clinical manifestations of severe SARS-CoV-2 infection (Jiang et al; Liu et al; Varga et al) and development of MIS-C (Consiglio et al, Gruber et al).

Acquired immune mechanisms:

  1. Antibody or T-cell recognition of self-antigens (viral mimicry of the host) resulting in autoantibodies (Gruber et al)
  2. Antibody or T-cell recognition of viral antigens expressed on infected cells (Waggoner et al)
  3. Formation of immune complexes which activate inflammation (Hoepel et al)
  4. Viral superantigen sequences which activate host immune cells (Cheng et al)

Clinical PresentationCopy Link!

SymptomsCopy Link!

Presenting Symptoms: Many children with MIS-C do not exhibit respiratory symptoms at any point in their course. Common symptoms include:

  • Fever
  • Gastrointestinal (GI) symptoms including abdominal pain, nausea, vomiting, and non-blood diarrhea (87%, Abrams et al). In some cases, GI symptoms occurred 1-2 weeks prior to presentation, and may represent the period of acute infection.
  • Dermatologic manifestations such as rash and malar erythema (73%, Abrams et al) that develops a mean of 2.7 days after onset of fever and lasts for a median of 5 days. Mucocutaneous features can be an important clue in recognizing MIS-C, but does not correlate with disease severity (Young et al).
  • Conjunctival Injection
  • Periorbital Edema and/or Distal Extremity Edema
  • Strawberry Tongue

Timing: MIS-C appears to be a late (i.e. post-viral) manifestation SARS-CoV-2 infection, as many patients presented 2-3 weeks after the peak of infection in their geographic area (Panupattanapong et al and Jamal et al).

Differential DiagnosisCopy Link!

MIS-C has manifestations that overlap with Kawasaki Disease (KD), Toxic Shock Syndrome, and Macrophage Activation Syndromes like Cytokine Storm. All of these can occur in a post-acute illness setting and involve fever, rash, erythema, edema, conjunctivitis, and oral mucosal changes (e.g. “strawberry tongue”). Severe disease can also cause multi-organ failure. However, there are key differences:

  • Age: KD tends to occur in very young children with a mean age of 2 years but almost always < 5 years old, whereas the average age of MIS-C patients is 7-9 years old (Whittaker et al; Feldstein et al; Jiang et al)
  • Race: MIS-C is seen in a higher proportion of children of Hispanic, African and Afro-Caribbean descent and a lower proportion in those of East Asian descent compared with KD (Whittaker et al; Feldstein et al).
  • Triggering event: MIS-C is associated with SARS-CoV-2 infection. TSS is associated with Staphylococcus and Group A Strep infection (risk factors include recent tampon use, surgery or infection, especially skin or soft tissue). KD is triggered by many different infections (Whittaker et al; Jiang et al).
  • Laboratory findings: MIS-C patients generally have a greater elevation of inflammatory markers such as CRP, IL-6 and fibrinogen than patients with KD or TSS (Whittaker et al; Jiang et al).
  • Organ involvement: MIS-C has more diffuse cardiovascular involvement than KD, which has a predilection for the coronary arteries (Whittaker et al; Panupattanapong et al).

ComplicationsCopy Link!

  • Cardiac: In one study, 71% had cardiovascular complications including coronary artery aneurysms, myocardial dysfunction, pericarditis, valvulitis, or coronary dysfunction (Abrams et al). Other common cardiac complications include: arrhythmias, pericardial effusion, coronary artery dilatation, and reduced LVEF potentially leading to cardiogenic shock (Valverde et al). Another study that analyzed echocardiographic findings in 28 children with MIS-C found that unlike in KD, coronary arteries may be spared in early MIS-C; however, myocardial injury is common. After approximately 5 days, children demonstrated good recovery of systolic function, but diastolic dysfunction persisted (Matsubara et al). Medium- and long-term sequelae, particularly cardiovascular complications, are not yet known (Alsaeid et al).
  • Renal: In one study, 41% had AKI (27.6% severe) (Deep et al).
  • Hematologic: In another study, 4% had coagulopathy and thrombus (Davies et al).

Workup and MonitoringCopy Link!

Case Definition: The case definition for MIS-C from the CDC and WHO requires:

  1. FeverCopy Link!

  2. Clinical signs of multi-system involvementCopy Link!

  3. Laboratory signs of inflammationCopy Link!

  4. No other plausible diagnosis such as bacterial sepsis or staphylococcal/streptococcal TSSCopy Link!

  5. Evidence of SARS-CoV-2 infection by PCR, antigen test or serology, or exposure to a SARS-CoV-2 infected personCopy Link!

Diagnosis may be particularly difficult in places that do not have easy access to testing, so clinicians should maintain a high level of suspicion for MIS-C and potentially treat patients for multiple potential etiologies.Copy Link!

SARS-CoV-2 Testing: Because of the late timing, not all patients are positive via PCR, and some may not yet be positive on serology. Pooled data shows that 60-80% of patients have positive SARS-CoV-2 serology with a smaller number positive via PCR. Up to 5-10% are negative on both tests (Feldstein et al; Jiang et al).

  • Children with MIS-C have significantly higher SARS-CoV-2 receptor binding domain (RBD) IgG antibody titers than children with KD or SARS-CoV-2 without MIS-C. RBD IgG titers also correlate with ESR and with hospital and ICU lengths of stay, suggesting that quantitative SARS-CoV-2 serology may help to distinguish MIS-C from similar clinical entities, and help to stratify risk for adverse outcomes (Rostad et al).

Laboratory evaluation: Laboratories should be followed as outlined for Cytokine Storm.

Imaging and Procedures:

Cardiac evaluation: Suspected cardiac involvement should be evaluated via ECG and echocardiogram (Kache et al; ACR).

  1. ECGs should be performed at a minimum every 48 hours in MIS-C patients who are hospitalized, and during follow-up visits
  1. In a single-center cohort study of 32 patients with MIS-C, 6 developed atrioventricular block a median of 8 days after initial symptom onset suggesting that patients admitted with MIS-C require close ECG monitoring during the acute phase (Choi et al).
  1. Echocardiogram should be conducted at diagnosis and at a minimum of 7-14 days and 4-6 weeks after presentation.
  2. Patients should be followed on telemetry per consultation with a cardiology specialist.

Other evaluation: A child under investigation for MIS-C should be evaluated for other infectious etiologies (i.e. Septic Shock, meningitis) and non-infectious etiologies (i.e. oncological, rheumatological, Cardiogenic Shock) that may have similar presentations.

ManagementCopy Link!

Tool: Management Guidelines for MIS-C (American College of Rheumatology Multidisciplinary Task Force)

General ManagementCopy Link!
  • Stabilize hemodynamics and Shock; judicious fluid resuscitation is recommended given the high incidence of cardiac involvement and the cardiogenic etiology of shock (see Undifferentiated Shock (describes how to tell them apart) and Septic and Cardiogenic Shock).
  • Consider transferring the patient to a care facility where subspecialty care and IVIG are available if these are not available at your institution.
  • Involve the pediatric ICU team, and infectious disease, cardiology, rheumatology, and hematology services, if available.
ImmunomodulatorsCopy Link!

Most of the treatment of MIS-C with immunomodulators is taken from evidence in treating Kawasaki Disease (KD). There is a paucity of evidence supporting the use of immunomodulators for patients with mild cases of MIS-C, and there is no evidence that use of immunomodulators can prevent coronary artery aneurysms or cardiac involvement. However, the American College of Rheumatology (ACR) Multidisciplinary Task Force strongly recommends use of immunomodulators in patients hospitalized with MIS-C. While there is no set treatment protocol, the following is recommended by the ACR and several pediatric institutions:

Mild to Moderate Disease

Patients with suspected MIS-C should be evaluated much as a patient with KD would be. For MIS-C patients meeting criteria for complete or incomplete KD without shock, myocardial dysfunction or coronary artery changes:

  1. Initiate high dose IVIG at 2 gm/kg based on ideal body weight up to 100gm for 1 dose, preferably in the first 7-10 days of illness (McCrindle). See MIS-C Medication Dosing.
  1. Consider monitoring patients with any myocardial dysfunction in an ICU setting during IVIG infusion
  1. Methylprednisolone 2 mg/kg/day for 2 weeks followed by a taper over 2-3 weeks (this is generally higher than doses used to treat hypoxemia from SARS-CoV-2 infection). See MIS-C Medication Dosing.
  1. Most evidence supporting use of corticosteroids in MIS-C is limited to case series in which ~50-60% of patients were treated with corticosteroids at varying doses with most patients responding rapidly (Feldstein et al; Dufort et al; Godfred-Cato et al; Kaushik et al).
  2. A retrospective cohort study of 111 patients showed treatment with IVIG and methylprednisolone vs. IVIG alone was associated with lower risk of treatment failure, lower risk of escalation to second-line therapy, lower risk of requiring hemodynamic support, lower risk of acute left ventricular dysfunction after initial therapy, and lower duration of stay in a pediatric ICU (Ouldali et al).
  3. Currently, corticosteroids are suggested when patients meet criteria for complete or incomplete KD with a risk factor for IVIG resistance (i.e. coronary artery enlargement, age <12 months) (McCrindle et al) or persistent fevers or rising inflammatory markers despite treatment with IVIG, which may suggest MAS or Cytokine Release Syndrome.
  4. If no IVIG is available, use methylprednisolone (or equivalent, see MIS-C Medication Dosing) alone.
  5. Consider adding a Proton Pump Inhibitor (PPI) for GI prophylaxis.
  6. Start low dose Aspirin. See MIS-C Medication Dosing.
  1. If refractory (i.e. continued fever >36 hours after IVIG, worsening clinical condition, new cardiac dysfunction or shock), consider biologics in consultation with a rheumatology and infectious disease specialist.

Severe Disease

For patients with signs of shock, coronary artery dilation, arrhythmia, or cardiac dysfunction even in the absence of Kawasaki-like features:

  1. Initiate high dose IVIG at 2 gm/kg based on ideal body weight up to 100gm for 1 dose
  2. Concomitant high dose methylprednisolone 30 mg/kg (up to 1000mg) daily for 1-3 days followed by 2mg/kg divided q8-q12. Continue high dose for 2 weeks (can consolidate to daily) then taper over 2-3 weeks.
  1. If no IVIG is available, use corticosteroids alone.
  2. Consider adding a PPI for GI prophylaxis.
  1. Discuss use of biological medications (e.g. Anakinra, Infliximab, Tocilzumab - see dosing table below) in consultation with a rheumatology specialist.
  2. Start low dose Aspirin; discuss high dose Aspirin with a cardiology specialist if there are coronary changes. See MIS-C Medication Dosing.
Antiplatelet and anticoagulation therapyCopy Link!
  1. Give low dose Aspirin (3-5 mg/kg/day; max 81 mg/day) until normalization of platelet count and confirmed normal coronary arteries by echocardiogram at >4 weeks after diagnosis in consultation with a cardiology specialist. If coronary artery aneurysm is identified, low dose Aspirin should be continued with possible therapeutic anticoagulation in consultation with a cardiology specialist.
  2. Additional anticoagulation or antiplatelet therapy may be recommended for patients with large coronary aneurysms, documented thrombosis, or reduced ejection fraction in consultation with a cardiology specialist.
  3. Anticoagulant thromboprophylaxis is recommended for hospitalized children with MIS-C per the clinical recommendations outlined in the VTE prophylaxis section below (Goldenberg et al).
MIS-C Medication Dosing TableCopy Link!

Medication

Dosing

Notes

Aspirin

Low dose (antiplatelet): 3-5mg/kg/dose once daily

High Dose (anti-inflammatory): 20-25 mg/kg/dose every 6 hours

Round Aspirin dose to nearest ½ 81 mg tablet size

IVIG

2gm/kg/dose IV (max 100 gm) for 1 dose

Retreatment may be considered in case of refractory disease (continued fever > 36 hours or worsening clinical condition)

Methylprednisolone

Low dose: 2mg/kg/day for 2 weeks followed by taper for over 2-3 weeks

High dose: 30mg/kg/day (max 1000mg/day) for 1-3 days followed by 2mg/kg/day divided q8-q12. Continue high dose for 2 weeks (can consolidate to daily) then taper over 2-3 weeks

Consider adding a PPI for patients receiving steroids + Aspirin to decrease risk for GI bleed

Anakinra

2-4mg/kg/dose (max 100mg/dose) SQ twice daily (may increase to 3 times daily) for 3 days

Infliximab

10mg/kg/dose IV once

Tocilizumab

<30kg: 12mg/kg IV once

>30kg: 8mg/kg IV once; Max 800mg

Post MIS-C CourseCopy Link!

Updated Date: June 1, 2021

In the acute phase of MIS-C, there is a comparable incidence of coronary artery aneurysm and dilation in MIS-C and Kawasaki disease (20 and 25%, respectively) (Henderson et al). that resolve in most patients in mid- and long-term follow up. Neurological complications such as headache, muscle weakness, reduced reflexes, altered mental status, encephalopathy, cranial nerve palsies, stroke, and seizure appear more frequent than in Kawasaki disease (Schupper et al, Jin et al) with persistence following the acute phase.

In a pooled meta-analysis of follow up MIS-C studies, recovery was reported in 91.1% and death in 3.5% of patients (Jiang et al). In a 6 month follow up study (N=45), systemic inflammation was resolved in all but one patient with 96% of patients with normal echocardiograms, gastrointestinal symptoms decreased by 75% and all renal, hematological and otolaryngological findings largely resolved. Minor neurological abnormalities persisted in 39% of patients compared to 52% in cohort at diagnosis with ~20% with severe emotional difficulties and 50% with persisting poor exercise tolerance. Ongoing studies will help define the extended natural history of MIS-C.(Penner et al)

TherapeuticsCopy Link!

Tool: COVID Drug Interaction Tracker

Symptomatic TreatmentsCopy Link!

Updated Date: Jan 22, 2021

CoughCopy Link!

Non-Pharmacologic Therapy

  1. Drink fluids (preferably warm)
  2. Honey (2.5-5mL [0.5-1 teaspoon]) given straight or diluted in liquid may help ease coughing symptoms and improve sleep for children >12 months old (Cohen et al). Using honey for infants aged <12 months is not advised due to risk of botulism.
  3. Cough drops may be used for school-aged children and adolescents; however, there is a risk of aspiration with use in younger children.

Pharmacologic Therapy

  1. The WHO recommends against the use of codeine preparations for cough in children. However, dextromethorphan-containing cough medications may be warranted in the unusual circumstance where severe prolonged cough interferes with feeding or sleeping (WHO).

Nasal Secretions / SuctioningCopy Link!

  1. There is currently no specific guidance on nasal suctioning in SARS-CoV-2.
  2. Extrapolating from literature on children hospitalized with bronchiolitis, saline nasal drops and mechanical aspiration of the nares can relieve nasal obstruction and decrease length of hospital stay (Mussman et al).
  3. There is insufficient evidence to support frequent “deep” suctioning of the oropharynx or larynx with a nasopharyngeal catheter (Ralston et al).

DyspneaCopy Link!

  1. Appropriate Oxygen therapy in conjunction with creating a calm and comforting environment is the mainstay of management of dyspnea in most pediatric patients (Mussman et al). See Non-opioid Management.
  1. Opioid Management (adult doses presented here) should only be used when survival is unlikely and treatment is focused solely on comfort and control of symptoms, or in cases of significant refractory dyspnea despite treatment.
  1. Respiratory Secretions can be managed in a manner similar to adults.

CorticosteroidsCopy Link!

Updated Date: Jan 22, 2021

  1. For respiratory indications: Corticosteroids have been shown to decrease mortality in adult patients with oxygen requirements (Review of Evidence in Adults) and are currently being studied through clinical trials in the pediatric population (WHO REACT Working Group). For MIS-C dosing, see MIS-C Medication Dosing.
  1. Low dose corticosteroids may be beneficial for select pediatric patients with severe or critical SARS-CoV-2 infection (i.e. requiring supplemental oxygen or mechanical ventilation) (Dulek et al).
  1. Dosing is per the chart below. Duration of therapy is up to 10 days or until discharge; shorter durations are preferable.
  1. Recommendations about the use of corticosteroids for children with oxygen requirements are uncertain due to underrepresentation of children in the clinical trials (WHO).
  1. Monitoring:
  1. Monitor glucose, WBC count, mental status, and blood pressure in adolescents.
  2. If the patient has other risk factors requiring initiation of stress ulcer prophylaxis, add famotidine or a PPI.

Pediatric Glucocorticoid DosingCopy Link!

Corticosteroid

Dose

Dexamethasone

0.15 mg/kg PO or IV daily (max dose 10mg)

Prednisolone

1 mg/kg PO daily (max dose 40mg)

Methylprednisolone

0.8 mg/kg IV daily (max dose 32mg)

Hydrocortisone

For neonates (<1 month of age): 0.5 mg/kg IV every 12 hours for 7 days followed by 0.5 mg/kg IV daily for 3 days

For children >1 month: 1.3 mg/kg IV every 8 hours (max dose 50mg)

Antibody TherapiesCopy Link!

Updated Date: January 24, 2021

Convalescent PlasmaCopy Link!

  1. In adults: The FDA has issued an Emergency Use Authorization (EUA) for use of convalescent plasma to treat patients hospitalized with SARS-CoV-2 infection. However, convalescent plasma is not routinely recommended due to insufficient data on safety and effectiveness. See Convalescent Plasma for further information.
  2. In children: There are no high quality studies investigating use of convalescent plasma in children and adolescents. A systematic review that included 8 case studies suggested a potential benefit. Currently there are ongoing clinical trials in pediatric patients that may help clarify whether convalescent plasma should be used for treatment in children (Zaffanello et al).
  1. The National Institute of Health (NIH) and Infectious Diseases Society of America Guidelines recommend use in hospitalized pediatric and adult patients in the setting of a clinical trial.

Monoclonal AntibodiesCopy Link!

  1. Outpatient: Outpatient treatment of SARS-CoV-2 in children with monoclonal antibodies is recommended only on a case-by-case basis, and ideally in the context of a clinical trial (Dulek et al). The FDA has issued an EUA for two investigational monoclonal antibody therapies - Bamlanivimab-Etesevimab (EUA) and Casirivimab-Imdevimab (EUA) - for treatment of non-hospitalized pediatric patients >12 years and >40 kg with mild to moderate SARS-CoV-2 who have certain risk factors for severe disease and/or hospitalization.
  1. Risk factors for children age 12-17 years include any of the following:
  1. BMI >85 percentile
  2. Sickle Cell Disease
  3. Congenital or acquired heart disease
  4. Neurodevelopmental disorders (e.g., Cerebral Palsy)
  5. Medical-related technological dependance (e.g., tracheostomy, gastrostomy)
  6. Chronic respiratory disease that requires daily medication for control (e.g. asthma)
  1. See Monoclonal Antibodies for further information on pharmacology, evidence for use in adults, dosing and monitoring and toxicity
  1. Inpatient: There is insufficient data to support the use of monoclonal antibodies in the inpatient setting. (In adults, use of monoclonal antibodies late in the disease course does not appear to be effective).

VTE prophylaxisCopy Link!

Updated Date: February 23, 2021

Thromboembolic Disease is a major complication of SARS-CoV-2 infection in adults. In general, children are at lower risk of thromboembolic disease, but the incidence of VTE in children with SARS-CoV-2 infection is uncertain. Pediatric intensivists, hematologists, and rheumatologist have published guidance on VTE prophylaxis (Goldenberg et al; Loi et al):

Outpatients:

  1. VTE prophylaxis is not recommended in the absence of indwelling central venous catheters and significant clinical risk factors for VTE (listed below), except for some post-discharge cases (see below).

Hospitalized patients:

  1. Lab monitoring: Obtain a CBC with platelet count, fibrinogen, prothrombin time (PT), partial thromboplastin time (PTT), fibrinogen, and D-dimer on admission and serially (schedule for suggested lab frequencies).
  1. Consider calculating a DIC score (ISTH scoring system) and evaluating for VTE when there is a rising D-Dimer and DIC score.
  2. Common findings include: Elevated D‐dimer, elevated fibrinogen, mildly decreased platelet count, evidence of DIC
  1. Indications: Anticoagulant thromboprophylaxis should not be routinely prescribed in hospitalized children who have asymptomatic SARS‐CoV‐2 infection in the absence of risk factors for hospital‐associated VTE (listed below). Hospitalized patients with SARS-CoV-2 infection in addition to the following risk factors for thrombosis and the absence of contraindications should be started on anticoagulant thromboprophylaxis (see dosing below) in combination with mechanical thromboprophylaxis with sequential compression devices:
  1. Personal or family history of VTE
  2. Presence of a central venous line
  3. Post-pubertal
  4. Decreased mobility from baseline
  5. Burns
  6. Active malignancy
  7. Evidence of venous stasis or cardiac low-flow state
  8. Estrogen therapy
  9. Systemic infection or flare of inflammatory disease
  10. Obesity (BMI>95th percentile)
  11. Severe dehydration
  12. Recent surgery or trauma
  13. Inherited thrombophilia (e.g., protein S, protein C, or antithrombin deficiency; factor V Leiden; factor II G20210A; persistent antiphospholipid antibodies)
  14. Sickle Cell Disease vaso-occlusive crisis
  15. Previous splenectomy for hemoglobinopathy
  16. Autoimmune disorder
  17. Nephrotic syndrome
  1. Contraindications: Thromboprophylaxis should be held in patients with active bleeding or with platelets < 20,000/uL or per provider discretion for procedures or risk of bleeding.
  1. Note: In the absence of other bleeding risk factors, low‐dose anticoagulant thromboprophylaxis is not believed to increase the risk of clinically significant bleeding in MIS‐C patients receiving Aspirin at doses ≤5 mg/kg/d.
  1. Duration: Prophylaxis is typically continued for the duration of hospitalization. Ongoing thromboprophylaxis following discharge may be considered for patients with additional risk factors for VTE such as persistently decreased mobility, active cancer, autoimmune disorders, recent surgery, or D-dimer > 2x upper limit of normal.
  1. If post-discharge prophylaxis is indicated, LMWH subcutaneously twice a day (as below) is recommended in children < age 18.
  1. DOACs (e.g, Rivaroxaban 10 mg po qd), can be used in young adults > age 18 as an alternative.
  2. Duration of post-discharge prophylaxis (whichever occurs first): clinical risk factor resolution or 30 days post-discharge

Summary of anticoagulant thromboprophylaxis recommendations in children hospitalized with asymptomatic SARS‐CoV‐2 infection and children hospitalized for SARS-CoV-2–related illness (Goldenberg et al):

Clinical scenario

D-dimer > 5X upper limit of normal

Non-SARS-CoV-2 VTE risk factors

Anticoagulant thrombophylaxis suggested

Hospitalized, asymptomatic

Not applicable

>3

Yes

Not applicable

<3

No

Hospitalized, symptomatic (including MIS-C)

Yes

Not applicable

Yes

No

>1

Yes

Prophylaxis dosing:

  1. Low-molecular weight heparin (LMWH; e.g., Enoxaparin) is used in patients who are clinically stable (e.g., without hemodynamic compromise, renal failure or significant risk of bleeding). LMWH Prophylactic Doses:
  1. Age < 2 months: 0.75 mg/kg bid SC
  2. Age > 2 months: 0.5 mg/kg bid SC
  3. Adjust dose to achieve a 4‐hour post‐dose anti‐Xa activity level of 0.2 to <0.5 units/mL
  1. Unfractionated heparin (UFH) is used in patients who are unstable (e.g., with hemodynamic compromise, renal failure, or high risk for bleeding). Patients who become unstable while on LMWH should be transitioned to UFH prophylactic dose:
  1. 10-15 units/kg/hour, with no loading dose. Heparin level should be 0.1-0.3 units/mL (equivalent to aPTT of 40-70 seconds).
  1. Note: aPTT at baseline may be elevated or low and may not correlate with heparin levels. In these cases, anti-Xa levels may be used.
  1. Direct oral anticoagulants (DOACs) (e.g., Rivaroxaban and Apixaban) are not recommended for inpatient VTE prophylaxis because of possible drug interactions with some medications used to treat SARS-CoV-2 infection (including Dexamethasone) and limited data in children with SARS-CoV-2. This COVID drug interaction tracker can help determine if there are relevant interactions.
  2. Antiplatelet agents are not recommended for VTE prophylaxis in patients with SARS-CoV-2 infection. However, antiplatelet agents may be indicated for patients with atypical KD or MIS-C per cardiology and/or rheumatology specialists.

Therapeutic dosing:

  1. Consider advancing dose to therapeutic-intensity anticoagulation (e.g., LMWH 1 mg/kg/dose q 12 hours for patients with normal renal function) in patients considered very high risk for VTE/microvascular thrombosis
  1. Very high-risk patients include: those receiving anticoagulation therapy prior to admission; those with a highly suspected or diagnosed VTE; those with high levels of D-dimer; those with abnormal coagulation parameters including prolonged PT, prolonged aPTT, or decreased fibrinogen; those with markedly elevated inflammatory markers; and/or those with multi-organ failure (Loi et al).

RemdesivirCopy Link!

Updated Date: February 23, 2021

Remdesivir has been FDA-approved for children ≥12 years old and ≥40 kg with confirmed SARS-CoV-2 infection requiring hospitalization. Remdesivir may also be available via Emergency Use Authorization (EUA) for pediatric patients <12 years old (≥3.5 kg) or <40 kg. Use of non-FDA approved Remdesivir under the EUA requires additional documentation and procedures. The recommended dosing for children <40 kg is 5 mg/kg IV loading dose on day 1 followed by 2.5 mg/kg IV q24h for 4 additional days (Garcia-Prats et al). Pharmacokinetic modeling and simulation has been used to extrapolate pediatric-specific dosing regimens for use of Remdesivir to treat SARS-CoV-2 infection; the dosing scheme provides weight-normalized dosages for patients weighing <60 kg (Maharaj et al).

TocilizumabCopy Link!

Updated Date: February 23, 2021

Tocilizumab is FDA-approved to treat cytokine storm in children 2 years of age and older, but its use in SARS-CoV-2 is highly debated (see Tocilizumab for a review of the literature in adults). As of December, 2020 it is not recommended by the American College of Rheumatology for SARS-CoV-2 infection in pediatric patients (Henderson et al.), though it is used in some cases of MIS-C.

  1. Though not specifically studied in pediatric patients with SARS-CoV-2 infection, studies of cytokine storm in other clinical settings showed that doses of 6.9 to 12 mg/kg were pharmacologically active and resulted in appropriate concentrations.
  2. There is an increased risk of developing TB while taking Tocilizumab, which may be a concern in areas where prevalence of TB is high (Garcia-Prats et al).

AntibioticsCopy Link!

Updated Date: February 23, 2021

A discussion on the risks and benefits of empiric antibiotics with recommendations of initial antibiotics is covered here: Bacterial Infections. Antibiotics in SARS-CoV-2 infection are indicated for suspected or confirmed bacterial co-infection or secondary infection, which appears to be infrequent (Rawson et al; Vaughn et al). However, empiric antimicrobials may still be indicated at least temporarily during workup for indications such as shock or in situations where there is a high risk of not treating empirically. The recommended initial regimen for community acquired pneumonia is Ampicillin if the patient is unable to take oral medications, or high-dose Amoxicillin if able to take oral medications with the addition of Azithromycin in school-aged and adolescent patients (Bradley et al) (IDSA Guidelines).

Chapter 13

Specialty Care

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

CardiologyCopy Link!

Updated Date: December 11, 2020
Literature Review:
Gallery View, Grid View

Acute IllnessesCopy Link!

Acute Cardiac InjuryCopy Link!

Literature Review (Myocardial Injury): Gallery View, Grid View

Acute cardiac injury, variably defined as increased circulating troponin levels and/or new abnormalities on ECG or echocardiography, were noted in 7-22% of hospitalized patients in early reports from Wuhan (Ruan et al; Wang et al; Chen et al; Shi et al; Guo et al, Zhou et al). When present, these findings were associated with increased risk of ICU admission and death.

Several mechanisms for have been proposed (Ruan et al; Hu et al; Zeng et al; Inciardi et al), including:

  • Direct SARS-CoV-2 infection of cardiac myocytes (myocarditis)
  • Demand ischemia, with either large or small vessel thrombosis
  • Stress (Takotsubo) cardiomyopathy
  • Pathological myocardial response to inflammation or cytokine storm

Specific cardiac pathologies include myocarditis, arrhythmia, and precipitation of an acute coronary syndrome. Hypercoagulability in COVID-19, including its impact on the heart, are discussed in Hematology.

MyocarditisCopy Link!

Literature Review: Gallery View, Grid View

Fulminant SARS-CoV-2 myocarditis was clinically suspected in some early case reports (Ruan et al; Zeng et al; Hu et al; Inciardi et al), based on pre-existing clinical criteria and, in some cases, suggestive findings on cardiac MRI (Inciardi et al, Kim et al). Subsequent examination of myocardial tissue in autopsy series (Fox et al; Elsoukkary et al; Basso et al) found direct evidence for viral myocarditis (e.g., lymphocytic infiltrates) were relatively rare (0-14%).

The clinical significance of direct SARS-CoV-2 myocarditis remains unclear. If a patient has elevated troponins with no evidence of obstructive coronary artery disease, it may be on the differential diagnosis but is unlikely to alter management.

  • Provide supportive care for heart failure (Zhang et al) or Cardiogenic Shock
  • Where possible, discuss with cardiology and/or infectious disease consultants to see if the patient might benefit from antivirals or steroids (benefit is unknown)
  • Endomyocardial biopsy is unlikely to be informative.
  • See Advanced CV Imaging below regarding uses of cardiac MRI.

ArrhythmiaCopy Link!

Literature Review: Gallery View, Grid View

Cardiac arrhythmias can occur in COVID-19. An early case series of 138 patients in Wuhan, China, (Wang et al) found evidence of some arrhythmias in 17% of hospitalized patients with COVID-19, rising to 44% in those transferred to the ICU. Another early study of 189 hospitalized patients noted nearly 6% of inpatients had an episode of ventricular fibrillation or sustained ventricular tachycardia (Guo et al).

  1. Atrial Fibrillation/Atrial Flutter
  1. Consider beta-blockers, if no evidence of heart failure or shock.
  2. If acute heart failure or concern for hypotension, use amiodarone if not otherwise contraindicated.
  3. If unstable (with a pulse), synchronized DC cardioversion with 200 joules (biphasic).
  1. Ventricular Tachycardia
  1. If unstable or without palpable pulses: initiate local advanced life support protocol (e.g., ACLS).
  2. If stable:
  1. Involve a cardiologist. If cardiologist is not available, involve a senior clinician.Consider a single IV dose of amiodarone 150mg or lidocaine 100mg

Acute Coronary SyndromesCopy Link!

Literature Review: Gallery View, Grid View

Myocardial infarction in COVID-19 may be triggered by a combination of hypercoagulability, cardiac expression of SARS-CoV-2 entry receptor ACE2, possible direct viral myocardial injury, increased myocardial demand, or toxicity from inflammation. Cardiac markers and ECG changes alone may not be able to determine whether an underlying obstructive lesion exists as evidenced by the fact that up to 45% of hospitalized COVID patients have elevated cardiac markers (Lombardi et al).

The diagnosis of an acute coronary syndrome depends on:

  • Symptoms (if able to communicate): worsening shortness of breath, chest pain, or other anginal equivalents
  • Regional changes in the ECG or wall motion abnormalities on echocardiography
  • Rate of change of troponin changes (rapid rise or fall suggests an acute event)

Tool: Life in the Fast Lane Acute Coronary Syndromes

When the diagnosis is not clear, a cardiologist should be consulted.

