Covid 19 Treatmentguidelines
Covid 19 Treatmentguidelines
Covid 19 Treatmentguidelines
In response to the rapidly evolving COVID-19 pandemic, the National Institutes of Health assembled
a panel of experts to provide practical recommendations for health care providers and issued the first
version of the Coronavirus Disease 2019 (COVID-19) Treatment Guidelines on April 21, 2020. For
close to 4 years, the COVID-19 Treatment Guidelines Panel (the Panel) has critically reviewed the
growing body of research data on COVID-19 and used that information to develop and revise their
recommendations for treating patients with this disease. The Panel has released a total of 72 versions of
the Guidelines.
The federal COVID-19 Public Health Emergency ended in May 2023, and several professional societies
currently provide COVID-19 treatment guidelines for their medical specialties or subspecialties.
Accordingly, this will be the final update of the COVID-19 Treatment Guidelines.
The Panel members hope these Guidelines have been of value to health care providers, and they
appreciate the support and input they have received over the past 4 years.
The COVID-19 Treatment Guidelines website will remain available until August 16, 2024, and will
provide a downloadable PDF of the final version of the Guidelines.
The COVID-19 Treatment Guidelines were developed in response to the COVID-19 Public Health
Emergency declared by the U.S. Department of Health and Human Services in late January 2020. The
goal of the Guidelines was to provide clinicians with guidance on caring for patients with COVID-19.
Because clinical information about the optimal management of COVID-19 evolved quickly, a
multidisciplinary panel of experts frequently updated the Guidelines based on their assessments of the
emerging evidence on treatments for this disease.
Panel Composition
The COVID-19 Treatment Guidelines Panel (the Panel) co-chairs appointed Panel members with clinical
experience and expertise in adult or pediatric patient management, translational and clinical science, or
the development of treatment guidelines. Panel members included representatives from federal agencies,
health care organizations, academic institutions, professional societies, and the community. Federal
agencies and professional societies represented on the Panel include:
• American Association for Respiratory Care
• American Association of Critical-Care Nurses
• American College of Chest Physicians
• American College of Emergency Physicians
• American College of Obstetricians and Gynecologists
• American Society of Hematology
• American Thoracic Society
• Biomedical Advanced Research and Development Authority
• Centers for Disease Control and Prevention
• Department of Defense
• Department of Veterans Affairs
• Food and Drug Administration
• Infectious Diseases Society of America
• National Institutes of Health
• Pediatric Infectious Diseases Society
• Society of Critical Care Medicine
• Society of Infectious Diseases Pharmacists
The inclusion of representatives from professional societies does not imply that these societies endorsed
all elements of the Guidelines.
Appendix A, Table 1, provides the names and affiliations of the Panel members, ex officio members,
consultants, and support team members on the Panel roster as of the final update of the Guidelines.
Financial disclosures for the Panel members can be found in Appendix A, Table 2.
Epidemiology
Individuals of all ages are at risk of SARS-CoV-2 infection. However, the probability of severe
COVID-19 is higher in people aged ≥65 years, those living in nursing homes or long-term care facilities,
those who are not vaccinated against COVID-19 or who have poor responses to COVID-19 vaccines,
and those with certain chronic medical conditions. Data on comorbid health conditions among patients
with COVID-19 indicate that patients with cardiovascular disease, chronic kidney disease, chronic
obstructive pulmonary disease, diabetes with complications, neurocognitive disorders, and obesity
are at increased risk of severe COVID-19. The risk appears to be higher in patients with multiple
comorbid conditions. Other conditions that may lead to a high risk of severe COVID-19 include cancer,
cystic fibrosis, immunocompromising conditions, liver disease (especially in patients with cirrhosis),
pregnancy, and sickle cell disease. Transplant recipients and people who are taking immunosuppressive
medications are also at high risk of severe COVID-19.1 See Clinical Spectrum of SARS-CoV-2 Infection
for a description of the clinical manifestations of SARS-CoV-2 infection and a discussion of the
spectrum of disease.
Although COVID-19 vaccination does not eliminate the risk of SARS-CoV-2 infection, vaccination does
significantly reduce the risk of COVID-19–related morbidity and mortality, particularly in individuals
who are at high risk of progressing to severe disease.2,3
Key Considerations
• The COVID-19 Treatment Guidelines Panel recommends that health care providers, health
care systems, and payers ensure equitable access to high-quality care and treatment for all
patients, regardless of race, ethnic identity, or other minoritized identity or social status (AIII).
“Minoritized” refers to social groups that have been deprived of power and status by the dominant
culture in society and encompasses not just racial identities but other identities as well, including
gender identity and sexual orientation.4
• Promoting equitable care for these groups must include considering the full range of medical,
demographic, and social factors that may negatively impact health outcomes.
• Clinicians should be aware that pulse oximeters may not accurately detect hypoxemia in people
with darker skin pigmentation.5,6 This may delay treatment and lead to worse clinical outcomes in
patients with COVID-19.7 See Clinical Spectrum of SARS-CoV-2 Infection for more information.
• Supporting equitable access to high-quality care and treatment for all patients is now an imperative
for all health care organizations accredited by the Joint Commission, as well as a priority for the
Centers for Disease Control and Prevention (CDC) and other public health agencies.
SARS-CoV-2 Variants
Like other RNA viruses, SARS-CoV-2 is constantly evolving through random mutations. New mutations
can potentially increase or decrease infectiousness and virulence. In addition, mutations can increase the
virus’ ability to evade adaptive immune responses from previous SARS-CoV-2 infections or vaccination.
This viral evolution may increase the risk of reinfection or decrease the efficacy of vaccines.22 There is
evidence that some SARS-CoV-2 variants have reduced susceptibility to plasma from people who were
previously infected or immunized, as well as to anti-SARS-CoV-2 mAbs.23-25
Since December 2020, the World Health Organization has assigned Greek letter designations to
several identified variants. A SARS-CoV-2 variant designated as a variant of concern displays certain
characteristics, such as increased transmissibility or virulence. In addition, vaccines and therapeutics
may have decreased effectiveness against variants of concern, and the mutations found in these variants
may interfere with the targets of diagnostic tests. The variant of interest designation has been used for
important variants that are not fully characterized; however, organizations do not use the same variant
designations, and they may define their variant designations differently.26,27
In September 2021, the CDC added a new designation for variants: variants being monitored.
The data on these variants indicate a potential or clear impact on approved or authorized medical
countermeasures, or these variants are associated with cases of more severe disease or increased
transmission rates. However, these variants are either no longer detected or are circulating at very low
levels in the United States; therefore, they do not pose a significant and imminent risk to public health in
the United States.
The Omicron variant was designated as a variant of concern in November 2021 and rapidly became the
dominant variant across the globe. The Omicron subvariants BA.1, BA.1.1, and BA.2 emerged in early
to mid-2022, followed by the subvariants BA.4, BA.5, BQ.1, BQ.1.1, XBB, EG.5, HV.1, and FL.1.5.1.
The newer Omicron subvariants are generally more transmissible than previous variants and are not
susceptible to any of the anti-SARS-CoV-2 mAbs that were previously authorized for the treatment and
prevention of COVID-19.24,25,28,29
Data on the emergence, transmission, and clinical relevance of these new variants are rapidly evolving;
this is especially true for research on how variants might affect transmission rates, disease progression,
vaccine development, and the efficacy of current therapeutics. Because the research on variants is
moving quickly and the classification of the different variants may change over time, websites such
as the CDC’s COVID Data Tracker, CoVariants.org, and the World Health Organization’s Tracking
SARS-CoV-2 Variants provide regular updates on data for SARS-CoV-2 variants.
References
1. Centers for Disease Control and Prevention. Underlying medical conditions associated with higher risk
for severe COVID-19: information for healthcare professionals. 2023. Available at: https://www.cdc.gov/
Reinfection
Reinfection has been reported in people after an initial diagnosis of SARS-CoV-2 infection. Because
reinfection can be difficult to distinguish from persistent shedding (i.e., positive NAAT results persisting
for weeks or months), the CDC recommends using an antigen test instead of a NAAT in patients who
have symptoms compatible with SARS-CoV-2 infection who are within 90 days of recovering from
a previous SARS-CoV-2 infection. Because intermittent detection of viral RNA can occur, a negative
result on an initial NAAT followed by a positive result on a subsequent test does not necessarily mean a
person has been reinfected.11 When the results for an initial and subsequent test are positive, comparative
viral sequence data from both tests are needed to distinguish between the persistent presence of viral
fragments and reinfection. In the absence of viral sequence data, the cycle threshold (Ct) value from a
positive NAAT result may provide information about whether a newly detected infection is related to
the persistence of viral fragments or to reinfection. The Ct value is the number of PCR cycles at which
the nucleic acid target in the sample becomes detectable. In general, the Ct value is inversely related
to the SARS-CoV-2 viral load. Because the clinical utility of Ct values is unclear, an expert should be
consulted if these values are used to guide clinical decisions.
References
1. Centers for Disease Control and Prevention. Overview of testing for SARS-CoV-2, the virus that causes
COVID-19. 2023. Available at: https://www.cdc.gov/coronavirus/2019-ncov/hcp/testing-overview.html.
Accessed December 7, 2023.
2. Centers for Disease Control and Prevention. Isolation and precautions for people with COVID-19. 2023.
Available at: https://www.cdc.gov/coronavirus/2019-ncov/your-health/isolation.html. Accessed December 5,
2023.
3. Food and Drug Administration. In vitro diagnostics EUAs. 2023. Available at: https://www.fda.gov/medical-
devices/covid-19-emergency-use-authorizations-medical-devices/in-vitro-diagnostics-euas. Accessed
December 4, 2023.
4. Centers for Disease Control and Prevention. CDC’s influenza SARS-CoV-2 multiplex assay. 2022. Available
at: https://www.cdc.gov/coronavirus/2019-ncov/lab/multiplex.html. Accessed December 12, 2023.
5. Centers for Disease Control and Prevention. Interim guidelines for collecting and handling of clinical
specimens for COVID-19 testing. 2023. Available at: https://www.cdc.gov/coronavirus/2019-ncov/lab/
guidelines-clinical-specimens.html. Accessed December 7, 2023.
6. Centers for Disease Control and Prevention. Considerations for SARS-CoV-2 antigen testing for healthcare
providers testing individuals in the community. 2023. Available at: https://www.cdc.gov/coronavirus/2019-
ncov/lab/resources/antigen-tests-guidelines.html. Accessed December 11, 2023.
7. Centers for Disease Control and Prevention. Nucleic acid amplification tests (NAATs). 2023. Available at:
https://www.cdc.gov/coronavirus/2019-ncov/lab/naats.html. Accessed December 11, 2023.
8. Food and Drug Administration. Genetic variants of SARS-CoV-2 may lead to false negative results with
molecular tests for detection of SARS-CoV-2—letter to clinical laboratory staff and health care providers.
2021. Available at: https://www.fda.gov/medical-devices/letters-health-care-providers/genetic-variants-sars-
cov-2-may-lead-false-negative-results-molecular-tests-detection-sars-cov-2. Accessed December 7, 2023.
9. Food and Drug Administration. SARS-CoV-2 viral mutations: impact on COVID-19 tests. 2023. Available at:
https://www.fda.gov/medical-devices/coronavirus-covid-19-and-medical-devices/sars-cov-2-viral-mutations-
Summary Recommendation
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends COVID-19 vaccination for everyone who is eligible
according to the Centers for Disease Control and Prevention’s Advisory Committee on Immunization Practices (AI).
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
COVID-19 Vaccines
Recommendation
• The Panel recommends COVID-19 vaccination for everyone who is eligible according to the
CDC’s Advisory Committee on Immunization Practices (AI).
Rationale
Vaccination is the most effective way to prevent COVID-19. Two 2023–2024 mRNA vaccines,
BNT162b2 (Pfizer-BioNTech) and mRNA-1273 (Moderna), and the 2023–2024 recombinant spike
protein with adjuvant vaccine NVX-CoV2373 (Novavax)4 are currently available in the United States.
The adenovirus vector vaccine Ad26.COV2.S (Johnson & Johnson/Janssen) is no longer available in the
United States.
COVID-19 vaccination is recommended for everyone aged ≥6 months in the United States. The Food
and Drug Administration (FDA) Emergency Use Authorization fact sheet and the product label for
each vaccine provide detailed information on the vaccination schedule and the doses that are approved
or authorized for that vaccine. The type and dose of vaccine and the timing of the doses depend
on the recipient’s age and underlying medical conditions. The CDC regularly updates the clinical
considerations for the COVID-19 vaccines currently approved by the FDA or authorized for use in the
United States.5
Adverse Events
COVID-19 vaccines are safe and effective. Local and systemic adverse events are relatively common
with these vaccines. Most of the adverse events that occurred during vaccine trials were mild or
COVID-19 Treatment Guidelines 18
Pre-Exposure Prophylaxis
As of January 2024, no biomedical intervention other than vaccines prevents COVID-19 disease.
Previously, the FDA authorized the use of the anti-SARS-CoV-2 monoclonal antibodies tixagevimab
plus cilgavimab (Evusheld) as pre-exposure prophylaxis (PrEP) of COVID-19 in people who were
not expected to mount an adequate immune response to COVID-19 vaccination and in people with
COVID-19 vaccine contraindications.19 Due to the increased prevalence of Omicron subvariants that are
not susceptible to tixagevimab plus cilgavimab, this combination is not currently authorized by the FDA
for use as PrEP of COVID-19.20 It remains critical that these individuals:
• Keep up to date with COVID-19 vaccination unless a contraindication exists.
• Take precautions to avoid infection. The CDC provides information on the prevention of
COVID-19 in people who are immunocompromised.
• Be tested for SARS-CoV-2 infection if they experience signs and symptoms consistent with
COVID-19 and, if infected, promptly seek medical attention.
References
1. Centers for Disease Control and Prevention. COVID-19 overview and infection prevention and control
priorities in non-U.S. healthcare settings. 2023. Available at: https://www.cdc.gov/coronavirus/2019-ncov/hcp/
non-us-settings/overview/index.html. Accessed December 1, 2023.
2. Centers for Disease Control and Prevention. How to protect yourself and others. 2023. Available at: https://
www.cdc.gov/coronavirus/2019-ncov/prevent-getting-sick/prevention.html. Accessed December 1, 2023.
3. Centers for Disease Control and Prevention. Interim infection prevention and control recommendations for
healthcare personnel during the coronavirus disease 2019 (COVID-19) pandemic. 2023. Available at: https://
www.cdc.gov/coronavirus/2019-ncov/hcp/infection-control-recommendations.html. Accessed December 1,
2023.
4. Food and Drug Administration. FDA authorizes updated Novavax COVID-19 vaccine formulated to
better protect against currently circulating variants. 2023. Available at: https://www.fda.gov/news-events/
press-announcements/fda-authorizes-updated-novavax-covid-19-vaccine-formulated-better-protect-against-
currently. Accessed December 1, 2023.
5. Centers for Disease Control and Prevention. Use of COVID-19 vaccines in the United States. 2023. Available
at: https://www.cdc.gov/vaccines/covid-19/clinical-considerations/covid-19-vaccines-us.html. Accessed
December 1, 2023.
6. Centers for Disease Control and Prevention. Interim considerations: preparing for the potential management of
anaphylaxis after COVID-19 vaccination. 2022. Available at: https://www.cdc.gov/vaccines/covid-19/clinical-
considerations/managing-anaphylaxis.html. Accessed December 1, 2023.
7. Food and Drug Administration. Fact sheet for healthcare providers administering vaccine (vaccination
providers): Emergency Use Authorization (EUA) of the Janssen COVID-19 vaccine to prevent coronavirus
disease 2019 (COVID-19). 2022. Available at: https://www.fda.gov/media/146304/download.
8. See I, Su JR, Lale A, et al. US case reports of cerebral venous sinus thrombosis with thrombocytopenia after
Ad26.COV2.S vaccination, March 2 to April 21, 2021. JAMA. 2021;325(24):2448-2456. Available at:
https://pubmed.ncbi.nlm.nih.gov/33929487.
9. See I, Lale A, Marquez P, et al. Case series of thrombosis with thrombocytopenia syndrome after COVID-19
vaccination—United States, December 2020 to August 2021. Ann Intern Med. 2022;175(4):513-522. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/35038274.
10. Centers for Disease Control and Prevention. Selected adverse events reported after COVID-19 vaccination.
2023. Available at: https://www.cdc.gov/coronavirus/2019-ncov/vaccines/safety/adverse-events.html.
Accessed December 1, 2023.
11. Hanson KE, Goddard K, Lewis N, et al. Incidence of Guillain-Barré syndrome after COVID-19 vaccination in
the Vaccine Safety Datalink. JAMA Netw Open. 2022;5(4):e228879. Available at:
https://pubmed.ncbi.nlm.nih.gov/35471572.
12. Abara WE, Gee J, Marquez P, et al. Reports of Guillain-Barré syndrome after COVID-19 vaccination
in the United States. JAMA Netw Open. 2023;6(2):e2253845. Available at: https://pubmed.ncbi.nlm.nih.
gov/36723942.
13. Centers for Disease Control and Prevention. COVID-19 vaccines while pregnant or breastfeeding. 2023.
COVID-19 Treatment Guidelines 20
Patients with SARS-CoV-2 infection can experience a range of clinical manifestations, from no
symptoms to critical illness. In general, adults with SARS-CoV-2 infection can be grouped into the
following severity of illness categories; however, the criteria for each category may overlap or vary
across clinical guidelines and clinical trials, and a patient’s clinical status may change over time.
• Asymptomatic or presymptomatic infection: Individuals who test positive for SARS-CoV-2 using
a virologic test (i.e., a nucleic acid amplification test [NAAT] or an antigen test) but have no
symptoms consistent with COVID-19.
• Mild illness: Individuals who have any of the various signs and symptoms of COVID-19 (e.g.,
fever, cough, sore throat, malaise, headache, muscle pain, nausea, vomiting, diarrhea, loss of taste
and smell) but do not have shortness of breath, dyspnea, or abnormal chest imaging.
• Moderate illness: Individuals who show evidence of lower respiratory disease during clinical
assessment or imaging and who have an oxygen saturation measured by pulse oximetry (SpO2)
≥94% on room air at sea level.
• Severe illness: Individuals who have an SpO2 <94% on room air at sea level, a ratio of arterial
partial pressure of oxygen to fraction of inspired oxygen (PaO2/FiO2) <300 mm Hg, a respiratory
rate >30 breaths/min, or lung infiltrates >50%.
• Critical illness: Individuals who have respiratory failure, septic shock, or multiple organ
dysfunction.
SpO2 is a key parameter for defining the illness categories listed above. However, pulse oximetry has
important limitations (discussed in more detail below). Clinicians who use SpO2 when assessing a
patient must be aware of those limitations and conduct the assessment in the context of that patient’s
clinical status.
The risk of progressing to severe disease increases with age and the number of underlying conditions.
Patients aged ≥50 years, especially those aged ≥65 years, and patients who are immunosuppressed,
unvaccinated, or not up to date with COVID-19 vaccinations are at a higher risk of progressing to severe
COVID-19.1,2 Certain underlying conditions are also associated with a higher risk of severe COVID-19,
including cancer, cardiovascular disease, chronic kidney disease, chronic liver disease, chronic lung
disease, diabetes, advanced or untreated HIV infection, obesity, pregnancy, cigarette smoking, and being
a recipient of immunosuppressive therapy or a transplant.3 Health care providers should closely monitor
patients who have COVID-19 and any of these conditions until clinical recovery is achieved.
The initial evaluation for patients may include chest imaging (e.g., X-ray, ultrasound or computed
tomography scan) and an electrocardiogram, if indicated. Laboratory testing should include a complete
blood count with differential and a metabolic profile, including liver and renal function tests. Although
inflammatory markers such as C-reactive protein (CRP), D-dimer, and ferritin are not routinely
measured as part of standard care, results from such measurements may have prognostic value.4-7
The definitions for the severity of illness categories also apply to pregnant patients. However, the
threshold for certain interventions is different for pregnant and nonpregnant patients. For example,
oxygen supplementation for pregnant patients is generally used when SpO2 falls below 95% on room air
at sea level to accommodate the physiologic changes in oxygen demand during pregnancy and to ensure
COVID-19 Treatment Guidelines 22
Mild Illness
Patients with mild illness may exhibit a variety of signs and symptoms (e.g., fever, cough, sore throat,
malaise, headache, muscle pain, nausea, vomiting, diarrhea, loss of taste and smell). They do not
have shortness of breath, dyspnea on exertion, or abnormal imaging. Most patients who are mildly ill
can be managed in an ambulatory setting or at home. No imaging or specific laboratory evaluations
are routinely indicated in otherwise healthy patients with mild COVID-19. Patients aged ≥50 years,
especially those aged ≥65 years, patients with certain underlying comorbidities, and patients who are
immunosuppressed, unvaccinated, or not up to date with COVID-19 vaccinations are at higher risk
of disease progression and are candidates for antiviral therapy.1,2 See Therapeutic Management of
Nonhospitalized Adults With COVID-19 for recommendations regarding anti-SARS-CoV-2 therapies.
Moderate Illness
Moderate illness is defined as evidence of lower respiratory disease during clinical assessment or
imaging, with an SpO2 ≥94% on room air at sea level. Given that pulmonary disease can progress
rapidly in patients with COVID-19, patients with moderate disease should be closely monitored. See
Therapeutic Management of Nonhospitalized Adults With COVID-19 for recommendations regarding
COVID-19 Treatment Guidelines 24
Severe Illness
Patients with COVID-19 are considered to have severe illness if they have an SpO2 <94% on room
air at sea level, PaO2/FiO2 <300 mm Hg, a respiratory rate >30 breaths/min, or lung infiltrates >50%.
These patients may experience rapid clinical deterioration and should be given oxygen therapy and
hospitalized. See Therapeutic Management of Hospitalized Adults With COVID-19 for treatment
recommendations.
Critical Illness
SARS-CoV-2 infection can cause acute respiratory distress syndrome, virus-induced distributive (septic)
shock, cardiac shock, an exaggerated inflammatory response, thrombotic disease, and exacerbation of
underlying comorbidities.
The clinical management of patients with COVID-19 who are in the intensive care unit should include
treatment with immunomodulators and, in some cases, the addition of remdesivir. These patients
should also receive treatment for any comorbid conditions and nosocomial complications. For more
information, see Critical Care for Adults and Therapeutic Management of Hospitalized Adults With
COVID-19.
References
1. Skarbinski J, Wood MS, Chervo TC, et al. Risk of severe clinical outcomes among persons with
SARS-CoV-2 infection with differing levels of vaccination during widespread Omicron (B.1.1.529) and Delta
(B.1.617.2) variant circulation in Northern California: a retrospective cohort study. Lancet Reg Health Am.
2022;12:100297. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35756977.
2. Taylor CA, Patel K, Patton ME, et al. COVID-19–associated hospitalizations among U.S. adults aged ≥65
Years—COVID-NET, 13 states, January–August 2023. MMWR Morb Mortal Wkly Rep. 2023;72(40):1089-
1094. Available at: https://www.ncbi.nlm.nih.gov/pubmed/37796744.
3. Centers for Disease Control and Prevention. Underlying medical conditions associated with higher risk
for severe COVID-19: information for healthcare professionals. 2023. Available at: https://www.cdc.gov/
coronavirus/2019-ncov/hcp/clinical-care/underlyingconditions.html. Accessed January 10, 2024.
4. Tan C, Huang Y, Shi F, et al. C-reactive protein correlates with computed tomographic findings and predicts
severe COVID-19 early. J Med Virol. 2020;92(7):856-862. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32281668.
5. Berger JS, Kunichoff D, Adhikari S, et al. Prevalence and outcomes of D-dimer elevation in hospitalized
patients with COVID-19. Arterioscler Thromb Vasc Biol. 2020;40(10):2539-2547. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32840379.
6. Casas-Rojo JM, Antón-Santos JM, Millán-Núñez-Cortés J, et al. Clinical characteristics of patients
hospitalized with COVID-19 in Spain: results from the SEMI-COVID-19 registry. Rev Clin Esp (Barc).