If a patient is diagnosed with ACS, management should be coordinated with a cardiologist if at all possible. Medical management typically includes:

  • Treatment with full dose aspirin, clopidogrel (if not bleeding), heparin, oxygen (if hypoxemic), high-dose statin, nitrates (if hypertensive), and opioids as needed for symptom control. Beta blockers should be used with caution, given the risk of concomitant myocarditis or decompensated heart failure
  • If cardiac catheterization is available, there are no fundamental contraindications for patients with COVID-19 as long as strict infection control precautions are followed.
  • If cardiac catheterization is not available or if constrained resources unacceptably prolong door-to-balloon time, thrombolytic medications may be considered in lieu of PCI.

Advanced Cardiac DiagnosticsCopy Link!

Literature Review (Echocardiography and Cardiac MRI): Gallery View, Grid View
Literature Review (Angiography):
Gallery View, Grid View

Laboratory markers of cardiac injury (troponins, CK-MB, BNP) and electrocardiography are appropriate for most COVID-19 patients admitted to the hospital. Depending on availability and expertise, point-of-care ultrasound can also be considered, particularly in patients with concerning symptoms, lab values or ECG.

The indications for more advanced cardiac diagnostics are similar to patients without COVID-19, but with additional consideration for infection control. Testing should be limited to cases where the results will alter management to avoid unnecessary risk to providers and other patients.

  • Transthoracic echocardiography
  • Do not obtain routinely.
  • When possible, a bedside provider should assess cardiac function with point-of-care ultrasound for the following indications:
  • Marked elevation in troponin or NTproBNP, or decline in ScvO2/MvO2
  • Shock
  • New heart failure
  • New persistent arrhythmia
  • Significant ECG changes
  • If abnormalities are identified on point-of-care ultrasound (such as a new decrease in LV ejection fraction to below < 50%) and the patient is stable, a formal echocardiogram should be obtained if possible.
  • Evaluate both left and right ventricular function.
  • The differential diagnosis for right ventricular dysfunction includes myocarditis, hypoxic vasoconstriction, pulmonary embolus, and cytokine mediated dysfunction.
  • The differential diagnosis for left ventricular dysfunction includes myocarditis, acute coronary syndrome, and stress-induced cardiomyopathy.
  • Regional wall motion abnormalities with elevated troponins suggest an acute coronary syndrome, though direct myocardial injury by the virus can also result in focal wall motion abnormalities.
  • Stress Testing:
  • Should not be commonly required in patients with active COVID. If needed (and if available), consider pharmacologic nuclear stress testing or coronary CT angiography rather than exercise stress test.
  • Transesophageal Echocardiogram (TEE)
  • Only request if absolutely necessary. Although unclear whether this generates aerosolized virus, it likely does represent increased risk to the patient and provider.
  • Consider alternative noninvasive imaging modalities (such as cardiac CT or PET/CT) if they are available and appropriate for the question being asked.
  • Cardiac CT
  • Can consider for selected patients with elevated cardiac biomarkers when there is a need to distinguish myocardial injury from acute coronary syndrome. The decision to use CT in this context should be discussed with a cardiologist.
  • Consider in selected patients as a substitute for TEE to rule out left atrial appendage clot or to evaluate for endocarditis.
  • In appropriate cases, multiphase data acquisition may be used to evaluate both left and right ventricular function while concurrently evaluating the lung parenchyma or pulmonary artery.
  • Cardiac MRI
  • Consider for selected patients with elevated cardiac biomarkers and concern for myocarditis, if this information will impact patient management.
  • In the acute and subacute periods, T1 and T2 mapping as well as assessment of extracellular volume fraction (ECV) may improve sensitivity for myocarditis. However, the prognostic significance of such abnormalities (especially in the presence of normal ventricular function or when late enhancement abnormalities are absent) remains unclear.
  • In selected patients who have recovered from COVID, cardiac MRI using late gadolinium enhancement may be useful for evaluating for residual scar tissue.
  • Nuclear Imaging
  • In COVID-19 patients who require stress testing, vasodilator stress testing is preferred over exercise testing.
  • In selected patients who have recovered from COVID-19, PET MPI using a quantitative assessment of myocardial blood flow may be useful for evaluating microvascular dysfunction.

Chronic ConditionsCopy Link!

Preexisting cardiovascular disease and metabolic disorders (including diabetes and hyperlipidemia) worsen prognosis in acute COVID-19 (Izcovich et al).

AnticoagulationCopy Link!

See Anticoagulation in the Hematology section below.

Heart FailureCopy Link!

Literature Review (Heart Failure):Gallery View, Grid View
Literature Review (Structural Heart Disease):
Gallery View, Grid View

Patients with pre-existing heart failure have a nearly two-fold increased mortality and over three-fold greater risk of mechanical ventilation when they develop COVID-19 (Alvarez-Garcia et al).

There are currently no specific medication changes recommended for patients with prior heart failure who develop COVID-19, though all medications should be titrated based on other clinical context (e.g., in a patient with fevers and decreased oral intake, home diuretic doses may need adjustment).

Patients with rheumatic heart disease or other conditions requiring surgical intervention should continue to be considered as a potential priority case during the COVID-19 pandemic.

HypertensionCopy Link!

Literature Review: Gallery View, Grid View

Patients with pre-existing hypertension have a significantly increased risk of developing severe COVID-19 disease or dying (Izcovich et al).

Antihypertensive medications, such as RAAS inhibitors (e.g. ACE inhibitors) were initially suspected to be harmful in COVID-19, but these harms have not been supported by subsequent data (e.g., Baral et al). Other classes, such as calcium channel blockers, were thought to possibly be beneficial, but this also remains unsubstantiated.

Unless new data become available, patients with well-controlled hypertension should continue their current anti-hypertensive medications, unless those drugs need to be stopped for other reasons (e.g. renal issues).

Heart TransplantCopy Link!

Literature Review: Gallery View, Grid View

The initial protocol used by Brigham and Women’s Hospital, USA, for patients with heart transplant who develop COVID-19 is available here.

EndocrinologyCopy Link!

Updated Date: December 11, 2020
Literature Review:
Gallery View, Grid View

Acute IllnessesCopy Link!

HyperglycemiaCopy Link!

Literature Review (Hyperglycemia): Gallery View, Grid View
Literature Review (Diabetic Ketoacidosis):
Gallery View, Grid View

COVID-19 can cause hyperglycemic crisis in patients with and without known insulin resistance (summarized in Rubino et al). A systematic review of 110 reported cases of diabetic ketoacidosis (DKA) or combined DKA/hyperglycemic hyperosmolar state (HHS), found that 10% of patients with these severe presentations did not have a prior diagnosis of diabetes (Pal et al).

All patients with moderate to severe COVID-19 should thus be evaluated for hyperglycemia. Patients who do have blood glucose levels over 10 mmol/L (180 mg/dl) are generally managed with insulin rather than oral agents. Targets are similar to the management in patients without COVID-19, while recognizing that insulin requirements may be labile.

Treatment of DKA and/or HHS also has the same goals as patients without COVID-19, while recognizing provider safety concerns and limited resources in a pandemic. An example protocol for mild-to-moderate DKA at Brigham and Women’s Hospital, USA, uses subcutaneous insulin with slightly less frequent monitoring than standard protocols, in an effort to minimize provider exposure and conserve PPE while maintaining patient safety.

Adrenal EffectsCopy Link!

Literature Review: Gallery View, Grid View

Section in process

Thyroid EffectsCopy Link!

Literature Review: Gallery View, Grid View

Section in process

Chronic ConditionsCopy Link!

DiabetesCopy Link!

Literature Review: Gallery View, Grid View

People with diabetes who develop COVID-19 have ~2-3 fold increases in both mortality and risk of severe COVID-19 (summarized in Izcovich et al).

This risk in part reflects the chronic effects of diabetes, including increased susceptibility to several infections and an association with other chronic diseases, such as dyslipidemia, hypertension, and obesity.

As noted above, however, Pal et al. systematically reviewed several case reports where patients with diabetes and COVID-19 developed severe hyperglycemic crises, including diabetic ketoacidosis (DKA) and hyperglycemic hyperosmolar syndrome (HHS).

  • Seventy-four the 110 cases who presented with DKA or combined DKA/HHS had a prior diagnosis of type 2 diabetes. Since these are case reports, however, they do not define how frequently DKA occurs in severe COVID-19 compared to other severe infections.

For outpatients,

  • Instruct them to continue their normal oral or insulin regimens and monitor their glucose more frequently than usual. Depending on their glucose, they may need to temporarily increase their regimen.
  • This includes those patients with type 2 diabetes not on insulin who may not be accustomed to monitoring their glucose -- they should check twice daily if possible.
  • Use caution with oral hypoglycemic agents such as sulfonylurea or SGLT2 inhibitors, which can lead to euglycemic DKA, in patients with decreased caloric intake.

For all patients,

  • Frequent blood glucose and/or ketone (blood or urine) monitoring should be performed.
  • Target blood glucose remains the same as without COVID-19; for hospitalized patients, the Joint British Diabetes Society recommends 6-10 mmol/L, while the American Diabetes Association targets 140-180 mg/dl (Corsino et al.). Blood ketones should be kept below 0.6 mmol/L
  • Do not stop basal insulin even if febrile in those with type 1 diabetes or those with type 2 diabetes and require basal insulin for glycemic control. COVID-19 can significantly increase baseline insulin requirements.

For inpatients,

  • Monitor and maintain appropriate salt and water balance.
  • For patients requiring systemic steroids, appropriate insulin adjustments are required.
  • Manage DKA and HHS as discussed above.

ObesityCopy Link!

A BMI over 25-30 (different cutoffs depending on the study) appears to increase the odds of mortality from COVID-19 by nearly 50% and the odds of severe disease by ~2-4 fold (Izcovich et al, Popkin et al). The latter of these two papers raises the concern that BMI may impact the response to vaccination for COVID-19, based on experience with influenza and other vaccines.

There are no specific recommendations for management of COVID-19 patients with elevated BMI, but providers should be cautious with drug dosing, remain aware of possibly altered respiratory mechanics, and stay vigilant for decompensation.

HematologyCopy Link!

Updated Date: December 11, 2020
Literature Review:
Gallery View, Grid View

Acute IllnessesCopy Link!

Disseminated Intravascular CoagulationCopy Link!

Literature Review: Gallery View, Grid View

Coagulopathy is common in patients with COVID-19; early reports found 8-10% of hospitalized patients met criteria for disseminated intravascular coagulation (DIC; Tang et al) 16 of 183 hospitalized patients in Wuhan , most commonly in the critically ill or those with multi-organ dysfunction (Zhou et al). However, the coagulopathy in COVID-19 differs from DIC in bacterial sepsis and may require different management (Merrill et al; Asakura et al).

Median time to diagnosis of DIC in the series by Tang et al was 4 days into hospital admission, and this diagnosis was associated with worse survival in COVID-19 patients. Out of 183 COVID-19 patients in Wuhan, 71% of non-survivors had ISTH score ≥ 5 compared to 0.6% of survivors..

Diagnosis:

To diagnose DIC you can use the ISTH DIC score (MDcalc online calculator)

  • If score < 5, no DIC; recalculate in 1-2 days
  • If the patient develops DIC, measure PT/INR, PTT, D-dimer, fibrinogen every 3 days until discharge or death. Elevated PT/PTT and D-dimer correlate with worse prognosis

Management If Not Bleeding:

  • See Blood Products. If fibrinogen < 150 mg/dl: use FFP, cryoprecipitate or fibrinogen concentrate if you are worried about infusion volume of other options and it is available (RiaSTAP or Fibryga)
  • If platelets <30 k/mcl, transfuse (Consider holding anticoagulation if the patient requires blood products if benefits outweigh risks)

Management if Bleeding:

  • See Blood Products. For elevated PT/PTT and bleeding, use FFP, cryoprecipitate or fibrinogen concentrate if you are worried about infusion volume of other options and it is available
  • Hold anticoagulation for active bleeding in most cases. Start systemic anticoagulation only if critical thromboembolism or organ failure due to clot (i.e., purpura fulminans). There has been no mortality benefit of therapeutic anticoagulation in DIC (Levi et al).

LeukopeniaCopy Link!

Literature Review: Gallery View, Grid View

Most patients with COVID-19 have either a normal white blood cell count (WBC). A small number may have elevated WBC or low WBC. Leukocytosis (>10,000/µL) in 13% and leukopenia (<4000/µL) in 15.5% (Goyal et al Lymphocytopenia, or lymphopenia, typically defined as an absolute lymphocyte count < 1000/µL, is the most common abnormality on the CBC in COVID-19 and is found in over 80% of hospitalized patients (Guan et al; Huang et al). Low lymphocytes are also associated with poor prognosis, with lymphocyte percentage <10% on the WBC differential is strongly associated with decreased survival. (Ruan et al; Tan et al; Yang).

Numerous possible explanations for lymphopenia in COVID-19 have been proposed including

  • Invasion/ destruction of lymphocytes via ACE2 receptor
  • Acidemia, nutrition, bone marrow suppression
  • Cytokine Storm
  • Lymphatic organ damage (thymus, spleen) This possibility still requires pathological evidence and remains speculative (Tan et al).
  • Host Endothelial function. With age and chronic disease, there is more leukocyte adhesion and extravasation (Bermejo-Martin)
  • Sequestration of lymphocytes. Cytokine release leads to movement of the lymphocytes to the site of infection, the lung tissue, which could contribute to peripheral lymphopenia (Rahimmanesh).

Any patient with low lymphocytes should be considered potentially infected with COVID unless there is an alternate explanation.

  • Please note that concurrent infection and the use of steroids may skew these results. See Secondary Infections.

No current treatment regimen management changes based on lymphopenia.

  • There is no evidence for giving pneumocystis jiroveci prophylaxis given the transient nature of lymphopenia with COVID-19

ThrombocytopeniaCopy Link!

Literature Review: Gallery View, Grid View

Thrombocytopenia affected ~12% of patients with COVID-19 in one large meta-analysis (Zong et al). Another early report found thrombocytopenia in 72.5% of hospitalized patients (Chang et al), and the degree of thrombocytopenia appears to correlate with worse prognosis (Yang et al). Multiple proposed mechanisms have been proposed (Xu et al; Amgalan et al). Click here for a flow chart summarizing possible mechanisms. Disseminated Intravascular Coagulation (DIC) may contribute as a related or independent process.

Diagnosis:

  1. Consider other potential contributing etiologies of thrombocytopenia. Medication, additional infection(s), liver disease, splenomegaly, heparin-induced thrombocytopenia (HIT), thrombotic microangiopathy (TTP, HUS, DIC), alcohol, malignancy, pregnancy, rheumatologic/autoimmune, bone marrow disorders
  2. Initial workup can include: Peripheral blood smear, PT, aPTT, fibrinogen, LDH, LFTs, B12, folate
  3. If concerned for Heparin Induced Thrombocytopenia, the pretest probability of HIT can be calculated by 4Ts score (MDCalc 4Ts Calculator)
  1. Laboratory testing for HIT should typically only be sent in patients with at least intermediate probability of HIT (4 or more points on 4Ts score), although need to consider clinical context.
  2. If sending PF4, use a non-heparin anticoagulant (e.g. bivalirudin or other direct thrombin inhibitor per institutional protocols) while awaiting PF4 results. Serotonin release assays may be necessary to confirm positive PF4 results.

Management:

  1. If concern for DIC, refer to DIC Protocol Section
  2. If not bleeding, transfuse platelets if < 10,000/µL
  3. If bleeding, transfuse platelets according to clinical situation

ThrombosisCopy Link!

Literature Review (Pulmonary Embolism): Gallery View, Grid View
Literature Review (Vascular and Endothelial Damage):
Gallery View, Grid View

Incidence

  1. In ICU patients, cumulative incidences range from 9% to 70% in patients on varying levels of prophylactic anticoagulation, and whether patients were screened with compression ultrasonography or imaged for change in clinical status (Klok et al; Middeldorp et al; Klok et al; Llitjos et al; Nahum et al; Moll et al). One study suggests COVID-19 patients at increased risk for thrombosis and bleeding (Xu et al).
  2. We found that in 102 COVID-19 positive ICU patients, there were 9 radiographically-confirmed DVT or PE, based on imaging obtained for a change in clinical status; all patients received standard dose prophylactic anticoagulation (enoxaparin 40 mg daily or unfractionated heparin 5000 IU three times daily). No events occured in 108 wards patients (Moll et al). Similar findings were reported in Indianapolis (Maatman et al).
  3. Higher D-dimer and FDP levels track with multi-organ dysfunction syndrome and poorer prognosis (Wang et al).

Pathophysiology

  1. The mechanism for VTE are unknown and likely multifactorial:
  1. Systemic inflammatory response as seen in sepsis
  2. Stasis/critical illness
  3. Possibly direct endothelial damage from viral injury/ACE2 binding
  1. An autopsy series of 10 patients from New Orleans reported thrombotic and microangiopathic pathology (and diffuse alveolar damage) (Fox et al). Our discussions with pathology colleagues indicate more cellular debris than microthrombi.
  2. There is a theory from the SARS epidemic that SARS-CoV1 Spike protein can be cleaved by FXa and FIIa. Cleavage of the Spike protein activates it which promotes infectivity (Du). By extension, it is hypothesized that anticoagulation might inhibit SARS-CoV-2 replication, however this remains unproven.
  1. There is a small case series suggesting dipyridamole may be useful, though anticoagulation and antiplatelet agents require further investigation prior to being used therapeutically (Liu et al; Lin et al).

Management:

  1. Management of known DVT/VTE is with standard anticoagulation regimens (typically this would qualify as a provoked DVT). See Therapeutic Anticoagulation.
  2. Prophylaxis for DVT/VTE is an evolving area and is addressed in Anticoagulation under Prophylaxis

Chronic ConditionsCopy Link!

AnticoagulationCopy Link!

In patients previously anticoagulated with vitamin K antagonists (e.g. warfarin), PT/INR should be monitored closely. Both fever and acute infection may result in increases in INR. Other medications that may be used in COVID-19, such as antibiotics for suspected coinfection, can also result in increased or decreased warfarin metabolism. If frequent INR measurement for dose-titration is not available, and switching to an alternative, parenteral anticoagulant is not feasible, the warfarin dose may be empirically reduced by approximately 10% (e.g. from 5mg daily to 4.5mg daily) when a patient develops fever.

Anticoagulation should not be stopped for patients with COVID-19 unless there is a different reason to do so. See prophylactic and therapeutic anticoagulation for full recommendations on the treatment of COVID patients with anticoagulation.

Tool: Medications That Can Affect Warfarin Dosing

Sickle Cell DiseaseCopy Link!

In sickle cell crisis, patients have to consider the risk of COVID-19 exposure when going to the hospital for management of sickle cell crisis. Based on the local risk of nosocomial infection, providers and patients should discuss in advance regarding the patient-specific criteria which would warrant hospital evaluation versus staying at home with oral pain medications, hydration, and rest.

Infectious DiseasesCopy Link!

Updated Date: December 11, 2020
Literature Review:
Gallery View, Grid View

Acute IllnessesCopy Link!

Data on coinfection and secondary infections in COVID-19 are limited.

Viral Co-InfectionsCopy Link!

Literature Review: Gallery View, Grid View

Rates: There is enormous variance in the rates of viral coinfection depending on location, season, and viral coinfection epidemiology. A study in San Francisco found ~20% of symptomatic COVID-19 patients were also PCR positive for another viral pathogen (Kim et al). A meta-analysis of 1014 hospitalized COVID-19 patients found a viral co-infection rate of 3% (95% CI 1-6%, I2=62.3%), with RSV and influenza being the most common coinfections (Lansbury et al). In contrast, two studies in San Francisco and Wuhan, China where hospitalized COVID-19 patients tested for influenza and RSV found that none of these patients had evidence of viral co-infection (Myers et al; Chen et al).

  • The decision to test for concurrent viral panels should be based on availability and local epidemiology. Many COVID testing locations do not have the ability to also test for respiratory viruses
  • All respiratory viral infections should be considered COVID until proven otherwise (see testing), even if they present with minimal symptoms.
  • Empiric oseltamivir is reasonable in some circumstances where influenza rates are high and the patient has tested negative for COVID infection

Bacterial Co-Infection and Secondary InfectionCopy Link!

Literature Review: Gallery View, Grid View

Most patients who have COVID do not have concurrent bacterial infections. However, as with other viral infections, impaired mucociliary clearance can make these patients susceptible to secondary bacterial infections.

  • Coinfection vs secondary infection: One meta-analysis of 3448 COVID-19 patients broke bacterial infections down into co-infection and secondary infection and found the risk of co-infection on presentation to be 3.5%, while the risk of secondary infection after presentation was 15.5%. In this same cohort, 71.3% of patients received antibiotics, despite only 7.1% of patients overall having a bacterial infection (Langford et al) Other studies of secondary bacterial infections show incidence of around 7-8% of hospitalized patients. One meta-analysis of 2183 hospitalized COVID-19 patients found 7% had a bacterial coinfection (95% CI 3-12%, I2=92.2%) (Lansbury et al) Another meta-analysis of 806 hospitalized COVID-19 patients found 8% developed bacterial and/or fungal infections during admission (Rawson et al).
  • Most common infections: pneumonia (32%), bacteremia (24%), and urinary tract infections (22%) (He et al).
  • Glucocorticoid treatment was also found to be positively associated with secondary infection (He et al). However, we do not recommend withholding steroids in patients who qualify, even if they have concurrent bacterial infection.

Tool: A group out of the University of Toronto created a Living Systematic Review of the Data

Whether to use Empiric AntibioticsCopy Link!

Literature Review: Gallery View, Grid View

Clinical reports indicate that rates of bacterial superinfection with COVID-19 are low, but when present, increase mortality risk. That said, unnecessary antibiotics carry risks of fluid overload and drug-resistance, as well as the possibility that antibiotics may become a limited resource. (Zhou et al; Yang et al; Lippi et al; WHO, COVID-19 Interim guidance, May 2020). The choice about whether to give empiric antibiotics will rely on whether or not secondary bacterial infection can be safely ruled out and the acuity of the patient.

  • If laboratory and imaging guidance is available, use this evidence to guide the choice about whether to use antibiotics. There is a disproportionate high use of antibiotics despite paucity of evidence for bacterial secondary infection (He et al; Zhou et al; Rawson et al).
  • Workup can include any or all of WBC count, left shift and bandemia, procalcitonin, sputum culture, urine analysis and color, cholangitic picture on liver function tests or RUQ ultrasound, urine strep + legionella antigen, blood cultures, stool or other relevant cultures
  • If laboratory and imaging is not available or cannot be used to rule out a concurrent bacterial infection, antibiotics should be considered depending on the clinician’s expectations about risks and benefits
  • On the one hand, not treating a bacterial co-infection (depending on the type) could be fatal in some patients. There is a strong association between nosocomial infection and mortality (He et al; Wang et al). On the other hand, in one study 75% of patients who developed secondary infection were already receiving prophylactic antibiotics, suggesting prophylactic agents may not prevent hospital-acquired infections and risk selecting for more drug-resistant pathogens (He et al).
  • If a patient has shock or multiorgan failure it is appropriate to give antibiotics for the first 24-48 hours until the source is identified
Choice of AgentCopy Link!
  1. If antibiotics are to be used, they should reflect guidelines based on presumed source and multi-drug resistant organism risk factors. Administer oral antibiotics (azithromycin, levofloxacin, ciprofloxacin, etc.) when possible to reduce volume load, unless concerns for poor oral absorption.
  1. Organisms reported for those with secondary bacterial infections included those commonly seen with hospital-acquired infections including Mycoplasma sp., Haemophilus influenzae, Pseudomonas aeruginosa, Klebsiella sp., Enterobacter sp., Staphylococcus aureus, Acinetobacter sp., and E.coli, and vancomycin-resistant Enterococcus sp. (Langford et al; Lansbury et al)
  2. For empiric coverage for a presumed pulmonary source of infection, we recommend using your institutions antibiogram if one is available. If not, some possibilities include:
  1. In patients without risk factors for methicillin-resistant Staphylococcus aureus (MRSA) or Pseudomonas (i.e., those living in community without a history of resistant organisms), initiate ceftriaxone and (azithromycin or doxycycline)
  2. In patients with risk factors for MRSA or Pseudomonas (i.e., chronic hospitalization, prior resistant infections), obtain a respiratory culture and a MRSA nares screen if available and initiate an antipseudomonal cephalosporin (e.g. cefepime) and vancomycin

Tool: IDSA Guidelines

Tool: Sanford Guide

DiscontinuationCopy Link!
  1. Unnecessary antibiotics should be discontinued as soon as possible (ideally, within 48 hours) upon culture maturation. Clinical judgement should prevail over any specific lab value, but we suggest discontinuing when the following criteria are met:
  1. Clinical status is not deteriorating
  2. Cultures do not reveal pathogens at 48 hours and/or procalcitonin and WBC are relatively stable from 0 to 48 hours

Malaria and Other Tropical InfectionsCopy Link!

Malaria often presents with fever and could be confused with COVID in some patients. Where testing is possible (RDT, blood smear), it is important to test for these and to plan for increased demands on testing (Dittrich et al).

  • Where incidence is high and testing is not available, empiric (presumptive) therapy with Artemisinin Combined Therapy or the locally-approved regimen is appropriate (even though in non-COVID times empiric treatment this is generally discouraged by the WHO). Chloroquine may be used, but only if it is part of the preferred regimen for Malaria, and not for COVID (see Hydroxychloroquine).
  • Corticosteroids in concurrent malaria and COVID infection is not yet studied (Brotherton et al). Despite this we recommend using them for COVID infection as you would if the patient was not co-infected.

Tool: COVID/ Malaria Coinfection WHO

Dengue fever, like malaria, should be on the differential for COVID in places where it is prevalent. Treatment is largely supportive (oral rehydration or IV rehydration therapy, analgesics, antipyretics).

Parasitic infections should be treated as they would normally (with normal dose antihelminthic agents). We do not support the use of High-Dose Ivermectin for COVID outside of clinical trials.

Strongyloidiasis is a parasitic infection that is often asymptomatic. However, a life-threatening hyperinfection syndrome can occur with immunosuppression, including the use of corticosteroids. Because Corticosteroids are a recommended therapy for COVID with hypoxemia or critical illness, we recommend the following: (Stauffer et al).

Confirmed COVID with asymptomatic, minimally symptomatic, or mild disease (not a current candidate for corticosteroids)

And

Birth, residence, or long-term travel in Asia, Oceania, Sub-Saharan Africa, South America, Caribbean, Mediterranean countries, Middle East, North Africa*

Screen for Strongyloides infection and treat with ivermectin if positive

Confirmed COVID and likely candidate for corticosteroid treatment

And

Birth, residence, or long-term travel in Asia, Oceania, Sub-Saharan Africa, South America, Caribbean, Mediterranean countries, Middle East, North Africa*

Empiric treatment with ivermectin

*These groups are at moderate to high risk for disseminated strongyloides infection with administration of corticosteroids (A K Boggild et al).

Fungal Co-InfectionCopy Link!

Literature Review (Fungal Infection): Gallery View, Grid View
Literature Review (PJP):
Gallery View, Grid View

Fungal pathogens such as Aspergillus sp., Candida albicans, and Pneumocystis jirovecii have been described in a subset of patients (Lansbury et al; Menon et al). Some case series have reported COVID-19 associated pulmonary aspergillosis rates of 20-35% (Arastehfar et al), while others are as low as 3.8% (Lamoth et al). Unsurprisingly, aspergillus infection appears to be associated with increased mortality (OR 3.53). In a prospective Italian cohort of 108 mechanically ventilated COVID-19 patients, probable pulmonary aspergillosis was diagnosed in 30 patients (27.7%) after a median of 4 days from ICU admission, and these patients had a much higher risk of 30-day mortality (OR 3.53 (95% CI 1.29-9.67, p=0.014). Of note, most patients received tocilizumab or steroids in this cohort (Bartoletti et al).

  • At this time we do not recommend screening all patients with galactomannan and beta glucan, but patients who are already immunosuppressed, BMT, or oncologic patients should be screened with weekly
  • Treatment, and choices around immunosuppression, in these cases is highly individualized and infectious disease consultation is suggested where available.

Chronic ConditionsCopy Link!

HIV infectionCopy Link!

Literature Review: Gallery View, Grid View

The interaction between HIV and SARS-CoV-2 remains poorly defined and is likely complex. It remains unclear if, and how, HIV infection affects risk or severity of COVID-19.

Risk of acquiring COVID infection and outcomes in those infected: Multiple studies from New York City (Richardson et al; Sigel et al; Karmen-Tuohy et al), Spain (Vizcarra et al), and China (Guo et al) have found that HIV-positive patients develop COVID-19 at a similar rate as the general population. However, the patients included in these studies were largely on antiretroviral therapy (ART) with well-controlled HIV. A large Spanish cohort study of people with well-controlled HIV found that the rate of COVID-19 diagnosis and hospitalization in HIV patients was decreased to 30.0 cases per 10,000, compared with 41.7 per 10,000 in the general population (Del Amo et al). There is very limited data on COVID-19 patients with poorly controlled HIV or AIDS. One study examined public healthcare data in South Africa, which has the highest rate of HIV in the world at about 20%, with about ⅔ of those on ART (UNAIDS). In this population, HIV infection conferred an adjusted hazard ratio of 2.75 for risk of death from COVID-19 (Davies M, presentation on behalf of Western Cape Department of Health). Detailed information, including the number of participants, CD4+ T cell counts, HIV viral loads, and ART treatment status, has not yet been made available. Another prospective multi-site study in ICUs in Africa found HIV infection was independently associated with risk of death from COVID-19 (odds ratio 1.91).(ACCCOS Investigators)

Antiretrovirals : Some antiretroviral therapies used for HIV may be protective against COVID-19, but this is not yet fully supported by the data. For further information about antiretroviral agents under investigation for treatment of COVID-19, please see Lopinavir-Ritonavir in the therapeutics chapter (this so far has not been shown to benefit patients (Cao et al) or reduce viral shedding (Cheng et al)). Tenofovir has also been hypothesized to have a protective benefit, but data seems to be confounded by age and health of participants. A large Spanish cohort study of over 77,000 people with HIV, 236 of whom were diagnosed with COVID-19, found that patients taking tenofovir disproxil fumarate (TDF)/emtricitabine (FTC) had a significantly decreased risk of COVID-19 diagnosis and hospitalization compared with those taking tenofovir alafenamide (TAF)/FTC or abacavir (ABC)/lamivudine (3TC). This may be an effect of increased blood concentrations of tenofovir with TDF compared with TAF, though may also reflect that patients taking TDF are typically younger and healthier than those on TAF (Del Amo et al). Conversely, a smaller observational Spanish study of 2873 HIV-positive individuals, 51 of whom had COVID-19, found that tenofovir (either TDF or TAF) use was disproportionately enriched among COVID-19 cases (Vizcarra et al).

Diagnosis:

  • Keep in mind that people with HIV may present differently. Fever may be less frequent.
  • HIV should not change the role of either NAAT or antigen testing.
  • The impact of prior HIV on immune response and development of antibodies is not yet known.

Management:

Studies to date suggest that well-controlled HIV does not substantially increase the risk or severity of COVID-19, but data on patients with low CD4+ counts remains sparse. Given the limitations of the existing evidence at this time, we recommend that HIV-positive patients be considered high risk and be counseled on precautions accordingly

  • Per existing standard of care, all patients with HIV should remain on a daily ART regimen under the supervision of a trained provider
  • There is speculation that lymphopenia and immune dysfunction in HIV-positive individuals may protect from the hyperinflammatory state thought to contribute to severe COVID-19 disease (Mascolo et al), but no evidence currently exists to support this theory. This is not a reason to stop HIV treatment.
  • We do not recommend changing an existing ART regimen for the purposes of prophylaxis or treatment of COVID-19 in HIV-positive patients
  • HIV-positive patients who develop COVID-19 do not require any change from standard protocol in management or treatment strategies
  • Given the high prevalence of malnutrition among patients with TB/HIV, ensuring continued social support including food packages is important for disease control.
  • Patients with HIV who present with respiratory symptoms should be evaluated for TB in addition to COVID-19 if clinically indicated.

Chagas DiseaseCopy Link!

The association of Chagas disease with socioeconomic vulnerability, its large disease burden in Latin America, and the possibility of often-undiagnosed chronic cardiac injury (PAHO) all raise concerns about co-infection of Trypanosoma cruzi and SARS-CoV-2, though data from 2020 are limited to a few case reports (e.g., Alberca et al; Kurizky et al). A recent consensus opinion (Zaidel et al) considering the pathophysiology of both diseases and the impact on the healthcare system recommends that a COVID-19 diagnosis should not delay urgent antiparasitic treatment in acute Chagas disease or in those with evidence of reactivation though they recognize that drug interactions and the clinical severity of COVID-19 must be taken into account. In individuals with known Indeterminate Chronic Chagas Disease, the benefits of antiparasitic therapy are more controversial and less urgent. When these patients are also diagnosed with COVID-19, the authors advise delaying treatment while closely monitoring for reactivation. They make an exception for patients who have already started antiparasitic therapy when testing positive for SARS-CoV-2 only if no COVID-19 symptoms are present.

TuberculosisCopy Link!

Literature Review: Gallery View, Grid View

Not enough is known about the incidence of COVID in patients with tuberculosis. One case-study of 49 patients with tuberculosis (eight with drug resistant TB) showed a high case fatality at 12.3% (Tadolini et al). Some have posited that the increased social distancing measures from COVID will decrease tuberculosis, however it is more likely that any benefit on TB deaths is likely to be outweighed by health service disruption (McQuaid et al).

Tool: This Multi-Institution Consensus Statement describes TB public health and treatment plans during the COVID epidemic in great detail. Core management issues described include medications, drug-drug interactions, novel therapies, and principles of infection control and workplace safety.

Viral Hepatitis B and CCopy Link!

Patients who are newly diagnosed with viral hepatitis B should initiate hepatitis treatment if they qualify, regardless of COVID-19 status. However, patients who are newly diagnosed with viral hepatitis C should defer treatment until after the COVID-19 infection has cleared. For patients with viral hepatitis B or C who are already on treatment, they should continue treatment while being monitored for drug-drug interactions (Reddy).

NephrologyCopy Link!

Updated Date: May 3, 2020
Literature Review:
Gallery View, Grid View

Acute IllnessesCopy Link!

Acute Kidney InjuryCopy Link!

Literature Review (Acute Kidney Injury):Gallery View, Grid View
Literature Review (FSGS):
Gallery View, Grid View
Literature Review (Apol1):
Gallery View, Grid View

Incidence of Acute Kidney Injury in COVID-19 varies widely, but estimates have ranged from 0.5% (Guan et al) to 27% (Diao et al). The wide range of estimates of AKI incidence may reflect different populations included in studies. The most likely etiology of AKI is acute tubular necrosis (ATN) based on autopsy series from China, but other findings including interstitial inflammation, thrombotic microangiopathy, complement-mediated injury and direct viral infection of tubular cells and podocytes has also been described (Su et al; Diao et al). Studies find variable onset of AKI, from 7 days (Cheng et al) to 15 days after illness onset (Zhou et al). Onset of AKI more rapid and severe in patients with underlying CKD (Cheng et al)

Role of the renin-angiotensin-aldosterone system and medications that target it on the severity of COVID-19 is a source of much speculation and research, since angiotensin-converting enzyme 2 (ACE2) is used by SARS-CoV-2 as a functional receptor to enter into cells (including type II pneumocytes and kidney tubular epithelial cells). There is no data to support issues with RAAS inhibitors during COVID at this time.

Diagnosis:

  1. Monitor serum creatinine and electrolytes at least daily where available
  2. In patients with AKI, order urine electrolytes (urine Na, urea and Cr) and urinalysis with sediment
  1. Patients may present with proteinuria (44%), hematuria (26.9%) (Cheng et al). For patients with proteinuria, quantify proteinuria with spot urine protein-to-creatinine and albumin-to-creatinine ratios
  1. Consider other common etiologies of AKI that can occur in patients who do not have COVID-19 (e.g. volume depletion, ATN from hypotension, contrast-associated nephropathy, acute interstitial nephritis and urinary tract obstruction)
  2. If laboratory testing for serum creatinine is unavailable, check urinalysis to identify proteinuria. Patients with proteinuria can be classified as possible AKI until proven otherwise (Rudd et al).

Management:

  1. Discontinue all medications that can contribute to AKI (e.g. NSAIDs, ACE inhibitors, ARBs, and diuretics in volume depleted patients) and avoid using iodinated contrast with CT imaging as much as possible
  2. Consider a gentle fluid challenge (e.g. 1 liter of isotonic crystalloid fluid) to determine if there is a pre-renal component to AKI, especially in patients with clinical or laboratory signs suggestive of intravascular volume depletion (e.g. hypotension, tachycardia, dry mucous membranes, FENa<1% and/or FEurea<35%).
  1. Be cautious with fluid administration in patients with severe hypoxemia
  1. If available, consult nephrology for patients with any of the following:
  1. Creatinine clearance <30 ml/min/1.73m2
  2. Oliguria: urine output <500cc/day or <0.5cc/Kg/hour
  3. Volume overload not improving with diuretics
  4. Hyperkalemia (>5.5) not responsive to dietary K restriction and diuretics

Renal Replacement Therapy and HemodialysisCopy Link!

Literature Review: Gallery View, Grid View

Estimates for RRT range from 0.8 to 5% of hospitalized patients (Guan et al; Zhou et al) in studies including floor patients. Among critically ill patients in the ICU, need for CRRT has been reported as high as 39% (Chen et al). Few studies have reported outcomes of RRT. One case series reported that out of 191 patients, 10 received CRRT, and all 10 died (Zhou et al). The nephrology consult service will determine the need, timing, and modality of renal replacement on a case-by-case basis. Indications for RRT in COVID-19 patients are the same as the indications for all patients.

Recommendations for RRT in LMICsCopy Link!

For patients in LMICs with COVID-19 without ARDS, consider peritoneal dialysis as first choice, where available and feasible. Locally produced peritoneal dialysis solutions can be used in situations where commercially produced solutions are unavailable or unaffordable. With ARDS, consider hemodialysis, where available and feasible, in order to optimize fluid removal (Rudd et al).

Muscle Injury and RhabdomyolysisCopy Link!

Mild creatinine kinase elevation is relatively common with SARS-CoV-2 infection, with muscle pain and elevated CK occuring in 11-45% of hospitalized patients, depending on the study. It is more common in severe disease (23.9%, median CK 525 U/L) vs non-severe disease (5.0%, median CK 230 U/L) (Mao; Wang)

  • Up to 10% of patients developed rhabdomyolysis complicated by acute renal failure (Chen). Other case reports of rhabdomyolysis in SARS have been published (Tsai; Huang; Wang)

The cause is likely to be some combination of direct viral myopathy, and critical illness/immobility myopathy (which can be made worse by corticosteroids, though do not discontinue them for this reason alone).

Diagnosis:

  • Mild muscle injury: myalgias, proximal weakness, and/or mildly elevated CK (100s U/L)
  • Rhabdomyolysis: myalgias, muscle weakness, myoglobinuria, moderate-marked elevation in CK (> 5x ULN, usually > 1500 U/L)
  • Also check: CK, BMP, Phosphate, LFTs, TSH, UA, strength exam. Muscle biopsy is rarely likely to change management

Management:

  • Mild muscle injury does not require specific intervention if renal function is normal
  • Rhabdomyolysis carries risk of AKI, usually associated with CK >15-20k U/L. Increased risk of AKI in the setting of sepsis, dehydration, or acidosis (Bosch).

Tool: Treatment of Rhabdomyolysis

Chronic ConditionsCopy Link!

Chronic Kidney DiseaseCopy Link!

This section is in progress

Renal TransplantCopy Link!

Literature Review: Gallery View, Grid View
This section is in progress

SurgeryCopy Link!

Updated Date: April 30, 2020
Literature Review (Surgery):
Gallery View, Grid View
Literature Review (Surgical Modifications):
Gallery View, Grid View

Acute IllnessesCopy Link!

Preoperative AlgorithmCopy Link!

Literature Review (Operating Room Guidance):Gallery View, Grid View

Depending on the severity of the COVID outbreak, some surgical centers are reducing or stopping elective cases. The following is based on former BWH Pre-operative decision pathway during severe outbreak and included suggestions about managing operating room screening/ testing.

If the case is elective:

  • Reschedule for a future date.

If the patient has no COVID-19 symptoms or high-risk features:

  • Proceed with standard precautions
  • Face shield, surgical mask
  • Double glove
  • Avoid contamination of work surfaces with secretions
  • Concerning symptoms include:
  • Fever
  • Cough, sore throat
  • Shortness of breath, respiratory failure
  • Muscle aches, fatigue
  • High-risk features include:
  • Difficulty differentiating symptoms from baseline in patients with thoracic or upper respiratory disease
  • Contact with known cases

If the case can not be delayed until COVID-19 test results are positive:

  • Proceed with increased precautions
  • Head cover, face shield, N95
  • Double glove
  • Avoid contamination of work surfaces with secretions
  • Experienced provider intubating
  • Minimize providers entering and exiting the OR/theatre
  • Perioperative droplet precautions (patient masked, in isolation room)

If COVID-19 test results are available prior to surgery:

  • If the patient has a single negative test,
  • Proceed with increased precautions, as above.
  • If the patient has a positive test,
  • Reconsider if the case needs to be done.
  • Consider delay for 14 days or repeat testing 24 hours after symptoms resolve.
  • Ok to proceed after 2 negative tests 24 hours apart
  • If delay may cause significant morbidity or mortality,
  • Proceed with COVID-19 precautions
  • All “increased precautions as above”
  • Patient should be on isolation precautions perioperatively
  • All aerosol generating portions of the case should ideally be done in a negative pressure room
  • Full PPE for all providers in the room

Perioperative Intubation and ExtubationCopy Link!

Perioperative intubation and extubation is largely similar to ICU Intubation and Extubation. To see an example of OR extubation guides, see BWH Operating Room COVID Intubation Protocol.

TracheostomyCopy Link!

Literature Review (Tracheostomy Management): Gallery View, Grid View

Tracheostomy for patients with COVID is a clinical challenge, as it can expose health care practitioners to significant aerosols.

The timing of tracheostomy in any patient is complex. Patients should have adequate time to recover and avoid patient/provider risk, and the course of COVID-19 is longer than many pneumonias. However, earlier tracheostomy can make it easier to wean sedation and mobilize patients. Different institutions require different timeframes for tracheostomy eligibility. Early in the pandemic many institutions were waiting a full 21 days, but now practice patterns are changing.

Tool: This Consensus Guidance covers timing and patient selection and management of tracheostomy
Tool: BWH’s Tracheostomy (Percutaneous and Open) Surgical Protocols

Endoscopic ProceduresCopy Link!

Literature Review (Surgical Modifications): Gallery View, Grid View
Literature Review (Endoscopy):
Gallery View, Grid View

Both upper and lower endoscopy are considered aerosolizing procedures.

Tool: A comparison of guidance from endoscopic societies worldwide, including Wuhan, Hong Kong, Australia, Canada, US, UK, and European societies, can be found here.

Guidance from the US GI societies (AGA, ACG, ASGE, AASLD) can be found here:

Guidance from the European Society of Gastroenterology and Endoscopy Nurses and Associates can be found here:

PulmonologyCopy Link!

Updated Date: December 11, 2020

Acute IllnessesCopy Link!

Parenchymal DiseaseCopy Link!

This is covered extensively elsewhere. Please see Oxygen Care, ARDS, Mechanical Ventilation, etc.

Tracheostomy ManagementCopy Link!

Tool: Multidisciplinary Tracheostomy Management Summary (Lancet)

Chronic ConditionsCopy Link!

AsthmaCopy Link!

Literature Review: Gallery View, Grid View

In one large retrospective study, asthma appeared less prevalent in those with COVID than in those without it (6.75 vs 9.72%), and hospitalization rates did not appear very different between each group. As with all retrospective studies, these are somewhat limited by confounding (Green et al). One large study of about 44,000 people looking at risk factors for severity and mortality in China did not find asthma as a risk factor for disease severity (Li et al). Multiple smaller studies have shown similar results. It is not yet known if or why asthma may be protective against infection and/or not linked to worse outcomes.

This Green et al study also did not find any difference between those using ICS or LABA. ICS use in asthma has been dose-dependently associated with lower ACE2 and transmembrane protease serine 2 mRNA expression, but the impact of this on disease status is unknown. (Peters et al). It has also been posited that ICS may reduce airways inflammation and thus offer some protection (Carli et al). At this time we do not recommend treating patients with comorbid asthma and COVID any differently than you would normally, save for avoiding nebulizers (aerosol generating) and favoring MDIs where possible.

Chronic Obstructive Pulmonary DiseaseCopy Link!

Literature Review: Gallery View, Grid View

In one large metanalysis of 22 studies involving 11,000 patients, COPD was associated with three-fold higher mortality in COVID infected patients (OR 3.23, 1.59-6.57; P<0.05), but it was not more prevalent (5% of the patients(437/9337) than it is in the global population (9%) (Vankata et al). The severity difference in this study did not appear to be related to active smoking. Currently we do not recommend treating patients with comorbid COPD and COVID any differently than you would normally save for avoiding nebulizers (aerosol generating) and favoring MDIs where possible.

Nebulizer UseCopy Link!

Literature Review: Gallery View, Grid View

Nebulizers can increase risk of transmission of COVID-19. Patients should avoid use. If nebulizer use is required, it should be done in isolation from others. COVID-19 virus may persist in droplets in the air for several hours.

SmokingCopy Link!

Literature Review: Gallery View, Grid View

The relationship between COVID-19 infection and smoking remains unclear. Metanalysis has shown that in terms of testing positive for disease, there may be a slight risk reduction in current smokers compared with nonsmokers (RR=0.73; 95% CI: 0.73–0.99). However, the evidence is not high-quality and may be confounded by other factors (testing and social behaviors may be very different in these groups).Test positivity risk amongst former smokers and never-smokers was similar (Farsalinos et al; Grundy et al). In most studies smoking is associated with more severe disease (OR rates in many metanalyses around ~2 (Grundy et al). Smoking poses multiple health risks and cannot be considered as a protective habit.

Pulmonary FibrosisCopy Link!

Literature Review: Gallery View, Grid View

Pulmonary Vascular DiseaseCopy Link!

Literature Review: Gallery View, Grid View
This section is in process

Lung TransplantCopy Link!

Literature Review: Gallery View, Grid View
This section is in process

OncologyCopy Link!

Updated Date June 19, 2020
Literature Review (Oncology):
Gallery View, Grid View
Literature Review (Cancer Care Delivery):
Gallery View, Grid View

Acute IllnessesCopy Link!

Febrile NeutropeniaCopy Link!

Patients with febrile neutropenia (ANC < 500 cells/mm3 AND T ≥ 101F or T ≥ 100.5 for 1hr) should be worked up for COVID infection at the same time as they are evaluated for other infections. In patients with heme malignancy or SCT: findings are more subtle or absent in neutropenic and immune suppressed patients.

  • Examine catheters (port, CVC, others) daily. Avoid rectal exams and any per-rectum therapies in neutropenic patients, but examine the perirectal area if symptoms or persistent fevers.

Tool: BWH guidance on Neutropenic Fever (workup, empiric antibiosis, line management, etc)

Chronic ConditionsCopy Link!

Solid and Liquid TumorsCopy Link!

Literature Review (Solid Oncology): Gallery View, Grid View
Literature Review (Liquid Oncology):
Gallery View, Grid View

Based on early descriptive studies from China, patients with cancer - particularly those on active treatment for cancer - appear to have a worse prognosis. This includes higher prevalence, higher risk of severe disease, and higher risk of death from COVID-19 in patients with cancer compared to those without. (WHO-China Joint Mission on COVID-19, Yu et al). Prognosis for various cancers is highly variable, and the patient’s oncologist should be involved in goals of care conversations.

In additional labs to standard workup, if available we recommend also obtaining:

  • Weekly glucan/galactomannan in neutropenic/transplant patients.
  • Specific patient populations may require additional monitoring (such as CMV, EBV monitoring in transplant patients – ask primary oncologist).

Patients with solid tumors are at very high risk of thrombosis but at lower risk of infection than most heme malignancy patients. Prophylactic anticoagulation is particularly important in this setting.

  • Hold pharmacologic prophylaxis if platelet count < 30K, use pneumoboots

Patients on Immune Checkpoint InhibitorsCopy Link!

Literature Review: Gallery View, Grid View

Immune Checkpoint Inhibitors (ICIs) Most common ICIs are CTLA-4 inhibitor (ipilimumab) and PD-1/PD-L1 inhibitors (pembrolizumab, nivolumab, durvalumab, atezolizumab and avelumab). are not immunosuppressive when used alone, but the steroid dosages used to treat immune toxicities are often immunosuppressive. If patient develops organ dysfunction, it may be due to immune toxicity and not COVID. Please see BWH guidance for more information.

DermatologyCopy Link!

Updated Date: May 11, 2020
Literature Review (Dermatology):
Gallery View, Grid View

Acute IllnessesCopy Link!

Tool: Please report cases at aad.org/covidregistry

RashesCopy Link!

Literature Review (Livedo): Gallery View, Grid View
Literature Review (Morbiliforme Eruptions):
Gallery View, Grid View
Literature Review (Viral Exanthem):
Gallery View, Grid View
Literature Review (Urticaria):
Gallery View, Grid View
Literature Review (Vesicular Eruptions):
Gallery View, Grid View

Earliest data out of China with rash in 0.2% of patients, without morphologic description (Guan et al). Later data from Italy collected by dermatologists on the front lines showed rash in 18/88 patients (20.4%), excluding patients on new drugs (Recalcati). Morphology and type are varied. In one case study of 375 cases of rashes suspected to be COVID related (Galvan Casas et al) 47% with “other maculopapules” including perifollicular lesions, pityriasis rosea-like lesions, non-palpable purpura and palpable purpura. Lesions lasted for a mean of 8.6 days, and appeared with the onset of other symptoms. Associated with more severe disease (2% mortality) 19% of cases with pseudo-Chilblains (“COVID toes”) Typically appearing in younger patients. Lesions lasted for a mean of 12.7 days and were found later in the course of disease. Associated with less severe disease 19% with urticaria. Lesions lasted for a mean of 6.8 days, and appeared with the onset of other symptoms. Associated with more severe disease (2% mortality) 9% with vesicular eruption. Middle-aged patients. Lesions lasted for a mean of 10.4 days, and appeared before other symptoms. Associated with intermediate severity. and 6% with livedo/necrosis. Found in older patients with most severe disease (10% mortality). See BWH Dermatology Guidance for information on morbilliform rashes, urticaria, vasculopathies, livedo, and vesicular eruptions

Perniosis, Pseudo-Chillblains, COVID ToesCopy Link!

Literature Review: Gallery View, Grid View

Clinically “Covid toes” presents as erythematous to violaceous papules over acral surfaces (usually the fingers and toes, less commonly the nose and ears) following exposure to cold (fittingly called “acrocyanosis”). The mechanism is unknown, but likely due to Type I interferon-mediated complement activation and resultant microangiopathy (Kolivras). They often cause pain, itching, or paresthesias. Blistering, crusting, and ulceration can occur in severe cases, and they may be accompanied by livedo-like changes in adjacent skin. They are generally self-resolving within 1-2 weeks, however recurrence is possible with repeated cold exposure (Fernandez-Nieto et al; Mazzatto et al). Generally, these do not need extensive workup if they occur in the setting of known COVID. If they occur without clear explanation or are very severe, consider the following workup: CBC with differential, ANA, RF, Cold agglutinins, Cryoglobulins, C3, C4, CH50, CRP, ESR, D-dimer, Fibrinogen, Antiphospholipid antibodies

Treatment involves avoiding cold exposure, wearing socks, smoking cessation, and aspirin for a possible vasculopathic etiology (with caution in the pediatric population given the risk of Reye’s syndrome). In some cases you can use topical steroids, pentoxifylline, hydroxychloroquine, and calcium channel blockers.

Pressure InjuriesCopy Link!

Bed-bound patients are at risk for a variety of pressure injuries including erythema, skin breakdown, ulcerations, gangrene, or frank necrosis. Highest risk areas include sites of repeated pressure. In the context of COVID-19 and proning, facial pressure injuries have frequently been reported. Avoid pressure injury by:

  • Minimizing pressure over bony prominences and face with dressings. Massachusetts General Hospital recommends the application of foam dressings to the upper chest/clavicles, shoulders, pelvis, elbows, knees, forehead, and dorsal feet. Gel pads may be used under the cheeks/nose (MGH, 2020)
  • Turning the head and reposition arms every 2 hours.
  • Frequently assessing for blanchable erythema, an early sign of pressure injury

Chronic ConditionsCopy Link!

Skin CancersCopy Link!

Literature Review (Outpatient Dermatology): Gallery View, Grid View

Management of suspected skin cancer in the ambulatory setting is challenging during COVID (NCCN Covid Resources). If skin cancer is suspected, a high resolution photograph should be taken and dermatology telemedicine referral placed where available. Please see additional guidance from BWH guidelines.

Tool: Skin conditions that warrant referral for in-person evaluation: Medical Dermatology Society Guidelines

ImmunosuppressionCopy Link!

See baseline immunosuppressants
See
BWH dermatology guidelines

RheumatologyCopy Link!

Updated Date: December 11, 2020
Literature Review:
Gallery View, Grid View

Acute IllnessesCopy Link!

Cytokine Storm SyndromeCopy Link!

This is covered Cytokine Storm Syndrome

MIS-CCopy Link!

This content will be covered in pediatrics

Rheumatologic SymptomsCopy Link!

COVID-19 can cause a number of symptoms that may overlap with those seen in rheumatologic diseases as outlined below. For patients with established rheumatologic disease who have confirmed or suspected COVID-19, careful evaluation will be required to determine if their symptoms are due to flare of the disease or are sequelae of viral infection.

  1. Arthralgias, Myalgias, Myositis
  1. Myalgia or arthralgia occur in approximately 15% of patients. 14.8% of patients (based on analysis as of 2/20/2020 on 55924 cases. (WHO-China Joint Mission on Coronavirus Disease 2019 (COVID-19) 2020
  2. Rhabdomyolysis is also a potential late complication of Covid-19. (Tong et al; Ronco et al). Please see Muscle Injury under Neuromuscular Disorders
  1. Lung Disease
  2. Pericarditis and Myocarditis
  3. Livedo Reticularis and Pernio- or Chilblain-like lesions of hands and feets (“COVID toes”)
  4. Fever
  5. Coagulopathy, Lymphocytopenia, and Thrombocytopenia
  6. Elevated levels of inflammatory markers. including CRP, ESR, and ferritin as well as elevated levels of cytokines including IL-1 and IL-6 (see Cytokine Storm Syndrome)

Chronic ConditionsCopy Link!

Underlying Rheumatologic DiseaseCopy Link!

Literature Review: Gallery View, Grid View

Please enroll patients with rheumatologic/autoimmune disease who are diagnosed with COVID-19 into the COVID-19 Global Rheumatology Alliance Registry or the EULAR – COVID-19 Database.

Patients with rheumatologic disease are not known to differ from other patients in terms of clinical presentation of COVID-19, though further information will be elucidated from the studies of the Global Rheumatology Alliance Registry. Early reports show that symptoms are similar in patients with rheumatic disease than in patients without. A report of 86 patients with immune-mediated inflammatory disease in New York reported high percentages of patients with fever (84%), cough (42%) and shortness of breath (41%), with lower rates of diarrhea, rhinorrhea and loss of taste and smell. Similar to reports from immunocompetent patients. (Haberman et al, NEJM, 2020) Similar findings were found in smaller studies of patients with rheumatic disease: 13 patients with chronic arthritis in Italy (Monti S et al) and 52 rheumatic disease patients in Boston (D’Silva K et al).

Studies looking at outcomes for COVID patients with rheumatic disease are limited to date, and major conclusions cannot be drawn at this time.

Incidence appears to be similar to the general population. In one prospective case series of 86 patients in New York City with a variety of immune-mediated inflammatory diseases. (Haberman et al). Hospitalization and mortality rates appear similar to the general population. In one comparative cohort study of 52 patients with rheumatic disease (75% on immunosuppressive medications) hospitalization and mortality rates were similar. However, rheumatic patients were more likely to require mechanical ventilation than healthy comparators, though the number of patients was low (11 patients [48%] vs 7 patients [18%], multivariable OR with 95% CI 1.07 to 9.05) (D’Silva K et al).

For management of patients on immunosuppressants see baseline Immunosuppressants.

Tool: Guidelines as developed by the American College of Rheumatology. The authors also have outlined a Helpful Resource on UptoDate.

GastroenterologyCopy Link!

Updated Date: June 10, 2020

Literature Review: Gallery View, Grid View

Acute IllnessesCopy Link!

Diarrhea and Abdominal PainCopy Link!

Literature Review: Gallery View, Grid View

About 10% to 60% patients have GI symptoms on presentation incidence appears to be much higher in the US as compared to China (Sultan et al; Redd et al). GI symptoms may be the initial or predominant presenting symptom in 15-20% of patients (Redd et al). Very few patients have only GI symptoms throughout the course of illness (Pan et al). Anorexia is the most common gastrointestinal symptom followed by diarrhea and nausea. Vomiting and abdominal pain are not as common (Luo et al; Pan et al). In an analysis of the presenting symptoms and clinical outcomes of 318 adult patients with COVID-19 who were admitted to 9 hospitals across Massachusetts, 61% reported at least one gastrointestinal symptom (35% w/anorexia, 34% w/diarrhea, 26% w/nausea, 15% w/vomiting, and 15% w/abdominal pain) (Redd et al, Gastroenterology, 2020). Different mechanisms may mediate the broad array of GI symptoms seen in COVID-19. Proposed mechanisms include direct damage to the intestinal mucosa (Xiao et al), microthrombi in the lamina propria and submucosa (Bhayana et al; Ignat et al) and indirect alterations in mucosal immunity via the “gut-lung axis” (Pan et al)

  • Diarrhea attributed to COVID-19 is often mild (mean # of bowel movements per day = 3-4, range = 2-10) and usually lasts for <1 week (mean # of days = 4-5, range = 1-14). (Han et al; Jin et al; Lin et al).
  • Abdominal pain has been described as “stomachache, epigastric pain, and abdominal discomfort;” further characteristics have not been reported (Sultan et al).

Management:

  • Treatment is largely supportive for diarrhea, nausea, and vomiting.
  • If severe symptoms, a more extensive laboratory evaluation (such as lipase, amylase, lactate, stool studies, C Diff) or imaging (such as KUB, CT abdomen / pelvis, abdominal US +/- dopplers, or pelvic US) may be indicated.

Bowel InfarctionCopy Link!

Literature Review: Gallery View, Grid View

Bowel infarction in COVID-19 appears to be rare, but cases reports are emerging (all in critically ill patients). In one retrospective study, abdominal imaging findings were analyzed in all patients admitted to the ICU (33%) or floor (67%) over a 2-week period. The most common indications to obtain a CT a/p were abdominal pain (33%) and sepsis (29%). Among the CT a/p scans performed in ICU patients (n = 20), the following findings were seen: fluid-filled colon in 65%, colonic or rectal thickening in 20%, small bowel wall thickening in 25%, pneumatosis or portal vein gas in 20% (n = 4), and perforation in 5% (n = 1). Among the CT a/p scans performed in non-ICU patients (n = 22), the following findings were seen: fluid-filled colon in 23% and colonic or rectal thickening in 14%; none of the non-ICU patients had findings of small bowel wall thickening, pneumatosis or portal vein gas, or perforation (Bhayana et al). Consider lactate and abdominal imaging in patients with severe abdominal pain. Management of thrombotic risk is discussed in Thrombosis.

Liver InjuryCopy Link!

Literature Review: Gallery View, Grid View

Elevated liver biochemistries are seen in 15-70% of patients on presentation (incidence appears to be higher in the US as compared to China). Liver injury (defined as AST or ALT > 3x ULN; ALP and/or bilirubin > 2x ULN) is uncommon in presentation (Bloom et al). Acute liver failure has not been reported, even in those who are severely ill and in those with chronic liver disease (AASLD May 14, 2020). Both the incidence and degree of elevation in liver biochemistries are higher in severe disease (as compared to non-severe-disease) (Lei et al). Liver injury is associated with increased length of admission, need for ICU admission, and mortality (Fan et al; Lei et al; Hajifathalian et al). Abnormalities are predominantly hepatocellular and mild (even in severe disease), often rising during the course of illness. (Lei et al; AASLD; Bloom et al)

  • AST > ALT. AST and ALT elevations are usually 1-2x ULN on presentation. Levels are higher in patients with severe disease. May rise to >3x ULN in 40% of patients.
  • GGT is often elevated, but ALP is normal.

Hypothesized pathways for liver injury include: a direct viral cytopathic effect (ACE2 is expressed on cholangiocytes and, to a lesser extent, hepatocytes); altered hepatic perfusion secondary to microthrombi; and cytokine-mediated injury (Zhang et al; Bloom et al). A limited number of post-mortem liver examinations have shown relatively non-specific findings, including: moderate microvesicular steatosis; mild, mixed lobular and portal activity; mild sinusoidal dilation with mildly increased small lymphocytes infiltration in sinusoidal spaces; and multifocal hepatic necrosis (Li and Xiao). In critically ill patients, liver injury may be secondary to ischemic / hypoxic hepatitis (“shock liver”); hepatic congestion; and cholestasis of sepsis. Hepatotoxic medications, such as Remdesivir, Hydroxychloroquine, and Tocilizumab, may also contribute.

In general, extensive workup and hepatic imaging is not needed in patients with asymptomatic, mild, hepatocellular-predominant elevations in liver biochemistries. If the patient has RUQ pain or cholestatic enzymes or AST or ALT >3x ULN; ALP or T-bili >2x ULN consider workup as below, and evaluate for drug toxicity. If the liver injury is mild and self-resolving. No specific therapy is typically needed (AASLD Clinical Insights)

Pancreatic InjuryCopy Link!

Literature Review: Gallery View, Grid View

Hyperlipasemia, defined as a lipase level above the upper limit of normal, has been seen in 12-18% of patients. However, a lipase level >3 times the upper limit of normal appears to be rare (seen in only 2-3% of patients in whom lipase was checked) and no cases meeting diagnostic criteria for acute pancreatitis (as per the Revised Atlanta Classification) have been reported. Gastrointestinal symptoms are common in patients with hyperlipasemia (11-67% with anorexia, 56% with nausea, 11-57% with diarrhea, 33% with general abdominal discomfort) (McNabb et al; Wang et al).

In patients with elevated lipase and abdominal pain / tenderness, CT Abdomen Pelvis is recommended to clarify the differential, including pancreatitis vs enteritis / colitis vs bowel ischemia or obstruction vs cholecystitis or other hepatobiliary process (all of which can cause elevated lipase and abdominal pain) (Hameed et al). If not available, RUQ ultrasound may be helpful in some circumstances.

Gastrointestinal BleedCopy Link!

Literature Review: Gallery View, Grid View

Chronic ConditionsCopy Link!

Cirrhosis and Chronic Liver DiseaseCopy Link!