2020;220(8):480-494. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32762922.
7. Smilowitz NR, Kunichoff D, Garshick M, et al. C-reactive protein and clinical outcomes in patients
with COVID-19. Eur Heart J. 2021;42(23):2270-2279. Available at: https://www.ncbi.nlm.nih.gov/
pubmed/33448289.
8. Society for Maternal-Fetal Medicine. Management considerations for pregnant patients with COVID-19.
2021. Available at: https://s3.amazonaws.com/cdn.smfm.org/media/2734/SMFM_COVID_Management_of_
Two main processes are thought to drive the pathogenesis of COVID-19. Early in the clinical course, the
disease is primarily driven by the replication of SARS-CoV-2. Later in the clinical course, the disease
is driven by a dysregulated immune/inflammatory response to SARS-CoV-2 infection that may lead
to further tissue damage and thrombosis. Based on this understanding, therapies that directly target
SARS-CoV-2 are anticipated to have the greatest effect early in the course of the disease, whereas
immunosuppressive, anti-inflammatory, and antithrombotic therapies are likely to be more beneficial
after COVID-19 has progressed to stages characterized by hypoxemia, endothelial dysfunction, and
immunothrombosis.
The clinical spectrum of SARS-CoV-2 infection includes asymptomatic or presymptomatic infection and
mild, moderate, severe, and critical illness. Table 2a provides guidance for clinicians on the therapeutic
management of nonhospitalized adults. This includes patients who do not require hospitalization or
supplemental oxygen and those who have been discharged from an emergency department or a hospital.
Table 2c provides guidance on the therapeutic management of hospitalized adults according to their
disease severity and oxygen requirements.
Summary Recommendations
• Management of nonhospitalized patients with acute COVID-19 should include providing supportive care, taking steps to
reduce the risk of SARS-CoV-2 transmission (including isolating the patient), and advising patients on when to contact a
health care provider and seek an in-person evaluation (AIII).
• Patients who are at high risk of progressing to severe COVID-19 may be eligible for pharmacologic therapy. See
Therapeutic Management of Nonhospitalized Adults With COVID-19 for specific recommendations.
• Patients with persistent or progressive dyspnea, especially those who have an oxygen saturation measured by pulse
oximetry (SpO2) ≤94% on room air at sea level or have symptoms that suggest high acuity (e.g., chest pain or tightness,
dizziness, confusion, other mental status changes), should be referred to a health care provider for an in-person
evaluation (AIII).
• Clinicians should be aware that using pulse oximeters to measure oxygen saturation has important limitations.
Therefore, SpO2 results should be considered in the context of the patient’s clinical condition. See Clinical Spectrum of
SARS-CoV-2 Infection for more information.
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
This section of the Guidelines is intended to provide general information to health care providers who
are caring for nonhospitalized adults with COVID-19. The COVID-19 Treatment Guidelines Panel’s (the
Panel) recommendations for pharmacologic management can be found in Therapeutic Management of
Nonhospitalized Adults With COVID-19.
This section focuses on the evaluation and management of:
• Adults with COVID-19 in an ambulatory care setting
• Adults with COVID-19 following discharge from the emergency department (ED)
• Adults with COVID-19 following inpatient discharge
Outpatient evaluation and management in each of these settings may include some or all of the
following: in-person visits, telemedicine, remote monitoring, and home visits by nurses or other health
care providers.
Data from studies in the United States show that certain racial and ethnic minorities experience higher
rates of COVID-19, hospitalization, and death in relation to their share of the total U.S. population.1-5
In addition, some studies have found that members of certain racial, ethnic, and socioeconomic groups
were less likely to receive COVID-19 treatments.6-8 The underlying causes of these observed disparities
may include inadequate insurance coverage, a lack of primary care providers, hesitancy about receiving
treatment, barriers to telehealth visits, and transportation challenges. To reduce COVID-19 treatment
disparities, providers must be aware of the problem and provide patient-centered care. The Panel
recommends that health care providers, health care systems, and payers ensure equitable access to high-
quality care and treatment for all patients, regardless of race, ethnic identity, or other minoritized identity
or social status (AIII).
References
1. Azar KMJ, Shen Z, Romanelli RJ, et al. Disparities in outcomes among COVID-19 patients in a large health
care system in California. Health Aff (Millwood). 2020;39(7):1253-1262. Available at: https://www.ncbi.nlm.
nih.gov/pubmed/32437224.
Symptom management should be initiated for all nonhospitalized adults with mild to moderate
COVID-19. For adults who are at high risk of progressing to severe disease, several antiviral therapeutic
options are available to reduce the risk of hospitalization or death. The COVID-19 Treatment Guidelines
Panel’s (the Panel) recommendations on the use of these drugs for the treatment of COVID-19 are
outlined in this section.
The main goal of therapeutic management for nonhospitalized patients is to prevent progression to
severe disease, hospitalization, or death. Other goals may include accelerating symptom recovery and
viral clearance and preventing long-term sequelae. Current data on the impact of therapy on these
secondary goals are limited.
Several factors affect the selection of the best treatment option for a specific patient. These factors
include the clinical efficacy and availability of the treatment option, the feasibility of administering
parenteral medications, the potential for significant drug-drug interactions, the patient’s pregnancy
status, the time from symptom onset, and the in vitro activities of the available products against the
currently circulating SARS-CoV-2 variants and subvariants.
Most of the data that support the use of the recommended treatment options come from clinical trials
that enrolled individuals who were at high risk of disease progression and who had no pre-existing
immunity from COVID-19 vaccination or prior SARS-CoV-2 infection. Accordingly, the proportion
of hospitalizations and deaths in the placebo arms of these trials was high compared to what has been
seen more recently in populations where most people are vaccinated or have had prior SARS-CoV-2
infection. Although these trials demonstrated the efficacy of using antiviral drugs in high-risk
populations, it is difficult to know their precise effectiveness in the current setting because of the low
rates of hospitalization and death among those who have been vaccinated.
Nevertheless, some patients continue to have an increased risk of disease progression, and it is in
those people that therapies are most likely to be beneficial. Patients who are at the highest risk are
older patients (i.e., those aged >50 years and especially those aged ≥65 years) and patients who are
unlikely to have an adequate immune response to COVID-19 vaccines due to a moderate to severe
immunocompromising condition or the receipt of immunosuppressive medications. Other risk factors
include lack of vaccination or incomplete vaccination; a prolonged amount of time since the most recent
vaccine dose (e.g., >6 months); and conditions such as obesity, diabetes, and chronic pulmonary, cardiac,
or renal disease.1
People who are members of racial and ethnic minority groups have higher rates of hospitalization and
death from COVID-19 than people who are White.2 Disparities in the use of antiviral treatments in
patients who are not White have been reported; therefore, attention to equitable access is critical.3,4
The Panel’s recommendations reflect the available data on the benefits of using antiviral therapies
to prevent progression to severe COVID-19. Table 2a outlines the Panel’s recommendations for the
therapeutic management of nonhospitalized adults with COVID-19. For the recommended doses for the
agents listed in Table 2a, refer to Table 2b below.
Immunomodulators
The Panel recommends against the use of dexamethasone or other systemic corticosteroids to treat
outpatients with mild to moderate COVID-19 who do not require hospitalization or supplemental
oxygen (AIIb). Patients with COVID-19 who are receiving dexamethasone or another corticosteroid for
an underlying condition should continue this therapy as directed by their health care provider (AIII).
Medicare and FDA data show a significant increase in the number of prescriptions for systemic
COVID-19 Treatment Guidelines 52
References
1. Centers for Disease Control and Prevention. Underlying medical conditions associated with higher risk
for severe COVID-19: information for healthcare professionals. 2023. Available at: https://www.cdc.gov/
coronavirus/2019-ncov/hcp/clinical-care/underlyingconditions.html. Accessed February 4, 2024.
2. Mackey K, Ayers CK, Kondo KK, et al. Racial and ethnic disparities in COVID-19-related infections,
hospitalizations, and deaths: a systematic review. Ann Intern Med. 2021;174(3):362-373. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33253040.
3. Wu EL, Kumar RN, Moore WJ, et al. Disparities in COVID-19 monoclonal antibody delivery: a retrospective
cohort study. J Gen Intern Med. 2022;37(10):2505-2513. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35469360.
4. Gold JAW, Kelleher J, Magid J, et al. Dispensing of oral antiviral drugs for treatment of COVID-19 by ZIP
code-level social vulnerability—United States, December 23, 2021–May 21, 2022. MMWR Morb Mortal Wkly
Rep. 2022;71(25):825-829. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35737571.
5. Hammond J, Leister-Tebbe H, Gardner A, et al. Oral nirmatrelvir for high-risk, nonhospitalized adults with
COVID-19. N Engl J Med. 2022;386(15):1397-1408. Available at:
https://pubmed.ncbi.nlm.nih.gov/35172054.
6. Gottlieb RL, Vaca CE, Paredes R, et al. Early remdesivir to prevent progression to severe COVID-19 in
outpatients. N Engl J Med. 2022;386(4):305-315. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34937145.
7. Ritonavir-boosed nirmatrelvir (Paxlovid) [package insert]. Food and Drug Administration. 2023. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/217188s000lbl.pdf.
8. Hiremath S, Blake PG, Yeung A, et al. Early experience with modified dose nirmatrelvir/ritonavir in dialysis
patients with coronavirus disease 2019. Clin J Am Soc Nephrol. 2023;18(4):485-490. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/36723285.
9. Toussi SS, Neutel JM, Navarro J, et al. Pharmacokinetics of oral nirmatrelvir/ritonavir, a protease inhibitor
for treatment of COVID-19, in subjects with renal impairment. Clin Pharmacol Ther. 2022;112(4):892-900.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/35712797.
10. University of Liverpool. Prescribing resources. 2022. Available at:
https://covid19-druginteractions.org/prescribing_resources. Accessed January 31, 2024.
11. Ontario Health. COVID-19 supplemental clinical guidance #4: nirmatrelvir/ritonavir (Paxlovid) use in patients
with advanced chronic kidney disease and patients on dialysis with COVID-19. 2022. Available at:
https://www.ontariohealth.ca/sites/ontariohealth/files/2022-04/PaxlovidClinicalGuide.pdf.
12. Food and Drug Administration Center for Drug Evaluation and Research. Antimicrobial drugs advisory
committee meeting. 2023. Available at: https://www.fda.gov/media/168508/download.
13. Huygens S, Gharbharan A, Serroukh Y, et al. High-titer convalescent plasma plus nirmatrelvir/ritonavir
treatment for non-resolving COVID-19 in six immunocompromised patients. J Antimicrob Chemother.
2023;78(7):1644-1648. Available at: https://www.ncbi.nlm.nih.gov/pubmed/37248664.
14. Brosh-Nissimov T, Ma’aravi N, Leshin-Carmel D, et al. Combination treatment of persistent COVID-19
in immunocompromised patients with remdesivir, nirmatrelvir/ritonavir and tixagevimab/cilgavimab. J
Microbiol Immunol Infect. 2023;Published online ahead of print. Available at:
https://pubmed.ncbi.nlm.nih.gov/37805361.
15. Mikulska M, Sepulcri C, Dentone C, et al. Triple combination therapy with two antivirals and monoclonal
antibodies for persistent or relapsed SARS-CoV-2 infection in immunocompromised patients. Clin Infect Dis.
2023;77(2):280-286. Available at: https://www.ncbi.nlm.nih.gov/pubmed/36976301.
Two main processes are thought to drive the pathogenesis of COVID-19. Early in the clinical course, the
disease is primarily driven by the replication of SARS-CoV-2. Later in the clinical course, the disease
is driven by a dysregulated immune/inflammatory response to SARS-CoV-2 infection that may lead
to further tissue damage and thrombosis. Based on this understanding, therapies that directly target
SARS-CoV-2 are anticipated to have the greatest effect early in the course of the disease, whereas
immunosuppressive, anti-inflammatory, and antithrombotic therapies are likely to be more beneficial
after COVID-19 has progressed to stages characterized by hypoxemia, endothelial dysfunction, and
immunothrombosis.
Currently, most people in the United States have some degree of immunity to SARS-CoV-2 due to
COVID-19 vaccination or SARS-CoV-2 infection. The increase in population immunity and the change
in variants have led to a decrease in the rate of severe disease caused by COVID-19. Because other
co-existing diseases can cause hypoxemia in patients who test positive for SARS-CoV-2 infection,
clinicians should perform the appropriate evaluations to rule out alternative diagnoses. However,
the ongoing evolution of SARS-CoV-2 can lead to immune escape, allowing the virus to continue
circulating in the community. Thus, COVID-19 remains a concern for public health.
Many of the patients who are hospitalized for COVID-19 are immunocompromised to some degree.
For most hospitalized patients with severe or critical COVID-19 who are immunocompromised,
the COVID-19 Treatment Guidelines Panel (the Panel) recommends using antiviral drugs and
immunomodulatory therapies at the doses and durations recommended for the general population (AIII).
Some patients who are immunocompromised have prolonged COVID-19 symptoms and evidence of
ongoing SARS-CoV-2 replication. See Special Considerations in People Who Are Immunocompromised
for additional guidance on managing these patients.
Below is a summary of the rationale for the Panel’s recommendations on the therapeutic management of
hospitalized patients with COVID-19. For dosing information for each of the recommended drugs, see
Table 2d below. For more information about these therapies and the evidence that supports the Panel’s
recommendations, please refer to the specific drug pages and clinical data tables.
Patients Who Are Hospitalized for Reasons Other Than COVID-19 and Who Do Not
Require Supplemental Oxygen
Hospitalized patients with COVID-19 who do not require supplemental oxygen are a heterogeneous
population. Some patients may be hospitalized for reasons other than COVID-19 but may also have mild
to moderate COVID-19 (see Clinical Spectrum of SARS-CoV-2 Infection). In these cases, patients who
COVID-19 Treatment Guidelines 59
Patients Who Are Hospitalized for COVID-19 and Who Do Not Require
Supplemental Oxygen
Recommendations
• The Panel recommends using remdesivir for the treatment of COVID-19 in hospitalized patients
who do not require supplemental oxygen and who are immunocompromised (BIIb) and for other
patients who are at high risk of progressing to severe disease (BIII).
• Remdesivir should be administered for 5 days or until hospital discharge, whichever comes first.
The rationale for using remdesivir in high-risk patients is based on several lines of evidence. In a
trial conducted predominantly among hospitalized patients with COVID-19 who were not receiving
supplemental oxygen at enrollment, a 5-day course of remdesivir was associated with greater clinical
improvement when compared with standard of care.1 Evidence from the PINETREE trial also suggests that
early therapy reduces the risk of progression, although that study was performed in high-risk, unvaccinated,
nonhospitalized patients with ≤7 days of symptoms who received a 3-day course of remdesivir.2
Other studies have not shown a clinical benefit of remdesivir in this group of patients. In the ACTT-1
trial, remdesivir showed no significant benefit in hospitalized patients with mild to moderate COVID-19;
however, only 13% of the study population did not require supplemental oxygen.3 In the large Solidarity
trial, the use of remdesivir was not associated with a survival benefit among the subset of hospitalized
patients who did not require supplemental oxygen.4 See Table 4a for more information.
The aggregate data on using remdesivir to treat all high-risk patients with COVID-19 show a faster time
to recovery in patients who received remdesivir but no clear evidence of a survival benefit. Therefore,
the Panel recommends using remdesivir in hospitalized patients with COVID-19 who do not require
supplemental oxygen and who are at high risk of progressing to severe disease (BIII).
In a large, retrospective cohort study of hospitalized patients with COVID-19 who were
immunocompromised (n = 28,338), patients who received remdesivir had a lower risk of mortality
than those who did not receive remdesivir.5 Forty percent of patients in this cohort were not receiving
supplemental oxygen at baseline; mortality was reduced in this subset of patients. Therefore, the Panel
recommends using remdesivir in hospitalized patients with COVID-19 who do not require supplemental
oxygen and who are immunocompromised (BIIb).
COVID-19 Treatment Guidelines 60
Recommendation
• The Panel recommends against the use of dexamethasone (AIIa) or other systemic corticosteroids
(AIII) for the treatment of COVID-19 in patients who do not require supplemental oxygen.
In the RECOVERY trial, no survival benefit was observed for dexamethasone among the subset of
patients with COVID-19 who did not require supplemental oxygen at enrollment.6 In an observational
cohort study of U.S. veterans, the use of dexamethasone was associated with higher mortality in
hospitalized patients with COVID-19 who did not require supplemental oxygen.7
There are insufficient data to inform the use of other systemic corticosteroids in hospitalized patients
with COVID-19 who do not require supplemental oxygen. See Table 5a for more information about the
clinical trials that have evaluated the use of these drugs in patients with COVID-19. Patients who are
receiving corticosteroid treatment for an underlying condition should continue to receive corticosteroids.
Recommendation
• For patients with COVID-19 who only require minimal conventional oxygen, the Panel
recommends using remdesivir without dexamethasone (BIIa).
In a subgroup analysis during the ACTT-1 trial, remdesivir significantly reduced the time to clinical
recovery and significantly reduced mortality among the subset of patients who were receiving
conventional oxygen at enrollment.3 Evidence from ACTT-1 and a pooled analysis of individual
data from 9 randomized controlled trials8 suggest that remdesivir will have its greatest benefit when
administered early in the clinical course of COVID-19 (e.g., within 10 days of symptom onset). See
Table 4a for more information.
Recommendations
• For most patients with COVID-19 who require conventional oxygen, the Panel recommends using
dexamethasone plus remdesivir (BIIa).
• If dexamethasone is not available, an equivalent dose of another corticosteroid (e.g., prednisone,
methylprednisolone, hydrocortisone) may be used (BIII).
The results of several studies suggest that the use of remdesivir plus dexamethasone improves clinical
outcomes among hospitalized patients with COVID-19. In the CATCO trial, in which 87% of patients
received corticosteroids and 54% were on conventional oxygen, remdesivir significantly reduced the
need for mechanical ventilation among the subset of patients who did not require mechanical ventilation
at enrollment when compared with standard of care.9 In the Solidarity trial, in which approximately
two-thirds of the patients received corticosteroids, remdesivir significantly reduced mortality among the
large subset of patients (n > 7,000) who were receiving conventional or HFNC oxygen at enrollment.4
See Table 4a for more information.
COVID-19 Treatment Guidelines 61
Recommendation
• If remdesivir is not available, the Panel recommends using dexamethasone alone in patients with
COVID-19 who require conventional oxygen (BI).
In the RECOVERY trial, the use of dexamethasone 6 mg once daily for 10 days or until hospital
discharge significantly reduced mortality among the subset of patients who were receiving oxygen
(defined as receiving oxygen supplementation but not mechanical ventilation or extracorporeal
membrane oxygenation [ECMO]) at enrollment.6 Remdesivir was administered to <1% of the study
participants. Results for patients who were only receiving conventional oxygen at enrollment were not
available. See Table 5a for more information.
Recommendations
• For patients with COVID-19 who have rapidly increasing oxygen needs and systemic
inflammation, the Panel recommends adding 1 of the following immunomodulators to
dexamethasone:
• Preferred Second Immunomodulators
○ Oral (PO) baricitinib (BIIa)
○ Intravenous (IV) tocilizumab (BIIa)
• Alternative Second Immunomodulators (Listed in Alphabetical Order)
○ IV abatacept (CIIa)
○ IV infliximab (CIIa)
If none of these options are available or feasible to use, the Janus kinase (JAK) inhibitor PO tofacitinib
(CIIa) or the interleukin (IL)-6 inhibitor IV sarilumab (CIIa) can be used in combination with
dexamethasone. Sarilumab is only commercially available as a subcutaneous (SUBQ) injection; see
Table 5e for information regarding the preparation of an IV infusion using the SUBQ product.
Several large randomized controlled trials have evaluated the use of dexamethasone in combination with
a second immunomodulator, including:
• Abatacept, a cytotoxic T-lymphocyte–associated antigen 4 agonist10
• Baricitinib, a JAK inhibitor11-13
• Infliximab, a tumor necrosis factor–alpha inhibitor10
• Tocilizumab, an IL-6 inhibitor14-16
These studies included patients who required conventional oxygen only, as well as those with increasing
oxygen needs and/or elevated levels of inflammatory markers. Subgroup analyses in these trials have not
clearly defined which patients in this heterogeneous group are most likely to benefit from adding a second
immunomodulator to corticosteroid therapy. The study endpoints for these trials included progression
to more severe disease, the need for mechanical ventilation, and death. Nonetheless, some trials suggest
that adding a second immunomodulator provides benefits to patients who require conventional oxygen,
COVID-19 Treatment Guidelines 62
Use of Anticoagulants
• The Panel recommends using a therapeutic dose of heparin for nonpregnant patients with
D-dimer levels above the upper limit of normal who require conventional oxygen and who do not
have an increased bleeding risk (CIIa).
• The Panel recommends the use of a prophylactic dose of heparin for patients who do not meet
the criteria for receiving therapeutic heparin or are not receiving a therapeutic dose of heparin for
other reasons, unless a contraindication exists (AI).
• The Panel recommends the use of a prophylactic dose of anticoagulation for pregnant patients who
are hospitalized for manifestations of COVID-19, unless a contraindication exists (BIII).
The Panel’s recommendations for the use of heparin are based on data from 3 open-label randomized
controlled trials that compared the use of therapeutic doses of heparin to prophylactic or intermediate
doses of heparin in hospitalized patients who did not require intensive care unit (ICU)-level care. Pooled
data from the ATTACC/ACTIV-4a/REMAP-CAP trials reported more organ support-free days (i.e., days
alive and free of ICU-based organ support) for patients in the therapeutic heparin arm than for those
in the usual care arm, but there was no difference between the arms in in-hospital mortality or length
of hospitalization.18 The RAPID trial compared a therapeutic dose of heparin to a prophylactic dose in
hospitalized patients with moderate COVID-19. There was no statistically significant difference between
the arms in the occurrence of the primary endpoint (which was a composite of ICU admission, NIV
or mechanical ventilation, or death by Day 28), but the therapeutic dose of heparin reduced the risk of
all-cause death.19 In the HEP-COVID trial, venous thromboembolism (VTE), arterial thromboembolism,
and death by Day 32 occurred significantly less frequently in patients who received a therapeutic dose of
heparin than in those who received a prophylactic dose of heparin, but there was no difference in mortality
COVID-19 Treatment Guidelines 63
Recommendations
• Dexamethasone should be administered to all patients with COVID-19 who require HFNC
oxygen or NIV (AI).
• If not already initiated, promptly add 1 of the following immunomodulators to dexamethasone:
• Preferred Second Immunomodulator
○ PO baricitinib (AI)
• Preferred Alternative Second Immunomodulator
○ IV tocilizumab (BIIa)
• Additional Alternative Second Immunomodulators (Listed in Alphabetical Order)
○ IV abatacept (CIIa)
○ IV infliximab (CIIa)
If none of these options are available or feasible to use, PO tofacitinib (CIIa) or IV sarilumab (CIIa)
can be used in combination with dexamethasone. Sarilumab is only commercially available as a SUBQ
injection; see Table 5e for information regarding the preparation of an IV infusion using the SUBQ
product.
Clinicians should make a significant effort to obtain and administer 1 of the recommended second
immunomodulators. However, dexamethasone should be initiated immediately, without waiting until the
second immunomodulator can be acquired. Dexamethasone was used as a single-agent immunomodulatory
strategy in the RECOVERY trial and demonstrated a survival benefit among patients who required
supplemental oxygen.6 In this trial, the treatment effect for dexamethasone was not evaluated separately for
those who required conventional oxygen and those who required HFNC oxygen or NIV.
Several large randomized controlled trials have demonstrated that patients with COVID-19 who require
HFNC oxygen or NIV benefit from combining dexamethasone with an additional immunomodulator,
such as a JAK inhibitor or an IL-6 inhibitor. The quality of the evidence and the totality of the data
support a stronger recommendation for baricitinib than for tocilizumab.