Literature Review: Gallery View, Grid View

Chronic liver disease (including NAFLD) is a risk factor for severe disease and increased mortality. Cirrhotics and liver transplant recipients are at particularly high risk of death (Ji et al; Singh et al; Lee et al). The SECURE-Cirrhosis and COVID-Hep registries are tracking data on patients (throughout the world) with cirrhosis, chronic liver disease, and liver transplant who are infected with COVID-19 (Update). Data thus far shows: Patients w/non-cirrhotic chronic liver disease: 18% required ICU admission and 6% died. Patients s/p liver transplant: 22% required ICU admission and 22% died. Patients w/cirrhosis: 24% required ICU admission and 37% died. Unfortunately, poorer outcomes in cirrhosis are not unexpected. Among patients with ARDS of any cause, cirrhotic patients are known to have poorer outcomes (increased 90-day mortality) as compared to non-cirrhotic patients (Gacouin et al).

Tool: The American Association for the Study of Liver Diseases (AASLD) has constructed a ‘living document’ on best clinical practices in hepatology during the COVID-19 pandemic: AASLD "Clinical Best Practice Advice for Hepatology and Liver Transplant Providers during the COVID-19 Pandemic"

Inflammatory Bowel DiseaseCopy Link!

Literature Review: Gallery View, Grid View

As of May 8, 2020, there were 1170 reported cases of COVID-19 in IBD patients. Of these, 32% required hospitalization, 6% required ICU admission, 5% required mechanical ventilation, and 4% died. Among the 1170 cases, 58% were in remission. Among those in remission, 28% required hospitalization; 30% and 44% of those with mild and moderate-severe disease activity required hospitalization, respectively. (SECURE-IBD Registry) Patients on prednisone (> 20 mg daily) are likely at increased risk of COVID-19. It is unclear if the risk and severity of infection are increased in patients on thiopurines (azathioprine, 6-mercaptopurine), methotrexate, anti-TNF therapies (infliximab, adalimumab, certolizumab, golimumab), vedolizumab, ustekinumab, and the JAK inhibitor tofacitinib (Rubin et al).

Tool: Recommendations From the International Organization for the Study for Inflammatory Bowel Disease on the Management of IBD During the COVID-19 Pandemic

Liver TransplantCopy Link!

Literature Review: Gallery View, Grid View
This section is in process

NeurologyCopy Link!

Updated Date July 13, 2020
Literature Review:
Gallery View, Grid View

Acute IllnessesCopy Link!

Neurologic Manifestations of COVIDCopy Link!

Literature Review (Brain MRI): Gallery View, Grid View
Literature Review (Headache):
Gallery View, Grid View

Neurologic manifestations may occur in 36.4%-69% of hospitalized COVID-19 patients (Mao; Helms). Severely ill patients are more likely to have neurologic symptoms (45.5% severe vs. 30.2% non-severe), stroke (5.7% severe vs. 0.8% non-severe), impaired consciousness (14.8% severe vs. 2.4% non-severe), and skeletal muscle injury (19.3% severe vs. 4.8% non-severe) (Mao) Manifestations can include:

  1. Delirium, confusion, or executive dysfunction (Helms). 69% of patients displayed agitation; 65% of patients assessed with CAM-ICU had confusion; 33% of discharged patients had inattention, disorientation, or poorly organized movements to command
  2. Smell or taste abnormalities (see Anosmia)
  3. Headache
  4. Corticospinal tract signs (67%) (Helms)
  5. Dizziness (16.8%) (Mao)
  6. Stroke (2.5-5%) (see Stroke).
  7. GBS, Miller Fisher syndrome (case reports) (see BWH Guidance on Neuromuscular Disorders)
  8. Encephalitis, acute necrotizing encephalopathy, myelitis, CNS demyelinating lesions (case reports) (see Meningoencephalitis)

Pathophysiology: Illness from SARS-CoV-2 can provoke states that increase risk of neurological disease. The pathophysiology of the various neurological manifestations of COVID-19 is currently unknown, but possible mechanisms include:

  1. Direct viral invasion of the nervous system, with potential transsynaptic spread
  2. Theoretical possibility of blood-brain barrier disruption secondary to SARS-CoV-2 binding to angiotensin-converting enzyme 2 (ACE2)
  3. Autoimmune sequelae
  4. Ischemic injury via systemic hypoxia or local vascular endothelial inflammation or thrombosis
  5. Toxic metabolic encephalopathies
  6. Long term impact of the systemic proinflammatory state
    (Zubair et al).

Altered Mental Status (Encephalopathy and Delirium)Copy Link!

Literature Review (Delirium): Gallery View, Grid View
Literature Review (Altered Mental Status):
Gallery View, Grid View

Rates of AMS (including delirium) have been relatively high (7.5-66%) in COVID, with variability likely related to differences in assessment of mental status and definitions of deficits. In a series of patients in Strasbourg, France, 66% displayed agitation, 65% of patients assessed with CAM-ICU had confusion, and 33% of discharged patients showed dysexectutive function (Helms). Delirium, characterized by waxing and waning arousal and impaired attention, is common in hospitalized patients of advanced age and with multiple comorbidities. One study of ICU patients (before the COVID pandemic) showed that 83.3% of patients develop delirium. Delirium was present in 39.5% of easily arousable patients and persisted in 10.4% of patients at discharge (Ely).

Encephalopathy in patients with COVID-19 may be caused by systemic infection, toxic-metabolic derangements (hypoxemia, hypercarbia, renal or hepatic dysfunction, nutritional deficiencies) or medication effects (sedation, cephalosporins/ quinolones), or primary CNS dysfunction (e.g. seizure, stroke). Delirium can happen even in the absence of these conditions due to sleep/wake disturbances and psychological stress.

Workup: Recommend performing a general workup for AMS as below. If these are unrevealing and the patient has significant abnormalities, in some patients it may be worth pursuing MRI brain for structural etiologies such as encephalitis or stroke (see Meningoencephalitis and Stroke sections), EEG for seizure, or LP for signs of meningitis or unexplained neurologic findings.

Tool: General evaluation for AMS, regardless of COVID-19 status
Tool:
Screen for Delirium Using the Confusion Assessment Method (CAM)

Management

  1. Treat specific causes as discovered in work-up
  2. Treat for Delirium
  3. Detailed guidelines regarding ICU treatment of sedation, pain, agitation, and delirium can be found in BWH’s Guidance.

Anosmia and Ageusia (Decreased Smell and Taste)Copy Link!

Literature Review: Gallery View, Grid View

Changes in smell and taste perception have been reported in many patients with COVID-19. A meta-analysis of 10 studies (1627 patients) demonstrated olfactory dysfunction in 53% and gustatory dysfunction in 44% of COVID-19 patients (Tong). Anosmia may precede COVID-19 diagnosis (Kaye), and when anosmia/ageusia occur they most frequently precede hospitalization (Giacomelli).

Pathophysiology is unknown. There is some evidence that supports direct neural invasion of the virus. Retrograde neuronal transport to CNS through peripheral nerves is documented in other viral illnesses rabies, HSV, murine counterparts to coronavirus (Perlman) and there are case report of MRI FLAIR hyperintensities in the olfactory bulbs and right rectus gyrus in COVID-positive patient presenting with isolated anosmia (Politi). However, ACE-2 is expressed in nasal epithelium, but not in olfactory sensory neurons, indicating epithelium may be the entry site (Gengler; Zubair).

Recovery: 66-80% of patients with COVID-19-associated smell impairment report spontaneous improvement or resolution within days to weeks of recovery from clinical illness (Yan; Lechien; Hopkins; Vaira). In a study of 3191 patients, median time to recovery for anosmia and ageusia was 7 days (Lee).

Management: No indication for corticosteroids to treat hyposmia/anosmia, as it frequently recovers without intervention

Meningoencephalitis and MyelitisCopy Link!

Literature Review (Encephalitis): Gallery View, Grid View
Literature Review (Myelitis):
Gallery View, Grid View

Encephalitis the inflammation of the brain parenchyma secondary to infection or autoimmune conditions. Diagnostic criteria (Venkatesan): AMS > 24 hours without alternative cause and 2 (possible encephalitis) or 3 (probable encephalitis) of the following: (a) fever > 100.4F within 72 hours of presentation, (b) generalized or partial seizures, (c) CSF leukocyte count > 5, (d) abnormality on imaging that is new and consistent with encephalitis. meningitis is the inflammation of the membranous coverings of the brain secondary to infection or autoimmune conditions, myelitis is the inflammation of the spinal cord parenchyma secondary to infection or autoimmune conditions, acute necrotizing encephalopathy (ANE) a rare type of brain injury that usually follows an acute febrile illness (potentially a post-infectious autoimmune condition) characterized by symmetric multifocal brain lesions, without inflammatory cells in the brain parenchyma (Poyiadji). and acute disseminated encephalomyelitis (ADEM) an immune-mediated inflammatory disorder characterized by wide-spread demyelination of the brain and spinal cord have all been reported with COVID infection.

MRI and CSF findings: MRI brain abnormalities are commonly present in patients with severe COVID with encephalopathy or neurologic symptoms, and display some characteristic findings. Of 13 patients with encephalopathy of unclear etiology, 8 (62%) displayed leptomeningeal enhancement. 100% of 11 patients who had perfusion imaging showed bilateral frontotemporal hypoperfusion (Helms). Of 27 ICU patients with neurologic symptoms, 10 (37%) had cortical FLAIR abnormalities (of these, 7 demonstrated cortical diffusion restriction, 5 had subtle leptomeningeal enhancement, and 3 had subcortical or deep white matter signal abnormalities) (Kandemirli). In a study of 37 patients with severe COVID with neurologic manifestations and abnormal MRI, 3 main patterns were seen: 43% had signal abnormalities in the medial temporal lobe, 30% had nonconfluent multifocal white matter hyperintense lesions with variable enhancement (the majority of these with associated hemorrhagic lesions), and 24% had extensive white matter microhemorrhages (Kremer et al). CSF abnormalities may be present patients with neurologic symptoms requiring LP, typically with elevated protein and variable pleocytosis, rarely positive PCR (see below). In study of 22 children with COVID and encephalitis in Wuhan, 10 had CSF pleiocytosis and 8 had elevated CSF protein (Li et al). In a study of 7 adults patients (unclear clinical presentation), CSF showed no pleocytosis in all patients, there was elevated CSF protein in 1 patient, and CSF RT-PCR for SARS-CoV-2 was negative in all patients (Helms).

Evidence suggestive of direct CNS invasion: Rarely CSF has been noted to be positive by PCR (in 2 of 578 samples in one study, but not at levels that are infectious (Destras et al). Additional case reports: (Moriguchi; Hanna Huang; Xiang et al) SARS-CoV-2 was also identified in 8/22 patient brains in one series by RT-PCR (Puelles). A case report identified SARS-CoV-2 viral particles on electron microscopy of the frontal lobe, in endothelial cells and neural cell bodies (Paniz-Mondolfi).

Evidence of Autoimmunity: Autoantibodies have been noted suggesting some patients may have an autoimmune meningoencephalitis (Lucchese). These are largely against unidentified neural autoantigens, though there have been a couple case reports of COVID-associated NMDA receptor encephalitis (Panariello et al; Monti et al).

Responses have been described with plasmapheresis (4/6 patients) (Dogan); rituximab and IVIG (case report) (Monti et al), and high-dose steroids (case report) (Pilotto et al).

Work-up and management: Consult neurology where possible for guidance.

Tool: BWH General Approach to Work-up and Management of Encephalitis and Myelitis

SeizureCopy Link!

Literature Review: Gallery View, Grid View

In early studies, the frequency of seizures appears low (<1%) in COVID-19 patients relative to other coronaviruses (8-9% for MERS-CoV and other HCoV (Saad; Dominguez). In a series of 214 patients, 1 patient had a generalized seizure lasting 3 minutes (Mao). Case reports exist of COVID-19 positive patients developing new-onset seizures (Moriguchi; Xiang; Duong; Zanin; Sohal; Bernard-Valnet; Karimi) though in most of these cases, seizures were presumed to be secondary to unmasking of an underlying seizure disorder. Limited evidence to date does not suggest that patients with epilepsy are at higher risk of COVID-19 infection or severe disease manifestations (French).

Work-up and management:

  1. General Seizure Work-up and Management Regardless of COVID-19 Status
  2. Note that convulsive seizure should be considered aerosol generating, providers should don appropriate PPE

StrokeCopy Link!

Literature Review (Stroke): Gallery View, Grid View
Literature Review (Intracerebral Hemorrhage):
Gallery View, Grid View

Patients with COVID-19 may have an increased risk of stroke related to a systemic inflammatory and prothrombotic state (Klok), possible endothelial dysfunction, and/or medical comorbidities. A systemic review of 39 studies comprising 135 patients (hospitalized, many with severe COVID) found a pooled incidence of acute ischemic stroke of 1.2%, with onset 10+/-8 days after COVID symptoms began. Most of these strokes were large vessel thrombosis, embolism, or stenosis (62%) or multiple vascular territory embolic (26%), rather than small vessel. 38% of patients died (Tan et al).

Workup and management: Protocols for acute stroke work-up and management remain largely the same as for non-COVID patients: IV tPA, Endovascular Therapy, and 2018 AHA Guidelines. Consult neurology for any acute stroke evaluation.

  1. tPA increases D-dimer levels and decreases fibrinogen levels for at least 24 hrs (Skoloudik). D-dimer should not be used for COVID-19 prognostication post-tPA.
  2. Given likely hypercoagulable state (see Thrombotic Disease) in many COVID-19 patients, consider therapeutic anticoagulation for confirmed stroke in a COVID-19 patient if stroke mechanism is unclear (discuss with neurology)

Tool: BWH Approach to Minimize TIA and Stroke-Related Admissions During the COVID-19 Pandemic

Tools: Multiple published guidelines exist addressing how to best manage acute stroke during the COVID-19 pandemic (Khosravani; AHA/ASA Stroke Council; Baracchini; Qureshi)

Guillain-Barré SyndromeCopy Link!

Literature Review: Gallery View, Grid View

Guillain Barré syndrome (GBS) (inclusive of acute inflammatory demyelinating polyneuropathy [AIDP], acute motor axonal neuropathy [AMAN], and acute motor and sensory axonal neuropathy [AMSAN]) have been reported in COVID-19 positive patients. Rare cases have been reported of neuromuscular symptoms presenting prior to COVID-19 symptoms, or as late as 24 days after COVID-19 symptoms.

Tool: BWH Management Guide for GBS in COVID-19 Positive Patients

Chronic ConditionsCopy Link!

DementiaCopy Link!

Literature Review: Gallery View, Grid View
This section is in process

Myasthenia GravisCopy Link!

Literature Review: Gallery View, Grid View

Patients with MG may be at higher risk of contracting COVID-19 or developing severe disease because they are often on immunosuppressive therapies and have respiratory muscle weakness. In a series of 15 hospitalized patients in Brazil, 13 developed an exacerbation during their hospitalization and 73% needed mechanical ventilation and 30% died (Camelo-Filho).

  • While we do not recommend them in general for COVID treatment, these medications are known to exacerbate MG flares: Chloroquine, hydroxychloroquine, azithromycin
  • Discuss whether or not to hold immunosuppressants with the patient’s prescribing physician

Tool: Please Consider Reporting Cases to CARE-MG, a Physician-Reported Registry

Tool: BWH Management Guidelines for all Inpatients with Myasthenia Gravis

Multiple SclerosisCopy Link!

Literature Review: Gallery View, Grid View

It is not known whether patients with MS have increased incidence of COVID-19. Patients with MS may be at higher risk of infection in general, including pneumonia and influenza, but do not appear to be at higher risk of all upper respiratory infections (Wijnands; Brownlee; Willis). It is not known if MS patients with COVID-19 have a more severe course of disease. In a French cohort, 3.5% of 347 patients died and 21% had severe COVID, though the study was registry-based (Loupre et al). Data from 1540 MS patients across 21 countries showed that progressive MS and worse disability score were associated with worse outcomes, and that anti-CD20 therapies were linked to increase risk of artificial ventilation but not death (Americas Committee for Treatment and Research in Multiple Sclerosis (ACTRIMS), First results of the COVID-19 in MS Global Data Sharing Initiative suggest anti-CD20 DMTs are associated with worse COVID-19 outcomes, 22-Sep-2020. See here.

Treatment: Please see baseline Immunosuppression for more information about the risk of infection associated with disease modifying therapies. In general, during mild viral infections DMTs are usually continued (Brownlee) but in severe disease consider temporarily suspending or delaying certain high-risk immunosuppressive DMTs. Recommendations by DMT type have been published - see for instance the European Academy of Neurology for Management of Patients with Neurological Diseases During the COVID-19 Pandemic. Do not change treatment without contacting the prescribing physician.

Tool: DMT Management (National Multiple Sclerosis Society)
Tool: Report Cases to COVIMS, a de-identified patient data repository.

Chapter 14

Post-COVID Care

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

Literature Review (Post-COVID Patient Care): Gallery View, Grid View

Post-Acute COVID Syndrome (PACS)Copy Link!

Updated Date: May 24, 2021

Course of RecoveryCopy Link!

Data on COVID-19 recovery are still emerging. However, it is clear that the course of recovery is highly variable among patients, may be prolonged over months, and is dependent on severity and particular manifestations of the initial illness. Anecdotal evidence suggests that recovery can be highly dynamic, with periods of improvement followed by periods of apparent acute worsening. Generally, recovery is thought to occur within about 2 weeks for mild infections and 2-3 months in severe infections (WHO).

Long term symptoms after COVID are often called "Long-COVID" in the popular media, but generally called post-acute COVID-19 syndrome (PACS), chronic COVID-19, post-COVID syndrome (PCS), or post-acute sequelae of SARS-CoV-2 infection (PASC) by the medical community. Various definitions for each of these exist, though increasingly they refer to symptoms occurring >4 weeks after COVID infection, without active viral activity or reinfection.

A recent Nature summary (Nalbandian et al) proposes two separate categories of PACS quoted here:

  1. Subacute or ongoing symptomatic COVID-19, which includes symptoms and abnormalities present from 4–12 weeks beyond acute COVID-19
  2. Chronic or post-COVID-19 syndrome, which includes symptoms and abnormalities persisting or present beyond 12 weeks of the onset of acute COVID-19 and not attributable to alternative diagnoses

Prevalence of PACSCopy Link!

In one study of 4,182 cases, 13.3% reported symptoms lasting ≥28 days, 4.5% for ≥8 weeks, and 2.3% for ≥12 weeks (Sudre et al). Amongst patients with prior hospitalization these are much higher: Patients were seen a mean of 60 days days after onset of symptoms. Only 12.6% of patients reported being asymptomatic, with particularly high rates of ongoing fatigue (53.1%), dyspnea (43.4%), joint pain (27.3%), and chest pain (21.7%) (Carfi et al). PACS is more common in people displaying a wider array of initial symptoms (>5 symptoms at time of diagnosis) (Sudre et al). It also appears to be more common in breakthrough cases (19% experience symptoms >6wks) (Bergwerk et al).

Demographics:

Longer term effects have been seen in persons of all demographic groups, though appear to be more common in women (14.9%) compared with men (9.5%), and older populations (ranging from 9.9% in 18–49 year olds to 21.9% in ≥70 year olds) (Sudre et al).

Tool: Table of Reported Prevalence of PACS (Nature Article, as of March 2021)

Symptom PatternsCopy Link!

Common reported symptoms are fatigue and dyspnea, joint pain, and chest pain (Carfi et al) In one survey of over 5000 patients with symptoms lasting >21 days, the most common symptoms were fatigue (79.0%), headache (55.3%), shortness of breath (55.3%), difficulty concentrating (53.6%), cough (49.0%), changed sense of taste (44.9%), diarrhea (43.9%), and muscle or body aches (43.5%). The timing of symptom onset varied and was best described as happening in waves. (Lambert et al)

One study found two different patterns of symptomatology (Sudre et al):

  1. Exclusively fatigue, headache and upper respiratory complaints
  2. Multisystem complaints (including fever and gastroenterological symptoms)

PathophysiologyCopy Link!

The causes of post-covid syndrome remain unknown at this time and are an area of active research. They include virus-specific pathophysiologic changes, immunologic aberrations and inflammatory damage and expected post-critical illness consequences. (Nalbandian et al)

  1. Virus-specific pathophysiologic changes. The virus is known to infect and harm multiple organs, as described in each subsection here. Further, the endothelial damage and pro-coagulability seen in COVID may cause microangiopathic changes that cause longer-term damage (Lerner et al).
  2. Immune and Inflammatory changes. Immune responses may have a role in the etiology of persistent symptoms. One analysis indicated that B-Cell responses may be similar to those described in autoimmune processes (activation of extrafollicular pathways) (Woodruf et al). T cell dysregulation may also be involved, given their role in acute disease. One study showed that coordinated CD4+ and CD8+ T-cell and antibody responses were associated with milder acute disease (Moderbacher et al). The relationship between antibody titers and viral load and post-COVID syndrome is unknown. In one study there was no statistically significant difference between initial viral titres or serial antibody levels between those who did and did not develop long term symptoms (Pereira et al).
  3. Expected Post-Critical Illness. Post ICU syndrome is a constellation of symptoms and sequelae that is common for many people who have been critically ill.

Outpatient WorkupCopy Link!

Updated Date: April 19, 2021

Post-COVID Clinic VisitsCopy Link!

In some locations there are dedicated post-COVID care clinics specializing in care for PACS. However, most patients are still seen by primary care clinicians as well as an array of subspecialists such as pulmonologists, cardiologists, neurologists, and psychiatrists. Generally subspecialty care is not needed, except in patients who have known complications.

  1. Frequency of Visits: Acute phase followup is discussed here. Visit frequency depends on many things, including severity of patient symptoms and initial disease, but generally for priorly hospitalized patients out of the acute phase and for those with ongoing symptoms we recommend a virtual check-in at 3 days after discharge, and again at 4-6 weeks. They should be seen in person at 12 weeks where possible. (Nalbandian et al)
  2. Infection control: Patients who are seen in a clinic should be cleared based on infection control practices at the institution before an appointment is made. Generally time-based clearance criteria are acceptable, though those at risk for persistent infection or reinfection may need retesting.

Work-UpCopy Link!

Clinical testing is not always needed. If performed, testing should target evaluation for non-COVID etiologies as well as serious sequelae of disease. Clinicians need to distinguish between non-life-threatening symptoms such as persistent dyspnea, fatigue, and neurocognitive issues and serious sequelae such as VTE and heart failure (Greenhaigh et al).

  1. Screen for “red flag” symptoms:
  1. New or worsening dyspnea
  2. Unexplained chest pain
  3. New confusion
  4. Focal weakness
  5. New or worsening lower extremity edema
  1. Screen for common neurocognitive symptoms:
  1. Anxiety
  2. Depression
  3. PTSD
  4. Sleep disturbances
  5. Cognitive impairment
  1. COVID Testing: SARS-CoV-2 PCR is discouraged as it can be positive for months (Katz). Reinfection is rare and generally there is not an indication for repeat testing. However if the patient has new symptoms of COVID or a concern for reinfection, it may be indicated in select cases. See here for guidance on repeat testing. Generally there is not an indication for serology, unless the initial diagnosis was not clear.
  2. Imaging and Pulmonary Testing. See Radiographic Abnormalities and Pulmonary Function Tests. Adapted from the British Thoracic Society’s followup algorithms (George et al).
  1. Mild disease without known pneumonia: Generally followup radiograph is not performed in patients with a mild course of disease, no prior positive imaging, and no worsening symptoms.
  2. Mild disease with persistent dyspnea, or history of mild to moderate pneumonia: Obtain a Chest Xray at 12 weeks.
  1. If normal and symptoms are resolved, no further workup is needed.
  2. If persistently abnormal or symptoms persist, obtain full PFTs and consider CTPE for pulmonary embolism.
  1. If PFTs are normal and the patient is improving, repeat Chest Xray.
  2. If PFTs are abnormal or the patient is not improving, consider high resolution CT (HRCT).
  1. Severe, or ICU- level disease: Obtain a Chest Xray at 12 weeks. Consider full pulmonary function tests and CTPE as well on a case-by-case basis.
  1. If Xray is normal and symptoms are resolved, no further workup is needed.
  2. If abnormal, get PFTs and a high resolution CT. Consider Echocardiogram and CTPE as well.
  1. Ambulatory saturations including exercise pulse oximetry can be very helpful in determining of a patient has tachycardia on exertion (suggestive of POTS or arrhythmia) or desaturations on exertion (suggestive of parenchymal or pulmonary vascular disease).
  1. 6 minute walk tests may help differentiate etiology of dyspnea in some patients
  1. General labs for patients with persistent symptoms >4 weeks after infection, a complete blood count, chemistry panel, and liver function tests are recommended.
  1. Additional labs will depend on symptoms, but could include cardiac enzymes and BNP for cardiac symptoms or prior cardiac concerns; D-dimer for concern for pulmonary embolism (although D-dimer may remain elevated up to four months following acute COVID-19 infection); thyroid function tests for fatigue symptoms CPK and ANA for joint or skin concerns. Routine coagulation markers are not needed. (Townsend et al.)
  2. EKGs are done on a case-by-case basis, and should be performed on patients with persistent cardiopulmonary symptoms.
  3. Echocardiograms are not routine except if concerned for heart failure. Cardiac MRI is typically experimental and not indicated clinically except by specialists for select indications (typically prior myocarditis).
  1. Other workup and treatment will depend on the presenting symptoms. See below for:
  1. Dyspnea and cough
  2. Interstitial lung disease or organizing pneumonia
  3. Fever or joint pain
  4. Anxiety and depression
  5. Fatigue and brain fog
  6. Anosmia and ageusia (difficulty smelling and tasting)
  7. Lightheadedness, sweating, racing heart or other symptoms of dysautonomia or postural orthostatic tachycardic syndrome (POTS)
  8. Chest pain or history of cardiac complications/ myocarditis
  9. Deep vein thrombosis
  10. Kidney dysfunction
  11. Post-ICU syndrome

General ManagementCopy Link!

  1. Optimize comorbidities and health behaviors including sleep hygiene, smoking cessation, and decreased alcohol intake.
  2. If the patient has residual oxygen requirement, have them keep a daily pulse oximetry journal to monitor for recurrent COVID-19
  3. Encourage gradual increase in exercise (Greenhaigh et al). Generally patients should exercise as much as tolerated with Sp02 >90%. If oximetry monitoring is not available, try using an oximeter in the office, and tell them to pay attention to symptoms. Transient desaturations are unlikely to have negative consequences, but persistent desaturation should be avoided.
  4. Please see all the specific subsections below (also linked just above) for specific management recommendations for common specific complaints, as well as indications for subspecialty referral.
  5. Consider early rehabilitation referral for the patient if it is available
  6. Educate patient on the typical course of recovery.
  7. Consider enrollment in patient studies if available and patient advocacy groups if desired.
  8. Make sure the patient still gets vaccinated for COVID.
  9. Generally we advise against extended thromboprophylaxis unless it would be otherwise indicated if the patient were hospitalized with an acute illness (see delayed thrombosis for risk calculators and a more extensive discussion), though this is an area of active research.

Vaccination after COVID InfectionCopy Link!

Vaccination is recommended for patients who have had COVID infection. Vaccine-based immunity may provide immunity against a more conserved part of the virus (e.g. the receptor binding domain) than native immunity, and thus may be more effective.

  1. Timing of vaccination for already infected patients is not determined, but many institutions recommend waiting 90 days after infection, especially if treated with monoclonal antibodies or convalescent plasma (as these may reduce the effectiveness of vaccines).
  2. Currently most institutions recommend giving both vaccinations for two-shot vaccines, though in the future this may change as there is some evidence that a single dose may provide adequate immunity (An NIH study showed that after a single dose of the Pfizer-BioNTech vaccine, people with prior infection had antibody levels similar to people receiving 2 doses. However we do not know how antibody levels correlate with immunity. (NIH)
  3. Warn patients that the injections may create more robust immune responses in them than they would in a never-infected patient, and thus they may experience more fevers and fatigue in the days following the injection

Anecdotal reports in COVID survivor communities that 30-40% of people may feel improved after COVID vaccination have not yet been empirically studied.

Pulmonary ConcernsCopy Link!

Dyspnea or CoughCopy Link!

Updated Date: April 19, 2021

Dyspnea is the most common persistent symptom beyond acute COVID-19, ranging from 42–66% prevalence at 60–100 d follow-up (Nalbandian et al). The need for supplemental oxygen due to persistent hypoxemia at 60 days was reported in 6.6 of patients in one US study (Chopra et al).

Differential: The differential for persistent dyspnea after COVID infection includes:

  1. Neuromuscular weakness associated with deconditioning, protein calorie malnutrition, or Post Intensive Care Syndrome including myopathy from corticosteroid or neuromuscular blockade
  2. Post-ARDS pulmonary fibrosis (see below on PFT and imaging findings)
  3. Pulmonary embolism. Pro-thrombotic effects of COVID infection and hospitalization may increase risk of pulmonary embolism. We do not yet know how long these effects last.
  4. Bacterial superinfection. As with other viral pneumonias, there is a theoretical increased risk of subsequent bacterial superinfection. About 3% of patients with COVID-19 who are admitted have a community acquired bacterial infection (Garcia-Vidal et al). Patients previously admitted may be at risk for drug-resistance bacterial infections in the future.
  5. Dysautonomia or neurologically-caused dyspnea.
  6. Cardiac dyspnea including myocarditis and arrhythmias.

Workup:

  1. In addition to a full history for cardiac and pulmonary symptoms, ascertain the patient’s tolerance for exertion and whether symptoms are worsening. Persistent or worsening symptoms should prompt workup for non-COVID causes.
  1. For fever, infectious workup should be performed (see fever)
  2. For signs of heart failure or tachyarrhythmia, see cardiac.
  3. For tachycardia, pleuritic chest pain, or other signs of PE consider EKG, D-Dimer, Echocardiogram, or contrast CT.
  4. If features of dysautonomia (orthostatic hypotension, rapid pulse fluctuations, temperature dysregulation, sweating changes, gastrointestinal concerns), workup and treat as below.
  1. Vitals signs for patients that have dyspnea should include:
  1. Ambulatory saturations
  2. Orthostatic vitals signs (which could indicate dysautonomia)
  1. Chest imaging indications are described here. Generally with persistent or worsening symptoms after 3 months imaging is merited.
  2. Pulmonary Function Tests are indicated as described here. Generally full PFTs should be considered at 3-6 months for persistent dyspnea.
  3. 6 minute walk tests may help differentiate etiology of COVID in some patients
  4. If despite this workup dyspnea remains unexplained, we recommend a gentle exercise routine as described below. For very complex or ill patients with multiple possible etiologies (e.g. cardiac, pulmonary, and/or dysautonomic), referral to pulmonary for consideration of cardiopulmonary exercise testing may be indicated to help differentiate between these causes, but generally these do not often elucidate a cause of ongoing dyspnea in otherwise well patients and should not be routine.

Treatment:

  1. Treatment consists of treating the underlying etiology
  1. Treatment of pulmonary embolism, secondary infection, post-ARDS ILD, etc are as they would be for any non-COVID patient
  2. Treatment of breathing concerns related to chest pain on deep inspiration after COVID is covered here.
  3. If the patient has features of dysautonomia, see here.
  1. If workup is normal and the patient had mild COVID we typically focus on physical therapy, breathing exercises, and reassurance. Many of these patients seem anecdotally to improve with time.
  1. Some specialized breathing-related therapy programs exist, and more are starting to treat post-COVID symptoms. Some online programs are also available for this purpose, such as Stasis Performance. More will no doubt become available as demand increases.
  2. Symptomatic treatment of dyspnea is described here
  3. Symptomatic treatment of cough is described here
  1. Unexplained persistent dyspnea may merit referral to pulmonary or dysautonomia treatment providers

Tool: Information on Six Minute Walk Tests from the American Thoracic Society

Pulmonary Function TestsCopy Link!

Updated Date: April 19, 2021
Literature Review (Pulmonary Function Tests):
Gallery View, Grid View

Testing options:

Generally full PFT (spirometry, lung volumes, and DLCO) are not indicated for all patients, but should be performed 3-6 months after infection if there are persistent pulmonary symptoms or persistent radiographic abnormalities after 12 weeks from infection. MIP/MEP in patients who have been ventilated or who have concern for steroid myopathy may help differentiate neuromuscular etiologies, especially in patients with restrictive disease.

PFT abnormalities:

Abnormalities in PFTs, specifically diffusion capacity (DLCO) and TLC, at time of hospital discharge appear to be common and correspond with severity of illness (Mo X et al). It is unclear how long reductions in pulmonary function persist, though DLCO reductions remain common at 30 days after symptom onset (Frija-Masson et al).

  • In one study of 57 patients 30 days after hospitalization: “Six (10.5%), 5(8.7%), 25(43.8%) 7(12.3%), and 30 (52.6%) patients had FVC, FEV1, FEV1/FVC ratio, TLC, and DLCO values less than 80% of predicted values, respectively… Compared with non-severe cases, severe patients showed higher incidence of DLCO impairment (75.6%vs42.5%, p = 0.019)” (Huang et al)
  • The median 6-min walking distance is low in approximately 1/4 of patients at 6 months. (Huang et al)

Radiographic AbnormalitiesCopy Link!

Updated Date: April 19, 2021

Timing of radiographs is described in the general workup section above.

Persistent radiographic abnormalities from COVID pneumonia may be present. These are likely to improve gradually over several months, but some dysfunction may persist, particularly in patients with ARDS. In a small study of 55 patients 3 months after discharge (64%) had persistent symptoms and 39 (71%) had radiologic abnormalities (interstitial changes or fibrosis) (Zhao et al) In a study of 114 patients with severe COVID pneumonia, fibrotic-like changes were found in 35% of patients on 6 month CT scans. The remainder had either complete radiological resolution (38%) or residual ground-glass opacification or interstitial thickening (27%) (Han et al).

Interstitial Lung DiseaseCopy Link!

There are case reports of COVID causing organizing pneumonia and interstitial lung disease, some of which appears to be independent of ARDS or mechanical ventilation (see radiographic abnormalities and pulmonary function tests above). The pathophysiology is likely a viral invasion of epithelial and endothelial cells, as well as immunologic damage, that cause endothelial/epithelial breakdown similar to other forms of ARDS. In history studies and autopsy studies, diffuse alveolar damage in multiple stages is seen, as are myofibroblasts, mural fibrosis, and honeycombing (Carsana et al). Fibrosis may be provoked by cytokines such as IL-6 and TGF-β, which have been found in non-COVID studies to contribute to pulmonary fibrosis.

There is not yet any consensus about whether treating patients who appear to have an inflammatory ILD or organizing pneumonia with corticosteroids is beneficial. At present, we recommend that clinicians weigh the risks and benefits of protracted corticosteroid treatment for each individualized patient case. There is early evidence to suggest it may help in a subset of cases: One study reviewed 837 patients with ongoing symptoms six weeks after infection. 35 patients were found to have persistent interstitial lung changes, predominantly organizing pneumonia (as reviewed by multidisciplinary specialists). Of these, 30 were offered corticosteroid therapy, resulting in a mean relative increase in transfer factor (DLCO) of 31.6% and forced vital capacity of 9.6% with significant symptomatic and radiological improvement (Myall et al).

Rheumatologic ConcernsCopy Link!

FeverCopy Link!

Updated Date: April 19, 2021

Persistent fevers can be seen in COVID infection, however fevers after 14 days (or new or severe fevers after improvement) should always prompt an infectious workup as these can be caused by secondary bacterial infections.

  • Infectious workup should include (where available) a CBC with differential, LFT, UA, blood culture, procalcitonin, Chest Xray, and sputum culture.

Joint PainCopy Link!

Updated Date: April 19, 2021

While there are case reports of reactive arthritis (Jali, Ono) and rheumatoid arthritis (Derksen) triggered by COVID infection, frank arthritis (inflammation of the joints) is very rare after infection with COVID-19. However, persistent polyarthralgias (i.e. joints that are achy but not inflamed on exam or on imaging) are common, as is myalgic encephalitis/ chronic fatigue syndrome. Clinicians should work up joint pain depending on the severity and chronicity, similar to other clinical contexts.

  1. Patients with signs of inflammation (swelling, erythema, effusion, etc) in one or more joints should be referred to a rheumatologist for work-up of alternate etiologies, such as septic arthritis, Lyme disease, gout, and rheumatoid arthritis. A joint aspiration can be helpful. If there is any suspicion for septic arthritis (e.g. sudden swelling and effusion, often with fever), joint aspiration should be performed emergently.
  2. Patients with symptoms of less than 3 months’ duration can be managed conservatively with NSAIDS (if not otherwise contraindicated).
  3. In patients with persistent joint pain after 3 months, or particularly debilitating joint pain or red flags such as visibly swollen joints, joint effusions, rashes, or daily fevers or drenching night sweats, we recommend workup with ESR, CRP, appropriate rheumatologic serologies (e.g. ANA, rheumatoid factor, CCP antibodies), a CBC with differential, complete metabolic panel, and urinalysis with sediment (to screen for protein or blood in urine). Patients should be referred to a rheumatologist for consultation.

Neurologic and Neuropsychiatric ConcernsCopy Link!

Updated Date: April 19, 2021
Literature Review (Post-COVID Neuropsychiatric Effects):
Gallery View, Grid View

IncidenceCopy Link!

Patients may have sequelae of direct neurologic effects of COVID infection, including encephalitis or stroke. Prolonged hospitalization, particularly ICU admission, can lead to cognitive deficits (see post-ICU syndrome). In addition, prolonged illness can manifest different psychiatric and neurologic sequelae. In a study of over 200,000 patients, the estimated incidence of a neurological or psychiatric diagnosis in the 6 months following a COVID diagnosis was 33.62%, for 12.84% it was their first such diagnosis. These effects were more common in COVID-19 than in those who had influenza (hazard ratio [HR] 1.44, 1.78 for first diagnosis. (Taquet et al). The incidence of several neuropsychiatric complications are provided below. Please see ‘Fatigue’ for discussion of chronic fatigue syndrome.

  • 0.56% for intracranial haemorrhage and 2.10% for ischaemic stroke (these are covered here)
  • 0.11% for parkinsonism and 0.67% for dementia
  • Mood disorder 13.66%; first diagnosis 4.22%
  • Anxiety disorder 17.39%; first diagnosis 7.11%
  • Psychotic disorder 1.40%; first diagnosis 0.42%
  • Substance use disorder 6.58%; first diagnosis 1.92%

Workup and treatment: Patients with known neurologic and/or psychiatric complications of COVID-19 should be followed by a neurologist and/or psychiatrist where available. Management of these complications following COVID-19 should be the same as it is in patients who have these disorders independent of COVID19.

Fatigue and “Brain Fog”Copy Link!

Updated Date: May 9, 2021

Fatigue is by far one of the most common PACS symptoms. Half of patients in one study reported ongoing fatigue, a median of 10 weeks after infection, with 32% reporting symptoms >12 weeks after (Townsend et al). Most patients' symptoms resolve on their own, though it may take many months. If symptoms last more than six months, some of these patients may meet criteria for Myalgic encephalitis/chronic fatigue syndrome (ME/CFS), which has been documented with many severe viral syndromes including COVID. More information about diagnosis of ME/CFS is available in the Institute of Medicine Clinician’s Report. There is some relationship between ME/CFS symptoms and dysautonomia/ POTS. For ME/CFS specifically:

  1. Diagnosis requires the following three symptoms Frequency and severity of symptoms should be assessed. The diagnosis of ME/CFS (SEID) should be questioned if patients do not have these symptoms at least half of the time with moderate, substantial, or severe intensity. :
  1. A substantial reduction or impairment in the ability to engage in pre-illness levels of occupational, educational, social, or personal activities, that persists for more than 6 months and is accompanied by fatigue, which is often profound, is of new or definite onset (not lifelong), is not the result of ongoing excessive exertion, and is not substantially alleviated by rest and
  2. Post-exertional malaise, and
  3. Unrefreshing sleep
  1. At least one of the two following manifestations is also required:
  1. Cognitive impairment or
  2. Orthostatic intolerance

Management:

There is no cure or medical treatment for myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Some patients, but not all, achieve some degree of relief with symptom management. There is some overlap between ME/CFS and dysautonomia (treatment is covered here).

  • Post-exertional malaise can be managed by pacing training (or activity management). Many patients describe a push/crash cycle where they feel exhausted after physical or mental exertion, and take a long time to recover and thus avoid exercise. However, gentle exercise anecdotal improves patients tolerance over time and improves quality of life. Pacing management is generally targeted at increasing mental and physical exertion slowly, but staying within an “exertion envelope” to avoid exceeding personal limits. See this tool.
  • Rehabilitation specialists or exercise physiologists with experience with ME/CFS can help develop strategies.
  • For treating insomnia, good sleep habits and medications are helpful
  • Pain should be managed with over the counter medications (generally NSAIDs and acetaminophen), gentle movement, gentle massage and other supportive treatments.
  • Memory and concentration problems can be hard to treat. Some patients may benefit from stimulant medications such as those used to treat ADHD. However, not all patients derive benefit from these, and they often can worsen the push/crash cycles described above. Medications such as those used for Alzheimer's Disease are not recommended.
  • Depression and anxiety are covered here.
  • Lightheadedness and orthostasis are covered here.
  • Disability from ME/CFS is complicated. In the USA, more information is available from the CDC website.

Tool: Institute of Medicine Clinician’s Report for ME/CFS diagnosis and treatment

Tool: Pacing Tutorial

Anxiety and DepressionCopy Link!

Updated Date: May 9, 2021

Recovered COVID patients are at increased risk of mood disorders including depression and anxiety, as well as consequences of isolation and social stigma. It is unclear as yet whether mood changes arise from increased stress, isolation, and trauma or whether there is an additional direct neurocognitive effect of the virus.

  • In a study of analyzing health records of more than 236,000 patients with COVID-19 found that six months after being diagnosed with COVID-19, 1 in 3 patients had experienced a psychiatric or neurological illness (Taquet et al). While anxiety, mood, and substance use disorders were most common, concern was raised about rates of serious neurological complications, especially in patients who had been severely ill with Covid-19. The data also indicated that compared to control groups of people who had the flu or other non-Covid respiratory infections, first-ever neuropsychiatric diagnoses were almost twice as high.
  • The presence of a mood disorder prior to admission is associated with greater likelihood of discharge to a skilled nursing facility or other rehabilitation facility rather than home (Castro et al).
  • Hospitalized individuals with a history of mood disorder may be at risk for greater COVID-19 morbidity and mortality and are at increased risk of need for postacute care. Further studies should investigate the mechanism by which these disorders may confer elevated risk.
  • ICU patients are at particular risk of post-traumatic stress disorder. See Post-ICU syndrome
  • ME/CFS can also be associated with mood changes.

Management:

In general, these are not treated differently than patients suffering with anxiety or depression from other etiologies (see here for treatment recommendations).

Prolonged Anosmia and AgeusiaCopy Link!

Updated Date: April 19, 2021

Loss of smell and taste have been observed to be prolonged, often many months. See anosmia and ageusia for a review of this topic. Some patients have phantom smells (parosmia). Overall, the loss of smell persists for more than 4 weeks in about 10% of patients (Walker et al).

Management:

  • Confirm that the olfactory disturbance is COVID-19 related. People with progressive smell loss/disturbance that does not line up chronologically with COVID-19 infection need to go through the typical workup for olfactory disturbance so that nothing is missed (typically nasal endoscopy and MRI if endoscopy is normal). Similarly, patients with anosmia associated with other neurological findings should be worked up for alternate causes.
  • There is currently no definitive recommendation for medical treatment of these effects. A synthesis of ENTUK and BRS expert guidance and recommends (Walker et al, Hopkins et al):
  • Olfactory (smell) training for patients with symptoms over 2 weeks. Training is available at AbScent and Fifth Sense.
  • Topical corticosteroid nasal drops (fluticasone or betamethasone typically) can be tried in patients with symptoms lasting more than 2 weeks. These tend to be low-risk, but efficacy is not yet proven, and they likely help mostly with nasal obstruction.
  • Oral steroids have minimal data, except some case reports (Le Bon et al). Do not offer oral corticosteroids within 2 weeks of infection unless otherwise indicated as the chance of spontaneous recovery is high, and they may cause delayed viral clearance. We do not recommend using these in patients who are still symptomatic. They are optional in patients with no other symptoms more than 2 weeks after infection.
  • Monitor patients’ weight, as smelling and tasting changes can trigger weight loss
  • Ensure loss-of smell safety measures: smoke detectors should be functional, if there is natural gas in the household then it should be removed or detectors obtained, and food safety should be assured with adequate refrigerator/freezer temperatures and expiration date checks
  • For patients with symptoms persisting after 3 months, consider referral to ear, nose, and throat physician.

HeadacheCopy Link!

Migraine-like headaches are relatively common, and late-onset headaches may be do to high cytokine levels. Many times these headaches are refractory to typical analgesics. About 38% of patients in one study had ongoing headaches after 6 weeks (Pozo-Rosich et al). Management of headache is the same as for non-COVID patients, and is covered in BWH’s COVID guidelines.

Dysautonomia and POTSCopy Link!

Updated Date: March 15, 2021

After COVID, some patients develop disorders of the autonomic nervous system, which most commonly include orthostatic hypotension, postural tachycardia of either CNS or peripheral etiology, gastrointestinal disorders, and sweating abnormalities, as well as other rarer symptoms. Evidence-based treatments exist for autonomic dysfunction, particularly for the cardiovascular manifestations.

  • Autonomic dysfunction has been associated with post-infectious syndromes in the past, and is thought to occur most likely either through direct viral sequelae or through autoimmune mechanisms (Dani et al).
  • In a retrospective study of all 841 COVID patients admitted to two Spanish hospitals in March 2020 (the ALBACOVID registry), 2.5% developed dysautonomia (Romero-Sanchez et al).
  • The literature includes multiple case reports and small case series of patients developing autonomic intolerance (most commonly postural orthostatic tachycardia syndrome (POTS), but also orthostatic hypotension, hyperhidrosis, nausea/constipation/dyspepsia, and pupillary abnormalities such as accommodation defects) after COVID infection (see e.g. Umapathi et al).

DiagnosisCopy Link!

  • Vitals: Initial steps include orthostatic vital signs, thorough cardiopulmonary and neurologic exams, and EKG.
  • POTS is diagnosed by an increase in HR by at least 30 bpm from supine to standing or by an increase to a HR of >120 within ten minutes of standing. Further testing can help distinguish a central etiology, which is generally accompanied by a hyperadrenergic state, from a peripheral autonomic neuropathy.
  • Orthostatic hypotension is diagnosed by a drop in blood pressure by >20 mmHg systolic or 10 mmHg diastolic within three minutes of standing.
  • Differential: Autonomic symptoms can overlap with other pathologies. For instance, patients can have shortness or breath (often orthostatic), palpitations, and poor exercise tolerance. Thus, investigating other etiologies with targeted testing such as outpatient rhythm monitoring, echocardiogram, chest imaging, pulmonary function tests, etc. is often warranted.
  • Laboratory tests to consider (Benarroch et al, Zadourian et al).
  • CBC to rule out significant anemia, chemistry panel for electrolyte disturbances, TSH, Early morning cortisol, Hemoglobin A1c, ANA, with subserologies if elevated
  • Supine/seated/standing plasma catecholamine levels (marked increases can point towards a central hyperadrenergic state as a cause of POTS, rather than peripheral nerve pathology)
  • Urinary catecholamines and metanephrines
  • If available, serum paraneoplastic antibody panel (Through Mayo Clinic in the U.S.)
  • Additional diagnostic tests to consider in select patients (most of these are not widely available outside of select centers, and often do not change management)
  • Skin biopsy for epidermal axonal density, as evidence for small fiber autonomic neuropathy
  • Non invasive and invasive cardiopulmonary exercise testing
  • Tilt table testing
  • 24-hour blood pressure monitoring
  • Formal autonomic testing (including cardiovagal and sudomotor testing, sometimes thermoregulatory testing as well)
  • Urodynamic testing, gastrointestinal motility testing, or ophthalmologic exam if indicated by symptoms

Treatment:Copy Link!

Common treatment options for postural orthostatic tachycardia and orthostatic hypotension include the following:

  • Elevation of head of bed
  • Avoiding prolonged recumbent position
  • Copious fluid intake (2-3 L daily)
  • Plenty of salt intake (10-12g daily)
  • Graded exercise regimen - can start with recumbent exercise such as seated bike/rowing machine)
  • Compression pants such as running compression pants, and/or abdominal binders
  • Small frequent meals
  • Avoid exacerbating factors such as dehydration, overheating, alcohol, prolonged standing
  • Salt tablets - e.g. 1g TID, though dietary salt preferable
  • Beta blockers - best evidence for propranolol, 10-40 mg three times daily, start at lower end of dosage range
  • Fludrocortisone - For volume expansion. Start with 0.1 mg PO daily, then increase up to 0.2 mg PO daily, monitoring after initiation and titration for hypokalemia
  • Pyridostigmine - Acetylcholinesterase inhibitor to facilitate cholinergic ganglionic nerve transmission and increase sympathetic vascular tone, particularly in patients suspected of autonomic neuropathy as primary etiology. 30-60 mg two to three times daily. Start at 30 mg twice daily.
  • Midodrine - For orthostatic hypotension. 2.5-10 mg three times daily, start at 2.5 mg three times daily during daytime hours. Note that some patients with POTS may have hypertension, and that midodrine can cause severe supine hypertension, so use in select patients, avoid <4 hours before bedtime, and monitor blood pressure after initiation/titration.
  • Clonidine - As a sympatholytic, especially in patients with elevated standing catecholamines. 0.1-0.4 mg PO twice daily, start at 0.1 mg at night and can titrate up by 0.1 mg each week
  • Ivabradine, a negative chronotropic, may be indicated in patients with hyperadrenergic POTS (plasma NE >600 pg/ml and abnormal tilt table test). (Taub et al)
  • Some data, particularly for small fiber autonomic neuropathy, exists for IVIG (Oaklander) and/or immunosuppression in cases with strong suspicion for autoimmune etiology. This should be pursued only in consultation with neurology.
  • Appropriate referrals include cardiology and neurology. Specialist autonomic neurologists exist as well.
  • There is inadequate data at present to assess the course of post-COVID autonomic dysfunction, though some patients have shown recovery (see e.g. Umpathi et al).

Hematologic ConcernsCopy Link!

Delayed ThrombosisCopy Link!

Updated Date: May 9, 2021

The pro-thrombotic state of acute COVID is well-described, but the duration of this state is unclear. Retrospective studies to date suggest that the rates of VTE following hospitalization for COVID-19 are similar to rates for other acute medical illnesses (Patell et al and Roberts et al). One study of 163 patients suggested a 2.5% incidence of thrombosis by 30 days, with mean occurrence at 23 days. However, there was a 3.7% incidence of bleeding, mostly due to fall (Patell et al). One registry study of COVID patients indicated that the 90 day post-discharge venous and arterial thromboembolism and all-cause mortality rates were 1.55%, 1.71%, and 4.83% respectively (Giannis et al). Delayed VTE events, even in patients who were never hospitalized, have been reported.

Extended thromboprophylaxis. Generally we do not recommend extended thromboprophylaxis past hospitalization for COVID patients, unless it would otherwise be indicated for non-COVID factors related to acute illness (such as protracted immobility). We recommend the IMPROVEDD score (Spyropoulous et al) to assess 42- and 77-day VTE risk, and to weigh this risk against bleeding risk when determining if extended prophylaxis is indicated. Some centers do recommend extended prophylaxis (generally up to 6 weeks after discharge). This remains an area of active investigation, and shared decision-making is appropriate.

Acute COVID-related VTE treatment after hospitalization. For thromboses associated with acute COVID infection, these are considered “provoked” thromboses and are treated with the same duration of anticoagulation as for non-COVID associated clots of a similar type (typically 3-6 months depending on resolution of the initial provocation, residual vein thrombosis, elevated inflammatory markers, other VTE risks including obesity, known inherited thrombophilia, or past VTE).

Delayed VTE. The duration of the hypercoagulable state from COVID is unknown. For patients who present later than 8-12 weeks after an COVID infection with a new VTE we would recommend considering this “unprovoked” and working up accordingly for hypercoagulable states. There is no data behind this specific time cutoff for post-COVID syndrome, this recommendation is based on the analogy of delayed VTE after surgery and may change in the future as more is learned. There is some evidence that antiphospholipid autoantibodies may be found in some cases (Zuo et al).

Arterial Thrombosis. For arterial clots without obvious structural abnormalities, cardioembolic source, or catheter-associated thromboses, hypercoagulability workup is generally warranted, regardless of COVID status or timing (for arterial clot workup and treatment see May et al).

Cardiac ConcernsCopy Link!

Chest Tightness and DiscomfortCopy Link!

Updated Date: April 19, 2021

Persistent chest discomfort following recovery is common (21.7%, Carfi et al) and can take quite some time to abate. The etiology is unknown but is hypothesized to be a combination of pleuritic or pericardial inflammation, costochondritis, musculoskeletal pain from coughing, or microvascular cardiac injury. Of course, more serious causes of chest pain are also possible, including myocarditis, myocardial infarction, spontaneous coronary artery dissection (Cannata et al), pulmonary embolism, new bacterial pneumonia, pleural effusion, and pneumothorax. Palpitations are also reported in about 9% of patients. These can be due to arrhythmias, but are often due to a debilitating but challenging to diagnose condition called Postural Orthostatic Tachycardia Syndrome (POTS) (which is addressed under dysautonomia). Frustrating for the provider and the patient is that standard testing, encompassing ECG, ECHO, stress test, heart rate monitors, and cardiac magnetic resonance imaging, tend to be normal in the majority of patients.

Management:

  1. New and worsening chest pain should always prompt workup for the above etiologies according to the nature and timeline of the pain. An exhaustive summary of the workup of chest pain is beyond the scope of this site, but generally include ECG, Echocardiogram, stress testing, Holter or event monitors, and cardiac MR depending on the indication.
  2. Ongoing but improving chest pain after concerning etiologies have been ruled out generally does not require treatment if it is not interfering with life. However, NSAIDs may be considered in the absence of contraindications. Colchicine (in half dose) can be used NSAIDs are ineffective. We do not recommend opiates.
  3. For Postural Orthostatic Tachycardia Syndrome see Dysautonomia and POTS.

Cardiac DysfunctionCopy Link!

Updated Date: April 19, 2021

Many patients who have recovered from COVID infection have signs of cardiac damage (see myocarditis). In individuals with no known cardiac involvement or specific complaints, studies of recovered COVID patients have documented changes in systolic function and elevated troponin (Puntmann et al; Huang et al). The long term implications of documented inflammation remain unknown. Patients with viral myocarditis will likely have some recovery over time, as is seen in other forms of virally-induced inflammation, but the extent of recovery is not yet known

  • In one study of 100 patients undergoing cardiac MRI 69-92 days after diagnosis, 78% of patients who had recovered had some signs of cardiac damage, and 60% had ongoing myocardial inflammation. Troponins were elevated in 76% of these patients, but cardiac function was preserved. This was independent of severity, pre-existing conditions, and the time of diagnosis (Puntmann et al).
  • This is true even among patients without prior cardiac disease. In one study of 26 college athletes diagnosed with COVID (none hospitalized, most asymptomatic), 46% showed evidence of myocarditis or prior myocardial injury by cardiac MRI ranging 12-53 days after diagnosis. (Rajpal et al)

Management:

  1. Whether screenings to detect cardiovascular damage should become a routine part of follow-up care for COVID-19 patients remains unclear.
  1. Patients with ongoing chest pain, palpitations, or signs of heart failure should be worked up for active disease as above.
  2. Completely asymptomatic patients with a history of mild disease likely do not need any post-COVID workup, though this may change as we learn more.
  3. Completely asymptomatic patients with underlying cardiac conditions or a history of severe COVID infection, comparing a pre-COVID and a post-COVID EKG is reasonable, as is obtaining troponin/ Nt-proBNP. If any abnormalities, work up as above.
  1. Patients who experienced a known cardiac injury (including MI, cardiac arrest, arrhythmia, or symptomatic myocarditis) due to COVID should be followed by a cardiologist.
  1. Serial echocardiogram and electrocardiogram at weeks 4-12 is generally indicated (George et al).
  2. Management of arrhythmia is covered in this article (Desai et al).
  3. Competitive athletes with cardiac damage should abstain from competitive sports or aerobic activity for 3–6 months until resolution of myocardial inflammation (by MRI or troponin). (Maron et al)
  4. Adhere to guideline-directed medical therapy. Despite initial concerns, RAAS inhibitors are considered safe to use in COVID patients.
  5. Standard lifestyle modifications are always recommended including
  1. Ensure daily physical activity, ideally following the American Heart Association Guidelines of at least 30 minutes of exercise per day for at least 6 days per week
  2. Smoking and alcohol cessation
  3. Lipid lowering medications if indicated
  4. Weight loss management for obese patients

Renal ConcernsCopy Link!

Updated Date: March 15, 2021

Incidence: Up to 40% of the hospitalized COVID-19 patients develop acute kidney injury (AKI) and 6.6% require renal replacement therapy (RRT). (Ng et al) Interestingly, one study found that 13% of patients in a cohort of hospitalized patients from Wuhan had normal eGFR on admission and no AKI while in hospital had reduced eGFR at 6 month follow up, suggesting a longer-term insult. (Huang et al).

Pathophysiology: The mechanism of renal injury is generally acute tubular necrosis on renal biopsies and autopsies. Microthrombi may play a role as well. A new entity, COVID-19-associated nephropathy (COVAN), is also possible. COVAN is characterized by a collapsing variant of focal segmental glomerulosclerosis with involution of the glomerular tuft. COVAN likely emerges from interferon and chemokine activation, with APOL1 risk alleles as a risk factor (similar to HIV) (Velez et al).

Management

  1. No known renal injury: Given the Huang et al study suggesting a possible late presentation of decreased eGFR, we recommend that where possible post-COVID patients who have not received a BMP get their eGFR tested prior to the prescription of renally-cleared medications. Similarly, patients with symptoms past 4 weeks or prior hospitalization may benefit from a basic metabolic panel on followup.
  2. Known renal injury unresolved at the time of discharge: AKI patients who do not recover to baseline kidney function should be followed by nephrology where possible after their discharge from the hospital for potential residual CKD.
  3. For patients on Renal Replacement Therapy (RRT): Although mortality is significantly higher in RRT requiring patients, 66% of the survivors have renal recovery to become RRT-independent (Ng et al) in the first month. Recovery rate rises to 92% with longer follow up at 150 days (Stockmann et al). Hence, these patients should be monitored closely by the dialysis units for signs of renal recovery such as lower pre-dialysis serum creatinine levels and increasing urine output. Hemodialysis patients should be directed into the dialysis units accepting COVID-19 patients until completion of COVID-19 quarantine period, whereupon they can return to their original units. These practices highly vary depending on location, available dialysis units and logistics.
  4. Transplant patients who had reduction in their immunosuppression regimen during COVID-19 pneumonia should have close follow up after recovery. If the immunosuppression was reduced during the infection, it can be increased back to pre-infection level cautiously within weeks to months depending on their clinical course (Kataria et al). However, clear data and guidelines remain absent about how to manage the immunosuppression in patients with COVID-19.

Post-ICU SyndromeCopy Link!

Updated Date: April 19, 2021

Patients admitted to ICU are at risk of post-ICU syndrome, a diverse constellation of physical, cognitive, and psychiatric deficits. In one study of 45 patients in New York 91% met criteria for post-ICU syndrome (Martillo et al).

  • 86.7 % had impairments in the physical domain
  • 58% had some degree of mobility impairment
  • 48% reported impairments in the psychiatric domain. 38% exhibited at least mild depression, and 18 % moderate to severe depression. 18% met criteria for PTSD.
  • This is also supported by a much larger study where post-ICU patients had a six-month estimated incidence of a neurologic or psychiatric diagnosis of 46%. This was the first such diagnosis for 25%. (Taquet et al).
  • 8% had impairments on cognitive screening.

Tracheostomy and Chronic ventilation: In a study of 1,800 patients requiring tracheostomies, only 52% were successfully weaned from mechanical ventilation after a month in a national cohort study from Spain (Martin-Villars et al).

Diagnostic Evaluation:

  • Cognitive: Patients with suspected PICS can undergo an assessment in the clinic, with referral to neuropsychiatry – if available – for further testing depending on results. It is not known to what extent results on this testing following hospital discharge predicts long-term impairment.
  • Montreal Cognitive Assessment (MOCA). This is a more sensitive test for cognitive impairment. Modified Mini-Mental State Examination (MMSE)
  • Mini-Cog
  • Mental health:
  • Hospital Anxiety and Depression Scale (HADS)
  • Impact of Events Scale-Revised (IES-R) for PTSD

Management:

Treatment for post-ICU syndrome is largely targeted at the specific physical and psychological needs of the individual. Some additional information is available at AfterTheICU.

Gastrointestinal ConcernsCopy Link!

Updated Date: October 1, 2021

Functional GI DisordersCopy Link!

Like other infections that are associated with transient gut inflammation, COVID-19 patients may experience persistent GI dysfunction after resolution of the acute infection, consistent with a post-infectious functional gastrointestinal disorder (FGIDs) / disorder of the gut-brain interaction (DGBI).

  1. Proposed criteria for diagnosis of post-COVID-19 FGID/DGBI can be found here in Table 2: Schmulson, The American Journal of Gastroenterology, 2021.
  2. Treatment is based on symptoms.

Chapter 15

Facilities Management and Operations

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

Facility LayoutCopy Link!

Literature Review (Hospital Operations): Gallery View, Grid View

Principles for Adapting Spaces for COVID-19Copy Link!

Updated Date: December 19, 2020

Designating COVID Care Areas: Where possible hospitals and clinics should establish designated areas for screening, triage, specimen collection, and COVID inpatient care. It is important to try to separate patients by how likely they are to have COVID to avoid putting patients who do not have COVID on wards or in waiting areas where they could contract it. Patients who have tested negative or who are not suspected to have COVID-19 should never be co-housed with COVID positive or patients under investigation (PUI) for COVID infection.

  • Distances between people should be at least 1 meter (WHO recommendation), and ideally 2 meters (CDC recommendation) in all contexts.
  • Using outdoor spaces and spaces with good filtration or air turnover can decrease risk. All indoor spaces should be sufficiently ventilated and COVID care areas should be negative pressure whenever possible.
  • Areas should be clearly marked with appropriate and standardized signage indicating the category of precaution and PPE that is required to enter.

Entrances, Screening and Testing AreasCopy Link!

Updated date: December 19, 2020

Entrances and Screening AreasCopy Link!

To ensure all persons entering a healthcare facility are screened for symptoms of COVID-19, most facilities have reduced the number of entrances and exits (see Screening and Triage). Infrastructure adaptations include:

  1. Designating separate entrances for healthcare workers and patients. This allows for all staff to pass through the same entrance and undergo active syndromic surveillance (WHO).
  2. Ensuring screening areas are open to air (e.g. vehicle pull ups or on walkway outside). If weather or infrastructure make this impossible, people should never need to cluster close together while waiting to be screened.