Two large randomized controlled trials (RECOVERY and COV-BARRIER) both reported a survival
benefit among hospitalized patients with COVID-19 who required HFNC oxygen or NIV and who
received baricitinib plus dexamethasone.12,13 Data from the ACTT-211 and ACTT-421 trials support the
overall safety of using baricitinib in combination with remdesivir and the potential for a clinical benefit
of this combination, but neither trial studied baricitinib in combination with dexamethasone as the
standard of care. A retrospective analysis of data from 11 U.S. health systems suggests that the use of
COVID-19 Treatment Guidelines 64
Recommendations
• For certain hospitalized patients who require HFNC oxygen or NIV, the Panel recommends adding
remdesivir to 1 of the recommended immunomodulator combinations. Examples of patients who
may benefit most from adding remdesivir include:
• Patients who are immunocompromised (BIIb);
• Patients with evidence of ongoing viral replication (e.g., those with a low cycle threshold [Ct]
value, as measured by a reverse transcription polymerase chain reaction [RT-PCR] result or with
a positive rapid antigen test result) (BIII); or
• Patients who are within 10 days of symptom onset (CIIa).
Clinical trial data have not clearly established that remdesivir reduces the time to recovery or improves
survival in patients who require HFNC oxygen or NIV. However, because clinical trials have found
that remdesivir prevents clinical progression in patients who are not on mechanical ventilation, some
patients receiving HFNC oxygen or NIV might benefit from receiving remdesivir. In the Solidarity trial,
remdesivir had a modest but statistically significant effect on reducing the risk of death or progression
to ventilation in patients who were receiving oxygen but who were not ventilated at baseline.4
However, these effects could not be evaluated separately for patients who required conventional oxygen
supplementation and those who required HFNC oxygen or NIV.4 In the CATCO trial, among the patients
COVID-19 Treatment Guidelines 65
Use of Anticoagulants
• The Panel recommends using a prophylactic dose of heparin as VTE prophylaxis, unless a
contraindication exists (AI); (BIII) for pregnant patients.
• The Panel recommends against the use of a therapeutic dose of anticoagulation for VTE
prophylaxis (BI).
• For patients who start on a therapeutic dose of heparin in a non-ICU setting due to COVID-19
and then transfer to the ICU, the Panel recommends switching from the therapeutic dose to a
prophylactic dose of heparin, unless VTE is confirmed (BIII).
The multiplatform randomized controlled trial REMAP-CAP/ACTIV-4a/ATTACC compared the
effectiveness of a therapeutic dose of heparin to standard care in critically ill patients with COVID-19.28
The study did not show an increase in the number of organ support-free days or the probability of
survival to hospital discharge among patients who received therapeutic doses of anticoagulation. See
Antithrombotic Therapy in Patients With COVID-19 for more information.
COVID-19 Treatment Guidelines 66
Use of Anticoagulants
• The Panel recommends using a prophylactic dose of heparin as VTE prophylaxis, unless a
contraindication exists (AI); (BIII) for pregnant patients.
• For patients who start on a therapeutic dose of heparin in a non-ICU setting due to COVID-19
and then transfer to the ICU, the Panel recommends switching from the therapeutic dose to a
prophylactic dose of heparin, unless there is another indication for therapeutic anticoagulation
(BIII).
• The Panel recommends against the use of a therapeutic dose of anticoagulation for VTE
prophylaxis (BI).
Patients who required mechanical ventilation or ECMO were included in the multiplatform
REMAP-CAP/ACTIV-4a/ATTACC trial that studied therapeutic doses of heparin.28 Because these
studies reported no benefits of using therapeutic doses of heparin, the recommendations for using
prophylactic doses of heparin in hospitalized patients who require mechanical ventilation or ECMO are
the same as those for patients who require HFNC oxygen or NIV.
Table 2d. Dosing Regimens for the Drugs Recommended in Table 2c
The drugs in this table are listed in alphabetical order.
Drug Name Dosing Regimen Comments
Abatacept Abatacept 10 mg/kg actual body • No adjustment based on eGFR
weight (up to 1,000 mg) administered
as a single IV dose
Baricitinib BAR dose is dependent on eGFR; • eGFR ≥60 mL/min/1.73 m2: BAR 4 mg PO once daily
duration of therapy is up to 14 days • eGFR 30 to <60 mL/min/1.73 m2: BAR 2 mg PO once daily
or until hospital discharge, whichever • eGFR 15 to <30 mL/min/1.73 m2: BAR 1 mg PO once daily
comes first.
• eGFR <15 mL/min/1.73 m2: Not recommended.
Dexamethasone DEX 6 mg IV or PO once daily for up • If DEX is not available, an equivalent dose of another
to 10 days or until hospital discharge, corticosteroid may be used.
whichever comes first • For more information, see Systemic Corticosteroids.
COVID-19 Treatment Guidelines 68
Key: BAR = baricitinib; DEX = dexamethasone; eGFR = estimated glomerular filtration rate; IV = intravenous; LMWH =
low-molecular-weight heparin; NaCl = sodium chloride; PO = oral; RDV = remdesivir; SUBQ = subcutaneous; UFH =
unfractionated heparin; VTE = venous thromboembolism
References
1. Spinner CD, Gottlieb RL, Criner GJ, et al. Effect of remdesivir vs standard care on clinical status at 11 days in
patients with moderate COVID-19: a randomized clinical trial. JAMA. 2020;324(11):1048-1057. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32821939.
2. Gottlieb RL, Vaca CE, Paredes R, et al. Early remdesivir to prevent progression to severe COVID-19 in
outpatients. N Engl J Med. 2022;386(4):305-315. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34937145.
3. Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the treatment of COVID-19—final report. N Engl J
Med. 2020;383(19):1813-1826. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32445440.
4. WHO Solidarity Trial Consortium. Remdesivir and three other drugs for hospitalised patients with
COVID-19: final results of the WHO Solidarity randomised trial and updated meta-analyses. Lancet.
2022;399(10339):1941-1953. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35512728.
5. Mozaffari E, Chandak A, Gottlieb RL, et al. Remdesivir reduced mortality in immunocompromised patients
hospitalized for COVID-19 across variant waves: findings from routine clinical practice. Clin Infect Dis.
2023;77(12):1626-1634. Available at: https://pubmed.ncbi.nlm.nih.gov/37556727.
6. RECOVERY Collaborative Group. Dexamethasone in hospitalized patients with COVID-19. N Engl J Med.
2021;384(8):693-704. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32678530.
7. Crothers K, DeFaccio R, Tate J, et al. Dexamethasone in hospitalised COVID-19 patients not on intensive
COVID-19 Treatment Guidelines 69
Data from the Centers for Disease Control and Prevention demonstrate that severe disease and death due to
COVID-19 occur less often in children than in adults.1-4 However, weekly hospitalization rates for children
aged <6 months are high, exceeded only by the rates for adults aged ≥75 years.5 The overall incidence of
SARS-CoV-2 infection and, by extension, COVID-19–related hospitalizations among children increased
substantially with the emergence of newer variants, particularly the Omicron variant and its subvariants.6,7
According to the Centers for Disease Control and Prevention, by December 2022, an estimated 96% of
children and adolescents had serologic evidence of prior SARS-CoV-2 infection.8 The high infection rates
among children makes the overall burden substantial despite the low rate of severe outcomes.9
Data on the pathogenesis and clinical spectrum of SARS-CoV-2 infection in children are still limited
compared to the data for adults. Although only a small percentage of children with COVID-19 will
require medical attention, the percentage of intensive care unit admissions among hospitalized children
is comparable to that for hospitalized adults with COVID-19.6,10-18
Observational studies and meta-analyses have found that children with certain comorbidities are at
increased risk of severe COVID-19. These comorbidities include cardiac disease, neurologic disorders,
prematurity (in young infants), diabetes, obesity (particularly severe obesity), chronic lung disease,
feeding tube dependence, and immunocompromised status.19-22 Demographic factors, such as age (<1
year and 10–14 years)23 and non-White race/ethnicity,15,24-26 have also been associated with severe
disease. However, many studies did not assess the relative severity of underlying medical conditions in
children with severe COVID-19.
In general, COVID-19 has similar clinical manifestations and disease stages in children and adults,
including an early phase driven by viral replication and a late phase that appears to be driven by a
dysregulated immune/inflammatory response to SARS-CoV-2 that leads to tissue damage. Respiratory
complications in young children that can occur during the early clinical phase include croup and
bronchiolitis. In addition, a small number of children who have recovered from acute SARS-CoV-2
infection develop multisystem inflammatory syndrome in children (MIS-C) 2 to 6 weeks after infection.
MIS-C is a postinfectious inflammatory condition that can lead to severe organ dysfunction.
Published guidance on the treatment of COVID-19 in children has been extrapolated mostly from
recommendations for adults with COVID-19, recommendations for children with other viral infections,
and expert opinion.27-29 Applying adult data from COVID-19 trials to children is a unique challenge
because most children experience a mild course of illness with COVID-19.30 Relative to adults, children
with COVID-19 have substantially lower mortality and less need for hospitalization.10,11,31 Because
of these differences in epidemiology and disease severity, the effect sizes of treatments for children
are likely to be smaller than those observed in adults. Therefore, to produce a beneficial outcome in
children, the number needed to treat is higher. Collectively, these factors influence the risk versus benefit
balance for pharmacologic therapies in children.
In the absence of sufficient clinical trial data on the treatment of children with COVID-19, the
COVID-19 Treatment Guidelines Panel’s (the Panel) recommendations for the therapeutic management
of hospitalized children are based largely on safety and efficacy data from clinical trials in adults, the
child’s risk of disease progression, and expert opinion.
In general, data from clinical trials in adults are most applicable to the treatment of older children
COVID-19 Treatment Guidelines 72
a
Molnupiravir is not authorized by the FDA for use in children aged <18 years and should not be used.
b
See Table 3b for the Panel’s framework for assessing the risk of progression to severe COVID-19 based on patient
conditions and COVID-19 vaccination status.
c
Initiate treatment as soon as possible after symptom onset.
d
The relative risk of severe COVID-19 for intermediate-risk patients is lower than the risk for high-risk patients but higher
than the risk for low-risk patients.
e
Low-risk patients include those with comorbid conditions that have a weak or unknown association with severe COVID-19.
COVID-19 Treatment Guidelines 73
Table 3b. The Panel’s Framework for Assessing the Risk of Progression to Severe COVID-19
Based on Patient Conditions and COVID-19 Vaccination Status
a
See the current COVID-19 vaccination schedule from the CDC.
b
Recent SARS-CoV-2 infection (i.e., within 3–6 months) may confer substantial immunity against closely related variants. A
patient’s recent infection history should be factored into the risk assessment.
c
The degree of risk conferred by obesity in younger children is less clear than it is in older adolescents.
d
This includes patients with a tracheostomy and those who require NIV.
e
The data for this group are particularly limited.
Key: BMI = body mass index; CDC = Centers for Disease Control and Prevention; NIV = noninvasive ventilation; the Panel =
the COVID-19 Treatment Guidelines Panel
References
1. Centers for Disease Control and Prevention. COVID-19 monthly death rates per 100,000 population
by age group, race and ethnicity, and sex. 2024. Available at: https://covid.cdc.gov/covid-data-
tracker/#demographicsovertime. Accessed February 15, 2024.
2. Centers for Disease Control and Prevention. Provisional COVID-19 deaths: focus on ages 0–18 years. 2023.
COVID-19 Treatment Guidelines 76
Key Considerations
• SARS-CoV-2 infection is generally milder in children than in adults, and a substantial proportion of children with the
infection are asymptomatic.
• Most nonhospitalized children with COVID-19 will not require any specific therapy.
• Children with ≥1 of the following comorbidities are at increased risk of severe COVID-19: cardiac disease, neurologic
disorders, prematurity (in young infants), diabetes, obesity (particularly severe obesity), chronic lung disease, feeding
tube dependence, and immunocompromised status. Age (<1 year and 10–14 years) and non-White race/ethnicity are
also associated with severe disease.
• Data on the pathogenesis and clinical spectrum of SARS-CoV-2 infection are more limited for children than for adults.
• Vertical transmission of SARS-CoV-2 appears to be rare.
• A small subset of children and young adults with SARS-CoV-2 infection may develop multisystem inflammatory
syndrome in children (MIS-C). Many patients with MIS-C require intensive care management. The majority of children
with MIS-C do not have underlying comorbidities.
• Data on the prevalence of post-COVID conditions in children are limited but suggest that younger children may have
fewer persistent symptoms than older children and adults.
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
This section provides an overview of the epidemiology and clinical spectrum of disease, including
COVID-19, multisystem inflammatory syndrome in children (MIS-C), and post-COVID conditions.
It also includes information on risk factors for severe COVID-19, vertical transmission, and infants
born to a birth parent with SARS-CoV-2 infection. Throughout this section, the term “COVID-19”
refers to the acute, primarily respiratory illness due to infection with SARS-CoV-2. MIS-C refers to the
postinfectious inflammatory condition.
Epidemiology
Data from the Centers for Disease Control and Prevention (CDC) demonstrate that severe disease and
death due to COVID-19 occur less often in children than in adults.1-4 However, weekly hospitalization
rates for children aged <6 months are high, exceeded only by the rates for adults aged ≥75 years.5 The
overall incidence of SARS-CoV-2 infection and, by extension, COVID-19–related hospitalizations
among children increased substantially with the emergence of newer variants, particularly the Omicron
variant and its subvariants.6,7 According to the CDC, by December 2022, an estimated 96% of children
and adolescents had serologic evidence of prior SARS-CoV-2 infection.8 The high infection rate among
children makes the overall burden substantial despite the low rate of severe outcomes.9
Data on the pathogenesis and clinical spectrum of SARS-CoV-2 infection in children are still limited
compared to the data for adults. Although only a small percentage of children with COVID-19 will
require medical attention, the percentage of intensive care unit (ICU) admissions among hospitalized
children is comparable to that for hospitalized adults with COVID-19.6,10-18
Children from some racial and ethnic groups experience disproportionate rates of COVID-19–related
hospitalization, which may be a result of barriers to accessing health care and economic and structural
inequities. From 2020 to 2021, Black/African American children with COVID-19 in the United States
Comorbidities
Several chronic conditions are prevalent in hospitalized children with COVID-19. When the Delta
variant was the dominant variant in the United States, 68% of hospitalized children had ≥1 underlying
medical conditions, such as obesity (32%), asthma or reactive airway disease (16%), or feeding tube
dependence (8%).26 Obesity was present in approximately a third of hospitalized children aged 5 to 11
years, 60% of whom had severe obesity (i.e., a body mass index [BMI] ≥120% of the 95th percentile).
For adolescents, 61% had obesity; of those patients, 61% had a BMI ≥120% of the 95th percentile.
Meta-analyses and observational studies identified risk factors for ICU admission, mechanical
ventilation, or death among hospitalized children with COVID-19.27-29 These risk factors included
prematurity in young infants, obesity, diabetes, chronic lung disease, cardiac disease, neurologic disease,
and immunocompromising conditions. Another study found that having a complex chronic condition
that affected ≥2 body systems or having a progressive chronic condition or continuous dependence on
technology for ≥6 months (e.g., dialysis, tracheostomy with ventilator assistance) was significantly
associated with an increased risk of moderate or severe COVID-19.34 The study also found that children
with more severe chronic diseases (e.g., active cancer treated within the previous 3 months or asthma
with hospitalization within the previous 12 months) had a higher risk of critical COVID-19 or death than
those with less severe conditions. The CDC has additional information on the underlying conditions that
are risk factors for severe COVID-19.
Having multiple comorbidities increases the risk of severe COVID-19 in children. A meta-analysis
of data from children hospitalized with COVID-19 found that the risk of ICU admission was greater
for children with 1 chronic condition than for those with no comorbidities, and the risk increased
substantially as the number of comorbidities increased.30
COVID-19 Vaccination
Staying up to date with COVID-19 vaccinations remains the most effective way to prevent severe
COVID-19. See the CDC webpages Stay Up to Date With COVID-19 Vaccines and Use of COVID-19
Vaccines in the United States for more information on COVID-19 vaccination schedules. Estimates of
COVID-19 Treatment Guidelines 81
Mortality
Death from COVID-19 is uncommon in children. Risk factors for death include having chronic
conditions, such as neurologic or cardiac disease, and having multiple comorbidities. Among children
aged <21 years in the United States, the number of deaths associated with COVID-19 has been higher
for children aged 10 to 20 years, especially for young adults aged 18 to 20 years, and for those who
identify as Hispanic, Black, or American Indian/Alaskan Native.41,42
A systematic review and meta-analysis reported that neurologic or cardiac comorbidities were associated
with the greatest increase in risk of death among hospitalized children with COVID-19.30 In the same
study, an individual patient data meta-analysis found that the risk of death related to COVID-19 was
greater for children with 1 chronic condition than for those with no comorbidities, and the risk increased
substantially as the number of comorbidities increased.
Table A. Clinical Manifestation Criteria for the 2023 CSTE/CDC MIS-C Surveillance Case Definition
Distinguishing MIS-C from other febrile illnesses in the community setting remains challenging,
particularly with the declining incidence of MIS-C, but the presence of persistent fever, multisystem
manifestations, and laboratory abnormalities could help early recognition.72 The clinical spectrum of
hospitalized cases has included younger children with mucocutaneous manifestations that overlap
with Kawasaki disease, older children with more multiorgan involvement and shock, and patients with
respiratory manifestations that overlap with COVID-19.
Patients with MIS-C are often critically ill, and up to 80% of children require ICU admission; however,
data collected while Omicron was the dominant variant in the United States suggest that the cases
of MIS-C reported during this period were less severe than those reported when other variants were
dominant.66,73,74 Most patients with MIS-C have markers of cardiac injury or dysfunction, including
COVID-19 Treatment Guidelines 84
Post-COVID Conditions
The persistent symptoms after COVID-19 that have been described in children are similar to those
seen in adults. The terminology for these collective symptoms is evolving and includes long COVID,
post–COVID-19 condition, and post-acute sequelae of SARS-CoV-2 infection (PASC). The data on
the incidence of post-COVID conditions in children are limited and somewhat conflicting, but the
overall incidence appears to be lower in children than in adults (see Clinical Spectrum of SARS-CoV-2
Infection).85-92
Case definitions for post-COVID conditions vary between studies, which makes determining the true
incidence of these conditions challenging. The incidence of post-COVID symptoms in children appears
to increase with age. The most common symptoms reported include persistent fatigue, headache,
shortness of breath, sleep disturbances, gastrointestinal symptoms, and an altered sense of smell.93
Cardiopulmonary injury, neurocognitive impairment, and new-onset diabetes may occur. However,
some studies did not include control groups of children who did not have SARS-CoV-2 infection, which
makes assessing the relative risk of these symptoms a challenge.
Details on the pathogenesis, clinical presentation, and treatment for post-COVID conditions in children
are beyond the scope of these Guidelines. The CDC provides additional information about the incidence,
presentation, and management strategies for post-COVID conditions in children as well as adults.
Additional research is needed to define the incidence, pathophysiology, spectrum, and severity of
post-COVID conditions in children and to identify the optimal strategies for the prevention, diagnosis,
and treatment of these conditions.
References
1. Centers for Disease Control and Prevention. COVID-19 monthly death rates per 100,000 population
by age group, race and ethnicity, and sex. 2024. Available at: https://covid.cdc.gov/covid-data-
tracker/#demographicsovertime. Accessed February 15, 2024.
COVID-19 Treatment Guidelines 85
This section outlines the COVID-19 Treatment Guidelines Panel’s (the Panel) recommendations for
the therapeutic management of nonhospitalized children (i.e., pediatric patients aged <18 years) with
mild to moderate COVID-19. These recommendations are also for children who have mild to moderate
COVID-19 and are hospitalized for reasons other than COVID-19. For patients aged ≥18 years, see
Therapeutic Management of Nonhospitalized Adults With COVID-19. Throughout this section, the term
“COVID-19” refers to the acute, primarily respiratory illness due to infection with SARS-CoV-2. For the
Panel’s recommendations for managing multisystem inflammatory syndrome in children (MIS-C), see
Therapeutic Management of Hospitalized Children With MIS-C, Plus a Discussion on MIS-A.
Treatment Considerations
Currently, no results from pediatric clinical trials that evaluated the treatment of COVID-19 have been
published. Data evaluating the use of pharmacologic therapy in children with COVID-19 are limited
largely to descriptive reports.1-3 Therefore, more high-quality randomized trials, observational studies,
and pharmacokinetic studies are urgently needed. Whenever possible, clinical trials of therapeutics and
multicenter observational cohorts should enroll children with COVID-19.
Published guidance on the treatment of COVID-19 in children has been extrapolated mostly from
recommendations for adults with COVID-19, recommendations for children with other viral infections,
and expert opinion.4-6 Applying adult data from COVID-19 trials to children is a unique challenge
because most children experience a mild course of illness with COVID-19. Relative to adults, children
with COVID-19 have substantially lower mortality and less need for hospitalization.7-9 Because of these
differences in epidemiology and disease severity, the effect sizes of treatments for children are likely
to be smaller than those observed in adults. Therefore, to produce a beneficial outcome in children, the
number needed to treat is higher. Collectively, these factors influence the risk versus benefit balance for
pharmacologic therapies in children.
Other challenges are the uncertainty about which comorbid conditions place children at the highest
risk of severe COVID-19 and the uncertainty about the absolute magnitude of the increased risk from
those comorbid conditions. For children with COVID-19, the type, number, and severity of comorbid
conditions influence decisions about pharmacologic treatments. For more information on risk factors for
children with COVID-19, see Special Considerations in Children.
Recommendations
In the absence of sufficient clinical trial data on the treatment of children with COVID-19, the Panel’s
recommendations for the therapeutic management of nonhospitalized children are based largely on
safety and efficacy data from clinical trials in adults (see Table 3a). No pediatric comparative studies
have been published; therefore, all the quality of evidence ratings for the Panel’s recommendations in
this section are based on expert opinion (i.e., a III rating).
The majority of children with mild to moderate COVID-19 will not progress to more severe illness;
therefore, the Panel recommends managing these patients with supportive care alone (AIII). The risks
and benefits of therapy should be assessed based on COVID-19 disease severity, age, vaccination status,
Panel’s Recommendations
Risk of Severe COVID-19
Aged 12–17 Years Aged <12 Years
Symptomatic, Regardless of • Provide supportive care (AIII). • Provide supportive care (AIII).
Risk Factors
• Use 1 of the following options (listed in • Ritonavir-boosted nirmatrelvir is
order of preference):c not authorized by the FDA for use in
• Ritonavir-boosted nirmatrelvir children aged <12 years.
High Riska,b (Paxlovid) within 5 days of symptom • There is insufficient evidence to
onset (BIII) recommend either for or against the
• Remdesivir within 7 days of routine use of remdesivir. Consider
symptom onset (CIII) treatment based on age and other risk
factors.
• There is insufficient evidence to • There is insufficient evidence to
recommend either for or against the recommend either for or against the
Intermediate Riskb,d use of any antiviral therapy. Consider routine use of remdesivir.
treatment based on age and other risk
factors.
• Manage with supportive care alone • Manage with supportive care alone
Low Riskb,e
(AIII). (AIII).
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
a
Molnupiravir is not authorized by the FDA for use in children aged <18 years and should not be used.
b
See Table 3b for the Panel’s framework for assessing the risk of progression to severe COVID-19 based on patient
conditions and COVID-19 vaccination status.
c
Initiate treatment as soon as possible after symptom onset.
d
The relative risk of severe COVID-19 for intermediate-risk patients is lower than the risk for high-risk patients but higher
than the risk for low-risk patients.
e
Low-risk patients include those with comorbid conditions that have a weak or unknown association with severe COVID-19.
Patients with no comorbidities are included in this group.
Key: FDA = Food and Drug Administration; the Panel = the COVID-19 Treatment Guidelines Panel
Health Disparities
COVID-19–related outcomes are worse among medically underserved populations, although this factor
is not strictly a comorbid condition. Some racial and ethnic minority groups experience disproportionate
rates of COVID-19 hospitalization and are less likely to receive specific therapies.23,35-37 These factors
may be relevant when making clinical decisions about treatment.38,39 See Special Considerations in
Children for more information.