Waiting AreasCopy Link!

After screening positive, individuals should be directed to a waiting area for Acuity Triage. Within the waiting area for those who screen positive, patients must be able to remain 2 meters from any other patient, or have physical barriers in between. The waiting room should be clearly visible from the triage area.

Specimen Collection AreasCopy Link!

Updated Date:

  1. COVID respiratory sample testing and sputum collection should be done outside in an area designated for sample collection where possible. Some places use outdoor stands, tents, and drive-throughs.
  2. If respiratory sampling must be done indoors, ensure adequate PPE and air turnover/filtration where available.
  3. Blood finger pricks and blood draws can be done in the consultative space.

Open Shared WardsCopy Link!

Updated Date: December 19, 2020

Ward DesignCopy Link!

In settings where private rooms are not feasible, patients with confirmed and suspect COVID are admitted to open wards, ideally stratified by likelihood of disease (see Case Definitions and Isolation).

Finding space: To create these new wards, some healthcare settings combine or convert wards typically used for other reasons into COVID-19 treatment wards

Designs for open shared wards: for COVID care should ensure that there are donning and doffing areas at the entrance to the ward, separate staff work and break areas, supply areas within the ward, and separate bathrooms for patients and staff. There should be sufficient spacing between patient beds to maintain physical distancing (minimum 1 meter, or 2 meters if health care workers will move between beds). Screen walls/ partitions between beds in open wards should be used to reduce particle transfer between patients.

Grouping by risk level: Ideally, separate wards will be created by Likelihood of COVID Disease. If separate wards are impossible, patients may be cohorted within different areas of the same ward, grouped according to likelihood level. Use physical distance (>2m) or barriers (designs available in the BHI guidelines below) between groups to minimize risk to PUI patients under investigation who do not have COVID (for detailed information on barriers see BHI Infrastructure Tool below). Strict decontamination and other Facility-based IPC Practices must be performed between patients, and practitioners should see patients from the lowest to the highest likelihood areas.

Tool: COVID Ward Plans Developed by PIH and Build Health International (BHI)
Tool:
BHI Infrastructure Guidelines
Tool: Ward Design Troubleshooting

Critical Care Units: Care of critically ill patients should ideally take place in intensive care wards or units, high dependency units (HDU) (designated units with increased personnel, equipment, and monitoring capacity), or in a designated high-dependency area of a larger ward with adequate resources to care for acute patients. Separate wards (and/or separate negative pressure rooms with donning/doffing antichambers) are particularly important for critical care activities to reduce the risk of aerosol spread as aerosols are generated from some critical care activities such as intubation and nebulization (see Aerosol Generating Procedures).

BathroomsCopy Link!

Wards should have dedicated separate bathrooms for suspected and confirmed COVID-19 patients.

Staff Work AreasCopy Link!

Wards should have designated staff work areas. In open wards, work areas near patient beds should be considered potentially dirty, and staff should remain in PPE in these areas. For many environments it may help to designate clean staff areas in nearby rooms where staff can take breaks.

Donning and Doffing StationsCopy Link!

At each patient room or in each ward, there should be a donning area separated from patients where healthcare workers can put on protective gear. There should also be doffing stations for each area where PPE can be removed. Buckets and bins for used material should separate material that will be decontaminated (such as goggles), incinerated (such as cloves), or laundered (such as reusable gowns and linens). Dirty material should be handled with appropriate PPE

Ventilation and FiltrationCopy Link!

In settings where air is recirculated (including air conditioning system), air filtration is important for infection control and prevention. Air recirculation without filtration may increase risk of transmitting airborne pathogens. (Mousavi et al).

Airborne Infection and Isolation Rooms (AIIR, or negative pressure rooms) are the ideal rooms for PUIs and COVID patients. This will typically be done by ventilation systems that ensure an adequate number of air exchanges per hour. Natural ventilation is not recommended for smaller airborne isolation rooms as it does not reliably achieve the needed air exchanges per hour. The efficacy of natural ventilation systems depends on both building design and outdoor air movement as well as human factors such as ensuring windows remain open (Nardell). For a discussion of the use of open wards versus private rooms see Open Shared Wards.

When AIIRs are not available, air quality and safety measures must be taken to reduce infection risk. Strategies in these situations include:

  1. Optimizing ventilation to achieve as close to 12 Air Changes per Hour (ACH) as possible, including the addition of mixed mechanical and natural ventilation schemes.
  2. Ensuring that all recirculated air is adequately filtered through viral filtration systems (like HEPA).
  3. If augmenting ventilation is not an option, such as in some resource limited settings, windows that open to the outdoors away from trafficked areas (and not into other patient care areas) can decrease transmission risk. Here is a New York Times visualization on the impact of opening windows.

Tool: The Epidemic Task Force established by ASHRAE (The American Society of Heating, Refrigerating and Air-Conditioning Engineers) technical guidance for ventilation and filtration healthcare settings
Tool
: The ASHRAE Epidemic Task Force website with additional resources.

Air turnover: Key recommendations include improving direction of airflow in the direction of “more clean” to “less clean” (more important than air exchange rate), increasing filtration, and maintaining humidity at 40-60%. Minimum recommendation for COVID wards and rooms is 12 total air exchanges per hour (ASHRAE, CDC).

In settings where air needs to be recirculated to maintain conditioned spaces for patient comfort or where outside air changes are not feasible, recirculated air should be filtered through a viral filter (e.g. HEPA filtration system). In these cases, upper room germicidal ultraviolet (GUV) air disinfection may also provide infection control benefits. See section on upper room GUV below.

Viral Filters: High efficiency particulate air (HEPA) filters are notably useful for enhancing filtration. If replacing existing filters with higher-efficiency ones, care must be taken to balance pressure drop and fan speed to avoid unintentionally reducing air flow to the area. Some facilities have installed portable plastic anterooms outfitted with HEPA filters to simultaneously filter areas with high viral load and create a negative pressure seal (Mousavi et al).

Upper Room Germicidal Ultraviolet (GUV): Upper room GUV is proven to reduce transmission of tuberculosis and other infectious diseases in spaces with high potential of transmission (Nardel et al). The effect of upper room GUV is most prominent in closed spaces that use recirculating air conditioning systems and have low air change rates. In order to achieve maximum effectiveness of upper room GUV, low speed air circulation fans should be utilized to help circulate air within the space and ensure that air is continuously moving through the GUV zone. Consult a professional for design and installation of upper room GUV systems.

Infrastructure Standards (Resource-Limited Settings)Copy Link!

Updated Date: December 19, 2020

The table below summarizes suggested basic facility needs and standards in resource-limited settings in relation to water supply, power supply, power distribution/lighting, wastewater treatment, hazardous waste, oxygen, ventilation, internet connectivity, and fire safety. Each situation and site is unique, and this should be taken as suggested guidance and adapted to local needs, regulations, and resources.

Facility Needs

COVID-19 Treatment Center Standards

Water Supply

1. Supply

2. Potability

3. Reserve Capacity

4. Redundancy

5. Access Points

Adequate water supply is required for laundry, hand washing, cleaning, patient bathing, and drinking water for patients, staff, and family caregivers.

Water should be tested for pathogens and then treated as required to make it potable. One common disinfection method is to treat it with chlorine, in which case the water should be tested for residual chlorine to maintain levels between 0.5 and 2 ppm. See WHO and CDC guidelines for more details. (WHO, CDC)

Power Supply

1. Reliability

2. Capacity

3. Redundancy

4. Controls

Consistent electrical power is needed for safe basic care. For non-ICU level care, two reliable sources of electricity are needed. They can be any combination of diesel generator, solar and batteries, or utility grid connection. In cases where diesel generators are the only source of electricity, one primary generator and two backup generators are recommended for redundancy. Whatever the energy sources are, they should include an automatic transfer switch between the two primary sources of power. If there is an ICU, we recommend that there be a UPS with a size of at least 20kVAto feed the receptacles and lights for the space. Main circuit breakers and electrical panels should be readily accessible but located outside of patient areas so that a technician can service without PPE. Generators should be equipped with an automatic start signal fed from the automatic transfer switch. Generators should also have an external fuel tank. This fuel tank should be sized to provide fuel for a minimum of seven days continuous generator use.

Power Distribution and Lighting

1. Ward level

2. Distribution level

3. Documentation

Treatment and administrative areas need a minimum of 40 foot-candles of illumination at 1 meter above the floor. All lighting should be LED strip lighting securely hung on chain or wire at a minimum of 2.6 meters above the floor. All treatment areas should have a minimum of two duplex receptacles for each bed or patient exam chair. There should be no more than five duplex receptacles on each 20 amp circuit breaker. For ICU, there should be three dedicated 20 amp duplex receptacles for each bed all fed from uninterruptible power supply (UPS).

Wastewater Treatment

1. Treatment Level

2. Capacity

3. Monitoring and Maintenance

Wastewater can be treated in many different ways (See Compendium of Sanitation Systems and Technologies), and care must be taken to prevent the contamination of groundwater and water sources. One common, simple treatment method is a septic tank and leach field. In this case wastewater from hand sinks, janitorial sinks, toilets, and showers would be discharged by gravity into a holding tank sized based on flow requirements for a 48-hour retention time, and then discharged into a soak pit or leaching field, built to WHO and MSF Guidelines. Toilets in temporary facilities should be dedicated pit latrines which discharge into a lined tight tank of sufficient size to require pumping no more than twice per month. The lined pit latrine should have access and inspection hatches and be vented to promote breakdown of solids (see: World Bank Guidelines for Ventilated Improved Pit Latrines). There should be an overflow pipe for liquid wastewater at least 15 cm below the floor running to a separate soak pit.

Biohazardous, Pharmaceutical, and Chemical Waste

1. Collection and Sorting

2. Disposal

3. Staff Safety

Sharps containers should be mounted at between 1.3 to 1.4 meters above the floor. The container should be placed in a visible location, within easy horizontal reach, and below eye level. The container should also be placed away from any obstructed areas, such as near doors, under sinks, near light switches, etc. Containers should be clearly visible to the health care worker. There should be one 5-liter sharps container for every 4 beds or patient exam stations, and no less than 1 sharps container per room. The collection, storage and disposal of unwanted pharmaceutical and chemical waste should also be considered. (See WHO’s Safe Management of Wastes from Health-Care Activities)

Ventilation

See ventilation and filtration. Either mechanical ventilation through the use of exterior exhaust fans and opposite wall/end air intake louvers to achieve 12 air changes/hour (ACH) by volume in the space. It may be possible in some locations and climates to achieve 12 ACH in a ward by using natural ventilation especially a scheme that utilizes low intake and high exhaust. If this method is employed, it is strongly recommended that a professional engineer be consulted and that the space be tested for CO2 build up and transfer prior to the space being used.

If isolation rooms are desired, additional ventilation needs to be provided to meet ventilation requirements. Through-wall exhaust fans are an effective way to create negative pressure in the isolation room and to provide the recommended 12 ACH, while natural ventilation is not generally an effective method for achieving air flow in small rooms.

Oxygen

1. Supply

2. Distribution

3. Reserve Capacity

4. Redundancy

Anticipate that up to 40% of patients with COVID-19 will require O2.

An oxygen quantity of 10 liters per minute (LPM) per bed is recommended for sizing piped oxygen planning. RESERVE oxygen is required as well as REDUNDANT CAPACITY if the primary supply fails. If a manifold with high pressure oxygen cylinders is used, then there should be an adequate supply for all the beds for 24 hours of use at 6 liters per minute. In a 16-bed ward this would translate to 96 LPM. A 0.75 demand factor then can be applied so the 24-hour supply would be 96 LPM x 60 minutes x 0.75= 4320 liters per hour. An H-cylinder yields approximately 7,000 usable liters; so, for a 24 hour supply you would need 15 full cylinders. A reserve manifold (in addition to the 15 cylinders) should have a minimum of 4 full H-cylinders. The manifold and zone valve must be connected to an audible (and if possible) visual alarm to notify if there is a drop in oxygen pressure below 40 PSI. Redundancy in O2 means having a second oxygen source if the first one fails. For example, bedside O2 concentrators could be a redundant oxygen for a cylinder based system run out.

Network/Internet Connectivity

1. Reliability

2. Speed

3. Availability

Ability to connect to the internet (via wire or wireless connection) wherever layout of the facility deems necessary. Suggest: Dual Wired RJ45 connections at each convenient and/or required location. Wireless Access Points placed throughout the facility positioned for complete and optimum coverage. Consider a backup internet source from 4G cellular routers, if available.

Fire Safety

1. Fire Extinguisher

2. Smoke Detectors

3. Fire Assembly Points

4. Fire Evacuation Plan

Fire safety is especially important in buildings where a high volume of compressed oxygen is stored and used. Fire safety in temporary facilities in countries with little or no fire safety training and standards is always challenging. We suggest a flexible commonsense approach. Within the temporary wards, try to avoid using sheets or other linens for shading as these are a fire hazard. Fire extinguishers should be hung on the walls in locations that are accessible and highly visible. We recommend a minimum of two per every 1000 square feet, plus one by each exit and entrance. There should be fire extinguishers not more than 50 feet (or 15 meters) apart throughout the facility. The fire extinguishers should be clearly marked with a sign and arrow in the appropriate language.

Visitation PoliciesCopy Link!

Updated Date: September 24, 2020
Literature Review:
Gallery View, Grid View

When community rates of COVID are high, institutions may need to restrict visitors from entering health facilities. This protects visitors, patients and staff from unnecessary exposure, and facilitates safe physical distancing within the facility. Visitor policies should define 1) how visitors are screened and what PPE they must wear, 2) who can visit and under what circumstances, 3) any restrictions on visitor movement within the facility, and 4) any exceptions to the policies.

General principles for visitor policies:

  1. COVID-19 positive or PUI inpatients should not be are not allowed any visitors unless absolutely necessary. Institutional policies should specify what circumstances exceptions should be made in.
  2. Visitors must stay in the room for the duration of the visit.
  3. In most cases, only one visitor is allowed per day.
  4. No children under the age of 16 may visit.
  5. Visitors must wash hands, and follow universal face covering guidelines, and pass COVID symptom screening.

Examples of Possible Visitor Exceptions (must be adapted to each individual facility and epidemiologic circumstance):

  1. Adult outpatients are not allowed visitors unless they have a medical need for 1 assistant or guardian. Providing a letter stating medical need at the time of visit is recommended.
  2. Patients who are under the age of 18 may have one parents or guardian visitor. If facility circumstances allow, both parents/guardians should be allowed to visit.
  3. Patients with disruptive behavior may be visited by 1 person important to their care.
  4. Patients who have altered mental status or developmental delays may have 1 caregiver visit to increase their safety.
  5. Patients who receive home care may have 1 person visit to receive care-related education or training.
  6. Patients giving birth may have 1 partner and 1 other support person (up to 2 visitors)
  7. Patients on the Nursery/Neonatal Care Unit may have 1 partner and 1 other support person.
  8. Patients who are at end-of-life may have two 2 visitors.
  9. Patients undergoing surgery or procedures may have 1 visitor. Visitor must leave as soon as possible after the procedure/surgery.

In many countries, family members play a significant role in the daily care of inpatients. If visitors cannot be safely allowed during periods of high community transmission, facilities should plan for additional support staff to help patients meet their routine activities of daily living, including personal hygiene and feeding/nutritional needs.

Tool: Current BWH Visitation Policies

Surge PlanningCopy Link!

Updated Date: September, 2020

Stages of CrisisCopy Link!

Literature Review (Ethics): Gallery View, Grid View

During a COVID outbreak, growing patient numbers (known as a surge) can outpace hospital capacity, requiring adjustments to operations. Healthcare facilities should plan for sequential adjustments to operating procedures in order to meet capacity needs as safely as possible over the course of the crisis. Many healthcare organizations plan ahead for multiple stages of a surge, developing plans for patient care and management of resources.

Preparation and planning for surge events requires establishing an incident management committee, establishing and training triage teams, and assessing resources and systems in order to provide the best possible response to increasing patient numbers and changes in severity of illness in the patient population. Crisis situations present new requirements for infection control as well as new ethical challenges.

Conventional

Contingency

Crisis

Space

Usual patient care space maximized

Patient care areas repurposed (PACU, monitored units for critical care)

Non-traditional areas used for critical care or facility damage does not permit usual critical care

Staff

Additional staff called in as needed

Staff extension (supervision of larger number of patients, changes in responsibilities, documentation, etc)

Insufficient ICU trained staff available/unavailable to care for volume of patients, care team model required & expanded scope

Supplies

Cached/on-hand supplies

Conservation, adaptation and substitution of supplies with selected re-use of supplies when safe

Critical supplies lacking, possible allocation/reallocation or lifesaving resources

Standard of care

Usual care

Minimal impact on usual patient care practices

Not consistent with usual standards of care (Mass Critical Care)

ICU expansion goal

1.2x usual capacity (20% increase)

2x usual capacity (100% increase)

3x usual capacity (200% increase)

Resources

Local

Regional/State

National

Source: Christian et al. 2014

Planning for Pandemic DemandsCopy Link!

Updated Date: September, 2020

Incident Management CommitteeCopy Link!

Hospitals will ideally already have an established incident command structure for responding to crises. If a structure is not already established, an ad hoc structure can be created. The incident management committee should systematically estimate maximum total demand and assess maximum total capacity, including space, supplies, and staff. Resource data should be updated regularly and made as transparent and accessible as possible to all staff.

The Incident Management Committee should be formed of members across a wide range of facility services and departments. Members of the Committee should be leaders who can actively implement committee decisions. Committee members will be more effective if they consult and collaborate with the staff in their departments who perform direct tasks (for example, disinfection and bedside nursing care).

Recommended Membership of Incident Management Committee:

  1. Administration
  2. Communication
  3. Medical personnel
  4. Nursing administration
  5. Bedside nursing team clinical leaders
  6. Infection control
  7. Respiratory therapy
  8. Human Resources
  9. Security
  10. Pharmacy
  11. Laboratory
  12. Maintenance and engineering
  13. Dietary services
  14. Laundry, cleaning, and waste management

Reorganizing Services to Increase Hospital CapacityCopy Link!

Reorganization of services may be necessary to increase hospital capacity in surge situations. However, this can have unintended consequences for the health of populations; these consequences can increase over time. Each strategy should be continuously re-evaluated for its impact and ongoing necessity.

  1. Anticipate and plan for increasing patient numbers and changes in location of admissions.
  1. Identify which services must be maintained at all times; consider prioritizing services and temporarily stopping some services if necessary.
  2. Cancel elective (non-urgent) procedures and surgeries at least 1 week prior to the expected surge to free inpatient beds used for post-operative care.
  3. Whenever possible, shift to remote services, home visits, and home medication delivery.
  4. Minimize the number of people accompanying patients.
  5. Modify outpatient services as described below (see Adapting Outpatient Services).
  6. Organize appointment schedules and seating to ensure physical distancing.
  7. Reassure patients that routine care is available, as data show decreases in health service availability (WHO) and utilization (Czeisler et al) during the pandemic.
  8. Shift non-COVID inpatients to alternate treatment sites. For example:
  1. Shift malnutrition treatment to health centers or local community settings.
  2. Transition patients on long courses of antibiotics to oral antibiotics and discharge home.
  3. Move all routine hospital outpatient visits (antenatal care, children under age 5) to primary care centers and repurpose outpatient space for inpatient care.
  4. Identify alternative treatment sites for mild cases (churches, hotels, schools, etc.) and work to establish them with local authorities.

StaffingCopy Link!

  1. Plan for staffing interruptions, increased patient acuity, and need for additional staff. Anticipate a possible increase in staff illnesses and exposures.
  2. Analyze staff skills to develop plans for staff redeployment to meet gaps caused by increasing demand or staffing interruptions due to staff illness or quarantine.
  3. Consider training or rapid capacity building to allow staff to redeploy if needed.
  1. Plan to meet staffing needs as effectively as possible in the event of a contingency or crisis situation.
  1. Consider ways to reduce staff exposure by combining tasks sometimes done by different cadres or workers to limit entry and exit to COVID care spaces (for example, have a nurse take routine vital signs while administering a medication, even if routine vitals are normally taken by an aide).
  2. Review existing occupational health systems and plan for increased occupational illnesses in order to maintain a healthy workforce. Develop standardized staff screening for COVID symptoms and establish systems for follow-up and testing.
  3. Make it clear to all that team members should not be asked to assume added risk for the explicit purpose of minimizing someone else’s exposure (for example, a physician should not ask a nurse to enter an isolation room solely to facilitate a phone call between the doctor and the patient).

Adapting Outpatient Health ServicesCopy Link!

Community level

  1. Ensure visiting health professionals, community health workers and traditional healers have adequate PPE.
  2. For stable patients, work with the pharmacy team to obtain extended medication supplies to prevent unnecessary facility visits. Also consider decentralized pharmacy pick-up points.
  3. Create lists of high-risk patients to provide remote care, safety plans, and no disruptions to medication supply.
  4. Recruit community health workers, visiting nurses, and other team members to distribute medication refills and check on high-risk patients (determine and deliver an essential social support package).
  5. Maintain physical distancing and universal face covering guidelines according to local policies. Additional measures to enable social distancing include: additional make-shift clinic space (e.g., tents) and weekend clinics.
  6. For routine follow-up of chronic patients, try to extend periods between visits where possible; If relevant, address medication, testing, home visits, and other needs as described previously.

Remote Services and SupervisionCopy Link!

  1. Determine the best and most accessible platform for remote services, considering costs and network reliability.
  2. Utilize private, secure, and appropriate channels of communication.
  3. Establish referral pathways with supervisors for those with severe psychological distress.
  4. Allocate resources for air time to ensure staff and patients connect through phone and video calls.
  5. Define clear times when providers will be available for sessions.
  6. When clinical supervision is part of normal care, schedule times for regular clinical supervision with staff.

Crisis Standard of CareCopy Link!

Updated Date: September, 2020
Literature Review (Resource Allocation):
Gallery View, Grid View

Literature Review (Triage): Gallery View, Grid View

In the event of a patient surge, allocation of advanced care services may become necessary for treatments such as oxygen therapy or ventilation.

  1. A “crisis standard of care” is a set of principles to help guide triage of resources when there are insufficient resources to meet medical needs(including ICU beds, ventilators, dialysis machines, etc.) (Institute of Medicine 2012).
  2. A crisis standard is triggered by “a substantial change in usual healthcare operations and the level of care it is possible to deliver, which is made necessary by a pervasive (for example, pandemic influenza) or catastrophic (for example, earthquake, hurricane) disaster” (Institute of Medicine 2012).
  1. It must be formally declared by regional/state authorities and hospital leadership.
  2. It typically involves contingencies for different stages of a crisis.
  1. Declaring a crisis standard allows transparency in decision making, which is essential when resources are scarce and cannot be provided to all who need them (Biddison et al).
  2. Planning for these possibilities in advance is critical to ensure equity and to protect individual healthcare workers from needing to make resource allocation decisions.
  3. For a comprehensive framework for critical care resource allocation, please see an Example Model Developed at the University of Pittsburgh that many institutions incorporate into their plans.

Goals and Principles when Triaging ResourcesCopy Link!

This section is in process

Principles in Triaging ResourcesCopy Link!

The following is adapted from WHO Guidance:

  1. Just, equitable, and non-discriminatory distribution of scarce resources.
  2. Transparency in protocols and decision-making, with information made accessible and understandable at an elementary-school level in all languages spoken by the patient population.
  3. Non-abandonment of patients needing care. Palliative care must be provided to all patients with respiratory failure not receiving life sustaining treatment.
  4. Duty of organizations to plan and implement equitable management of resources in advance, in order to avoid placing the burden of triage decision-making on frontline healthcare workers. Committees or triage officers who are not providing direct patient care should be designated to guide these decisions.
  5. Standardized assessment: Triage officers and or committees should have a standardized approach to using clinical data for making triage decisions. One example is the Sequential Organ Failure Assessment (SOFA) calculator.
  6. Reassessment at routine intervals: Decisions should be ongoing as clinical parameters change.

Tool: WHO Guidance on Ethics and COVID-19

Resource Triage TeamsCopy Link!

  1. Consensus guidelines suggest that all decisions about triage should be made by a Triage Officer, not the bedside clinicians caring for patients (Christian).
  2. The Triage Officer should be a physician with critical care training.
  3. Decisions about triage should be made based on protocols established by the hospital. These protocols should be evidence-based and nondiscriminatory (Gostin et al).
  4. Bedside clinicians, patients, and families should have ways for appealing (challenging) triage decisions.
  5. An oversight committee should be established to review decisions made by Triage Officers to ensure consistent application of the triage protocol and to make decisions about appeals.

Equipment AdaptationsCopy Link!

Updated Date: July, 2020

Shared Ventilators and Ventilator AlternativesCopy Link!

Literature Review (Alternative Ventilator Options): Gallery View, Grid View

Use of Anesthesia Machine for Prolonged ICU VentilationCopy Link!
  1. In the event of shortage of ICU ventilators, anesthesia machines may be used for prolonged ICU ventilation (ASA/ASPF Ventilator Guidance)
  2. A quick reference sheet and hotline to set up and monitor a repurposed anesthesia machine are provided (ASA/APSF Quick Setup Instructions)(1-800-224-1001)
  3. Draeger and GE have provided specific guidance for their anesthesia machines (GE Guidance)(Draeger Guidance)
Use of Adapted BIPAP machinesCopy Link!

This section is forthcoming

Use of Single Ventilator Multiple PatientsCopy Link!

The ASA, SCCM, APSF, AARC, AACN, and CHEST societies have issued a joint consensus statement against using a single ventilator for multiple patients (Joint Statement On Multiple Patients Single Ventilator). Splitting of ventilators comes with many risks, including infection transfer between patients, difficulty positioning essential equipment, difficulty adjusting set respiratory parameters to meet individual patients’ needs and different clinical courses, difficulty controlling for sensed parameters (e.g. spontaneous respiration), alarm failures, measurement error in ventilator self-checks, and ethical dilemmas in prioritizing different patients’ treatment plans.

Supply Chain, Logistics, and ProcurementCopy Link!

Updated Date: December 19, 2020

Supply chain, logistics and procurement are critical aspects of a COVID-19 response. In some settings, particularly resource-limited settings, supply chain systems are underdeveloped; the guidance below reflects general principles for supply chain management in an emergency setting. In addition, operations staff, including logisticians, couriers, drivers, and warehouse workers, should receive basic education about COVID-19, including modes of transmission, proper hand hygiene, and any precautions required (wiping down equipment, wearing gloves to handle boxes, wearing masks, etc.)

GovernanceCopy Link!

  1. Clearly identify a specific individual to lead emergency supply chain efforts.
  2. If applicable, implement the use of emergency procurement processes to get necessary commodities and services where needed in time. Emergency procurement processes might include simplified bid solicitation or alternate approval procedures to ensure the most efficient and fastest response.
  3. Develop protocols for how the emergency and routine supply chains should interact, including storage, information systems, and purchasing.
  1. Identify regularly stocked items whose availability may be affected by COVID due to increased demand or global shortages and plan for additional procurement. It may also be necessary to identify items that can be used as substitutes for regularly stocked items.
  2. Commonly, emergency and routine supply chains are not separated at the beginning of the response and are later separated as dedicated funding becomes available. More restrictive funders, such as bilateral agencies (USAID, DFID, etc.) will require this.
  3. Clearly identify and authorize specific funds for emergency procurement in the earliest stages of the response and identify ongoing funding approval processes. Clearly identify a specific individual to lead clinical decisions for emergency supply chain efforts, including product specifications, substitutions, and forecasting.
  4. Ensure that supply chain governance is aligned with international and national institutions.
  1. For individual institutions and/or organizations in resource limited settings, clarify country-level governance in partnership with the Ministry of Health.
  2. Determine whether the UN Logistics Cluster System (Log Cluster) has been established and identify how the organization will engage.
  1. Among other things, this global mechanism coordinates requests for supplies and transportation across local, regional, and global implementers of all sizes, including donors (e.g. USAID, DFID) and local governments. LogCluster reports contain important information on these topics.
  2. If Log Cluster is not active, identify mechanisms to coordinate efforts with other partners involved in emergency response.
  1. Often this happens through the Ministry of Health and/or the implementing partner for bilaterally or multilaterally-funded supply chain projects.

Mapping and InterventionsCopy Link!

  1. Map current supply systems and available resources in health and other sectors.
  2. If UN LogCluster has been activated, utilize their mapping, which will include local & global partners (including military, private, and public agencies).
  3. Coordinate closely with district and national Ministry of Health Colleagues to understand public supply chain processes, even if they are seemingly sparse.
  4. Review PIH’s in-country supply chain management systems to identify risks that could disrupt existing systems (i.e. importation and customs, strategic stockpiling, storage, security, transportation, distribution, information systems, and requisition processes) for clinical and other essential supplies and equipment and create contingency plans.
  5. Identify the list of items considered essential to the response, including clinical and non-clinical supplies and equipment.
  6. Conduct review of stock levels and orders already in the pipeline of all items on a facility’s formulary. Use relevant procurement mechanisms and partnerships to address low stock levels, prioritizing essential items.
  7. Conduct rapid assessment of essential clinical and non-clinical equipment/systems and spare parts (oxygen, generators, incinerators, patient monitoring equipment, etc.). Use relevant procurement mechanisms and partnerships to increase access to needed equipment and stock spare parts.
  8. Conduct rapid assessment of vehicle fleet and stocks of spare parts (SUVs, ambulances, motos, etc.) as well as access to fuel. Use relevant procurement mechanisms and partnerships to avoid disruption to transportation and distribution networks.
  9. Map existing qualified suppliers, considering countries of origin for manufacturing and suppliers’ own emergency response procedures, including ability to obtain needed quality assurance documentation and navigate the changing environment around exportation. It may be necessary to expand sourcing to include new suppliers to decrease the risk of order delays or cancellations. For all items procured, ensure suppliers can provide the required quality assurance documentation.
  10. Work with suppliers and international bodies like the Log Cluster to identify items that may be in global shortage due to increased demand caused by the pandemic, or countries of origin that may experience delays in export. Consider placing additional orders with a diverse list of qualified suppliers to avoid disruptions to supply.
  11. Assess the capacity of the local market to meet increased demand for clinical and other essential supplies. Encourage preference for qualified local sources when possible.
  12. Assess systems providing visibility into stock levels and rates of consumption. Ensure clear communication systems are in place to communicate shortages and urgent needs.
  13. Assess storage and warehousing infrastructure and capacity including for cold chain products. It may be necessary to identify additional temporary storage for the items needed for the response, such as PPE.
  14. Assess basic in-country logistics and identify any major (or potential) infrastructure breakdowns (port closures, road or bridge disruptions, etc.) and create contingency plans.
  15. Map existing or anticipated disruptions to international shipping routes. Consider identifying additional logistics providers.
  16. Work with donors to ensure in-kind donations are aligned with identified needs.
  17. Assess likely funders of medium and long-term response efforts to anticipate allowable commodities and other restrictions.

Example Supply Lists:

Items needed in different areas of COVID-19 treatment centers. In addition, see PPE for a full list of necessary PPE.

Durable Equipment

Secondary Screening Area

Presumptive COVID-19 Area

COVID-19 Ward

Critical Care Area

Scales, adult and pediatric

X

X

X

Thermometers, infrared

X

X

X

X

Pulse oximeters

X

X

X

X

Vital signs machines, blood pressure

cuffs

X

X

X

X

Cardiac monitors

X

Stethoscopes

X

X

X

X

Ultrasound

X

X

Intravenous Infusion pumps

X

Beds with washable mattress

X

X

X

Bed linens

X

X

X

Pillows (washable)

X

X

X

Plastic chairs

X

X

X

X

Waste bins

X

X

X

X

Nurse / Provider station table

X

X

X

X

Pharmacy tables

X

X

X

Pharmacy cabinet

X

X

X

Shelves

X

X

X

Medication trolley

X

X

X

Refrigerator

X

X

X

White board with markers for patient tracking

X

X

X

Laryngoscope, various sizes

X

Hemocue

X

X

X

Glucometer

X

X

X

X

Light for clinical exams

X

X

X

X

Clock with second hand

X

X

X

X

Suction machine

X

Oxygen source – O2 tanks vs O2 concentrator

X

X

X

Ventilators

X

Consumables (products and disposable items)

Secondary Screening Area

Presumptive COVID-19 Area

COVID-19 Ward

Critical Care Area

Non-sterile gloves - small

X

X

X

X

Non-sterile gloves - medium

X

X

X

X

Non-sterile gloves – large

X

X

X

X

Bar soap + water source

X

X

X

X

Hand sanitizer

X

X

X

X

Bleach/Chlorine

X

X

X

X

Biohazard bag

X

X

X

X

Sterile gloves, assorted sizes

X

IV cannulae 18 – 24 gauge

X

X

X

IV tubing, 15-20 drops/mL

X

X

X

IV tubing, 60 drops/mL

X

X

X

Nasal cannula, pediatric and adult

X

X

X

O2 masks, pediatric and adult

X

X

X

O2 masks, non-rebreather, pediatric and adult

X

X

X

Adhesive tape

X

X

X

X

Bags, Urinal drainage, with non-return valve and tap, Sterile, 85cm tube, 2000mL

X

X

X

Nasogastric tubes for adults and children

X

X

X

Foley Catheters 12Fr and 16Fr

X

X

X

Needles, 18G, 21G and 25G

X

X

X

Spacers for inhalers

X

X

X

Syringes, 5ml, 10ml and 20 ml

X

X

X

Cotton wool, 500g roll

X

X

X

Wristbands, Patient Identification

X

X

X

Body bags

X

X

X

Sharps containers, 3 gallon

X

X

X

Tablet bags, Resealable, with Pictogram, 80mm x 100mm

X

X

X

Underpads, Tissue, 3 ply, 17in x 24in (chux)

X

X

X

Bags, Specimen transport, 6in x 9in

X

X

X

Tubes, Blood collection, Heparin (Green) Vacutainer Tubes Case

X

X

X

Tubes, Blood collection, K3 EDTA (K3E), 15% solution, Lavender, 6mL

X

X

X

Tubes, Blood collection, Serum, Silicone coated, Red, 6mL

X

X

X

HIV rapid tests

X

X

X

Hemocue microcuvettes

X

X

X

Glucometer strips

X

X

X

Lancets

X

X

X

Consumables for Mechanical Ventilation (if applicable)

Secondary Screening Area

Presumptive COVID-19 Area

COVID-19 Ward

Critical Care Area

Viral filter for ventilator circuit

X

Ventilator circuit

X

Ambu bag, adult, pediatric and neonatal

X

Endotracheal tube - 4.0 - 8.0

X

Water Sanitation, Hygiene, and Waste ManagementCopy Link!

Updated Date: September 24, 2020

WaterCopy Link!

  1. Persistence of SARS-CoV-2 in drinking-water is possible.
  2. There is no evidence to date about survival of SARS-CoV-2 in water or sewage, but this virus is likely to become inactivated significantly faster than non-enveloped human enteric viruses with known waterborne transmission (such as adenoviruses, norovirus, rotavirus and hepatitis A).

Disinfection and CleaningCopy Link!

FrequencyCopy Link!

COVID-19 virus can likely survive on surfaces, for variable amounts of time (hours to a few days). Survival time can depend on the type of surface, temperature, relative humidity and specific strain of the virus. Surfaces should be cleaned periodically with soap and water, and with disinfectant. The following is a suggested guide developed by Partners In Health as an example for some settings. Individual hospitals may have different policies. Always follow your hospital’s policy.

Offices and NON-CLINICAL areas

Recommended Minimum Cleaning and Disinfecting Frequencies

Type of Surface

Examples

Clean with Soap and Water

Clean with Disinfectant*

Minimally Touched Surfaces

Floors

Ceilings

Walls

Windows

When dirty

(At least 3 times/week)

After human contact When Dirty

(At least weekly)

Frequently Touched Surfaces

Door handles

Table tops / Desks

Light switches

Computers

Sinks/basins

Daily

Daily

CLINICAL AREAS (Including Isolation Units)

Recommended Minimum Cleaning and Disinfecting Frequencies

Type of Surface

Examples

Clean with Soap and Water

Clean with Disinfectant*

Minimally Touched Surfaces

Floors

Ceilings

Walls

Blinds

3 times daily + any known COVID-19 exposure

3 times daily + any known COVID-19 exposure

Frequently Touched Surfaces

Door handles

Table tops / Desks

Light switches

Computers

Sinks/basins

3 times daily + between each patient

3 times daily + between each patient

*Effective inactivation of the virus can be achieved with a 10 minute dwell time for chlorine bleach solutions. Other disinfectants may vary, follow the guidance on the packaging

Worker SafetyCopy Link!

  1. See these tables for PPE needed for clinical and support staff.
  1. Wear disposable gloves for all tasks in the cleaning process, including handling trash.
  2. Wash hands with soap and water immediately after gloves are removed. If water is unavailable, clean hands with alcohol-based hand rub.
  3. Gloves should be worn when handling and preparing bleach solutions, and eye protection should be worn in case of splashing.
  1. Close off areas to be cleaned and wait as long as practical before beginning cleaning and disinfection to minimize the potential for exposure to respiratory droplets.
  1. See air turnover and ventilation to know how long to wait before entering
  2. Open outside doors and windows to increase air circulation in the area.
  1. NEVER MIX SOLUTIONS

Soap and Water CleaningCopy Link!

  1. Always clean surfaces using a detergent or soap and water before disinfection.
  2. Remove visible pollutants (blood, secretions, excreta) completely.
  3. Damp mopping is preferable to dry mopping.
  4. Surfaces should be disinfected if they have come into direct human contact or are frequently touched.
  5. Always sterilize washing cloths, mops and other supplies used during cleaning.

DisinfectingCopy Link!

  1. Use freshly made solutions or premix. Follow manufacturer’s instructions or table below for appropriate dilution.
  2. Use proper safety measures (manufacturer guidance and worker safety measures)
  3. Wipe the area with the disinfectant solution using a cloth.
  1. Wipe cleaner regions first, then more contaminated regions.
  2. Dispose of or sterilize cloth immediately after use.
  1. Leave the disinfectant solution on the surface for sufficient time required to kill the virus (a minimum of 10 minutes for chlorine bleach).
  2. Always rinse chlorine/bleach with water after 10 minutes.
  1. Hydrogen peroxide and alcohol-based cleaners do not need to be rinsed.
Choosing the Right DisinfectantCopy Link!

Always note the listed concentration when using this product. Due to limited and changing availability of disinfectants, stocked items may change. Ensure staff working with disinfectants have knowledge for using each product correctly.

  1. For non-porous surfaces such as floors, sinks, toilets, walls: use chlorine bleach
  1. Be aware that chlorine can damage or stain computers, plastic, fabric and metal
  2. A solution of water and regular household chlorine bleach (sodium hypochlorite) can be used to create a disinfectant. Use 1 part 5% household bleach and 9 parts water (WHO). Other concentrations are below.
  1. For biomedical equipment: consult maintenance manuals to determine the best cleaning solution
  1. If maintenance manual is not available, use an Alcohol-based cleaner (if available) on biomedical equipment, electronics, computers, phones, screens, etc. If not available, clean with bleach.

Disinfecting Solution

Concentration

Directions

OK to use on

Do NOT use on

Diluted chlorine bleach (5.25% sodium hypochlorite)

0.5% (1:50)

Apply, leave for 10 minutes, rinse

Floors, desks, non-porous surfaces

Computers, phones, screens, fabric, can discolor plastic, metal

Chlorine (see table below to mix chlorine)

0.5%

Apply, leave for 10 minutes, rinse

Floors, desks, non-porous surfaces

Computers, phones, screens, fabric. Can discolor plastic, metal

Hydrogen Peroxide

0.5%

Apply

Floors, desks, non-porous surfaces, metal

Fabric

Ethanol / Ethyl Alcohol

62% minimum

Apply

Computers, Phones, Non-porous surfaces

Can discolor plastic

Isopropyl Alcohol

70% minimum

Apply

Computers, Phones, Non-porous surfaces

Can discolor plastic

Propanol

70% minimum

Apply

Computers, Phones, Non-porous surfaces

Can discolor plastic

Do NOT: use ammonia or vinegar; Do NOT: mix multiple disinfectants

Preparing Liquid Chlorine solutionsCopy Link!

% Solution

0.05 %

0.5 %

2 %

Use for:

Hands, skin, laundry, clothes

Floors, walls, equipment

Disinfection of stool, vomit, blood. Disinfection of corpses.

Bleach, 5% sodium hypochlorite

(5% active chlorine)

10 milliliters in

10 liters of water

1 liter in

10 liters of water

4 liters in

6 liters of water

Chlorine laundry powder

(30% active chlorine)

16 grams

(1 tablespoon) in

10 liters of water

16 grams

(1 tablespoon) in

1 liter of water

64 grams

(4 tablespoons) in

1 liter of water

Chlorine granules(HTH)

(70 % active chloride)

8 grams

(1/2 tablespoon) in 10 liters of water

8 grams

(1/2 tablespoon) in 1 liter of water

32 grams

(2 tablespoons) in

1 liter of water

ALWAYS label solutions using a permanent marker

Note: WaterGuard brand is 1.25% Sodium Hypochlorite. If this is used, different ratios are required.

Disposal of ExcretaCopy Link!

Updated Date: September 24, 2020

  1. Feces must be treated as a biohazard and handled as little as possible.
  2. Anyone handling feces should follow WHO contact and droplet precautions and use PPE, including long-sleeved gowns, gloves, boots, surgical masks, and goggles or a face shield.
  3. Conduct hand hygiene when there is suspected or direct contact with feces. Soap and water are preferred to the use of an alcohol-based hand rub if hands are visibly dirty.
  4. Excreta collected in diapers or bedpan should immediately be disposed of in a separate toilet or latrine that is used only by suspected or confirmed COVID-19 patients.
  5. Dispose of diapers as infectious waste, as they would be in all situations.
  6. Bedpans should be cleaned with a neutral detergent and water, disinfected with a 1% chlorine or 0.5% sodium hypochlorite solution, and rinsed with clean water. Dispose of rinse water in a drain, toilet, or latrine.

LaundryCopy Link!

  1. For handling soiled laundry used by COVID-19 patients, wear appropriate PPE, including heavy duty gloves, mask, eye protection, a long-sleeved gown, an apron if the gown is not fluid resistant, and boots or closed shoes.
  2. Place soiled laundry in clearly labelled, leak-proof bags or containers after carefully removing any solid excrement and putting it in a covered bucket to be disposed of in a toilet or latrine.
  3. Machine wash with warm water at 60−90° C with laundry detergent. The laundry can then be dried according to routine procedures.
  1. If machine washing is not possible, use a large drum to soak in hot water. Use a stick to stir and avoid splashing. Empty drum and soak linens in 1% chlorine for approximately 30 minutes. Rinse laundry with clean water and allow to dry fully in sunlight.
  2. Carefully remove excreta from surfaces (such as linens or floor) with a towel and immediately place it in a toilet or latrine. Treat soiled disposable towels as infectious use and reusable towels as soiled linens.

Dead Body ManagementCopy Link!

Updated Date: September 24, 2020

There is a risk of transmission of COVID-19 post-mortem. While taking appropriate precautions during the post-mortem period, it is essential to maintain the dignity of the deceased and respect relevant cultural and religious traditions.

Preparation of body for transfer from inpatient isolation ward

  1. All staff should perform hand hygiene before and after contact with the body.
  2. Ensure proper use of PPE, including gown, goggles/face shield, surgical mask and gloves.
  3. Remove all tubes, IVs and other lines from the patient.
  4. Wrap the body in cloth for transfer to the mortuary area.
  5. Ensure that all used equipment including the patient’s bed are cleaned, as per cleaning and disinfection guidelines.
  6. In the following circumstances a leak-proof or a double plastic bag may be necessary:
  1. Excessive fluid leakage
  2. Management of a large number of bodies
  3. Other situations where use of a body bag is recommended by standard mortuary procedures

Mortuary procedures

  1. Ensure that mortuary staff wear appropriate PPE, including gown, goggles/face shield, surgical mask and gloves. If activities have the potential to generate aerosols (such as autopsy), particulate respirators (N95 or FFP2 or its equivalent) should be worn.
  2. Ensure daily cleaning of the mortuary with chlorine or bleach per instructions in previous sections.

Transfer to family

  1. If culturally appropriate, place the body in a leak-proof or a double plastic bag.
  2. Alternatively, cover the body with a sheet. Plastic or cloth sheeting can be used.
  3. If covering the body is not possible, place a non-medical/fabric mask on the deceased before any movement or manipulation of the body

Counseling for the family

  1. Recommend that gloves be worn by people with physical contact with the deceased.
  2. Those preparing the body should instruct family and friends not to kiss or touch the deceased.
  3. Anyone who has assisted in preparing the body should wash hands thoroughly with soap and water when finished.
  1. Clothing worn during contact with the body should be immediately removed and washed after the procedure, or an apron or long-sleeved fluid resistant gown should be worn.
  1. If any ceremonial or burial activity may involve the splashing of bodily fluids, eye protection and medical masks should be word

Laboratory GuidelinesCopy Link!

Updated Date: September 24, 2020

The most commonly used and reliable assays for diagnosis of COVID-19 have been those based on molecular testing (NAAT), mainly RT-PCR (reverse transcriptase polymerase chain reaction). However, this technology requires highly skilled personnel, well-controlled laboratory environments, more expensive equipment, and in some instances long turnaround times. On the other hand, Rapid Tests are faster, less costly, simpler and easier to use, though their sensitivity and specificity are generally lower than those reported for RT-PCR assays. Choice between the two is covered in Testing (including WHO guidance on when it is acceptable to use RDTs). The WHO Interim Guidance also provides guidance on selection of tests for procurement and implementation. Ten factors are listed for consideration when selecting rapid Antigen tests such as reported performance, cost, kit contents, quality of available validation data, etc. It is noteworthy to mention that the antigen test procured by PIH, STANDARD Q COVID-19 Ag test is included in the validation studies performed by FIND as well as in the Emergency Use Listings assessment pipeline from WHO. Based on several verification studies performed at different countries, the overall sensitivity of this Ag test is 92.66% and the specificity is 99.25% under the conditions tested.

General aspects of the laboratory procedures:

  1. Controls: Laboratory procedure for external quality controls for antibody and antigen rapid tests. Good laboratory practices recommend the use of control materials. Users should follow the appropriate guidelines concerning the frequency and use of external control materials.
  2. Instructions: The package insert must be read completely before performing the test. Failure to do so may yield inaccurate test results.
  3. PPE. PPE must be worn correctly. See PPE During Testing.
  4. Biosafety measures: Observe biosafety measures and good laboratory practices when handling specimen or performing the test, such as:
  1. Clean work surface with disinfectant available before starting work.
  2. Place absorbent bench liner on work surface to capture potential splatters and splashes.
  3. Clean up spills thoroughly using an appropriate disinfectant.
  4. Handle all specimens as if they contain infectious agents
  5. Dispose of all specimens and test materials as biohazard waste.
  6. Laboratory chemical and biohazard wastes must be handled and discarded in accordance with all local, state, and national regulations.
  7. Clean the workbench and all non-disposable materials with disinfectant at the end of the work.
  1. Test kit and reagent storage: Store kits and reagents according to manufacturers’ specifications.
  1. Do not freeze or thaw unless instructed to
  2. Do not reuse elements that are not marked for reuse
  3. Do not mix buffers or reagents from different lots
  4. Do not use expired testing materials
  5. Do not use any damaged or unsealed products

PIH has offered specific SOPs for rapid tests and corresponding external controls that contain: Product Description, Test Principle, Warnings and Precautions, Sample Collection, Test Procedure, External Quality Control and Interpretation and Limitation of Test.

Chapter 16

Contact Tracing

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

Contact TracingCopy Link!

What is Contact Tracing?Copy Link!

Updated: February 1, 2021

Definition: Contact tracing is a process through which we can break chains of COVID-19 transmission, connect people to care, and identify clusters or outbreaks.

A comprehensive contact tracing program includes: conducting case investigation, identifying known exposed contacts, providing support for both positive cases and exposed individuals to safely isolate or quarantine until no longer infectious, and connecting unvaccinated individuals with vaccination resources.

Each person who is newly diagnosed with COVID-19 as a “case” is interviewed to enumerate their close contacts, e.g. those who have been within six feet for 15+ minutes over a 24-hour period. Those people who are identified as close contacts are notified and entered into a care management system, so that they can:

  1. Stay in safe quarantine according to CDC guidelines, supported by resources to protect themselves and their families
  2. Be tested for COVID
  3. Be monitored for symptoms

This process is the prospective component of contact tracing. Another important aspect of case investigation is called “source investigation”, or understanding where the person may have been infected with COVID-19. This is also called retrospective tracing. It is an important part of identifying clusters and is discussed further in Outbreak Response.

Contact tracing is not new for Departments of Public Health in the US. Local health departments have historically conducted contact tracing for other diseases, such as tuberculosis and sexually transmitted infections, but additional support is essential to manage the volume of cases and contacts associated with the novel threat of COVID-19. COVID-19 is also rapidly transmissible and can cause large outbreaks and move through communities quickly, introducing a higher degree of time pressure to testing and tracing than with some other conditions.

Contact tracing is a core public health tool used to respond to infectious disease outbreaks. It was identified early on as a way to combat COVID 19. Contact tracing, supplemented with community protection strategies, widespread testing, supported isolation and quarantine, and vaccination are the essential components of an effective pandemic response and of the broader public health toolkit for epidemic control and care delivery.

Previous contact tracing efforts:

  • Contact tracing helped prevent Ebola from spreading in other West African countries during the 2014-2016 outbreak. Studies describe the importance of contact tracing in also identifying gaps that can be addressed for future response.
  • Contact tracing was one of several interventions that helped control the SARS epidemic in 2003.
  • Contact tracing was a core component of smallpox eradication and is frequently used for HIV, STIs, and tuberculosis.

Contact Tracing in COVID-19Copy Link!

Since people can transmit COVID before or without symptoms, contact tracing combined with quarantine is most effective.

Evidence supporting contact tracing in COVID-19 includes:

Contact tracing is not only a purely epidemiological exercise to stop the spread of COVID-19, but also a care exercise that connects cases and contacts to social and material support. Given the potential logistical, emotional, and financial difficulties of quarantining or isolating for several days, effective contact tracing programs must also be able to connect cases and contacts to food, housing, medicine or other needs that arise over the quarantine and isolation period, like diapers or rent. Without contact tracing programs that can provide social support, people will have to choose between quarantining to stop the spread or continuing with their daily lives, thus perpetuating the pandemic. The inability to quarantine or isolate without direct support disproportionately affects poorer people and communities of color. Incorporating social support mechanisms into contact tracing programs also introduces the possibility of connecting cases and contacts to longer term support programs. This could include long-term enrollment in benefits programs for food, rental assistance or housing, or other resources in their community.

Core ElementsCopy Link!

Effective contact tracing programs are guided by four core elements:

  1. Technical expertise: Effective contact tracing programs that successfully reach cases and contacts require epidemiological knowledge, logistical capacity, clinical support guidelines, robust training and mentorship programs, and program management expertise. Programs should be set up by or in conjunction with public health departments.
  2. Adequate Staffing: Without adequate staffing and flexibility, contact tracing programs will be unable to flex up and down as the course of the disease changes. A nimble, trained workforce is necessary to create an effective contact tracing program.
  3. Leadership: In order to implement a contact tracing program at the state or local level, effective government leadership is necessary to champion and manage the program, combat misinformation, build community buy-in, and properly finance tracing efforts.
  4. Equity agenda: Equity must be built into any COVID-19 contact tracing program to combat the systemic racism that puts Black, Latinx, and Native American populations at a higher risk of contracting the virus and suffering poor outcomes including death; these populations also have higher needs for social support in order to safely quarantine or isolate. Contact tracing programs must address these needs.

Governance and PartnershipsCopy Link!

  • Building a robust COVID-19 contact tracing program requires high-level political support and significant resources.
  • COVID-19 contact tracing programs should be integrated within or in close partnership with public health departments who know the context of a region and have been doing this work for years. Contact tracing programs should amplify the existing work of local public health departments rather than centering themselves as the primary source of knowledge.
  • Strong leadership and buy-in at both the senior policymaker and programmatic levels is necessary to a successful contact tracing program. Without buy-in and political will at the highest level, contact tracing programs will often fail to achieve public buy-in or meet funding needs. Strong technical and operational leadership at the program level is needed to guide rapid decision-making about the evolving demands on CT programs.
  • Contact tracing programs should be well integrated within a state or local jurisdiction’s larger public health response to COVID-19, with strong operational integration and strategic alignment with testing, mitigation policy, supported isolation/quarantine and vaccine rollout efforts.
  • Clear definition of roles, responsibilities and decision rights are essential within complex partnership structures supporting contact tracing; a regular review and adaptation of governance arrangements is essential to keep up with rapidly evolving epidemic dynamics and partner ecosystems.

Roles and WorkflowsCopy Link!

An overview of the contract tracing process can be found here.

Key ComponentsCopy Link!

Effective contact tracing programs include 6 pillars. Some programs include all pillars within a central program, others rely on close partnership with separate workstreams:

  • Testing – Testing should be widespread and decentralized. A successful and timely contact tracing program relies on tight coordination with testing in order to quickly contact anyone who has tested positive for COVID-19 and to refer contacts for testing.
  • Case Investigation is the first step in the process after an individual is diagnosed with COVID-19, Case investigation involves interviewing a newly diagnosed person with COVID-19 to enumerate their contacts, ask about exposure details, monitor their symptoms over time, and ensure ability to safely isolate throughout their illness and provide connections to social support.
  • Contact Tracing – After cases identify their contacts, contact tracing involves contacting and interviewing those people identified as exposed contacts through case investigation, and monitoring their symptoms, recommending testing, ensuring their ability to safely isolate throughout quarantine and provide connections to social support.Contacts of positive cases should all be referred to testing.
  • Safe Isolation/Quarantine and Care Coordination – In order to successfully isolate/quarantine, CT programs need a coordinated approach to care and social support. Cases and contacts should be screened for social needs during case investigation and contact tracing. Some will require support to safely isolate or quarantine, and programs should mobilize or refer to different social resources including food, housing, transportation, economic support, and mental health and addiction resources.
  • Outbreak Investigation – As case investigators uncover sources of exposure through source investigation, contact tracing programs are often the first source of information that a cluster may exist. This can trigger further investigation of the cluster and the outbreak response.
  • Vaccination Resources -- COVID-19 identifies pockets of unvaccinated individuals, spreading more easily through unvaccinated populations than those that are vaccinated. By coupling contact tracing with vaccination resources, individualized education is provided to unvaccinated cases and contacts, followed by their connection to vaccination resources if willing. This also helps to prevent future outbreaks and severe disease.

Workforce considerations are described below.

Contact Tracer RolesCopy Link!

Contact tracers and case investigators are expected to provide a number of services to cases and contacts, including health education on COVID-19; collection of demographic, clinical and exposure information; isolation and quarantine advising; and resource assessment.

  • Education – What is COVID-19? How is it spread? What are the symptoms? How can I keep myself and my community safe and healthy? ​
  • Epidemiological Data Collection – Demographic information; clinical information (symptoms, hospitalizations, etc.); exposure information (to identify clusters/multiple exposures); contacts; resource assessment ​
  • Advising on Isolation/Quarantine – Importance of isolation for positive cases; importance of testing/quarantine for contacts; regular follow up and monitoring of symptoms; eventual clearing of people from isolation/quarantine​
  • Assessment for and Connection to Social Needs – Resource assessment during initial intake; continued assessment during follow up calls; liaising with community partners/care resource coordinators to arrange for resource delivery​

WorkflowsCopy Link!

Contact tracing includes definitive workflows between all pillars of the COVID response, including testing, treatment, and social support. This contact tracing workflow (page 13) can serve as a model for contact tracing programs creating their own workflows.

Care Resource CoordinationCopy Link!

Care resource coordination (CRC) is the process of identifying the needs and providing the social, material, and other supports needed to allow cases and contacts to safely isolate or quarantine. Social determinants of health and disparities in health outcomes are exacerbated by the fragmented social support landscape in the U.S. and care resource coordination is a must-have to address this problem. Many cases and contacts cannot isolate or quarantine without support.

CRC work is needed because:

  • Isolation (cases) and quarantine (contacts) break the chain of COVID-19 transmission.
  • For cases and contacts to isolate and quarantine for 7-14+ days, support systems need to be in place to allow for everyone to quarantine and isolate safely and effectively. Without ensuring that everyone can quarantine and isolate safely, people may be forced to choose between meeting basic needs and quarantining, thereby perpetuating the pandemic.
  • CRC work is essential to address health-related social needs and social determinants of health that directly impact individual health outcomes and population health.

Integrating with Contact TracingCopy Link!

Tool: PIH guide on the Components of CRC Programs

  • Case investigators and contact tracers should complete an initial needs identification for all cases and contacts.
  • Anyone with support needs should be referred to a care resource coordinator and a more detailed needs assessment is conducted.
  • CRCs then refer to or provide resources, using warm handoffs and ensuring linkage, in order to support cases or contacts to isolate/quarantine safely. CRCs also help connect individuals to those who can link them to long term benefits if needed.
  • Regular follow-up is required to assure additional resources are provided when necessary and symptoms are monitored.

Best PracticesCopy Link!

  • Screening for social support should be done early and often. Screening should start at testing sites and support should be offered throughout quarantine and isolation at regular intervals. All contact tracing staff should be trained to ask these questions in a non-threatening and culturally appropriate way as well as understand the variety of the types of support people may need.
  • CRCs should have local experience and work closely with local partners in order to properly connect cases and contacts with the necessary resources. Diverse language abilities are necessary to reach everyone who may be infected.
  • Establish clear referral protocols and lists of social support resources. Incorporate screening questions into contact tracing scripts. Ensure contact tracers and case investigators are trained on requirements for isolation/quarantine, common social needs, specific needs assessment scripts, and how to refer patients to a CRC.
  • The overarching goal of care resource coordination is linkage to care, but linkage assumes supply. Creative solutions are required to ensure sufficient referral pathways, including adaptive systems to facilitate referrals between CRCs and community based organizations and updated resource databases organized geographically and thematically.
  • Flag potential clusters for outbreak investigations. These clusters often identify pockets of unvaccinated individuals, who can then be connected with education, testing, and vaccination resources.

Workforce and TrainingCopy Link!

Key Roles and ManagementCopy Link!

There are three primary roles within a contact tracing program: case investigators, contact tracers, and care resource coordinators.

  • Case investigators engage with newly diagnosed COVID-19 patients, explain diagnosis and facilitate safe isolation; offer assistance; identify people they may have exposed to the virus (contacts); collect key information about the person; identify where the person may have been exposed and if they could be part of a cluster.
  • Contact tracers engage with the contacts of people diagnosed with COVID-19; explain their risk of infection and of transmitting the disease; assess symptoms; explain and facilitate safe quarantine; assess support needs; collect key information about the person.
  • Care resource coordinators engage with COVID-19 patients and their contacts to understand their resource needs to safely quarantine and isolate; connect them to essential material, financial, and social supports.

Programs should strive to combine the case investigator and contact tracer roles into one cross-trained role. This is for two major reasons: (1) to build a nimble workforce that can withstand large fluctuations in cases and contacts as the pandemic ebbs and flows and (2) to prepare employees for inevitable household transmission that requires them to speak to cases and contacts in the same phone call.

Support structures are necessary to manage the workforce of case investigators, contact tracers, and care resource coordinators, promoting productivity and wellbeing as well as improving program metrics and design.

  • Management and Leadership Team: Responsible for oversight and accountability of the program, as well as troubleshooting. Consists of program leadership, responsible for implementation & design, data, strategy, policy, communications, HR, clinical, and coordination with the government and DPH. Should continuously track program metrics to improve program efficacy.
  • For a contact tracing program to properly respond to an evolving pandemic and manage ongoing programmatic issues, a strong workforce management team must lead the program.
  • Training Team: Responsible for training all new CIs/CTs/CRCs and offering continued education for all employees. Training programs should be nimble and tightly connected to implementation updates.
  • Mentors and Peer Support: Given the numerous changing protocols needed in an adaptive CT program, mentors should be continuously available for questions and training of all employees. Peer support programs should be created to offer emotional assistance to employees given the taxing nature of the work.

Adapting to Evolving EpidemicsCopy Link!

Workforce quantification

  • A nimble workforce that can fluctuate in size depending on the course of the pandemic is necessary for a program to be effective. The number of contact tracers and case investigators needed will vary, depending on:
  • The number of new positive tests (cases)
  • The number of contacts per case
  • The average duration for each initial case investigation and contact tracing call
  • The number of follow up calls per case and contact, and their average duration
  • The responsibilities for outbreak investigation and response and linking with businesses or other places where larger exposures may have occurred
  • Workforce estimation tools can be useful to estimate how many CTs/CIs/CRCs are needed at a given time
  • Partners In Health and the Analysis Group created a workforce quantification tool to model the exact number of contact tracers and case investigators needed based on the epidemiological curve of the disease
RecruitingCopy Link!
  • Engage and reinforce existing community health workforce members and cadres, including Community Health Workers, and engage FQHCs as key staffing partners where possible.
  • Prioritize hiring from hardest-hit and most vulnerable communities; ensure recruiting and workforce partners have clear accountability for equity and diversity in hiring.
  • Prioritize hiring of staff with diverse language capacities to address the diverse language needs of your constituency.
  • For call center-based contact tracing programs, recruiting contact tracers with basic to moderate tech skills will increase the speed of training.
  • The program should clearly communicate to recruits the evolving nature of the program based on the spread of the infectious disease and the resulting contract length -- a need for flexibility should be a key component of recruiting.
Surge StaffingCopy Link!
  • Essential to rapidly respond to hotspots and outbreaks
  • Depending on local financing and governance (home-rules vs. more centralized approach to public health), options for how to ramp up surge staffing vary; approaches include:
  • Hire a centralized surge workforce to be deployed upon request to local departments, or operate virtual call centers with full statewide coverage
  • Provide grant funding to local health departments to hire and manage surge staff locally
  • Contract local CBOs to provide local health department staff with surge support

Community EngagementCopy Link!

ApproachCopy Link!

  • Contact tracing communications campaigns should be hyperlocal with connections to local leaders and influencers.
  • Building trust and sharing knowledge are the most important goals of any communications program.
  • Information should be accessible to everyone in a community – extensive multi-media campaigns, broad coalitions of community organizations, and engagement in representative languages are needed.
  • Information should be thoughtfully placed in relevant parts of the process, meaning the places people will likely go before being called by a case investigator if they do test positive (e.g. clinical officers, testing sites, etc.).

GoalsCopy Link!

  • Generate awareness about contact tracing.
  • Dispel misinformation by pointing the public to official and comprehensive sources of verified facts.
  • Rebuild trust in the public healthcare system among communities that have been historically marginalized.
  • Destigmatize COVID-19 by ensuring cases and contacts feel safe when they are contacted and are comfortable sharing their contacts.
  • Communicate that contact tracers are calling to help, connect people to medical services and social supports; they are not authority figures seeking to get them in trouble or affecting their immigration status.

StrategiesCopy Link!