Corticosteroids
Corticosteroids are not indicated for the treatment of COVID-19 in nonhospitalized children. However,
corticosteroids should be used per usual standards of care in children with asthma and croup triggered by
SARS-CoV-2 infection. Children with COVID-19 who are receiving corticosteroids for an underlying
condition should continue this therapy as directed by their health care providers.
References
1. Schuster JE, Halasa NB, Nakamura M, et al. A description of COVID-19-directed therapy in children admitted
to US intensive care units 2020. J Pediatric Infect Dis Soc. 2022;11(5):191-198. Available at: https://www.
ncbi.nlm.nih.gov/pubmed/35022779.
2. Goldman DL, Aldrich ML, Hagmann SHF, et al. Compassionate use of remdesivir in children with severe
COVID-19. Pediatrics. 2021;147(5):e2020047803. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33883243.
This section outlines the COVID-19 Treatment Guidelines Panel’s (the Panel) recommendations for the
therapeutic management of children (i.e., pediatric patients aged <18 years) who are hospitalized for
COVID-19. Throughout this section, the term “COVID-19” refers to the acute, primarily respiratory
illness due to infection with SARS-CoV-2. Multisystem inflammatory syndrome in children (MIS-C)
refers to the postinfectious inflammatory condition.
Recommendations
In the absence of sufficient clinical trial data on the treatment of children with COVID-19, the Panel’s
recommendations for the therapeutic management of hospitalized children are based largely on safety
and efficacy data from clinical trials in adults, the child’s risk of disease progression, and expert
opinion (see Table 3c). For the Panel’s recommendations for adults, see Therapeutic Management of
Hospitalized Adults With COVID-19.
In general, data from clinical trials in adults are most applicable to the treatment of older children
with severe COVID-19 and predominantly lower respiratory tract disease. Using data from clinical
trials in adults to develop recommendations for children with SARS-CoV-2 infection who have
clinical syndromes associated with other respiratory viruses (e.g., bronchiolitis, croup, asthma) is a
challenge. No evidence suggests that these syndromes should be managed differently when caused by
COVID-19 Treatment Guidelines 102
Dexamethasone
Dexamethasone was evaluated in the RECOVERY trial, which was an open-label, randomized trial
conducted in the United Kingdom.27 The trial compared the use of up to 10 days of dexamethasone 6
mg, administered by intravenous injection or orally, with usual care among hospitalized adults with
COVID-19. The primary outcome was all-cause mortality at 28 days, which occurred in 22.9% of
patients randomized to receive dexamethasone versus 25.7% of patients randomized to receive usual
care (age-adjusted rate ratio 0.83; 95% CI, 0.75–0.93; P < 0.001). Patients who required mechanical
ventilation or extracorporeal membrane oxygenation (ECMO) had the greatest effect size (29.3% vs.
41.4%; rate ratio 0.64; 95% CI, 0.51–0.81). No difference in outcomes was observed in patients who did
not require supplemental oxygen (17.8% vs. 14.0%; rate ratio 1.19; 95% CI, 0.92–1.55). For the 28-day
mortality outcome, a difference between arms was observed in patients who required supplemental
oxygen (23.3% vs. 26.2%; rate ratio 0.82; 95% CI, 0.72–0.94). However, it should be noted that these
patients were a heterogeneous group, including those who received either conventional oxygen or NIV.
See Systemic Corticosteroids for detailed information.
The safety and efficacy of using dexamethasone or other corticosteroids for the treatment of COVID-19
have not been evaluated in pediatric patients. Given that the mortality for adults in the placebo arm in
the RECOVERY trial was substantially greater than the mortality generally reported for children with
COVID-19, caution is warranted when extrapolating from recommendations for adults and applying
them to patients aged <18 years.
However, because of the effect size observed in the RECOVERY trial, the Panel recommends the use
of dexamethasone for children who require mechanical ventilation or ECMO (AIII). The Panel also
recommends the use of dexamethasone, with or without concurrent remdesivir, for children who
require oxygen through a high-flow device or NIV (BIII). The Panel does not recommend routine
use of corticosteroids for children who require only conventional oxygen, but corticosteroids can be
considered in combination with remdesivir for patients with increasing oxygen needs, particularly
adolescents (BIII).
Evidence has demonstrated that the use of corticosteroids does not benefit infants with viral bronchiolitis
not related to COVID-19, and current American Academy of Pediatrics guidelines recommend against
the use of corticosteroids in this population.28 There are no data specific to COVID-19 that support
the use of corticosteroids in children with bronchiolitis due to SARS-CoV-2 infection. Corticosteroids
should be used per the usual standards of care in children with asthma or croup triggered by SARS-CoV-
2.
The use of dexamethasone for the treatment of severe COVID-19 in children who are profoundly
Baricitinib
The Janus kinase inhibitor baricitinib was approved by the FDA for the treatment of COVID-19 in
hospitalized adults. An FDA Emergency Use Authorization for baricitinib remains active for the
treatment of COVID-19 in hospitalized children aged 2 to 17 years who require supplemental oxygen,
NIV, mechanical ventilation, or ECMO.29
In the COV-BARRIER trial, adults with COVID-19 pneumonia were randomized to receive baricitinib
or standard care. Patients treated with baricitinib showed a reduction in mortality when compared
with those who received standard care.30 The reduction was greatest in patients who received
high-flow oxygen or NIV. Similarly, the ACTT-2 trial in adults showed that patients who received
baricitinib plus remdesivir had improved time to recovery when compared with patients who received
remdesivir alone.31 This effect was most pronounced in patients who received high-flow oxygen or
NIV. In the ACTT-4 trial, 1,010 patients were randomized 1:1 to receive baricitinib plus remdesivir or
dexamethasone plus remdesivir.32 The study reported no difference between the arms for the outcome of
mechanical ventilation–free survival.
In the RECOVERY trial, 8,156 patients, including 33 children aged 2 to 17 years, were randomized
to receive baricitinib or usual care (95% received corticosteroids).33 Treatment with baricitinib was
associated with a 13% proportional reduction in mortality, with the greatest effect size occurring in
patients who received NIV. The RECOVERY investigators included these patients in a meta-analysis
and found that treatment with baricitinib was associated with a 20% relative reduction in mortality (rate
ratio 0.80; 95% CI, 0.72–0.89; P < 0.0001). See Janus Kinase Inhibitors and Therapeutic Management
of Hospitalized Adults With COVID-19 for additional information. These data in adults indicate that
baricitinib is likely to be most beneficial for patients receiving noninvasive forms of respiratory support.
Several open-label trials and cohort studies have evaluated baricitinib in children with autoinflammatory
and rheumatic diseases, including many children aged <5 years, and found the treatment was well
tolerated; however, the pharmacokinetics of baricitinib in younger children are not well studied.34-37
Information on the safety and effectiveness of the use of baricitinib in children with COVID-19 is
limited to case reports.
In contrast to the strong recommendation for its use in adults, baricitinib is not considered the standard
of care for all children who require high-flow oxygen or NIV because of the low mortality in children
with COVID-19 (especially young children) and the limited data on the use of baricitinib in these
children.
Extrapolating from clinical trials among adults with COVID-19, the Panel recommends that:
• For children who require oxygen through a high-flow device or NIV and do not have rapid (e.g.,
within 24 hours) improvement in oxygenation after initiation of dexamethasone, baricitinib can
be considered for children aged 12 to 17 years (BIII) and for children aged 2 to 11 years (CIII).
• For children who require mechanical ventilation or ECMO and do not have rapid (e.g., within
24 hours) improvement in oxygenation after initiation of dexamethasone, baricitinib may be
Tofacitinib
There are no data on the efficacy of tofacitinib in pediatric patients with COVID-19; the Panel’s
recommendation is extrapolated from data in adults. The STOP-COVID trial compared tofacitinib to
the standard of care in adults hospitalized for COVID-19 pneumonia.38 The standard of care included
glucocorticoids for most patients. The study demonstrated a reduction in mortality and respiratory failure
at Day 28 for the tofacitinib arm when compared with the placebo arm. Tofacitinib has been studied
less extensively than baricitinib for the treatment of COVID-19. Thus, tofacitinib, as an alternative to
baricitinib, is recommended to be used in combination with dexamethasone in adults with COVID-19
who require high-flow oxygen or NIV. See Janus Kinase Inhibitors and Therapeutic Management of
Hospitalized Adults With COVID-19 for additional information.
No trials have evaluated the safety of using tofacitinib in children with COVID-19. Overall, there has
been more clinical experience with the use of tofacitinib than baricitinib in children, particularly when
used in children with juvenile idiopathic arthritis (JIA) as young as 2 years of age. A Phase 1 study
was conducted to define the pharmacokinetics and safety of using tofacitinib in children,39 and a Phase
3, double-blind, randomized, placebo-controlled trial investigated the efficacy of using tofacitinib in
children with JIA.40 Tofacitinib is available as a liquid formulation for children.
Given the established safety of tofacitinib in the pediatric population, tofacitinib can be considered an
alternative for children hospitalized for COVID-19 if baricitinib is not available (BIII). The dose of
tofacitinib that should be used to treat hospitalized children with COVID-19 has not been established.
As with baricitinib, the dose of tofacitinib for hospitalized children with COVID-19 likely needs to be
higher than the dose typically used to treat pediatric rheumatologic diseases. Therefore, clinicians should
consult with specialists experienced in treating children with immunosuppression (e.g., in pediatric
infectious disease, pediatric rheumatology) when considering administering tofacitinib to hospitalized
children with COVID-19.
Tocilizumab
Tocilizumab is an interleukin-6 inhibitor that has received an FDA Emergency Use Authorization for
the treatment of hospitalized adults and children with COVID-19 who are aged ≥2 years; receiving
systemic corticosteroids; and require supplemental oxygen, NIV, mechanical ventilation, or ECMO.41
Two large randomized controlled trials (REMAP-CAP and RECOVERY) conducted among hospitalized
adults with COVID-19 have demonstrated reductions in mortality with the use of tocilizumab.42,43 See
Interleukin-6 Inhibitors and Therapeutic Management of Hospitalized Adults With COVID-19 for
additional information.
The RECOVERY trial was an open-label study that included hospitalized adults who had an oxygen
saturation of <92% on room air or were receiving supplemental oxygen therapy; patients also had
C-reactive protein levels ≥75 mg/L.43 Patients were randomized to receive tocilizumab plus usual care
or usual care alone. Mortality at 28 days was significantly lower in the tocilizumab arm compared to
the usual care arm. The REMAP-CAP trial included adults with suspected or confirmed COVID-19
who were admitted to an intensive care unit and received either respiratory (i.e., NIV or mechanical
ventilation) or cardiovascular organ (i.e., vasopressor/inotrope) support.42 Patients were randomized
COVID-19 Treatment Guidelines 107
Sarilumab
Sarilumab, a monoclonal antibody that blocks interleukin-6 receptors, is not authorized by the FDA
for the treatment of COVID-19. Data on the efficacy of sarilumab for the treatment of COVID-19
hyperinflammation are limited, and there is a lack of pediatric dosing information. Therefore, the Panel
recommends against the use of sarilumab in hospitalized children with COVID-19, except in a clinical
trial (AIII).
References
1. Schuster JE, Halasa NB, Nakamura M, et al. A description of COVID-19-directed therapy in children admitted
to US intensive care units 2020. J Pediatric Infect Dis Soc. 2022;11(5):191-198. Available at: https://www.
ncbi.nlm.nih.gov/pubmed/35022779.
2. Goldman DL, Aldrich ML, Hagmann SHF, et al. Compassionate use of remdesivir in children with severe
COVID-19. Pediatrics. 2021;147(5):e2020047803. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33883243.
3. Chan-Tack K, Sampson M, Earp J, et al. Considerations and challenges in the remdesivir COVID-19 pediatric
development program. J Clin Pharmacol. 2023;63(2):259-265. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/36149807.
4. Kautsch K, Wiśniowska J, Friedman-Gruszczyńska J, Buda P. Evaluation of the safety profile and therapeutic
efficacy of remdesivir in children with SARS-CoV-2 infection—a single-center, retrospective, cohort study.
Eur J Pediatr. 2023;Published online ahead of print. Available at: https://pubmed.ncbi.nlm.nih.gov/37864601.
5. Food and Drug Administration. NDA 214787/S-11: Combined cross-discipline team leader, clinical, clinical
pharmacology, and division director review. 2022. Available at: https://www.fda.gov/media/166514/download.
6. Chiotos K, Hayes M, Kimberlin DW, et al. Multicenter initial guidance on use of antivirals for children
with coronavirus disease 2019/severe acute respiratory syndrome coronavirus 2. J Pediatric Infect Dis Soc.
2020;9(6):701-715. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32318706.
7. Dulek DE, Fuhlbrigge RC, Tribble AC, et al. Multidisciplinary guidance regarding the use of
immunomodulatory therapies for acute coronavirus disease 2019 in pediatric patients. J Pediatric Infect Dis
Soc. 2020;9(6):716-737. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32808988.
8. Wolf J, Abzug MJ, Anosike BI, et al. Updated guidance on use and prioritization of monoclonal antibody
therapy for treatment of COVID-19 in adolescents. J Pediatric Infect Dis Soc. 2022;11(5):177-185. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/35107571.
9. Dong Y, Mo X, Hu Y, et al. Epidemiology of COVID-19 among children in China. Pediatrics.
2020;145(6):e20200702. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32179660.
10. Delahoy MJ, Ujamaa D, Whitaker M, et al. Hospitalizations associated with COVID-19 among children and
adolescents—COVID-NET, 14 states, March 1, 2020–August 14, 2021. MMWR Morb Mortal Wkly Rep.
This section outlines the COVID-19 Treatment Guidelines Panel’s (the Panel) recommendations for
the therapeutic management of pediatric patients with multisystem inflammatory syndrome in children
(MIS-C). The case definition for MIS-C from the Council of State and Territorial Epidemiologists
(CSTE) and the Centers for Disease Control and Prevention includes individuals aged <21 years.1 The
recommendations in this section encompass this age group. There are few randomized controlled trials
that directly compared different treatment approaches for MIS-C, and these trials have limitations.
Because of those limitations, the Panel’s recommendations for the therapeutic management of
patients with MIS-C are primarily based on data from large descriptive and observational comparative
effectiveness studies. For information on the clinical manifestations of MIS-C, see Special
Considerations in Children.
Panel’s Recommendations
Initial treatment for MIS-C includes both immunomodulatory and antithrombotic therapy.
Initial Immunomodulatory Therapy
• IVIG 2 g/kg IBW IV (up to a maximum total dose of 100 g) plus low to moderate dose
methylprednisolone (1–2 mg/kg/day IV)a or another glucocorticoid at an equivalent dosea
(AIIb).
• Glucocorticoid monotherapy, only if IVIG is unavailable or contraindicated (BIIa).
• IVIG monotherapy, only if glucocorticoids are contraindicated (BIIb).
Intensification Immunomodulatory Therapy
• Intensification therapy is recommended for children with refractory MIS-C who do not improve
within 24 hours of receiving initial immunomodulatory therapy (AIII). One of the following can
be used (listed in alphabetical order):
• High-dose anakinra 5–10 mg/kg/day IV (preferred) or SUBQ in 1–4 divided dosesb (BIIb)
• Higher-dose glucocorticoid (e.g., methylprednisolone 10–30 mg/kg/day IV for 1–3 days,
MIS-C up to a maximum of 1,000 mg/day, or equivalent glucocorticoid for 1–3 days)a,b (BIIb)
• Infliximab 5–10 mg/kg IV for 1 doseb,c (BIIb)
Antithrombotic Therapy
• Low-dose aspirin (3–5 mg/kg PO once daily, up to a maximum dose of 81 mg) for all patients
without risk factors for bleeding (AIII), AND
• Anticoagulation for patients who fall under 1 of the following clinical scenarios:
• Therapeutic anticoagulation for patients with large CAAs according to the American Heart
Association guidelines for Kawasaki disease (AIII).
• Therapeutic anticoagulation for patients with moderate to severe LV dysfunction who have
no risk factors for bleeding (AIII).
• For patients with MIS-C who do not have large CAAs or moderate to severe LV dysfunction,
consider prophylactic or therapeutic anticoagulation on an individual basis, taking
into consideration risk factors for thrombosis and bleeding. See Table 3e for additional
information.
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
a
T he duration of glucocorticoid therapy may vary. When a patient shows clinical improvement (e.g., resolution of fever,
improvement of organ function, reduction of levels of inflammatory markers), a steroid taper should be initiated. Typically,
the patient’s clinical status guides the taper, and it continues for several weeks to avoid rebound inflammation. See Table
3e and text below.
b
In certain patients with severe illness, intensification therapy may include dual therapy with higher-dose glucocorticoids
plus anakinra (BIII) or higher-dose glucocorticoids plus infliximab (BIII). Anakinra and infliximab should not be used in
combination.
c
Infliximab should not be used in patients with macrophage activation syndrome.
Key: CAA = coronary artery aneurysm; IBW = ideal body weight; IV = intravenous; IVIG = intravenous immunoglobulin; LV =
left ventricular; MIS-C = multisystem inflammatory syndrome in children; PO = oral; SUBQ = subcutaneous
References
1. Centers for Disease Control and Prevention. Information for healthcare providers about multisystem
inflammatory syndrome in children (MIS-C). 2023. Available at: https://www.cdc.gov/mis/mis-c/hcp_cstecdc/
index.html. Accessed February 20, 2024.
2. Centers for Disease Control and Prevention. Multisystem inflammatory syndrome in adults (MIS-A) case
definition and information for healthcare providers. 2021. Available at:
https://www.cdc.gov/mis/mis-a/hcp.html. Accessed February 20, 2024.
3. Bamrah Morris S, Schwartz NG, Patel P, et al. Case series of multisystem inflammatory syndrome in adults
associated with SARS-CoV-2 infection—United Kingdom and United States, March–August 2020. MMWR
Morb Mortal Wkly Rep. 2020;69(40):1450-1456. Available at: https://pubmed.ncbi.nlm.nih.gov/33031361.
4. Kunal S, Ish P, Sakthivel P, Malhotra N, Gupta K. The emerging threat of multisystem inflammatory syndrome
in adults (MIS-A) in COVID-19: a systematic review. Heart Lung. 2022;54:7-18. Available at:
https://pubmed.ncbi.nlm.nih.gov/35306376.
5. Melgar M, Abrams JY, Godfred-Cato S, et al. A multicenter retrospective cohort study to characterize patients
hospitalized with multisystem inflammatory syndrome in adults and coronavirus disease 2019 in the United
States, 2020–2021. Clin Infect Dis. 2023;77(10):1395-1405. Available at:
https://pubmed.ncbi.nlm.nih.gov/37384794.
6. Verdoni L, Mazza A, Gervasoni A, et al. An outbreak of severe Kawasaki-like disease at the Italian epicentre
of the SARS-CoV-2 epidemic: an observational cohort study. Lancet. 2020;395(10239):1771-1778. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/32410760.
Summary Recommendations
Hemodynamics
• For adults with COVID-19 and shock, the COVID-19 Treatment Guidelines Panel (the Panel) recommends using dynamic
parameters, capillary refill time, and/or lactate levels over static parameters to assess fluid responsiveness (BIIa).
• For acute resuscitation in adults who have COVID-19 and shock, there is insufficient evidence for the Panel to
recommend either for or against the use of balanced crystalloids, such as Ringer’s lactate solution, over normal saline.
• For acute resuscitation in adults with COVID-19 and shock, the Panel recommends against the initial use of albumin
(BI).
• For adults with COVID-19 and shock, the Panel recommends norepinephrine as the first-choice vasopressor (AI).
• For adults with COVID-19 and shock, the Panel recommends titrating vasoactive agents and targeting a mean arterial
pressure (MAP) of 60 to 65 mm Hg over targeting a higher MAP (BI).
• The Panel recommends against using hydroxyethyl starches for intravascular volume replacement in adult patients
with COVID-19 and sepsis or septic shock (AI).
• As a second-line vasopressor, the Panel recommends adding either vasopressin (up to 0.03 units/min) (BIIa) or
epinephrine (BIIb) to norepinephrine to raise MAP to the target or adding vasopressin (up to 0.03 units/min) (BIIa) to
decrease the norepinephrine dose.
• The Panel recommends against using low-dose dopamine for renal protection (AI).
• The Panel recommends using dobutamine in adult patients with COVID-19 who show evidence of cardiac dysfunction
and persistent hypoperfusion despite adequate fluid loading and the use of vasopressor agents (BIII).
• The Panel recommends that all adult patients with COVID-19 who require vasopressors have an arterial catheter placed
as soon as practical, if resources are available (BIII).
• For adult patients with refractory septic shock who have completed a course of corticosteroids to treat COVID-19, the
Panel recommends using low-dose corticosteroid therapy (“shock-reversal”) over no corticosteroid therapy (BIIa).
Oxygenation and Ventilation
• For adults with COVID-19 and acute hypoxemic respiratory failure despite conventional oxygen therapy, the Panel
recommends starting therapy with high-flow nasal cannula (HFNC) oxygen; if patients fail to respond, noninvasive
ventilation or intubation and mechanical ventilation should be initiated (BIIa).
• For adults with COVID-19 and acute hypoxemic respiratory failure despite conventional oxygen therapy who do not
have an indication for endotracheal intubation and for whom HFNC oxygen is not available, the Panel recommends
performing a closely monitored trial of noninvasive ventilation (BIIa).
• For adults with persistent hypoxemia who require HFNC oxygen and for whom endotracheal intubation is not indicated,
the Panel recommends a trial of awake prone positioning (BIIa).
• The Panel recommends against the use of awake prone positioning as a rescue therapy for refractory hypoxemia to
avoid intubation in patients who otherwise meet the indications for intubation and mechanical ventilation (AIII).
• If intubation becomes necessary, the procedure should be performed by an experienced practitioner in a controlled
setting due to the enhanced risk of exposing health care practitioners to SARS-CoV-2 during intubation (AIII).
• For mechanically ventilated adults with COVID-19 and acute respiratory distress syndrome (ARDS):
• The Panel recommends using low tidal volume (VT) ventilation (VT 4–8 mL/kg of predicted body weight) over higher
VT ventilation (VT >8 mL/kg) (AI).
• The Panel recommends targeting plateau pressures of <30 cm H2O (AIIa).
• The Panel recommends using a conservative fluid strategy over a liberal fluid strategy (BIIa).
• The Panel recommends against the routine use of inhaled nitric oxide (AIIa).
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
COVID-19 can progress to critical illness, including hypoxemic respiratory failure, acute respiratory
distress syndrome (ARDS), septic shock, thromboembolic disease, renal and hepatic dysfunction,
cardiac dysfunction, central nervous system disease, and exacerbation of underlying comorbidities in
both adults and children.
In these Guidelines, many of the early recommendations for managing critically ill adults with
COVID-19 were extrapolated from experience with other causes of sepsis and respiratory failure.1
However, the amount of research on the management of these patients has grown, and the COVID-19
Treatment Guidelines Panel’s (the Panel) current recommendations have been informed by that research.
Treating patients with COVID-19 in the intensive care unit (ICU) often requires managing underlying
illnesses or COVID-19–related morbidities. Clinicians also need to focus on preventing ICU-related
complications, as they would for any patient admitted to the ICU.
Inflammatory Response
Many critically ill patients with COVID-19 meet the criteria for virus-induced sepsis because they have
life-threatening organ dysfunction related to a dysregulated host response to SARS-CoV-2 infection.2
Patients with COVID-19 may express increased levels of pro-inflammatory cytokines and anti-
inflammatory cytokines, a condition that has been called “cytokine release syndrome” or a “cytokine
storm,” but these terms are imprecise and misnomers. The magnitude of cytokine elevation in many
critically ill patients with COVID-19 is modest compared to the levels in patients with sepsis and ARDS
not related to COVID-19.3,4
Thromboembolic Events
Prothrombotic states and higher rates of venous thromboembolic disease have been observed in
adults who are critically ill with COVID-19. In some studies, thromboemboli were diagnosed in
patients who received prophylactic doses of heparin.5-7 Autopsy studies provide additional evidence of
thromboembolic disease and microvascular thrombosis in patients with COVID-19.8 See Antithrombotic
Therapy in Patients With COVID-19 for a more detailed discussion.