  • All community engagement and education should be available in local languages.
  • Communicate with faith-based organizations and work with religious leaders to educate communities and build trust.
  • Partner with community organizations and support public events (e.g. food distributions, education sessions, etc.).
  • Coordinate with the DPH for events and campaigns around health promotion (e.g. mobile testing, flu vaccination).

Best PracticesCopy Link!

  • Public-facing communications for contact tracing programs should include multi-channel media campaigns in order to reach the highest number of people (i.e. TV, radio, Facebook/Instagram, Google Ads, and town halls).
  • Programs should develop partnerships at the community level (i.e. mayors/local councils, health centers, community centers, faith groups, immigrant groups, school superintendents, food pantries, etc.) in order to build trust and combat disinformation.
  • Anticipate transmission events and conduct proactive outreach to high risk locations to build trust, reinforce communication, and encourage safe practices (ex. Places of Worship ahead of religious holidays, bars ahead of St. Patrick’s Day/New Years, etc.).
  • All media should be developed in local languages and delivered at a hyper-local level (i.e. grocery stores, churches, etc.).
  • Consider working with communications companies (cell providers) for increased visibility on CallerID/getting through mobile spam filters.

Metrics and MonitoringCopy Link!

Defining and Tracking SuccessCopy Link!

Metrics should guide every contact tracing program in improving quality and ensuring program effectiveness

  • Collecting comprehensive data and ensuring quality reporting and dashboards are essential to monitor delivery across 4 key dimensions of an effective program: Scale, Speed, Retention, and Equity.
  • Demographic metrics provide a profile of the contract tracing program – all key performance indicators (KPIs) should be disaggregated by key demographic variables (i.e. gender, race, ethnicity, and language) and analyzed for differences.
  • All metrics should have defined targets and progress tracked against these targets; leadership should actively manage progress.

Effective contact tracing programs focus on 4 critical dimensions: equity, scale, speed, and retention.

  1. Equity: Are we responding to all unique needs with a social justice lens, and prioritizing the most vulnerable groups? Are CRC referrals at the optimized level?
  2. Scale: Has the response built up the infrastructure to meet demand (e.g., staffing capacity, social support resources)?
  3. Speed: Is the response happening quickly enough to drive the rate of infection below 1: < 3 days for the full cascade?
  4. Retention: Where is loss-to-follow-up occurring at each stage in the cascade (i.e. testing to case investigation to contact tracing to follow-up)?

Example MetricsCopy Link!

Tool: Partners In Health’s COVID-19 Data Evaluation: Metrics and KPIs, Section 3

Example metrics

  • COVID-19 response metrics: # of cases reached, # of cases identified and supported to isolate, # of contacts identified and supported to quarantine, # of educational materials provided
  • Social support metrics: % of individuals identified with resource need, referred, and received resource
  • Clinical support metrics: % of individuals identified with pre-existing conditions in need of healthcare, referred, and connected to clinical care
  • Timing metrics: show how long the entire cascade takes (from time of test to isolation/quarantine) with the goal of <3 days to drive R0 < 1

Tool: Sample Priority Metrics from PIH’s program in Immokalee, Florida

Tool: KPI guidance from PIH - these example key performance indicators (KPIs) provide an example of what contact tracing programs should be measuring and monitoring

Example Monitoring DashboardsCopy Link!

Tool: Partners In Health’s COVID-19 Data Evaluation: Metrics and KPIs, Sections 2 and 4

  • Case investigation metrics track case status and help identify problems in retention and scale
  • Care resource coordination metrics map vulnerability and equity among contacts, particularly demonstrating those who need support to quarantine

Digital and Technology SolutionsCopy Link!

Technology can be deployed at multiple times during the contact tracing process. However, technology requires humans to make it work. Contact tracing is not just an epidemiological exercise, it is an exercise in care that cannot be effective through technology alone.

Key digital and technology solutions for contact tracing include

  • Local epi surveillance systems: These are the systems of records for all communicable diseases. Positive lab tests are received here. COVID cases can then be transferred to the case & contact management platform. Local or state epi systems may not be able to support the scale of COVID-19 contact tracing.
  • Case and contact management platforms (CRMs): CRMs should be able to execute case investigation and contact tracing workflows at scale while collecting and storing data from calls. Data integrity and synchronization between the local epi system and CRM is of utmost importance, as duplicate cases and contacts should be minimized where possible, cases and contacts must be linked to capture the chain of transmission, and case data must be synced rapidly from the epi system to the CRM to ensure swift follow up.
  • Proximity tracking tools: Digital tracking systems, often on mobile devices, are used to determine contact between an infected patient and a user. These programs often use Bluetooth or GPS. In the US, adoption of proximity tracking tools has been slow due to privacy concerns. Notably, these automated tools cannot provide care resource coordination and though they can supplement manual contact tracing, they should not replace it.
  • Medical monitoring tools: These tools enable remote symptoms monitoring and referral to care and testing. Public health departments can enroll at-risk individuals in the app and monitor symptoms based on patient reports. Thus far, there has been limited data on adoption rates and effectiveness. These tools also lack functionality for referral to social support services and must be integrated into the CRM platform to connect patients to care.

Ensuring the privacy of a case and contact data is of the utmost importance. Contact tracing programs will not work if people don’t trust contact tracers to guard their information correctly and safely. Protected health information should only be used in reference to COVID-19 public health and individual care needs.

Tech DecisionsCopy Link!

  • Will the local disease surveillance database be used for the system of record for case data (i.e. MAVEN in MA, NEDSS elsewhere) or will a new system be deployed?
  • What case and contact management platform (CRM) will be used to centralize case investigation and contact tracing workflow (i.e. SalesForce, CommCare)?
  • What will the intersection be between the CRM and the epidemiological surveillance system? How will the CRM or epi system be updated to reflect the unique COVID-19 needs and considerations?
  • Will Bluetooth/GPS proximity tracking or symptom monitoring tools be used widely? If so, how will the contact tracing program interact with those tools?

Systems StrengtheningCopy Link!

Contact tracing systems have the opportunity to effect long-term system change.

  • Build community health programs that can also contribute to long-term healthcare system strengthening efforts
  • Contact tracing programs can build upon existing community health worker programs or become the foundation of a new one
  • Train contact tracers and community health workers to refer and accompany people to clinical care to improve access to healthcare even outside of the COVID-19 pandemic
  • Combine COVID-19 response strategies with other health promotion activities to increase trust in the health care system
  • Flu vaccination, basic primary care services, mobile COVID-19 testing, and COVID-19 vaccination

Chapter 17

COVID Outbreak Response

Please note that as of April 2023, this website is no longer actively being updated.Copy Link!

Outbreak ResponseCopy Link!

What is Outbreak Response?Copy Link!

Updated: February 2022

A COVID-19 cluster is commonly defined as when 2 or more people from different households share the same exposure source within a 14 day period. This definition may vary based on cluster type or jurisdiction. We define an outbreak as one or more related COVID-19 clusters where 4+ households share an exposure source at an organized social or community event, business, educational institution, or congregate setting.

For the purposes of this chapter, we discuss the application of outbreak investigation and response strategies regularly used in congregate settings in the context of non-congregate, community-based settings to reduce community transmission of COVID-19.

An outbreak response comprises all the steps necessary to investigate and contain an outbreak. This comprehensive strategy is designed to stop transmission by focusing on COVID-19 clusters.

  • Outbreak investigation refers to uncovering when and where a case was infected and who else may have been exposed there; mapping out the extent of the cluster(s) including all cases, close contacts, and casual contacts; identifying key areas for risk mitigation
  • Outbreak containment includes all the interventions (e.g. notifications, testing, vaccination, social support, preventive measures, referrals for treatments) put into action to suppress the cluster(s) and prevent future ones

Outbreak response addresses both close contacts and casual contacts of infected individuals. True close contacts are at highest risk for COVID-19 infection, but casual contacts can also be at risk for infection and therefore can propagate transmission if not tested following a possible exposure. Outbreak response is conducted regardless of vaccination status.

  • Close contacts are those with likely exposure that meet the criteria of ‘close contact’ (see exposure definition here). They typically need to be quarantined.
  • Casual contacts are people who do not meet the strict definition of close contact and those who do not need to quarantine but may have been exposed through sharing common spaces. For example, coworkers in a workplace outbreak who likely share airspace, people worshiping at the same church, people who attended the same party, etc.

Core Elements: Effective outbreak responses require a full range of COVID-19 public health interventions including: widespread notifications, contact tracing for cases, care resource coordination for cases and their close contacts in order to support isolation and quarantine, testing, vaccination, and treatment. The process as a whole is described in more detail in the Core Elements section.

Leadership: Ideally local health departments in conjunction with a well-trained contact tracing program lead outbreak response. (See Governance and Partnerships and Workforce and Training). This could mean the outbreak response team is integrated with the contact tracing workforce, or it could mean they communicate and collaborate with a separate contact tracing team.

An appropriate and comprehensive outbreak response strategy is:

  • Equitable: COVID-19 finds and thrives in environments with high social vulnerability. By bringing essential resources and outreach to high-risk settings where cases are already occurring, the strategy invests in marginalized communities to ensure equity in response.
  • Comprehensiveness and coordination: It unifies key pillars of response (testing, tracing, vaccines, treatment, supported isolation/quarantine), and the same team oversees performance of, or linkage to, each.
  • Efficient: It identifies locations that have cases for a reason—low vaccination rates, facility features, activities taking place—and targets interventions to them.
  • Impact: By working to identify and contain clusters early, interventions prevent spread in both the immediate setting and in the wider community. It uses existing public health infrastructure and workforce and unifies the outbreak control strategies already in place across the US. As testing volume drops, investigating clusters and intervening is even more critical to identify undiagnosed infections.
  • Flexible and Adaptable: Easily integrating into varied public health structures, outbreak response teams are part of the solution to the transition from emergency response to long-term systems improvement.

Outbreak Response in COVID-19Copy Link!

Updated: February 2022

COVID-19 is a clustering disease, and people are believed to be most infectious prior to symptom onset. This results in exposures often occurring in workplaces, schools, recreational activities, and other public-facing locations prior to individuals knowing that they are sick.

There are generally two types of COVID-19 outbreaks:

  1. Slower, ongoing chains of transmission
  1. These are seen frequently in workplaces where extended exposures result in many people being exposed, sometimes even despite efforts to implement preventive measures. These outbreaks can produce several cases each week and span over the course of weeks or months.
  1. Larger, more explosive outbreak events
  1. These can also occur in workplaces, but are frequently seen in public-facing locations, such as restaurants or nightclubs, places of worship, and high-risk exposure activities, such as youth sports and recreational activities where exertion and heavy breathing may increase exposure risk or where preventive measures are more difficult to implement.

Outbreak Response and EquityCopy Link!

COVID-19 has and will continue to exploit vulnerability within communities, exacerbating existing inequities. These vulnerabilities are often due to geographic, demographic, or socioeconomic characteristics of affected communities, but also extend into populations who are vulnerable to COVID-19 due to high risk behaviors. By following COVID in these vulnerable populations and focusing the public health response on outbreaks, departments can tailor their response to meet the needs of these specific communities, increasing access to resources essential for treatment of COVID-19 and minimizing the risk of ongoing transmission in the community. The vulnerability that rendered these populations susceptible to an outbreak in the first place is likely to render them susceptible to another outbreak in the future; acting quickly to provide education, tests, vaccines, and other resources reduces these chances while the community is most likely to heed public health advice.

Unvaccinated people are at highest risk of both contracting COVID and suffering negative health outcomes. Additionally, people who are unvaccinated may belong to social networks who share their hesitations about vaccination. Another strength of outbreak response is identifying pockets of unvaccinated people such that public health officials have an opportunity to engage, build trust, educate, and provide resources to this community and minimize their risks for subsequent outbreaks or exposures. Core Elements

Updated: December, 2021

There are five core elements of outbreak response:

  1. Widespread Notification: Notification to individuals extending farther than the close contact definition is critical in stopping the spread of COVID-19. Close contacts are those at highest risk for contracting COVID-19; however, exposures frequently occur over extended periods of time or involve activities that can increase the risk of transmission of COVID-19, despite those individuals not being strictly considered a ‘close contact’. Ensuring there is a system in place to notify these casual contacts is another critical piece to outbreak response. Widespread notifications are important in both high and low transmission settings, but the process by which people are notified of their potential exposure may change from setting to setting. For example, it may involve notifying employees at a workplace, members of a church, notices at a gym, or patrons at a restaurant, etc., and may employ phone calls, email, social media, or group text messages.. These widespread notifications help people make informed decisions about their health, symptom monitoring, and test seeking behaviors following a possible exposure.
  2. Testing: COVID transmits rapidly, and people are infectious prior to the development of symptoms. To get ahead of secondary COVID cases and ongoing transmission, a wide net should be cast when recommending and facilitating testing for those linked to a cluster, including close and casual contacts and household members. Testing should be recommended regardless of vaccination status. Those who test positive will enter the contact tracing program, where source investigation can take place. A process should be in place to be able to record, and include for investigations, home-based rapid test results. Mobile testing units, knowledge of nearby testing locations, and education for organization leaders are all good cluster-based testing strategies. When possible, sequencing the virus from positive tests can yield additional crucial information (see Sequencing below).
  3. Contact Tracing: Close contacts discovered through cluster investigation should be quarantined and entered into the contact tracing program. If resources are limited, high-risk close contacts can be prioritized to facilitate resource provision and treatment referrals.
  4. Tactical vaccination: Tactical vaccination involves deploying vaccination efforts specifically to people and communities who are part of an outbreak (close and casual contacts, or other connected people who may be at risk due to the outbreak, e.g. place of employment, place of worship, other places with indoor shared airspace, etc). Tactical vaccination often focuses education and outreach efforts on people who are unvaccinated or under-vaccinated, as they are most likely to become infected and infectious in the short- and long-term. It can include entire social networks who need access to vaccination. Tactical vaccination can be supported through mobile vaccination units, helping people register for local vaccine appointments, and educating individuals and trusted authorities linked to the cluster.
  5. Social support services: Those infected with or exposed to COVID should be connected to support services if they need help with accessing testing, resources for safely isolating/quarantining, vaccines, or treatment referrals. Because outbreaks tend to occur in vulnerable populations, incorporating resource coordination can connect people to the social safety net and – for example – reduce food insecurity by enrolling families in SNAP (previously called food stamps) and connecting them to food banks.

Comprehensive Outbreak ResponseCopy Link!

Updated: February 2022

A comprehensive outbreak response will include additional epidemiological, clinical, and public health measures to ensure the full scope of the outbreak is understood, contained, and future outbreaks are mitigated.

  • Sequencing: Where possible, all positive test samples should be sequenced for variants. This is especially important for clusters in communities with known or suspected variants or unusual outcomes. Sequencing can also be used to associate additional cases with clusters or even form the basis of detection of clusters when sequencing data is available in real time. When used in conjunction with information from source investigation, sequencing can be a powerful tool for understanding where transmission is occurring, which in turn can lead to more appropriate mitigation recommendations.
  • Therapeutics: Link people infected with or exposed to COVID, especially those at risk of severe outcomes, to monoclonal antibody therapy (as therapy or prophylaxis) and other COVID-19 therapeutics (paxlovid and malnupirovir). Treatment options continue to be developed and are becoming much more widely available. Outbreak response is an opportunity for early identification of contacts who may benefit from monoclonal antibody therapy and could be directly linked to providers for evaluation.
  • Location-specific guidance: Guidance tailored to the outbreak location should be given, including for ventilation, collection of patron lists, linkage with testing and vaccination services, and other measures that help with cluster prevention and investigation.
  • Community outreach: The outbreak response team should proactively conduct community outreach based on lessons from outbreak investigation and response. This can range from contacting similar locations to recommending preventive measures, talking to trusted authorities in the local community, providing educational materials, and local testing or vaccination resources directly to the locations.

Governance and PartnershipsCopy Link!

Updated: December, 2021

Coordination and communication

  • Outbreak response may involve coordination amongst a wide group of individuals and groups, including the outbreak team, local health departments (LHDs), state-level Department of Public Health (DPH) and those involved in liaising with the communities, businesses, mayoral offices, and others who are involved in the response to the outbreak.
  • Although LHDs and state-level DPHs are jurisdictional, COVID-19 is not. Communication across LHDs, and between LHDs and DPHs, is critical in responding to clusters.
  • Outbreak specialists should be in constant communication with all departments involved in a cluster; the jurisdiction of the cluster event itself and the jurisdictions of residence for each individual exposed or possibly exposed in the cluster event. Depending on the size and scope of the cluster, frequent communication with the DPH is recommended as well.
  • The ability to communicate clearly with business owners and community members is necessary to ensure swift exposure notifications and recommendations for testing and vaccination.

Investigation and response

  • During the cluster response, outbreak specialists can work with the LHDs to collect close contacts, casual contacts, and provide appropriate guidance to the business owners or hosts of social events throughout the cluster.
  • LHDs can deploy their testing and vaccination resources to identified cluster locations to mitigate spread and future outbreaks.
  • Following the investigation and completion of the cluster response, outbreak specialists can present a summary of the cluster, including a case map if available, to local officials involved in the response. These presentations and resulting discussion can help to glean lessons learned, prevent clusters in the same location or similar sectors, and can help guide policy decisions.
  • In addition to outbreak specialist-identified clusters, LHDs and other state officials can share tips with the cluster team to get ahead of transmission before significant transmission occurs in high-risk locations.

Workforce and TrainingCopy Link!

Key Roles and ManagementCopy Link!

Updated: February 2022

Outbreak Response teams can be as small as a few people or inclusive of complex team/unit structures with sector-specific focuses and a variety of workflows. Some settings will identify ‘flex’ team members from a regionally based pool that can be activated to support local outbreaks when needed.

The key roles of the outbreak investigation team itself include:

  • Outbreak Specialists:
  • Identify and investigate potential outbreaks: review exposure sources provided by COVID-19 cases; triage exposure sources and identify clusters; collaborate with specialized case investigators to conduct location outreach and exposure notifications;
  • Respond to outbreaks: collaborate with Care Resource Coordinators to identify and address social support needs and bottlenecks to testing, treatment, vaccination, and isolation/quarantine; connect cluster locations with testing and vaccination resources to prevent future outbreaks; provide guidance and technical support to the location in accordance with local guidelines
  • Document and report: map and document cluster across all cases and contacts; record data and metrics around cluster investigation; report to local and state governments swiftly and accurately;
  • Train: teach and mentor contact tracing staff on source investigation, epidemiology of clusters. The Outbreak Specialists often rely on the larger contact tracing workforce. It is critical that the bulk of the contact tracing workforce be trained at least on the basics of source investigation, epidemiological principles of clustering, and basic cluster investigation. The more well trained the contact tracing workforce is, the smaller and more agile the outbreak investigation team itself can be.
  • Specialized Case Investigators: Collaborate with outbreak specialists to support cluster investigations; conduct location outreach for exposure notifications; assist with widespread notification of individuals possibly exposed during outbreaks; explain risk of infection and transmitting COVID-19; connect the location and individuals with testing and vaccination resources; refer cases and contacts to Care Resource Coordinators if more complex resource needs are identified
  • Management and Leadership Team: Responsible for oversight and accountability of the team; review metrics to ensure appropriate prioritization; encourage collaborative workflow development and team building; adapt to evolving epidemics; HR and administrative support

These additional teams can be integrated within the outbreak response team, or work in partnership with them:

  • Contact Tracing Workforce: The outbreak-specific workforce outlined above relies on a strong contact tracing workforce, including contact tracers, case investigators, and care resource coordination. Communication pathways between these two teams are critical to identify outbreaks and provide continual training on epidemiology, clear data priorities, and standards on how data is recorded.
  • Care Resource Coordinators: Care Resource Coordinators (CRCs) provide social support and are often embedded within the contact tracing workforce to ensure cases and contacts have the material support they need for isolation/quarantine. Integrating resource coordination within outbreak response is important given the high number of cases/contacts that may be identified, often with a high degree of vulnerability. In addition, specifically putting resource coordination within the outbreak team means that these personnel can assist with community partnerships, navigate barriers to testing, vaccination, and treatment, and work with community partners on culturally appropriate communication strategies (including specific language needs).

Best Practices for HiringCopy Link!

Updated: December, 2021

  • Diverse workforce: Outbreaks occur in high-risk groups; these groups tend to be demographically, geographically, or behaviorally similar. An outbreak response workforce that is representative of the community will understand the cultural and societal contexts of the outbreaks. Language skills are critical for tracing and outbreak response; hiring a team that speaks the languages in the community is necessary.
  • Varied backgrounds: Knowledge of public health systems and disease control measures are beneficial in outbreak response programs, but are not the sole areas of expertise valuable to the response. Many other skillsets, such as communication, public relations, health education, and a variety of employment backgrounds are beneficial to outbreak investigation and response as these outbreaks are not restricted to typical public health environments. The ability to communicate and build trusting relationships with community members, businesses, and health departments is often a skill that can’t be easily taught, and is incredibly valuable in relationship building and mitigating transmission.

TrainingCopy Link!

Updated: February 2022

Tool: Training on ‘Introduction to COVID-19 Source Investigation & Clusters’

In ideal circumstances, a specialized outbreak response team can be built from the existing contact tracing workforce, but this may vary depending on the context. Outbreak specialists and specialized case investigators should have a basic understanding of case investigation and contact tracing. While not necessary, a moderate understanding of epidemiological and clinical principles is helpful for a fast-paced and quickly evolving outbreak response team.

Extensive academic backgrounds or first-hand knowledge of epidemiology beyond contact tracing experience is not required for most roles in the outbreak response team, however a leadership team that is able to guide new team members through necessary concepts is beneficial to the growth of the team.

Once the team has received the baseline COVID-19 contact tracing training modules (advanced case investigation, source investigation, retrospective contact tracing, basic COVID-19 epidemiology, introduction to clusters, methods for location outreach, etc.; view Training on ‘Introduction to COVID-19 source investigation & clusters for examples of training modules); strategic development should be ongoing for both the outbreak specialists and specialized case investigators, as disease and transmission dynamics evolve frequently, and should include:

  • Trend assessment (geographical, sector-specific, demographic, etc.)
  • Development of sector-specific strategies for communicating with businesses and other public locations
  • COVID-19 guidelines as they evolve for tracing, testing, and vaccination
  • Basics on COVID-19 testing options (pooled testing, outbreak testing, individual testing, PCR, antigen, etc.)
  • Basics on COVID-19 treatment options (monoclonal antibody therapies, COVID-19 antivirals, etc.)

Training for the outbreak response team should be ongoing. While formal initial trainings can be helpful to get the team started, informal and conversational seminars can be effective in facilitating ongoing training and in-depth understanding of public health and COVID-19 transmission dynamics

Workflows and AdaptabilityCopy Link!

WorkflowsCopy Link!

Updated: February 2022

Tool: Asking about and documenting COVID-19 exposure sources

Tool: Working with COVID-19 exposures in public or crowded locations
Tool: Widespread notification following COVID-19 exposures

Tool: How to make a case map for outbreak investigations

Outbreak response workflows are generally dependent on the level of transmission within the community. Source investigation is critical at all stages of an epidemic, but the specific outbreak response can be adapted based on high and low levels of COVID-19 community transmission and local prioritization.

At any time, the core workflow elements of outbreak response rely on:

  • Source Investigation: Understanding where COVID-19 comes from is critical to interrupting ongoing chains of transmission and preventing future outbreaks. Source investigation should be conducted with each confirmed, probable, or suspected case of COVID-19, and includes baseline questions case investigators and contact tracers use to assess possible high-risk exposure sources within the 14 days prior to symptom onset. As COVID-19 is a clustering disease, it’s likely that more than one person was exposed at that same exposure event. Source investigation is necessary in both high and low transmission settings. An approach for source investigation is included in Asking about and documenting COVID-19 exposure sources.
  • Location Outreach: Once an exposure source has been identified, the location needs to be informed of the potential exposure as quickly as possible. Conducting outreach to these locations serves as a touch point with management, the business owner, or other point person for the organization where preventive measures can be reinforced and any misunderstandings can be addressed. Asking general questions, such as “How many people have been out sick recently?” can expand the conversation and may provide information more than just the exposure the outbreak team has identified. The importance of widespread notification, testing, and vaccination can also be discussed with the point of contact. Rosters of employees, guests, patrons, or congregants can be collected so the outbreak team can assist in widespread and anonymous notifications of possible exposures and recommendations for testing. Guidance for making these notifications and starting investigations is given in Working with COVID-19 exposures in public or crowded locations. Should the point of contact prefer to do these notifications internally, the lead outbreak investigator can provide the necessary information for them to do so, examples provided in Widespread notification following COVID-19 exposures
  • Cluster Mapping: Mapping chains of transmission and outbreak events can often highlight transmission dynamics and help to visualize where preventive measures may have broken down. Maps can be simple, but are invaluable tools to help share the lessons learned with community partners and local or state government officials who may not be as closely involved in COVID-19 transmission dynamics as public health officials are. Cluster maps are valuable in both high and low transmission settings. Examples and instructions on cluster mapping can be found in How to make a case map for outbreak investigations

If a high percentage of cases can be thoroughly and completely interviewed by available staff, workflow modifications can include:

  • Individual Case-Linkages: Large clusters may not be as frequently identified in areas or seasons with low levels of community transmission, but cases are either imported into or spread within a community. Strategies should be developed to understand COVID-19 case demographic and geographic data to identify hidden linkages between cases. This strategy should be coupled with widespread testing and vaccination campaigns to protect these social groups and geographical areas from outbreaks in the event of an increase of COVID-19 community transmission. Real-time genomic sequencing, when available, can be very effective at uncovering some of these linkages if available.

Additionally, some elements can be added to these core workflows as resources allow and when needed based on the types of transmission that are most common in the area:

  • Community Outreach: Increases in transmission can be anticipated ahead of social events, holidays, and religious celebrations. Sector-specific community outreach is an effective tool for increasing messaging and education related to preventive measures. For example, if a church leader were inclined to send a COVID-19 prevention message to their congregation ahead of a large religious holiday, their community is likely to heed their advice much more seriously than a public health official. Every opportunity to engage community leaders and leverage their channels of communication should be used to mitigate outbreak events, in both high and low transmission settings.
  • Super-Spreading Event Response: COVID-19 is a clustering disease, and in areas with high levels of community transmission, larger outbreaks are common in workplaces and public-facing locations. Quick identification of these outbreaks is crucial, as workplaces and public-facing locations act as amplifiers for COVID-19 transmission, creating an opportunity for extensive household and community spread. Strategies should be developed to quickly identify these mega-clusters, coupled with swift sector-specific mitigation techniques and widespread exposure notifications. Testing and vaccination should be encouraged for staff and any community member associated with the outbreak location.

Adapting to the Evolving EpidemicCopy Link!

Updated: December, 2021

A core element of effective outbreak response is to remain dynamic and flexible, and to use the information that is being gathered in ongoing outbreaks to better discover and respond to future outbreaks. Some elements of this include:

  • Shifting trends: Outbreak response teams should continuously be monitoring for changes in disease transmission, severity, and affected populations. These shifts in COVID-19 trends should be monitored and reported to local and state public health authorities, and prioritization adjusted to accommodate as necessary. For example, shifts may occur in age patterns, types of exposure settings, vaccine breakthroughs, geography of outbreaks, etc.
  • Prioritization: As transmission dynamics shift, prioritization should also shift to ensure outbreak response is being targeted to the highest-risk populations. These priorities could be demographic, geographic, or sector-specific.
  • Surges: Numerous outbreaks are occurring during COVID-19 surges when community transmission is at its highest, but the types and size of outbreaks shift with transmission levels. Outbreak response teams should be dynamic and able to identify high priority outbreaks, defined by extent or risk for poor clinical outcome. These outbreaks should have resources made available as fast as possible for widespread testing and vaccination.

Data use and cluster detectionCopy Link!

Updated: December, 2021

Tool: Proposed Data Queries to Assist in Outbreak Investigations (provides a list of standard data queries that can be useful in cluster detection and identification)

Sophisticated tools like algorithmic cluster detection and text-based data mining can be helpful in cluster detection, but they should not form the basis of investigation. Standard methods of manual data review by outbreak specialists and case investigators, as well as clear communication across the teams and high-quality data collection, are a much more important starting point.

One of the key tools to detecting clusters is a data system that allows all users to both record key individual-level information related to clusters (e.g. source investigation results) and to ‘flag’ cases that are potentially part of cluster events. This can be done with the use of specific variables in the data system, or through external methods such as spreadsheets to track potential cluster leads. This allows cases of interest to be reviewed by the outbreak team, without requiring all contact tracing staff to be fully trained on investigation techniques.

Another key tool is the ability to generate data queries or perform complex searches in your data system. At minimum, outbreak specialists should be able to generate data queries to view the demographics (date of birth, language, gender, address, city of residence), basic clinical information (vaccination status, presence of symptoms, symptom onset date), and contact tracing data (exposure source, household connections, employer name/address). Additionally, the ability to perform keyword searches or filter these fields is required (e.g., searching for all cases in the data query among a given employer).

Metrics and MonitoringCopy Link!

Updated: December, 2021

Outbreak response metrics and evaluation should be conducted in tandem with evaluation of the standard contact tracing system. Much of the work of the outbreak response team will be dependent on the quality and availability of data collected through normal contact tracing. In particular, exposure sources and employer information are critical foundations of an outbreak response team.

Much like building the outbreak investigation team and outbreak investigation itself, monitoring of the program should be flexible and adapt to what is possible with the current team. Metrics and outcomes can range from simple (eg. number of clusters created, number of cases associated) to more complex (eg. percent of transmission explained, average cluster attack rates, and vaccine effectiveness).

Examples of metrics:

  • Number of clusters created in a time period
  • Number of clusters per transmission sector
  • Number of cases and contacts associated with clusters
  • Percent of contacts and casual contacts reached during cluster investigation
  • Numbers of people connected to testing, vaccination, and other social support services

Examples of exploratory data analysis to identify trends, confirm or readdress priorities, and identify gaps:

  • Proportion of cases coming from each language group
  • Proportion of cases coming from each age group
  • Rates of cases over time and by geography
  • Searching for zip codes, streets, or specific addresses with high case rates

The process of cluster identification, investigation, and analysis is a cycle where any one step should be informing the others. At a minimum, the following should be assessed regularly:

  • Proportion of an age group’s cases that are connected to clusters
  • Proportion of cases reporting a given primary language that are connected to clusters
  • Vaccination status of cluster identified and all cases identified
  • Attack rates and total sizes of clusters per sector and over time

Systems StrengtheningCopy Link!

Updated: February 2022

A fully integrated and comprehensive outbreak response team helps to target resources (human, time, and material) to populations that are the most vulnerable to COVID-19. Whether by geographic or demographic characteristic, or shared social beliefs on COVID-19 and/or vaccination, COVID-19 identifies individuals and locations that need the resources the most.

People are often more willing to discuss testing and vaccination following an exposure or outbreak to COVID-19, even if previously hesitant. Mobilizing education campaigns, testing, vaccination, treatment, and social support resources available to populations as they are personally impacted by COVID-19 increases their interest in using these resources. Improving vaccination in these populations prevents severe disease and future outbreaks, lessening the burden on the healthcare infrastructure and allowing Health Departments to prioritize other public health interventions in addition to COVID-19 response. Those who choose to get tested and vaccinated through an outbreak response can also in turn be trained to become vaccination ambassadors for their family, friends, wider social networks, and communities.

Community-wide mitigation measures are highly effective when the baseline knowledge in the community about disease prevention is low. As the community learns how to live with COVID-19, shifting the public health response to focus on those most vulnerable, or locations at highest risk for outbreaks, narrows the scope of the public health response while simultaneously focusing where it is most needed.