Additional Considerations
Drug-Drug Interactions
All patients in the ICU should be routinely monitored for drug-drug interactions. The potential for
drug-drug interactions between investigational medications or any medications used off-label to treat
COVID-19 Treatment Guidelines 128
Sedation Management
International guidelines provide recommendations on the prevention, detection, and treatment of pain,
sedation, and delirium in ICU patients.25,26 Sedation management strategies, such as maintaining a light
level of sedation (when appropriate) and minimizing sedative exposure, have shortened the duration of
mechanical ventilation and the length of ICU stay among patients without COVID-19.27,28
The Society of Critical Care Medicine (SCCM) ICU Liberation Campaign promotes the ICU Liberation
Bundle (A–F) to improve post-ICU patient outcomes.29 The A–F Liberation Bundle includes the
following elements:
A. Assess, prevent, and manage pain
B. Both spontaneous awakening trials and breathing trials
C. Choice of analgesia and sedation
D. Delirium: assess, prevent, and manage
E. Early mobility and exercise
F. Family engagement and empowerment
The A–F Liberation Bundle provides frontline staff with practical application strategies for each
element, and an interprofessional team model should be used to incorporate the elements.29 This
approach helps standardize communication among team members, improves survival, and reduces
long-term cognitive dysfunction of patients.30 The elements of the A–F Liberation Bundle represent the
steps required to implement the SCCM Clinical Practice Guidelines for the Prevention and Management
of Pain, Agitation/Sedation, Delirium, Immobility, and Sleep Disruption in Adult Patients in the ICU.25
Despite the known benefits of the A–F Liberation Bundle,29 its impact has not been directly assessed in
patients with COVID-19. However, implementing the elements of the bundle should be encouraged to
improve patient outcomes in the ICU.
Some factors may impede routine implementation of the A–F Liberation Bundle and increase the risk of
ICU and post-ICU complications. For example, staff workload increases when patients with COVID-19
require prolonged mechanical ventilation, deep sedation, or neuromuscular blockade. In addition, drug
shortages, which were common early in the pandemic, may affect the choice of analgesia or sedation.
During a drug shortage, older sedatives that have prolonged durations of action and active metabolites
are more likely to be prescribed.
Acknowledgments
The Surviving Sepsis Campaign (SSC), an initiative supported by SCCM and the European Society
of Intensive Care Medicine, issued the Guidelines on the Management of Critically Ill Adults with
Coronavirus Disease 2019 (COVID-19) in March 2020, and a revised version was published in March
2021.1 The Panel based its recommendations for the care of critically ill adults with COVID-19 on the
SSC COVID-19 guidelines with permission, and the Panel gratefully acknowledges the work of the SSC
COVID-19 Guidelines panel. The Panel also acknowledges the contributions and expertise of Andrew
Rhodes, MBBS, MD, of St. George’s University Hospitals in London, England, and Waleed Alhazzani,
MBBS, MSc, of McMaster University in Hamilton, Canada.
References
1. Alhazzani W, Evans L, Alshamsi F, et al. Surviving Sepsis Campaign guidelines on the management of adults
with coronavirus disease 2019 (COVID-19) in the ICU: first update. Crit Care Med. 2021;49(3):e219-e234.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/33555780.
2. Merad M, Blish CA, Sallusto F, Iwasaki A. The immunology and immunopathology of COVID-19. Science.
2022;375(6585):1122-1127. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35271343.
3. Leisman DE, Ronner L, Pinotti R, et al. Cytokine elevation in severe and critical COVID-19: a rapid
systematic review, meta-analysis, and comparison with other inflammatory syndromes. Lancet Respir Med.
2020;8(12):1233-1244. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33075298.
Most of the hemodynamic recommendations below are similar to those published in Surviving Sepsis
Campaign: International Guidelines for Management of Sepsis and Septic Shock 2021.1 Ultimately,
adult patients with COVID-19 who require fluid resuscitation or hemodynamic management of shock
should be treated and managed as adult patients with septic shock.
Recommendation
• For adults with COVID-19 and shock, the COVID-19 Treatment Guidelines Panel (the Panel)
recommends using dynamic parameters, capillary refill time, and/or lactate levels over static
parameters to assess fluid responsiveness (BIIa).
Rationale
In a systematic review and meta-analysis of 13 randomized clinical trials in intensive care unit (ICU)
patients without COVID-19 (n = 1,652), the use of dynamic assessment to guide fluid therapy reduced
mortality (risk ratio 0.59; 95% CI, 0.42–0.83), ICU length of stay (weighted mean difference -1.16
days; 95% CI, -1.97 to -0.36), and duration of mechanical ventilation (weighted mean difference -2.98
hours; 95% CI, -5.08 to -0.89).2 The dynamic parameters used in these trials included stroke volume
variation, pulse pressure variation, and stroke volume change after a passive leg raise or fluid challenge.
Passive leg raising followed by assessment of pulse pressure variation and stroke volume variation
appears to predict fluid responsiveness with the greatest accuracy.3 The static parameters included some
components of early goal-directed therapy (e.g., central venous pressure, mean arterial pressure [MAP]).
In patients who did not have COVID-19, resuscitation therapies for shock, as indicated by serum lactate
levels, were summarized in a systematic review and meta-analysis of 7 randomized controlled trials
(n = 1,301).4 When compared with therapy guided by central venous oxygen saturation levels, therapy
directed by early lactate clearance was associated with a reduction in mortality (relative ratio 0.68;
95% CI, 0.56–0.82), shorter ICU stay (mean difference -1.64 days; 95% CI, -3.23 to -0.05), and shorter
duration of mechanical ventilation (mean difference -10.22 hours; 95% CI, -15.94 to -4.50).
Recommendation
• For acute resuscitation in adults who have COVID-19 and shock, there is insufficient evidence
for the Panel to recommend either for or against the use of balanced crystalloids, such as Ringer’s
lactate solution, over normal saline.
Rationale
The composition of sodium, potassium, and chloride found in balanced crystalloids, such as Ringer’s
lactate solution, is similar to the composition found in extracellular fluid. The use of balanced
crystalloids may prevent hyperchloremic metabolic acidosis, which has been associated with
administration of large quantities of normal saline.5 Observational data have suggested an association
between normal saline and acute kidney injury and higher risk of death,6 and many Panel members with
experience in acute fluid resuscitation use balanced crystalloids.
A pragmatic randomized trial compared the use of balanced and unbalanced crystalloids for intravenous
fluid administration in critically ill adults without COVID-19 (n = 15,802).7 The rate of the composite
COVID-19 Treatment Guidelines 134
Recommendation
• For acute resuscitation in adults with COVID-19 and shock, the Panel recommends against the
initial use of albumin (BI).
Rationale
A meta-analysis of 20 non–COVID-19 randomized controlled trials (n = 13,047) that compared the use
of albumin or fresh-frozen plasma to crystalloids in critically ill patients found no difference in all-cause
mortality between the treatment arms.12 In contrast, a meta-analysis of 17 non–COVID-19 randomized
controlled trials (n = 1,977) that compared the use of albumin to crystalloids in patients with sepsis
reported a reduction in mortality among the patients who received albumin (OR 0.82; 95% CI, 0.67–1.0;
P = 0.047).13 Given the higher cost of albumin and the lack of a definitive clinical benefit, for acute
resuscitation in adults with COVID-19 and shock, the Panel recommends against the initial use of
albumin (BI).
Recommendation
• For adults with COVID-19 and shock, the Panel recommends norepinephrine as the first-choice
vasopressor (AI).
Rationale
Due to its vasoconstrictive effects, norepinephrine increases MAP with little change to heart rate and
less increase in stroke volume than dopamine. Dopamine increases MAP and cardiac output, primarily
due to increases in stroke volume and heart rate. Norepinephrine is more potent than dopamine and may
be more effective at reversing hypotension in patients with septic shock. Dopamine may be particularly
useful in patients with compromised systolic function, but it causes more tachycardia and may be more
arrhythmogenic than norepinephrine.14 Dopamine may also influence the endocrine response via the
hypothalamic pituitary axis and have immunosuppressive effects.15 A systematic review and meta-
analysis of 11 non–COVID-19 randomized controlled trials that compared vasopressors used to treat
patients with septic shock found that norepinephrine use resulted in lower all-cause mortality (risk ratio
0.89; 95% CI, 0.81–0.98) and a lower risk of arrhythmias (risk ratio 0.48; 95% CI, 0.40–0.58) than
dopamine use.16 Although the beta-1 activity of dopamine would be useful in patients with myocardial
dysfunction, the greater risk of arrhythmias limits its use.17,18
Rationale
A meta-analysis of individual patient data from 2 non–COVID-19 randomized controlled trials (n =
894) compared higher versus lower blood pressure targets for vasopressor therapy in adult patients with
shock.19 The study reported no significant differences between the higher-target and lower-target arms
for 28-day mortality (OR 1.15; 95% CI, 0.87–1.52), 90-day mortality (OR 1.08; 95% CI, 0.84–1.44),
myocardial injury (OR 1.47; 95% CI, 0.64–3.56), or limb ischemia (OR 0.92; 95% CI, 0.36–2.10). The
risk of arrhythmia was increased in the higher-target arm (OR 2.50; 95% CI, 1.35–4.77).
Similarly, the 65 trial, a randomized controlled trial in patients without COVID-19 (n = 2,463), reported
no significant difference in mortality between patients that received vasopressor therapy guided by a
target MAP of 60 to 65 mm Hg and those that received treatment guided by a higher, standard-of-care
target MAP (41% vs. 43.8%; relative risk 0.93; 95% CI, 0.85–1.03).20 Given the indication of improved
outcome with lower MAP targets (and no firm indication of harm), the Panel recommends titrating
vasoactive agents and targeting a MAP of 60 to 65 over targeting a higher MAP (BI).
References
1. Evans L, Rhodes A, Alhazzani W, et al. Surviving Sepsis Campaign: international guidelines for management
of sepsis and septic shock 2021. Intensive Care Med. 2021;47(11):1181-1247. Available at: https://www.ncbi.
nlm.nih.gov/pubmed/34599691.
2. Bednarczyk JM, Fridfinnson JA, Kumar A, et al. Incorporating dynamic assessment of fluid responsiveness
into goal-directed therapy: a systematic review and meta-analysis. Crit Care Med. 2017;45(9):1538-1545.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/28817481.
COVID-19 Treatment Guidelines 136
The COVID-19 Treatment Guidelines Panel’s (the Panel) recommendations in this section were
informed by the Surviving Sepsis Campaign guidelines for managing sepsis and guidelines for managing
COVID-19 in adults.
Severe illness in people with COVID-19 typically occurs approximately 1 week after the onset of
symptoms. The most common symptom is dyspnea, which is often accompanied by hypoxemia. Patients
with severe disease typically require supplemental oxygen and should be monitored closely for worsening
respiratory status, because some patients may progress to acute respiratory distress syndrome (ARDS).
Goal of Oxygenation
The optimal oxygen saturation measured by pulse oximetry (SpO2) in adults with COVID-19 who are
receiving supplemental oxygen is unknown. However, a target SpO2 of 92% to 96% seems logical,
considering that indirect evidence from patients without COVID-19 suggests that an SpO2 <92% or
>96% may be harmful.1,2 Special care should be taken when assessing SpO2 in patients with darker skin
pigmentation, as recent reports indicate that occult hypoxemia (defined as arterial oxygen saturation
[SaO2] <88% despite an SpO2 >92%) is more common in these patients.3,4 See Clinical Spectrum of
SARS-CoV-2 Infection for more information.
The potential harm of maintaining an SpO2 <92% was demonstrated during a trial that randomly assigned
patients with ARDS who did not have COVID-19 to either a conservative oxygen strategy (target SpO2 of
88% to 92%) or a liberal oxygen strategy (target SpO2 ≥96%).1 The trial was stopped early due to futility
after enrolling 205 patients, but increased mortality was observed at Day 90 in the conservative oxygen
strategy arm (between-group risk difference 14%; 95% CI, 0.7% to 27%), and a trend toward increased
mortality was observed at Day 28 (between-group risk difference 8%; 95% CI, -5% to 21%).
The results of a meta-analysis of 25 randomized trials that involved patients without COVID-19
demonstrated the potential harm of maintaining an SpO2 >96%.2 This study found that a liberal oxygen
supplementation strategy (a median fraction of inspired oxygen [FiO2] of 0.52) was associated with an
increased risk of in-hospital mortality (relative risk 1.21; 95% CI, 1.03–1.43) when compared with a
more conservative SpO2 supplementation strategy (a median FiO2 of 0.21).
References
1. Barrot L, Asfar P, Mauny F, et al. Liberal or conservative oxygen therapy for acute respiratory distress
syndrome. N Engl J Med. 2020;382(11):999-1008. Available at: https://www.ncbi.nlm.nih.gov/
pubmed/32160661.
2. Chu DK, Kim LH, Young PJ, et al. Mortality and morbidity in acutely ill adults treated with liberal versus
conservative oxygen therapy (IOTA): a systematic review and meta-analysis. Lancet. 2018;391(10131):1693-
1705. Available at: https://www.ncbi.nlm.nih.gov/pubmed/29726345.
3. Chesley CF, Lane-Fall MB, Panchanadam V, et al. Racial disparities in occult hypoxemia and clinically based
mitigation strategies to apply in advance of technological advancements. Respir Care. 2022;67(12):1499-
1507. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35679133.
4. Valbuena VSM, Seelye S, Sjoding MW, et al. Racial bias and reproducibility in pulse oximetry among
medical and surgical inpatients in general care in the Veterans Health Administration 2013–19: multicenter,
retrospective cohort study. BMJ. 2022;378:e069775. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35793817.
5. Frat JP, Quenot JP, Badie J, et al. Effect of high-flow nasal cannula oxygen vs standard oxygen therapy on
mortality in patients with respiratory failure due to COVID-19: the SOHO-COVID randomized clinical trial.
JAMA. 2022;328(12):1212-1222. Available at: https://pubmed.ncbi.nlm.nih.gov/36166027.
6. Frat JP, Thille AW, Mercat A, et al. High-flow oxygen through nasal cannula in acute hypoxemic
respiratory failure. N Engl J Med. 2015;372(23):2185-2196. Available at: https://www.ncbi.nlm.nih.gov/
pubmed/25981908.
7. Ni YN, Luo J, Yu H, et al. The effect of high-flow nasal cannula in reducing the mortality and the rate of
endotracheal intubation when used before mechanical ventilation compared with conventional oxygen therapy
COVID-19 Treatment Guidelines 144
Rationale
Variable rates of community- and hospital-acquired infections have been reported in adult patients
with COVID-19. Bacterial coinfection at the time of hospitalization has been reported in 1% to 3.5%
of patients with COVID-19.1,2 Secondary infections have been reported in 14% to 37% of patients in
intensive care units, but the reported rates have been influenced by differences in the severity of illness,
duration of hospitalization, method of diagnosis, and time period studied.3,4
No clinical trials have evaluated the use of empiric broad-spectrum antimicrobials in patients with
severe or critical COVID-19 or other coronavirus infections. Routine, empiric use of antimicrobials
in patients with severe or critical COVID-19 is not recommended (BIII). This recommendation is
intended to mitigate the unintended consequences of antimicrobial side effects and resistance. The use
of antimicrobials may be considered in specific situations, such as the presence of a lobar infiltrate on a
chest X-ray, leukocytosis, an elevated serum lactate level, microbiologic data, or shock.
The use of antimicrobials in patients with severe or critical COVID-19 should follow guidelines
established for other hospitalized patients (i.e., for hospital-acquired pneumonia, ventilator-associated
pneumonia, or bloodstream infections associated with central lines). It is unclear whether using
corticosteroids or other immunomodulatory agents recommended in the Guidelines should alter such
approaches.
Immune-Based Therapy
For recommendations on the use of immunomodulators in patients with COVID-19, see
Immunomodulators.
Antithrombotic Therapy
For the Panel’s recommendations regarding the use of antithrombotic therapy in critical care settings,
see Antithrombotic Therapy in Patients With COVID-19 and Therapeutic Management of Hospitalized
COVID-19 Treatment Guidelines 147
References
1. Langford BJ, So M, Raybardhan S, et al. Bacterial co-infection and secondary infection in patients with
COVID-19: a living rapid review and meta-analysis. Clin Microbiol Infect. 2020;26(12):1622-1629. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/32711058.
2. Garcia-Vidal C, Sanjuan G, Moreno-García E, et al. Incidence of co-infections and superinfections in
hospitalized patients with COVID-19: a retrospective cohort study. Clin Microbiol Infect. 2021;27(1):83-88.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/32745596.
3. Zangrillo A, Beretta L, Scandroglio AM, et al. Characteristics, treatment, outcomes and cause of death of
invasively ventilated patients with COVID-19 ARDS in Milan, Italy. Crit Care Resusc. 2020;22(3):200-211.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/32900326.
4. Yang X, Yu Y, Xu J, et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia
in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med. 2020;8(5):475-
481. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32105632.
Recommendation
• There is insufficient evidence for the COVID-19 Treatment Guidelines Panel to recommend
either for or against the use of extracorporeal membrane oxygenation (ECMO) in adults with
COVID-19–associated acute respiratory distress syndrome (ARDS) and refractory hypoxemia.
Rationale
ECMO has been used as a rescue therapy in patients with ARDS caused by COVID-19 and refractory
hypoxemia. However, there is no conclusive evidence that ECMO is responsible for better clinical
outcomes, regardless of the cause of hypoxemic respiratory failure.1-4
The clinical outcomes for patients with ARDS who are treated with ECMO are variable and depend
on multiple factors, including the etiology of hypoxemic respiratory failure, the severity of pulmonary
and extrapulmonary illness, the presence of comorbidities, and the ECMO experience of the individual
center.5-7 Several multicenter, observational cohort studies from the first half of 20208-10 reported that
patients who required ECMO for COVID-19 had similar mortality to patients in a 2018 randomized
study who did not have COVID-19 but had ARDS and received ECMO.3
However, subsequent observational studies reported that in patients who required ECMO for COVID-19,
outcomes in late 2020 and early 2021 were worse than outcomes in spring 2020.11,12 The largest analysis
used data from 4,812 patients in the international Extracorporeal Life Support Organization Registry
who had COVID-19 and received ECMO in 2020.11 At centers that provided ECMO throughout 2020,
patients who started ECMO before May 1, 2020, had a 90-day mortality of 36.9% after ECMO initiation
(95% CI, 34.1% to 39.7%). At the same centers, patients who initiated ECMO between May 2 and
December 31, 2020, had a 90-day mortality of 51.9% (95% CI, 50.0% to 53.8%). Furthermore, at
centers that started using ECMO for patients with COVID-19 after May 1, 2020, the 90-day mortality
after ECMO initiation was 58.9% (95% CI, 55.4% to 62.3%). These observational data should be
interpreted with caution, as they may reflect a changing case mix of patients with COVID-19 who were
referred for ECMO.
Three target emulation trials compared the efficacy of ECMO and conventional mechanical ventilation
in patients with severe COVID-19–associated ARDS.10,13,14 The largest of these trials included 844
patients with COVID-19 who had hypoxemic respiratory failure and were receiving ECMO.14 The
study reported that the patients who received ECMO had lower 60-day mortality than the patients who
received only conventional mechanical ventilation (26% vs. 33.2%; risk difference −7.1%; 95% CI,
−8.2% to −6.1%; risk ratio 0.78; 95% CI, 0.75–0.82). Favorable ECMO outcomes were associated with
the following factors: aged <65 years, a ratio of arterial partial pressure of oxygen to fraction of inspired
oxygen <80 mm Hg, ≤10-day duration of mechanical ventilation, and >15 cm H2O driving pressure.
Ultimately, the benefits of ECMO cannot be clearly defined for patients with COVID-19 and severe
ARDS because no randomized controlled trials have evaluated the use of ECMO in this population.
Clinicians interested in pursuing ECMO for patients with COVID-19 and severe ARDS should consider
transferring care to high-volume ECMO centers. These patients should be entered into clinical trials or
COVID-19 Treatment Guidelines 149
References
1. Peek GJ, Mugford M, Tiruvoipati R, et al. Efficacy and economic assessment of conventional ventilatory
support versus extracorporeal membrane oxygenation for severe adult respiratory failure (CESAR): a
multicentre randomised controlled trial. Lancet. 2009;374(9698):1351-1363. Available at: https://www.ncbi.
nlm.nih.gov/pubmed/19762075.
2. Pham T, Combes A, Rozé H, et al. Extracorporeal membrane oxygenation for pandemic influenza A(H1N1)-
induced acute respiratory distress syndrome: a cohort study and propensity-matched analysis. Am J Respir Crit
Care Med. 2013;187(3):276-285. Available at: https://www.ncbi.nlm.nih.gov/pubmed/23155145.
3. Combes A, Hajage D, Capellier G, et al. Extracorporeal membrane oxygenation for severe acute respiratory
distress syndrome. N Engl J Med. 2018;378(21):1965-1975. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29791822.
4. Munshi L, Walkey A, Goligher E, et al. Venovenous extracorporeal membrane oxygenation for acute
respiratory distress syndrome: a systematic review and meta-analysis. Lancet Respir Med. 2019;7(2):163-172.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/30642776.
5. Bullen EC, Teijeiro-Paradis R, Fan E. How I select which patients with ARDS should be treated with
venovenous extracorporeal membrane oxygenation. Chest. 2020;158(3):1036-1045. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32330459.
6. Henry BM, Lippi G. Poor survival with extracorporeal membrane oxygenation in acute respiratory distress
syndrome (ARDS) due to coronavirus disease 2019 (COVID-19): pooled analysis of early reports. J Crit Care.
2020;58:27-28. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32279018.
7. Mustafa AK, Alexander PJ, Joshi DJ, et al. Extracorporeal membrane oxygenation for patients with
COVID-19 in severe respiratory failure. JAMA Surg. 2020;155(10):990-992. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32780089.
8. Barbaro RP, MacLaren G, Boonstra PS, et al. Extracorporeal membrane oxygenation support in
COVID-19: an international cohort study of the Extracorporeal Life Support Organization Registry. Lancet.
2020;396(10257):1071-1078. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32987008.
9. Schmidt M, Hajage D, Lebreton G, et al. Extracorporeal membrane oxygenation for severe acute
respiratory distress syndrome associated with COVID-19: a retrospective cohort study. Lancet Respir Med.
2020;8(11):1121-1131. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32798468.
10. Shaefi S, Brenner SK, Gupta S, et al. Extracorporeal membrane oxygenation in patients with severe
respiratory failure from COVID-19. Intensive Care Med. 2021;47(2):208-221. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33528595.
11. Barbaro RP, MacLaren G, Boonstra PS, et al. Extracorporeal membrane oxygenation for COVID-19:
evolving outcomes from the international Extracorporeal Life Support Organization Registry. Lancet.
2021;398(10307):1230-1238. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34599878.
12. Broman LM, Eksborg S, Lo Coco V, et al. Extracorporeal membrane oxygenation for COVID-19 during first
and second waves. Lancet Respir Med. 2021;9(8):e80-e81. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34146489.
13. Hajage D, Combes A, Guervilly C, et al. Extracorporeal membrane oxygenation for severe acute respiratory
distress syndrome associated with COVID-19: an emulated target trial analysis. Am J Respir Crit Care Med.
2022;206(3):281-294. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35533052.
14. Urner M, Barnett AG, Bassi GL, et al. Venovenous extracorporeal membrane oxygenation in patients with
acute COVID-19 associated respiratory failure: comparative effectiveness study. BMJ. 2022;377:e068723.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/35508314.
COVID-19 may lead to critical illness in children, including hypoxemic respiratory failure, acute
respiratory distress syndrome, septic shock, cardiac dysfunction, thromboembolic disease, hepatic or
renal dysfunction, central nervous system disease, and exacerbation of underlying comorbidities. In
addition, multisystem inflammatory syndrome in children (MIS-C) is a rare, postinfectious complication
of SARS-CoV-2 and is frequently associated with critical illness.
Data informing the optimal management of children with acute COVID-19 or MIS-C are limited. In
general, care for children with acute COVID-19 or MIS-C should follow the usual principles of pediatric
critical care, such as the 2023 Pediatric Acute Lung Injury Consensus Conference (2023 PALICC-2)
recommendations and the Surviving Sepsis Campaign guidelines for pediatric sepsis. For patients with
COVID-19 in the intensive care unit (ICU), treatment often requires managing underlying illnesses
other than COVID-19 that may have contributed to the need for ICU admission, as well as managing
COVID-19 complications. Finally, prevention of ICU-related complications is critical to achieving
optimal clinical outcomes for any patient admitted to the ICU.
Thromboembolic Events
Limited data characterize the prevalence of thromboembolic disease in children with COVID-19 or
MIS-C. In a multicenter, retrospective cohort study including 814 hospitalized patients with COVID-19
or MIS-C, thromboembolic events were detected in 2.1% of patients with COVID-19 and 6.5% of
patients with MIS-C.6 The same study conducted a multivariable analysis and found that the following
COVID-19 Treatment Guidelines 151
Neurologic Involvement
Neurologic involvement is common in children with COVID-19 or MIS-C and is estimated to occur
in approximately 30% to 40% of children hospitalized with these conditions.2,11 Severe neurologic
manifestations, including severe encephalopathy, stroke, demyelinating conditions, cerebral edema, and
Guillain-Barré syndrome, have also been described.11
Additional Considerations
Considerations for the care of children with COVID-19 or MIS-C should generally follow the usual
principles of pediatric critical care. Sedation management and considerations related to post-intensive
care syndrome–pediatric (PICS-p) are discussed below. See Oxygenation and Ventilation for Children,
Hemodynamic Considerations for Children, and Extracorporeal Membrane Oxygenation for Children
for more information on pediatric critical care.
Sedation Management
Guidelines for the management of pain, agitation, neuromuscular blockade, delirium, and early mobility
(PANDEM) in infants and children admitted to the pediatric ICU have recently been published.12 In
general, children with COVID-19 or MIS-C who require mechanical ventilation should be managed
per the usual principles of critical care for patients with respiratory failure who require mechanical
ventilation. The usual care includes sedation with the minimal effective dose required to tolerate
mechanical ventilation, optimize gas exchange, and minimize the risk of ventilator-induced lung injury.
Using validated pain and sedation scales, the critical care team should set a sedation/pain target based on
the phase of ventilation.
Two large randomized controlled trials examined the use of protocols to manage sedation titration
in children receiving mechanical ventilation.13,14 In both studies, participants received usual care
or protocol-driven care implemented by nurses. Use of the protocols did not significantly reduce
the duration of ventilation or affect other study outcomes. However, a patient’s risk of harm from
protocolized sedation is generally low, which led the Society of Critical Care Medicine to issue a
conditional recommendation, based on low-level evidence, in its PANDEM clinical practice guidelines
for the use of protocolized sedation in children who are critically ill and receiving mechanical
ventilation.12
Studies evaluating data on the effect of early mobility protocols on critically ill children are limited.
One trial evaluated the safety and feasibility of early mobilization in 58 patients who were randomized
to receive usual care or early physical therapy, occupational therapy, and speech therapy consultation
within 72 hours of admission to the pediatric ICU.15 Although no differences between the arms
were demonstrated for clinical, functional, or quality of life outcomes, the study found that the early
rehabilitation consultations were safe and feasible.
Ongoing trials are measuring the effect of early mobilization on patient-centered outcomes in children
receiving mechanical ventilation. The PANDEM guideline statement issued by the Society of Critical
COVID-19 Treatment Guidelines 152
Acknowledgments
For these pediatric recommendations, the COVID-19 Treatment Guidelines Panel integrated the
recommendations from pediatric-specific guidelines, including the European Society of Paediatric and
Neonatal Intensive Care’s recommendations20 for the care of critically ill children with COVID-19
and the Surviving Sepsis Campaign’s perspective on managing sepsis in children with COVID-19.21
In addition, recommendations from several treatment guidelines not related to COVID-19, such as the
Surviving Sepsis Campaign International Guidelines for the Management of Septic Shock and Sepsis-
Associated Organ Dysfunction in Children,22 the 2023 PALICC-2 recommendations,23 and the Society of
Critical Care Medicine’s PANDEM guidelines,12 were integrated.
References
1. Abrams JY, Oster ME, Godfred-Cato SE, et al. Factors linked to severe outcomes in multisystem
inflammatory syndrome in children (MIS-C) in the USA: a retrospective surveillance study. Lancet Child
Adolesc Health. 2021;5(5):323-331. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33711293.
2. Feldstein LR, Tenforde MW, Friedman KG, et al. Characteristics and outcomes of US children and adolescents
with multisystem inflammatory syndrome in children (MIS-C) compared with severe acute COVID-19.
JAMA. 2021;325(11):1074-1087. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33625505.
3. Alsaied T, Tremoulet AH, Burns JC, et al. Review of cardiac involvement in multisystem inflammatory
syndrome in children. Circulation. 2021;143(1):78-88. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33166178.
4. Valverde I, Singh Y, Sanchez-de-Toledo J, et al. Acute cardiovascular manifestations in 286 children
with multisystem inflammatory syndrome associated with COVID-19 infection in Europe. Circulation.
2021;143(1):21-32. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33166189.
5. Jain SS, Steele JM, Fonseca B, et al. COVID-19 vaccination-associated myocarditis in adolescents. Pediatrics.
2021;148(5):e2021053427. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34389692.
6. Whitworth H, Sartain SE, Kumar R, et al. Rate of thrombosis in children and adolescents hospitalized with
COVID-19 or MIS-C. Blood. 2021;138(2):190-198. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33895804.
7. Raina R, Chakraborty R, Mawby I, et al. Critical analysis of acute kidney injury in pediatric COVID-19
patients in the intensive care unit. Pediatr Nephrol. 2021;36(9):2627-2638. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33928439.
8. Kari JA, Shalaby MA, Albanna AS, et al. Acute kidney injury in children with COVID-19: a retrospective
study. BMC Nephrol. 2021;22(1):202. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34059010.
9. Basalely A, Gurusinghe S, Schneider J, et al. Acute kidney injury in pediatric patients hospitalized with acute
COVID-19 Treatment Guidelines 153
Children with acute COVID-19 infrequently experience shock requiring hemodynamic support.
However, similar to children with sepsis or septic shock from other causes, children with COVID-19 and
shock should be evaluated and managed per the Surviving Sepsis Campaign International Guidelines for
the Management of Septic Shock and Sepsis-Associated Organ Dysfunction in Children.1,2
Shock occurs in approximately half of the patients with multisystem inflammatory syndrome in
children (MIS-C); reported prevalence ranges from 35% to 80%.3-5 Limited data inform the optimal
hemodynamic management for MIS-C. Given that the physiology observed in patients with MIS-C
results from a combination of distributive, cardiogenic, and, occasionally, hypovolemic shock, the
COVID-19 Treatment Guidelines Panel (the Panel) suggests that clinicians use the management
principles outlined in the Surviving Sepsis Campaign’s guidelines for children, as well as the
principles for clinical management of heart failure and general critical care, as appropriate. The Panel’s
recommendations apply to the care of children and infants >37 weeks gestational age.
Recommendation
• For children with COVID-19 or MIS-C and shock, the Panel recommends targeting a mean arterial
pressure (MAP) between the 5th and 50th percentiles for age or >50th percentile for age (AIII).
Rationale
No clinical trials support specific hemodynamic targets for children with septic shock due to COVID-19,
MIS-C, or any other etiology. The panel members for the pediatric Surviving Sepsis Campaign
guidelines were divided on the most appropriate MAP target and made no specific recommendation
for a target MAP.2 Therefore, for children with COVID-19 or MIS-C and shock, clinicians should use
the same approach used for children without COVID-19 and target a MAP between the 5th and 50th
percentiles for age or >50th percentile for age. When MAP cannot be reliably measured, systolic blood
pressure is a reasonable alternative.
Recommendation
• The Panel recommends that, when available, a combination of serial clinical assessments; cardiac
ultrasound or echocardiography; and laboratory markers, including lactate levels, should be used
to monitor the response to resuscitation in children with COVID-19 or MIS-C and shock (BIII).
Rationale
Observational data from children with sepsis not related to COVID-19 suggest that using clinical
assessment alone limits the ability to classify patients with sepsis as having “warm” (i.e., likely to
require fluid or vasopressors) or “cold” (i.e., likely to require inotropes) shock, when compared with
assessments that include objective measures of cardiac output/index or systemic vascular resistance.6,7
Cardiac ultrasonography can be performed at the bedside and serially, and it may provide additional
clinical data on volume responsiveness and cardiac function.8 Data from studies evaluating the use
of cardiac ultrasound in children with COVID-19 and MIS-C are limited to reports from case series.9
However, cardiac ultrasonography may have particular value when patients with MIS-C are being
monitored, as these patients can exhibit a wide range of hemodynamic perturbations, and approximately
Recommendation
• The Panel recommends administration of balanced crystalloids rather than 0.9% saline for the
initial resuscitation of children with shock due to COVID-19 or MIS-C (BIIa).
Rationale
In a randomized trial in India, 708 children with septic shock received either a balanced crystalloid
solution (e.g., Plasma-Lyte) or 0.9% saline.13 The incidence of the primary outcome of new or
progressive acute kidney injury was lower in the balanced crystalloid solution arm than in the saline arm
(21% vs. 33%; relative risk 0.62; 95% CI, 0.49–0.80; P < 0.001). There was no difference in mortality
between the arms. Another randomized trial comparing balanced fluids to 0.9% saline is ongoing
(ClinicalTrials.gov Identifier NCT04102371).
Two observational studies used administrative data to compare the use of balanced/buffered crystalloids
with 0.9% saline in propensity-matched cohorts of children with severe sepsis or septic shock not
related to COVID-19.14,15 One of the studies compared patients who received any or only Ringer’s
lactate solution in the first 3 days of admission with patients who received only normal saline.14 The
study demonstrated no differences between the arms for 30-day mortality or frequency of acute kidney
injury. The other study compared patients receiving only balanced fluids with those receiving only 0.9%
saline.15 The study demonstrated that the balanced fluid arm had lower mortality (12.5% vs. 15.9%; OR
0.76; 95% CI, 0.62–0.93; P = 0.007), reduced acute kidney injury (16.0% vs. 19.2%; OR 0.82; 95%
CI, 0.68–0.98; P = 0.028), and fewer days on vasoactive infusions (3.0 days vs. 3.3 days; P < 0.001)
than the saline arm. No published studies focused on patients with COVID-19 or MIS-C, although
hyponatremia is common in patients with MIS-C, and decisions about the type of fluid therapy used
should be individualized for this population.
Recommendations
• The Panel recommends the use of epinephrine or norepinephrine rather than dopamine in
children with COVID-19 or MIS-C and shock (BIIa).
• There is insufficient evidence to differentiate between norepinephrine and epinephrine as a
first-line vasoactive drug in children with COVID-19 or MIS-C. The choice of vasoactive agent
should be individualized and based on clinical examination, laboratory data, and data from cardiac
ultrasound or echocardiography.
Recommendation
• There is insufficient evidence for the Panel to recommend either for or against the use of
inodilators (including dobutamine or milrinone) in children with COVID-19 or MIS-C who show
evidence of cardiac dysfunction and persistent hypoperfusion despite adequate fluid loading and
the use of vasopressor agents.
Rationale
Data from studies evaluating the use of inodilators in children in COVID-19, MIS-C, and non-COVID-
19-related sepsis are limited to reports from case series. However, the majority of the pediatric Surviving
Sepsis Campaign guidelines panel (77%) would use an inodilator at least some of the time for patients
with sepsis not related to COVID-19, cardiac dysfunction, and persistent hypoperfusion despite adequate
fluid loading and the use of vasopressor agents.2 Expert consultation from specialists in pediatric
cardiology and critical care medicine is recommended in this scenario.
Additional Recommendations
• For the acute resuscitation of children with COVID-19 or MIS-C and shock, the Panel
recommends the use of crystalloids rather than albumin (AIIb).
References
1. Weiss SL, Peters MJ, Agus MSD, et al. Perspective of the Surviving Sepsis Campaign on the management
of pediatric sepsis in the era of coronavirus disease 2019. Pediatr Crit Care Med. 2020;21(11):e1031-e1037.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/32886460.
2. Weiss SL, Peters MJ, Alhazzani W, et al. Surviving Sepsis Campaign international guidelines for the
management of septic shock and sepsis-associated organ dysfunction in children. Pediatr Crit Care Med.
2020;21(2):e52-e106. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32032273.
3. Abrams JY, Oster ME, Godfred-Cato SE, et al. Factors linked to severe outcomes in multisystem
inflammatory syndrome in children (MIS-C) in the USA: a retrospective surveillance study. Lancet Child
Adolesc Health. 2021;5(5):323-331. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33711293.
4. Feldstein LR, Tenforde MW, Friedman KG, et al. Characteristics and outcomes of US children and adolescents
with multisystem inflammatory syndrome in children (MIS-C) compared with severe acute COVID-19.
JAMA. 2021;325(11):1074-1087. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33625505.
5. Godfred-Cato S, Bryant B, Leung J, et al. COVID-19-associated multisystem inflammatory syndrome
in children—United States, March–July 2020. MMWR Morb Mortal Wkly Rep. 2020;69(32):1074-1080.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/32790663.
6. Egan JR, Festa M, Cole AD, et al. Clinical assessment of cardiac performance in infants and children
following cardiac surgery. Intensive Care Med. 2005;31(4):568-573. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15711976.
7. Razavi A, Newth CJL, Khemani RG, Beltramo F, Ross PA. Cardiac output and systemic vascular resistance:
clinical assessment compared with a noninvasive objective measurement in children with shock. J Crit Care.
2017;39:6-10. Available at: https://www.ncbi.nlm.nih.gov/pubmed/28088009.
8. Ranjit S, Aram G, Kissoon N, et al. Multimodal monitoring for hemodynamic categorization and management
of pediatric septic shock: a pilot observational study. Pediatr Crit Care Med. 2014;15(1):e17-e26. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24196006.
9. Kennedy TM, Dessie A, Kessler DO, et al. Point-of-care ultrasound findings in multisystem inflammatory
syndrome in children: a cross-sectional study. Pediatr Emerg Care. 2021;37(6):334-339. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33871226.
10. Scott HF, Brou L, Deakyne SJ, et al. Association between early lactate levels and 30-day mortality in clinically
suspected sepsis in children. JAMA Pediatr. 2017;171(3):249-255. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28068437.
11. Bai Z, Zhu X, Li M, et al. Effectiveness of predicting in-hospital mortality in critically ill children by
assessing blood lactate levels at admission. BMC Pediatr. 2014;14:83. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24673817.
12. Scott HF, Brou L, Deakyne SJ, et al. Lactate clearance and normalization and prolonged organ dysfunction in
pediatric sepsis. J Pediatr. 2016;170:149-155.e1-4. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26711848.
13. Sankar J, Muralidharan J, Lalitha AV, et al. Multiple electrolytes solution versus saline as bolus fluid
COVID-19 Treatment Guidelines 158
The COVID-19 Treatment Guidelines Panel’s (the Panel) recommendations in this section were
informed by recommendations from the Surviving Sepsis Campaign guidelines for managing adult
sepsis, pediatric sepsis, and COVID-19, as well as by recommendations from the 2023 Pediatric Acute
Lung Injury Consensus Conference (2023 PALICC-2).
Goal of Oxygenation
Recommendations
• A target oxygen saturation measured by pulse oximetry (SpO2) of 92% to 97% is recommended
for most children with COVID-19 who require supplemental oxygen (AIIb).
• For children with severe pediatric acute respiratory distress syndrome (PARDS; i.e., with an
oxygenation index ≥16 or an oxygen saturation index ≥12), an SpO2 <92% can be considered to
minimize exposure to a high fraction of inspired oxygen (FiO2), but prolonged periods of an SpO2
<88% should be avoided (CIII).
Rationale
The optimal SpO2 in children with COVID-19 is unknown. However, there is no evidence that the
target SpO2 should differ from the 2023 PALICC-2 recommendation.1 An SpO2 of 92% to 97% is
recommended for most children with COVID-19 who require supplemental oxygen. The potential harm
of hyperoxia in children was demonstrated in a meta-analysis of 11 observational studies of children
without COVID-19.2 The study demonstrated that critically ill children with hyperoxia had greater odds
of mortality than those without hyperoxia (OR 1.59; 95% CI, 1.00–2.51). However, across the included
studies, there was significant heterogeneity for populations, definitions of hyperoxia, and the timing of
assessments for mortality outcomes. For children with severe PARDS, an SpO2 <92% can be considered
to minimize exposure to a high FiO2.1 Although no evidence clearly identifies a safe minimum SpO2
in children, prolonged exposure to an SpO2 <88% should be avoided. When a patient’s SpO2 is <92%,
monitoring oxygen delivery markers, including central venous SpO2, is suggested.3
The limitations of currently available measurement devices should be considered when using pulse
oximetry to manage children with COVID-19 or PARDS. Observational studies in children have
reported that pulse oximetry may be inaccurate, particularly at lower oxygen saturations (≤90%) and
for children who are Black.4,5 These reports are consistent with several observational studies in adults
that identified inaccuracies in pulse oximetry measurements, particularly for patients with darker skin
pigmentation.6-8 See Clinical Spectrum of SARS-CoV-2 Infection for more information.
Although procedures vary across institutions, the treatment of most patients with PARDS is managed
without the use of arterial lines or arterial blood gas testing because arterial line placement in children,
especially young children, can result in complications.9-11 Clinicians should monitor for adequate
delivery of oxygen or consider lowering the threshold for arterial line placement if a patient’s SpO2
measurements could be unreliable (e.g., for children who have darker skin or low SpO2 levels).
Monitoring methods could include observing the patient for altered mentation, measuring venous
oxygen saturation, or using near-infrared spectroscopy.
Rationale
No high-quality studies have evaluated the use of HFNC oxygen or NIV in children with COVID-19.
Therefore, when choosing a mode of respiratory support for children with COVID-19, the principles
of management used for patients without COVID-19 should be followed. Both the Surviving Sepsis
Campaign International Guidelines for the Management of Septic Shock and Sepsis-Associated
Organ Dysfunction in Children and the 2023 PALICC-2 recommend the use of NIV for children with
respiratory failure who have no indication for intubation.12,13
Furthermore, the response to NIV, particularly in children with more severe hypoxemia or high work
of breathing, should be gauged early (within the first several hours).13 If the patient does not show
improvement, intubation should be considered. To unload respiratory muscles, bilevel modes of NIV (with
inspiratory pressure augmentation, such as BiPAP), if tolerated, are preferred over the use of continuous
positive airway pressure (CPAP) alone, although CPAP is an alternative for children who cannot achieve an
adequate seal with the NIV interface or who have significant patient-ventilator asynchrony.
HFNC oxygen is a relatively new, but increasingly used, mode of respiratory support for infants and
children with acute respiratory failure.14 Data from studies evaluating the effectiveness of HFNC
oxygen relative to NIV or conventional oxygen are limited to studies of children with pneumonia in
limited-resource settings and to studies of children with bronchiolitis. Two randomized controlled trials
of children with pneumonia were conducted in limited-resource settings. One study demonstrated a
slightly lower relative risk of mortality with the use of HFNC oxygen when compared with conventional
oxygen therapy (aHR 0.79; 95% CI, 0.54–1.16), although the results were not statistically significant.15
The other trial randomized patients to receive bubble CPAP, low-flow oxygen, or HFNC oxygen.16
The children who received bubble CPAP demonstrated a lower risk of mortality than the children who
received low-flow oxygen (relative risk 0.25; 95% CI, 0.07–0.89; P = 0.02). The results also indicated
that for the composite outcome of treatment failure, there was no difference between the use of bubble
CPAP and HFNC oxygen (relative risk 0.50; 99.7% CI, 0.11–2.29).
A randomized, noninferiority trial compared HFNC oxygen (2 L/kg/min) and nasal CPAP among
142 infants aged <6 months with bronchiolitis not caused by COVID-19.17 The primary outcome was
treatment failure within 24 hours, defined as an increase of ≥1 point in the modified Wood’s Clinical
Asthma Score or Échelle Douleur Inconfort Nouveau-Né (EDIN) score (a neonatal pain and discomfort
scale), a respiratory rate >60 breaths/min and an increase of >10 breaths/min from baseline, or >2 severe
apnea episodes per hour. Treatment failure occurred more often in the HFNC oxygen arm than in the
nasal CPAP arm (51% vs. 31%), a result that failed to meet the prespecified noninferiority margin.
Notably, in the HFNC oxygen arm, 72% of the patients who had treatment failure were managed
successfully with nasal CPAP, and there were no differences between the arms for intubation rates or
length of stay in the pediatric intensive care unit (ICU).
Rationale
There are no high-quality pediatric data from studies that evaluated the effect of awake prone positioning
on clinical outcomes in children with COVID-19 or in children with illness not related to COVID-19.
Awake prone positioning may be considered for older children and adolescents. See Oxygenation and
Ventilation for Adults for more information on the use of awake prone positioning in adults. In addition,
pediatric clinicians should consider a child’s developmental stage and ability to comply with the
protocols for awake prone positioning.
General Considerations for Children With COVID-19 and PARDS Who Require
Mechanical Ventilation
Recommendations
For children with COVID-19 and PARDS who require mechanical ventilation:
• The Panel recommends using low tidal volume (VT) ventilation (VT 4–8 mL/kg of predicted body
weight) over higher VT ventilation (VT >8 mL/kg) (AIIb).
• The Panel recommends targeting plateau pressures of ≤28 cm H2O in children with normal chest
wall compliance and ≤32 cm H2O in those with impaired chest wall compliance (AIII).
• The Panel recommends using positive end-expiratory pressure (PEEP) at or above the level
recommended in the Acute Respiratory Distress Syndrome (ARDS) Network’s PEEP/FiO2 table
and titration of PEEP based on observed responses in oxygenation, hemodynamics, and respiratory
system compliance (AIIb).
• The Panel recommends permissive hypercapnia (i.e., 7.2–7.3 pH) to remain within lung-protective
strategies and to minimize ventilator-associated lung injury, provided the patient does not have a
coexisting condition that would be worsened by acidosis (e.g., severe pulmonary hypertension,
ventricular dysfunction, intracranial hypertension) (AIII).
• The Panel recommends limiting driving pressure as part of a lung-protective ventilation strategy
(BIIb).
• The Panel recommends against the routine use of inhaled nitric oxide (AIII).
Rationale
There is no evidence that ventilator management in patients with PARDS due to COVID-19
should differ from ventilator management in patients with PARDS due to other causes. The Panel’s
recommendations are derived from the 2023 PALICC-2 recommendations.1
A large observational study conducted in 71 international pediatric ICUs reported that for patients with
mild to moderate ARDS, less adherence to the recommended VT of 5 to 8 mL/kg (or 3 to 6 mL/kg for
patients with severe ARDS) was associated with higher mortality and with more time on ventilation.25
In general, supraphysiologic VT ventilation (>8 mL/kg) should not be used in patients with PARDS, and
VT should be adjusted within the acceptable range to maintain other lung-protective ventilation targets
(e.g., maintaining ≤28 cm H2O plateau pressure). The use of ultra-low VT ventilation (<4 mL/kg) has
not been systematically studied in children, so it should be used with caution.
The ARDS Network established a ventilation protocol that includes suggested low PEEP/high FiO2
values.26 Two observational studies reported better clinical outcomes associated with use of the
suggested (or higher) PEEP levels when compared with lower PEEP levels.25,27 The multicenter studies,
which included nearly 1,500 pediatric patients with ARDS, demonstrated that PEEP levels lower than
those suggested by the ARDS Network were associated with increased mortality.
Driving pressure (i.e., the difference between plateau pressure and PEEP) is a marker for lung strain.
COVID-19 Treatment Guidelines 163
Rationale
There is no evidence that fluid management in patients with PARDS due to COVID-19 should differ from
fluid management in patients with PARDS due to other causes. Therefore, the Panel’s recommendation
aligns with the 2023 PALICC-2 recommendations.1 No pediatric randomized trials have directly compared
a liberal fluid strategy to a conservative fluid strategy in patients with PARDS of any etiology. Several
observational studies have demonstrated an association between greater fluid overload and worse clinical
outcomes, including fewer ventilator-free days and increased mortality.31-33
In a multicenter study of 168 children with acute lung injury, daily and cumulative fluid balance
were measured over the first 7 days after participants met the inclusion criteria.31 After adjusting for
demographic characteristics, pediatric risk of mortality III (PRISM III) scores, vasopressor use, and the
ratio of arterial partial pressure of oxygen to fraction of inspired oxygen (PaO2/FiO2), the study found that
an increasing cumulative fluid balance on Day 3 was associated with fewer ventilator-free days, but no
association with mortality was detected.
A more recent single-center study that included 732 children with acute lung injury demonstrated
an association between higher cumulative fluid balance on Days 5 to 7 and increased mortality (OR
1.34 for 100 mL/kg on Day 5; 95% CI, 1.11–1.61) after adjusting for oxygenation index, the number
of nonpulmonary organ failures, immunocompromised status, and vasopressor scores.33 Also, greater
cumulative fluid balance on Days 4 to 7 was associated with a lower probability of successful extubation
by Day 28.
Collectively, the findings from these pediatric observational studies demonstrate the potential harm of
fluid overload in patients with PARDS, particularly after 3 to 4 days of illness. These results are consistent
with the findings from FACTT, a trial of conservative versus liberal fluid management strategies in
adults.34 In adults, FACTT found no difference between the arms for 60-day mortality, but the conservative
strategy arm demonstrated improved oxygenation and less time on mechanical ventilation and in the ICU
when compared with the liberal strategy arm. However, no analysis of data from prospective pediatric
COVID-19 Treatment Guidelines 164
Rationale
There is no evidence that the use of neuromuscular blockade in children with COVID-19 should differ
from practices used for severe PARDS from other causes. Therefore, the Panel’s recommendation aligns
directly with the 2023 PALICC-2 recommendation.1
Therapies for Mechanically Ventilated Children With Severe PARDS and Refractory
Hypoxemia
Recommendations
For children with severe PARDS and refractory hypoxemia after other oxygenation strategies have been
optimized:
• The Panel recommends the use of inhaled nitric oxide as a rescue therapy; if a patient’s
oxygenation does not improve rapidly, the inhaled nitric oxide should be discontinued (BIIb).
• The Panel recommends prone positioning for 12 to 16 hours per day over no prone positioning
(BIII).
• There is insufficient evidence for the Panel to recommend either for or against the use of
recruitment maneuvers, but if they are used in children, slow incremental and decremental
adjustments in PEEP are preferred over sustained inflation maneuvers.
• There is insufficient evidence for the Panel to recommend either for or against the use of high-
frequency oscillatory ventilation (HFOV) in patients with PARDS.
Rationale
There is no evidence that the use of inhaled nitric oxide, prone positioning, or HFOV in children with
COVID-19 should differ from practices used for severe PARDS from other causes. Therefore, the
Panel’s recommendations are based on the 2023 PALICC-2 recommendations.1
One randomized controlled trial and 2 propensity-matched, observational studies in the past 10 years
evaluated the use of inhaled nitric oxide in patients with PARDS.30,35,36 The randomized controlled trial
included 55 patients and found that the use of inhaled nitric oxide resulted in no statistical difference
between the arms for 28-day mortality (8% mortality in the inhaled nitric oxide arm vs. 28% in the
placebo arm), although the trial was underpowered for this outcome.30 However, the inhaled nitric oxide
arm had approximately 5 more ventilator-free days than the placebo arm, a result that was primarily
mediated by avoiding the use of ECMO.
Results from observational studies have shown that patients who received inhaled nitric oxide did
not improve or have fewer ventilator-free days.35,36 A meta-analysis of randomized controlled trials
COVID-19 Treatment Guidelines 165
References
1. Emeriaud G, López-Férnandez YM, Iyer NP, et al. Executive summary of the second international guidelines
for the diagnosis and management of pediatric acute respiratory distress syndrome (PALICC-2). Pediatr Crit
Care Med. 2023;24(2):143-168. Available at: https://www.ncbi.nlm.nih.gov/pubmed/36661420.
2. Lilien TA, Groeneveld NS, van Etten-Jamaludin F, et al. Association of arterial hyperoxia with outcomes
in critically ill children: a systematic review and meta-analysis. JAMA Netw Open. 2022;5(1):e2142105.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/34985516.
3. Fernández A, Modesto V, Rimensberger PC, et al. Invasive ventilatory support in patients with pediatric acute
respiratory distress syndrome: from the Second Pediatric Acute Lung Injury Consensus Conference. Pediatr
Crit Care Med. 2023;24(12 suppl 2):S61-S75. Available at: https://www.ncbi.nlm.nih.gov/pubmed/36661436.
4. Ross PA, Newth CJ, Khemani RG. Accuracy of pulse oximetry in children. Pediatrics. 2014;133(1):22-29.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/24344108.
5. Andrist E, Nuppnau M, Barbaro RP, Valley TS, Sjoding MW. Association of race with pulse oximetry
COVID-19 Treatment Guidelines 166
Recommendation
• The COVID-19 Treatment Guidelines Panel recommends that the use of extracorporeal membrane
oxygenation (ECMO) should be considered for children with acute COVID-19 or multisystem
inflammatory syndrome in children (MIS-C) who have refractory hypoxemia or shock when
hemodynamic parameters cannot be maintained or lung-protective strategies result in inadequate
gas exchange (CIII). Candidacy for ECMO should be determined on a case-by-case basis by the
multidisciplinary team.
Rationale
ECMO is used as a rescue therapy for children with refractory hypoxemia or shock. Similar to outcomes
for adults, outcomes for children managed with venovenous ECMO are variable and influenced by the
etiology and duration of the respiratory failure and by underlying, comorbid medical conditions.1,2 In
addition, studies have shown that pediatric centers that treat fewer patients with ECMO have worse
outcomes than facilities that treat a high volume of patients with ECMO.3,4 Other than studies of
neonates, no randomized trials have evaluated the efficacy or benefit of ECMO for the treatment of
hypoxemic respiratory failure in children without COVID-19. In an observational study of 122 children
with severe pediatric acute respiratory distress syndrome (PARDS), children who received ECMO and
those supported without ECMO had similar 90-day mortality (25% vs. 30%).5
The 2023 Pediatric Acute Lung Injury Consensus Conference suggests that patients with severe PARDS
from potentially reversible causes and children who are candidates for lung transplantation be evaluated
for management with ECMO if lung-protective strategies result in inadequate ventilation (conditional
recommendation, very low quality of evidence).6 The Surviving Sepsis Campaign International
Guidelines for the Management of Septic Shock and Sepsis-Associated Organ Dysfunction in Children
issued a weak recommendation, based on very low-quality evidence, for the use of venovenous ECMO
in children with PARDS and refractory hypoxemia.7
Venoarterial ECMO has been used successfully for the treatment of refractory shock in children,
although no trials have evaluated this approach, and the potential benefits must be weighed against risks
of bleeding and thromboembolic events.8-10 The Surviving Sepsis Campaign guidelines for children
issued a weak recommendation, based on very low-quality evidence, for the use of venoarterial ECMO
in children with refractory shock who have not improved with any other treatments.7 However, a
standardized definition for refractory shock in children is not available.
Studies that have evaluated data on the use of ECMO in children with COVID-19 and MIS-C have
suggested that these patients have outcomes similar to patients who have received ECMO for illnesses
not related to COVID-19.11-15 The Extracorporeal Life Support Organization published guidelines for
the use of ECMO in patients with COVID-19.16 In general, children with COVID-19 or MIS-C who are
candidates for ECMO should be assessed using criteria similar to those used for children with severe
respiratory failure or shock due to other causes. Cannulation approaches and management principles
should follow published international guidelines and local protocols for patients who do not have
COVID-19. Pediatric clinicians should consider entering patients into clinical trials or registries to
COVID-19 Treatment Guidelines 170
References
1. Zabrocki LA, Brogan TV, Statler KD, et al. Extracorporeal membrane oxygenation for pediatric respiratory
failure: survival and predictors of mortality. Crit Care Med. 2011;39(2):364-370. Available at: https://www.
ncbi.nlm.nih.gov/pubmed/20959787.
2. Gow KW, Heiss KF, Wulkan ML, et al. Extracorporeal life support for support of children with malignancy
and respiratory or cardiac failure: the extracorporeal life support experience. Crit Care Med. 2009;37(4):1308-
1316. Available at: https://www.ncbi.nlm.nih.gov/pubmed/19242331.
3. Freeman CL, Bennett TD, Casper TC, et al. Pediatric and neonatal extracorporeal membrane oxygenation:
does center volume impact mortality? Crit Care Med. 2014;42(3):512-519. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24164955.
4. Gonzalez DO, Sebastião YV, Cooper JN, Minneci PC, Deans KJ. Pediatric extracorporeal membrane
oxygenation mortality is related to extracorporeal membrane oxygenation volume in US hospitals. J Surg Res.
2019;236:159-165. Available at: https://www.ncbi.nlm.nih.gov/pubmed/30694751.
5. Barbaro RP, Xu Y, Borasino S, et al. Does extracorporeal membrane oxygenation improve survival in pediatric
acute respiratory failure? Am J Respir Crit Care Med. 2018;197(9):1177-1186. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29373797.
6. Rambaud J, Barbaro RP, Macrae DJ, et al. Extracorporeal membrane oxygenation in pediatric acute respiratory
distress syndrome: from the Second Pediatric Acute Lung Injury Consensus Conference. Pediatr Crit Care
Med. 2023;24(12 suppl 2):S124-S134. Available at: https://www.ncbi.nlm.nih.gov/pubmed/36661441.
7. Weiss SL, Peters MJ, Alhazzani W, et al. Surviving Sepsis Campaign international guidelines for the
management of septic shock and sepsis-associated organ dysfunction in children. Pediatr Crit Care Med.
2020;21(2):e52-e106. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32032273.
8. Schlapbach LJ, Chiletti R, Straney L, et al. Defining benefit threshold for extracorporeal membrane
oxygenation in children with sepsis—a binational multicenter cohort study. Crit Care. 2019;23(1):429.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/31888705.
9. Ramanathan K, Yeo N, Alexander P, et al. Role of extracorporeal membrane oxygenation in children with
sepsis: a systematic review and meta-analysis. Crit Care. 2020;24(1):684. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33287861.
10. Oberender F, Ganeshalingham A, Fortenberry JD, et al. Venoarterial extracorporeal membrane oxygenation
versus conventional therapy in severe pediatric septic shock. Pediatr Crit Care Med. 2018;19(10):965-972.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/30048365.
11. Di Nardo M, Hoskote A, Thiruchelvam T, et al. Extracorporeal membrane oxygenation in children with
coronavirus disease 2019: preliminary report from the collaborative european chapter of the extracorporeal life
support organization prospective survey. ASAIO J. 2021;67(2):121-124. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33009172.
12. Alfoudri H, Shamsah M, Yousuf B, AlQuraini N. Extracorporeal membrane oxygenation and extracorporeal
cardiopulmonary resuscitation for a COVID-19 pediatric patient: a successful outcome. ASAIO J.
2021;67(3):250-253. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33627597.
13. Feldstein LR, Tenforde MW, Friedman KG, et al. Characteristics and outcomes of US children and adolescents
with multisystem inflammatory syndrome in children (MIS-C) compared with severe acute COVID-19.
JAMA. 2021;325(11):1074-1087. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33625505.
14. Watanabe A, Yasuhara J, Karube T, et al. Extracorporeal membrane oxygenation in children with COVID-19:
a systematic review and meta-analysis. Pediatr Crit Care Med. 2023;24(5):406-416. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/36516348.
15. Bembea MM, Loftis LL, Thiagarajan RR, et al. Extracorporeal membrane oxygenation characteristics and
Remdesivir and ritonavir-boosted nirmatrelvir (Paxlovid) are approved by the Food and Drug
Administration for the treatment of COVID-19.
Molnupiravir and high-titer COVID-19 convalescent plasma (CCP) are available only under Food and
Drug Administration Emergency Use Authorizations for the treatment of COVID-19.
Summary Recommendations
Recommendations for Treating Nonhospitalized Adults
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends the following anti-SARS-CoV-2 therapies as
preferred treatments for COVID-19. These drugs are listed in order of preference:
• Ritonavir-boosted nirmatrelvir (Paxlovid) (AIIa)
• Remdesivir (BIIa)
• The Panel recommends molnupiravir as an alternative therapy when neither of the preferred therapies are available,
feasible to use, or clinically appropriate (CIIa).
• For more information on using these agents in nonhospitalized adults, see Therapeutic Management of Nonhospitalized
Adults With COVID-19.
Recommendations for Treating Nonhospitalized Children
• For recommendations on using antiviral therapy in nonhospitalized children, see Therapeutic Management of
Nonhospitalized Children With COVID-19.
Recommendations for Treating Hospitalized Adults or Children
• See Therapeutic Management of Hospitalized Adults With COVID-19 and Therapeutic Management of Hospitalized
Children With COVID-19 for recommendations on using remdesivir with or without immunomodulators in certain
hospitalized patients.
Antiviral Treatments With Insufficient Evidence
• There is insufficient evidence for the Panel to recommend either for or against the use of high-titer CCP for the
treatment of COVID-19 in hospitalized or nonhospitalized patients who are immunocompromised.
• Some people who are immunocompromised have prolonged, symptomatic COVID-19 with evidence of ongoing
SARS-CoV-2 replication. For the Panel’s recommendations for managing these patients, see Special Considerations in
People Who Are Immunocompromised.
• There is insufficient evidence for the Panel to recommend either for or against the use of high-titer CCP for the
treatment of COVID-19 in nonhospitalized patients who are immunocompetent.
Antiviral Treatments That the Panel Recommends Against
• The Panel recommends against the use of the following drugs for the treatment of COVID-19, except in a clinical trial:
• Interferon alfa or beta in nonhospitalized patients (AIIa)
• Systemic interferon alfa in hospitalized patients (AIIa)
• Nitazoxanide (BIIa)
• The Panel recommends against the use of the following drugs for the treatment of COVID-19:
• Anti-SARS-CoV-2 monoclonal antibodies (AIII)
• Chloroquine or hydroxychloroquine and/or azithromycin in hospitalized patients (AI) and nonhospitalized patients
(AIIa)
• CCP in hospitalized patients who are immunocompetent (BIIa)
• Lopinavir/ritonavir and other HIV protease inhibitors in hospitalized patients (AI) and nonhospitalized patients (AIII)
• Interferon beta in hospitalized patients (AI)
Remdesivir is a nucleotide prodrug of an adenosine analog. It binds to the viral RNA-dependent RNA
polymerase and inhibits viral replication by terminating RNA transcription prematurely. Remdesivir has
demonstrated in vitro and in vivo activity against SARS-CoV-2.1
Intravenous remdesivir is approved by the Food and Drug Administration (FDA) for the treatment
of COVID-19 in adults and pediatric patients aged ≥28 days and weighing ≥3 kg. In nonhospitalized
patients with mild to moderate COVID-19 who are at high risk of progressing to severe disease,
remdesivir should be started within 7 days of symptom onset and administered for 3 days. Hospitalized
patients should receive remdesivir for 5 days or until hospital discharge, whichever comes first.2 The
FDA prescribing information for remdesivir indicates that if a patient does not clinically improve within
5 days, clinicians may extend the treatment course for up to 5 additional days (for a total duration of 10
days). See Table 4e for more information on administering remdesivir.
Remdesivir has been studied in several clinical trials for the treatment of COVID-19. The
recommendations from the COVID-19 Treatment Guidelines Panel (the Panel) are based on the results
of these studies. See Table 4a for more information.
Recommendations
• For the Panel’s recommendations and information on the clinical efficacy of using remdesivir
to treat high-risk, nonhospitalized patients with mild to moderate COVID-19, see Therapeutic
Management of Nonhospitalized Adults With COVID-19.
• For the Panel’s recommendations and information on the clinical efficacy of using remdesivir with
or without immunomodulators to treat certain hospitalized patients, see Therapeutic Management
of Hospitalized Adults With COVID-19.
Considerations in Children
See Therapeutic Management of Nonhospitalized Children With COVID-19 and Therapeutic
Management of Hospitalized Children With COVID-19 for the Panel’s guidance regarding the use of
remdesivir in children.
References
1. Wang M, Cao R, Zhang L, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel
coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30(3):269-271. Available at: https://www.ncbi.nlm.nih.gov/
pubmed/32020029.
2. Remdesivir (Veklury) [package insert]. Food and Drug Administration. 2023. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/214787s019lbl.pdf.
3. Devgun JM, Zhang R, Brent J, et al. Identification of bradycardia following remdesivir administration
through the US Food and Drug Administration American College of Medical Toxicology COVID-19 Toxic
Pharmacovigilance Project. JAMA Netw Open. 2023;6(2):e2255815. Available at: https://www.ncbi.nlm.nih.
gov/pubmed/36787141.
The clinical trials described in this table do not represent all the trials that the Panel reviewed while developing the recommendations for
RDV. The studies summarized below are those that have had the greatest impact on the Panel’s recommendations.
Key: AE = adverse event; ALT = alanine transaminase; AST = aspartate aminotransferase; AZM = azithromycin; CQ = chloroquine; CrCl = creatinine clearance; DM =
diabetes mellitus; ECMO = extracorporeal membrane oxygenation; eGFR = estimated glomerular filtration rate; HCQ = hydroxychloroquine; HFNC = high-flow nasal
cannula; HTN = hypertension; IV = intravenous; IL = interleukin; LOS = length of stay; LPV/RTV = lopinavir/ritonavir; MV = mechanical ventilation; NIV = noninvasive
ventilation; OS = ordinal scale; the Panel = the COVID-19 Treatment Guidelines Panel; RCT = randomized controlled trial; RDV = remdesivir; RT-PCR = reverse
transcription polymerase chain reaction; SAE = serious adverse event; SCr = serum creatinine; SOC = standard of care; SpO2 = oxygen saturation; ULN = upper limit of
normal; WHO = World Health Organization
Nirmatrelvir is an oral protease inhibitor that is active against MPRO, a viral protease that plays an
essential role in viral replication by cleaving the 2 viral polyproteins.1 It has demonstrated antiviral
activity against all coronaviruses that are known to infect humans.2 Nirmatrelvir is packaged with
ritonavir (as Paxlovid), a strong cytochrome P450 (CYP) 3A4 inhibitor and pharmacokinetic boosting
agent that has been used to boost HIV protease inhibitors. Coadministration of ritonavir is required to
increase nirmatrelvir concentrations to the target therapeutic range.
Ritonavir-boosted nirmatrelvir is approved by the Food and Drug Administration (FDA) for the
treatment of mild to moderate COVID-19 in adults who are at high risk of progressing to severe
COVID-19.3 The Emergency Use Authorization (EUA) for ritonavir-boosted nirmatrelvir will continue
to authorize the use of the EUA-labeled product for the treatment of nonhospitalized adolescents aged 12
to 17 years and weighing ≥40 kg who are at high risk of progressing to severe COVID-19.
Recommendations
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends using nirmatrelvir 300 mg
with ritonavir 100 mg (Paxlovid) orally (PO) twice daily for 5 days in nonhospitalized adults
with mild to moderate COVID-19 who are at high risk of disease progression (AIIa). Treatment
should be initiated as soon as possible and within 5 days of symptom onset. For information
on medical conditions that confer high risk, see the Centers for Disease Control and Prevention
webpage Underlying Medical Conditions Associated With Higher Risk for Severe COVID-19.
• Ritonavir-boosted nirmatrelvir is available through an FDA EUA for the treatment of mild to
moderate COVID-19 in nonhospitalized adolescents aged 12 to 17 years and weighing ≥40 kg.4
For recommendations on using ritonavir-boosted nirmatrelvir in nonhospitalized children with
COVID-19, see Therapeutic Management of Nonhospitalized Children With COVID-19.
• See the section titled Considerations in Patients With Severe Renal Insufficiency below for
information on managing patients with COVID-19 and severe renal insufficiency.
• For a discussion of the treatment of prolonged, symptomatic COVID-19 in patients with evidence
of ongoing SARS-CoV-2 replication, see the section titled Patients Who Are Immunocompromised
and Have Prolonged COVID-19 Symptoms and Evidence of Ongoing Viral Replication below.
Rationale
The EPIC-HR trial enrolled nonhospitalized adults with mild to moderate COVID-19 who were not
vaccinated and who were at high risk of progressing to severe disease.5 The trial demonstrated that
starting ritonavir-boosted nirmatrelvir within 5 days of symptom onset in these patients reduced the risk
of hospitalization or death through Day 28 by 89% compared to placebo. This efficacy is comparable
to remdesivir (87% relative reduction)6 and greater than the efficacy reported for molnupiravir (31%
relative reduction).7 However, these agents have not been directly compared in clinical trials.
Although ritonavir-boosted nirmatrelvir demonstrated a clinical benefit during the EPIC-HR trial, the
benefits in unvaccinated people who are at low risk of progression to severe disease or in vaccinated
people who are at high risk of progression to severe disease are unclear. The EPIC-SR trial, which
included both of these populations, found that ritonavir-boosted nirmatrelvir did not reduce the duration
of symptoms and did not have a statistically significant effect on the risk of hospitalization or death
Drug-Drug Interactions
The FDA prescribing information for ritonavir-boosted nirmatrelvir includes a boxed warning about
significant drug-drug interactions between ritonavir-boosted nirmatrelvir and other medications. These
interactions are primarily caused by the ritonavir component of the combination. Ritonavir, a strong
CYP3A4 inhibitor and a P-glycoprotein inhibitor, may increase the blood concentration of certain
concomitant medications and increase the potential for serious drug toxicities. Before prescribing
ritonavir-boosted nirmatrelvir, clinicians should carefully review the patient’s concomitant medications,
including over-the-counter medications, herbal supplements, and recreational drugs, to evaluate
potential drug-drug interactions. Clinicians should consider both the potential benefits of treatment with
ritonavir-boosted nirmatrelvir and the potential risks related to drug-drug interactions. Many drug-drug
interactions between ritonavir-boosted nirmatrelvir and concomitant medications (e.g., certain statins,
calcium channel blockers, or direct oral anticoagulants) can be safely managed. For the Panel’s
recommendations on preferred and alternative antiviral therapies for outpatients with COVID-19, see
Therapeutic Management of Nonhospitalized Adults With COVID-19. Clinicians should be aware that
the drug-drug interaction potential of ritonavir-boosted nirmatrelvir may change if it is used for extended
durations (i.e., ≥10 days).
The following resources provide information on identifying and managing drug-drug interactions.
• Quick reference lists:
• Drug-Drug Interactions Between Ritonavir-Boosted Nirmatrelvir (Paxlovid) and Concomitant
Medications. Box 1 lists select outpatient medications that are not expected to have clinically
relevant interactions with ritonavir-boosted nirmatrelvir. Box 2 lists select outpatient
medications that have clinically relevant drug-drug interactions with ritonavir-boosted
nirmatrelvir.
• Web-based drug-drug interaction checker:
• The Liverpool COVID-19 Drug Interactions website
• Tables with guidance on managing specific drug-drug interactions:
• The University of Waterloo/University of Toronto drug interaction guide for ritonavir-boosted
nirmatrelvir
• The FDA prescribing information for ritonavir-boosted nirmatrelvir
SARS-CoV-2 Resistance
Viral mutations that lead to substantial resistance to nirmatrelvir have been selected for in in vitro
studies; the fitness of these mutations is unclear. Surveillance for the emergence of significant resistance
to nirmatrelvir is critical, particularly in patients who are severely immunocompromised and who
experience prolonged replication of SARS-CoV-2.
Additional Considerations
• Nirmatrelvir must be administered with ritonavir to achieve sufficient therapeutic plasma
concentrations.
• In patients with moderate renal impairment (i.e., those with an estimated glomerular filtration rate
[eGFR] of ≥30 to <60 mL/min), the dose should be reduced to nirmatrelvir 150 mg with ritonavir
100 mg PO twice daily.
• Ritonavir-boosted nirmatrelvir is not recommended for patients with known or suspected severe
hepatic impairment (i.e., Child-Pugh Class C), and it should be used with caution in patients with
pre-existing liver diseases, liver enzyme abnormalities, or hepatitis. No pharmacokinetic or safety
data are available for this patient population.
• Patients should complete the 5-day treatment course of ritonavir-boosted nirmatrelvir because
COVID-19 Treatment Guidelines 188
Considerations in Children
Ritonavir-boosted nirmatrelvir is available through an FDA EUA for the treatment of mild to
moderate COVID-19 in nonhospitalized adolescents aged 12 to 17 years and weighing ≥40 kg. For
recommendations on using ritonavir-boosted nirmatrelvir in nonhospitalized children with COVID-19,
see Therapeutic Management of Nonhospitalized Children With COVID-19.
Clinical Data
The EPIC-HR study was a multinational randomized trial that compared the use of ritonavir-boosted
nirmatrelvir PO twice daily for 5 days to placebo in nonhospitalized patients aged ≥18 years with mild to
moderate COVID-19 who were at high risk of clinical progression.5 Eligible patients were randomized
within 5 days of symptom onset, were not vaccinated against COVID-19, and had at least 1 risk factor
for progression to severe disease. Patients were excluded if they used medications that were either
COVID-19 Treatment Guidelines 189
References
1. Pillaiyar T, Manickam M, Namasivayam V, Hayashi Y, Jung SH. An overview of severe acute respiratory
syndrome-coronavirus (SARS-CoV) 3CL protease inhibitors: peptidomimetics and small molecule
chemotherapy. J Med Chem. 2016;59(14):6595-6628. Available at: https://www.ncbi.nlm.nih.gov/
pubmed/26878082.
2. Owen DR, Allerton CMN, Anderson AS, et al. An oral SARS-CoV-2 MPRO inhibitor clinical candidate for
the treatment of COVID-19. Science. 2021;374(6575):1586-1593. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34726479.
3. Ritonavir-boosed nirmatrelvir (Paxlovid) [package insert]. Food and Drug Administration. 2023. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/217188s000lbl.pdf.
4. Food and Drug Administration. Fact sheet for healthcare providers: Emergency Use Authorization for
Paxlovid. 2023. Available at: https://www.fda.gov/media/155050/download.
5. Hammond J, Leister-Tebbe H, Gardner A, et al. Oral nirmatrelvir for high-risk, nonhospitalized adults with
COVID-19. N Engl J Med. 2022;386(15):1397-1408. Available at:
https://pubmed.ncbi.nlm.nih.gov/35172054.
6. Gottlieb RL, Vaca CE, Paredes R, et al. Early remdesivir to prevent progression to severe COVID-19 in
outpatients. N Engl J Med. 2022;386(4):305-315. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34937145.
7. Jayk Bernal A, Gomes da Silva MM, Musungaie DB, et al. Molnupiravir for oral treatment of COVID-19 in
nonhospitalized patients. N Engl J Med. 2022;386(6):509-520. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34914868.
8. Pfizer. Pfizer reports additional data on PAXLOVID supporting upcoming new drug application submission to
U.S. FDA. 2022. Available at: https://www.pfizer.com/news/press-release/press-release-detail/pfizer-reports-
additional-data-paxlovidtm-supporting. Accessed January 26, 2024.
9. Dryden-Peterson S, Kim A, Kim AY, et al. Nirmatrelvir plus ritonavir for early COVID-19 in a large U.S.
health system: a population-based cohort study. Ann Intern Med. 2023;176(1):77-84. Available at: https://
www.ncbi.nlm.nih.gov/pubmed/36508742.
COVID-19 Treatment Guidelines 190
References
1. Katzenmaier S, Markert C, Riedel KD, et al. Determining the time course of CYP3A inhibition by potent
reversible and irreversible CYP3A inhibitors using a limited sampling strategy. Clin Pharmacol Ther.
2011;90(5):666-673. Available at: https://www.ncbi.nlm.nih.gov/pubmed/21937987.
2. Stader F, Khoo S, Stoeckle M, et al. Stopping lopinavir/ritonavir in COVID-19 patients: duration of the drug
interacting effect. J Antimicrob Chemother. 2020;75(10):3084-3086. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32556272.
3. Food and Drug Administration Center for Drug Evaluation and Research. Antimicrobial drugs advisory
committee meeting. 2023. Available at: https://www.fda.gov/media/168508/download.
4. Food and Drug Administration. FDA requiring Boxed Warning updated to improve safe use of benzodiazepine
drug class. 2020. Available at: https://www.fda.gov/media/142368/download.
5. Li M, Zhu L, Chen L, Li N, Qi F. Assessment of drug-drug interactions between voriconazole and
glucocorticoids. J Chemother. 2018;30(5):296-303. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30843777.
6. Foisy MM, Yakiwchuk EM, Hughes CA. Induction effects on ritonavir: implications for drug interactions. Ann
Pharmacother. 2008;42(7):1048-1059. Available at: https://pubmed.ncbi.nlm.nih.gov/18577765.
7. University of Liverpool. Evaluating the interaction risk of COVID-19 therapies. 2022. Available at:
https://covid19-druginteractions.org/prescribing_resources. Accessed February 22, 2024.
8. Ramsden D, Fung C, Hariparsad N, et al. Perspectives from the innovation and quality consortium induction
working group on factors impacting clinical drug-drug interactions resulting from induction: focus on
cytochrome 3A substrates. Drug Metab Dispos. 2019;47(10):1206-1221. Available at:
https://pubmed.ncbi.nlm.nih.gov/31439574.
9. Marzolini C, Kuritzkes DR, Marra F, et al. Recommendations for the management of drug-drug interactions
between the COVID-19 antiviral nirmatrelvir/ritonavir (Paxlovid) and comedications. Clin Pharmacol Ther.
2022;112(6):1191-1200. Available at: https://pubmed.ncbi.nlm.nih.gov/35567754.
Recommendations
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends using molnupiravir 800
mg orally (PO) twice daily for 5 days as an alternative therapy in nonhospitalized patients aged
≥18 years with mild to moderate COVID-19 who are at high risk of disease progression when
ritonavir-boosted nirmatrelvir (Paxlovid) and remdesivir are not available, feasible to use, or
clinically appropriate; treatment should be initiated as soon as possible and within 5 days of
symptom onset (CIIa).
• The Panel recommends against the use of molnupiravir for the treatment of COVID-19 in
pregnant patients unless there are no other options and therapy is clearly indicated (AIII). For
more details, see Considerations in Pregnant and Lactating People below.
• People who engage in sexual activity that may result in conception should use effective
contraception during and following treatment with molnupiravir. For more details, see
Considerations in Sexually Active Individuals below.
Molnupiravir may be used in patients who are hospitalized for a diagnosis other than COVID-19,
provided they have mild to moderate COVID-19 and are at high risk of progressing to severe disease.
For the Panel’s recommendations on preferred and alternative antiviral therapies for outpatients with
COVID-19, see Therapeutic Management of Nonhospitalized Adults With COVID-19.
Rationale
The MOVe-OUT trial enrolled high-risk, unvaccinated, nonhospitalized adults and reported that
molnupiravir reduced the rate of hospitalization or death among these patients by 31% compared to
placebo.7 A secondary analysis of patients who required hospitalization during the trial found a reduced
need for respiratory interventions among those who received molnupiravir compared to those who
received placebo.8 MOVe-OUT was conducted before the emergence of the Omicron subvariants.
The PANORAMIC trial enrolled nonhospitalized adults with COVID-19 who were at high risk of severe
disease during a period when the Omicron variant was circulating.9 Ninety-four percent of the patients
had received at least 3 doses of a COVID-19 vaccine. The study found that the use of molnupiravir plus
usual care did not reduce the occurrence of the primary composite outcome of hospitalization or death
Additional Considerations
• Patients should complete the 5-day treatment course of molnupiravir. It is unknown whether
a shorter course is less effective or associated with the emergence of molnupiravir-resistant
mutations.
• If a patient requires hospitalization after starting treatment, the full treatment course of
molnupiravir can be completed at the health care provider’s discretion.
• The FDA EUA for molnupiravir provides instructions for preparing and administering capsule
contents through orogastric or nasogastric tubes.6
• There are no data on using combinations of antiviral therapies to treat nonhospitalized patients
with COVID-19. Clinical trials are needed to determine whether combination therapy has a role in
the treatment of SARS-CoV-2 infection.
• Patients who are severely immunocompromised can experience prolonged periods of
SARS-CoV-2 replication, which may lead to rapid viral evolution. There are theoretical concerns
that using a single antiviral agent in these patients may produce antiviral-resistant viruses.
Additional studies are needed to assess this risk. The role of combination antiviral therapy
in treating patients who are severely immunocompromised is not yet known. See Special
Considerations in People Who Are Immunocompromised for more information.
• There are limited data on the frequency of SARS-CoV-2 rebound in patients who have completed
treatment with molnupiravir.13 During the MOVe-OUT trial, rates of symptomatic SARS-CoV-2
rebound were low (approximately 1%) in both those who received molnupiravir and those who
received placebo.6
• SARS-CoV-2 sequences with molnupiravir-induced mutations have been detected in many
countries, but the clinical implications of these mutations remain unclear.14
Considerations in Children
The MOVe-OUT and PANORAMIC trials excluded participants aged <18 years. There are no data
available on the use of molnupiravir in children aged <18 years. Molnupiravir is not authorized for use
in those aged <18 years due to potential effects on bone and cartilage growth.
Clinical Data
MOVe-OUT
MOVe-OUT was a Phase 3 trial that evaluated the use of molnupiravir in unvaccinated, nonhospitalized
adults with mild to moderate COVID-19 who were at high risk of progressing to severe COVID-19 and
who were enrolled within 5 days of symptom onset.7 Patients were randomized to receive molnupiravir
800 mg PO every 12 hours for 5 days or placebo. The primary composite endpoint was a hospital stay
>24 hours or death by Day 29.
Results
• The final analysis included 1,433 patients.
• The median age was 43 years (with 17% aged >60 years); 49% of patients were men, 57%
were White, 50% were Hispanic/Latinx, and 5% were Black or African American.
• 4% had a body mass index ≥30, and 16% had diabetes.
• The time from the onset of COVID-19 symptoms to randomization was ≤3 days in 48% of
patients.
• By Day 29, 48 of 709 patients (6.8%) in the molnupiravir arm and 68 of 699 (9.7%) in the placebo
arm experienced hospitalization or death (adjusted difference -3.0%; 95% CI, -5.9% to -0.1%).
• One death occurred in the molnupiravir arm, and 9 deaths occurred in the placebo arm.
COVID-19 Treatment Guidelines 202
PANORAMIC
PANORAMIC was a multicenter, open-label, adaptive platform trial conducted in the United Kingdom
that evaluated the use of molnupiravir in nonhospitalized adults who were at high risk of progressing to
severe COVID-19.9 The participants were aged ≥50 years or ≥18 years with comorbid conditions and
were enrolled within 5 days of symptom onset. Patients were randomized to receive molnupiravir 800
mg PO twice daily for 5 days plus usual care or usual care alone. The primary endpoint was a composite
of all-cause hospitalization (defined as ≥1 overnight hospital stay, ≥1 night at home with care and
monitoring by hospital clinicians, or an overnight stay in an emergency room) or death within 28 days.
The trial was conducted when the Omicron variant was the dominant variant.
Results
• The final analysis included 25,708 patients.
• The mean age was 56.6 years (26.5% aged ≥65 years); 94% were White; 59% were women.
• 94% received ≥3 doses of a COVID-19 vaccine.
• 69% of patients had comorbidities: 25% with lung disease; 15% with obesity; 12% with
diabetes; 8% with heart disease; 8.5% were immunocompromised.
• 24% were taking inhaled corticosteroids.
• The mean time from symptom onset to starting molnupiravir was 3 days (range 3–5 days). Among
the patients who provided information, 95% reported completing the 5-day treatment course.
• The study reported 103 hospitalizations and 3 deaths in the molnupiravir arm and 96
hospitalizations and 5 deaths in the usual care alone arm (aOR 1.06; 95% CrI, 0.81–1.41;
probability of superiority 0.33).
• The time to self-reported first recovery among those who received molnupiravir (median of 9
days) was shorter than among those who received usual care alone (median of 15 days).
• No serious adverse events were related to molnupiravir; 145 patients (1.1%) withdrew because of
adverse events attributed to molnupiravir.
Limitations
• Because the PANORAMIC trial was an open-label study and the patients knew whether they were
receiving molnupiravir or not, this may have affected their reported symptoms. As a result, these
findings are less reliable than those from a placebo-controlled trial.
References
1. Fischer WA II, Eron JJ Jr, Holman W, et al. A Phase 2a clinical trial of molnupiravir in patients with
COVID-19 shows accelerated SARS-CoV-2 RNA clearance and elimination of infectious virus. Sci Transl
Med. 2022;14(628):eabl7430. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34941423.
2. Zou R, Peng L, Shu D, et al. Antiviral efficacy and safety of molnupiravir against Omicron variant infection:
COVID-19 Treatment Guidelines 203
Monoclonal antibodies (mAbs) that target the SARS-CoV-2 spike protein have been shown to have
clinical benefits in treating SARS-CoV-2 infection. However, laboratory studies have found that the
activity of anti-SARS-CoV-2 mAbs against specific variants and subvariants can vary dramatically. The
anti-SARS-CoV-2 mAb products that have received Emergency Use Authorizations (EUAs) from the
Food and Drug Administration (FDA) are not expected to be effective against the currently circulating
SARS-CoV-2 variants and subvariants. As a result, these products are not currently recommended by the
COVID-19 Treatment Guidelines Panel (the Panel) for the treatment or prevention of COVID-19.
See Table 4b for information on the clinical trials that have evaluated the safety and efficacy of using
anti-SARS-CoV-2 mAbs in patients with COVID-19.
Recommendation
• The Panel recommends against the use of anti-SARS-CoV-2 mAbs for the treatment or
prevention of COVID-19 (AIII).
References
1. Food and Drug Administration. Fact sheet for health care providers: Emergency Use Authorization (EUA) of
bamlanivimab and etesevimab. 2022. Available at: https://www.fda.gov/media/145802/download.
2. Food and Drug Administration. Fact sheet for health care providers: Emergency Use Authorization (EUA) of
REGEN-COV (casirivimab and imdevimab). 2021. Available at:
https://www.fda.gov/media/145611/download.
3. Food and Drug Administration. Fact sheet for healthcare providers: Emergency Use Authorization (EUA) of
sotrovimab. 2023. Available at: https://www.fda.gov/media/149534/download.
4. Food and Drug Administration. Fact sheet for healthcare providers: Emergency Use Authorization for
bebtelovimab. 2022. Available at: https://www.fda.gov/media/156152/download.
Key: BAM = bamlanivimab; bpm = beats per minute; BEB = bebtelovimab; CAS = casirivimab; DM = diabetes mellitus; ETE = etesevimab; IMD = imdevimab; IV =
intravenous; mAb = monoclonal antibody; PHVL = persistently high viral load; RCT = randomized controlled trial; RT-PCR = reverse transcription polymerase chain
reaction; SOT = sotrovimab; SpO2 = oxygen saturation; VL = viral load
References
1. Dougan M, Azizad M, Mocherla B, et al. A randomized, placebo-controlled clinical trial of bamlanivimab and etesevimab together in high-risk
ambulatory patients with COVID-19 and validation of the prognostic value of persistently high viral load. Clin Infect Dis. 2022;75(1):e440-e449.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/34718468.
2. Food and Drug Administration. Fact sheet for healthcare providers: Emergency Use Authorization for bebtelovimab. 2022. Available at:
https://www.fda.gov/media/156152/download.
3. Weinreich DM, Sivapalasingam S, Norton T, et al. REGEN-COV antibody combination and outcomes in outpatients with COVID-19. N Engl J Med.
2021;385(23):e81. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34587383.
4. Gupta A, Gonzalez-Rojas Y, Juarez E, et al. Effect of sotrovimab on hospitalization or death among high-risk patients with mild to moderate
COVID-19: a randomized clinical trial. JAMA. 2022;327(13):1236-1246. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35285853.
Plasma from donors who have recovered from COVID-19 (regardless of vaccination status) may contain
antibodies to SARS-CoV-2 that could help suppress viral replication.1 In August 2020, the Food and
Drug Administration issued an Emergency Use Authorization (EUA) for COVID-19 convalescent
plasma (CCP) for the treatment of hospitalized patients with COVID-19. The EUA was subsequently
revised. The current EUA limits the authorization to the use of CCP products that contain high levels of
anti-SARS-CoV-2 antibodies (i.e., high-titer products) for the treatment of COVID-19 in outpatients or
inpatients who have immunosuppressive disease or who are receiving immunosuppressive treatment.
The testing criteria used to identify high-titer CCP products was also revised.2
The use of CCP should be limited to high-titer products. Products that are not labeled “high titer” should
not be used.
Recommendations
Patients Who Are Immunocompromised
• There is insufficient evidence for the COVID-19 Treatment Guidelines Panel (the Panel) to
recommend either for or against the use of high-titer CCP for the treatment of COVID-19 in
hospitalized or nonhospitalized patients who are immunocompromised.
• Some people who are immunocompromised have prolonged, symptomatic COVID-19 with
evidence of ongoing SARS-CoV-2 replication. The data for these approaches are not definitive,
but some Panel members would use 1 or more of the following treatment options:
• Longer and/or additional courses of ritonavir-boosted nirmatrelvir (Paxlovid)
• Longer and/or additional courses of remdesivir
• High-titer CCP from a vaccinated donor who recently recovered from COVID-19 likely caused
by a SARS-CoV-2 variant similar to the variant causing the patient’s illness
Rationale
Patients Who Are Immunocompromised
This section pertains to people who are moderately or severely immunocompromised. For examples of
moderately or severely immunocompromising conditions and for a broader discussion on the therapeutic
management of COVID-19 in people who are immunocompromised, see Special Considerations in
People Who Are Immunocompromised.
Patients who are immunocompromised are at risk of having reduced antibody responses to SARS-CoV-2
infection and COVID-19 vaccination, having suboptimal control of viral replication, and progressing to
Considerations in Children
The safety and efficacy of CCP have not been systematically evaluated in pediatric patients. The
published literature on its use in children is limited to case reports and case series. A few clinical trials
evaluating the use of CCP in children are ongoing. The use of high-titer CCP may be considered on a
case-by-case basis for hospitalized children who are immunocompromised and meet the EUA criteria
for its use. CCP is not authorized by the Food and Drug Administration for use in patients who are
immunocompetent.
Several other antiviral therapies are available for the treatment of children with COVID-19 who are at
high risk of progressing to severe disease. See Therapeutic Management of Nonhospitalized Children
With COVID-19 and Therapeutic Management of Hospitalized Children With COVID-19 for more
information.
References
1. Wang X, Guo X, Xin Q, et al. Neutralizing antibody responses to severe acute respiratory syndrome
coronavirus 2 in coronavirus disease 2019 inpatients and convalescent patients. Clin Infect Dis.
2020;71(10):2688-2694. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32497196.
2. Food and Drug Administration. Fact sheet for health care providers: Emergency Use Authorization (EUA) of
COVID-19 convalescent plasma for treatment of coronavirus disease 2019 (COVID-19). 2021. Available at:
https://www.fda.gov/media/141478/download.
3. Cattaneo C, Masina L, Pagani C, et al. High mortality in fully vaccinated hematologic patients treated with
anti-CD20 antibodies during the “Omicron wave” of COVID-19 pandemic. Hematol Oncol. 2023;41(1):205-
207. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35933702.
4. Shahzad M, Chaudhary SG, Zafar MU, et al. Impact of COVID-19 in hematopoietic stem cell transplant
recipients: a systematic review and meta-analysis. Transpl Infect Dis. 2022;24(2):e13792. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35030267.
5. Denkinger CM, Janssen M, Schäkel U, et al. Anti-SARS-CoV-2 antibody-containing plasma improves
outcome in patients with hematologic or solid cancer and severe COVID-19: a randomized clinical trial. Nat
Cancer. 2023;4(1):96-107. Available at: https://pubmed.ncbi.nlm.nih.gov/36581734.
6. Writing Committee for the REMAP-CAP Investigators. Effect of convalescent plasma on organ support-free
days in critically ill patients with COVID-19: a randomized clinical trial. JAMA. 2021;326(17):1690-1702.
The studies described in this table are those that had the greatest impact on the Panel’s recommendations. The Panel reviewed other clinical
studies of CCP for the treatment of COVID-19.1-5 However, those studies have limitations that make them less definitive and informative than
the studies summarized in the table.
References
1. Agarwal A, Mukherjee A, Kumar G, et al. Convalescent plasma in the management of moderate COVID-19 in adults in India: open label Phase II
multicentre randomised controlled trial (PLACID Trial). BMJ. 2020;371:m3939. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33093056.
2. Simonovich VA, Burgos Pratx LD, Scibona P, et al. A randomized trial of convalescent plasma in COVID-19 severe pneumonia. N Engl J Med.
2021;384(7):619-629. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33232588.
3. O'Donnell MR, Grinsztejn B, Cummings MJ, et al. A randomized double-blind controlled trial of convalescent plasma in adults with severe
COVID-19. J Clin Invest. 2021;131(13):e150646. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33974559.
4. Li L, Zhang W, Hu Y, et al. Effect of convalescent plasma therapy on time to clinical improvement in patients with severe and life-threatening
COVID-19: a randomized clinical trial. JAMA. 2020;324(5):460-470. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32492084.
5. Diago-Sempere E, Bueno JL, Sancho-López A, et al. Evaluation of convalescent plasma versus standard of care for the treatment of COVID-19 in
hospitalized patients: study protocol for a phase 2 randomized, open-label, controlled, multicenter trial. Trials. 2021;22(1):70. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33472681.
6. Writing Committee for the REMAP-CAP Investigators. Effect of convalescent plasma on organ support-free days in critically ill patients with
COVID-19: a randomized clinical trial. JAMA. 2021;326(17):1690-1702. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34606578.
7. Bégin P, Callum J, Jamula E, et al. Convalescent plasma for hospitalized patients with COVID-19: an open-label, randomized controlled trial. Nat Med.
2021;27(11):2012-2024. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34504336.
8. RECOVERY Collaborative Group. Convalescent plasma in patients admitted to hospital with COVID-19 (RECOVERY): a randomised controlled,
open-label, platform trial. Lancet. 2021;397(10289):2049-2059. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34000257.
9. Denkinger CM, Janssen M, Schäkel U, et al. Anti-SARS-CoV-2 antibody-containing plasma improves outcome in patients with hematologic or solid
cancer and severe COVID-19: a randomized clinical trial. Nat Cancer. 2023;4(1):96-107. Available at: https://pubmed.ncbi.nlm.nih.gov/36581734.
10. Misset B, Piagnerelli M, Hoste E, et al. Convalescent plasma for COVID-19-induced ARDS in mechanically ventilated patients. N Engl J Med.
2023;389(17):1590-1600. Available at: https://www.ncbi.nlm.nih.gov/pubmed/37889107.
11. Sullivan DJ, Gebo KA, Shoham S, et al. Early outpatient treatment for COVID-19 with convalescent plasma. N Engl J Med. 2022;386(18):1700-1711.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/35353960.
12. Alemany A, Millat-Martinez P, Corbacho-Monné M, et al. High-titre methylene blue-treated convalescent plasma as an early treatment for outpatients
COVID-19 Treatment Guidelines 226
Interferons are a family of cytokines with in vitro and in vivo antiviral properties. Interferon beta-1a
has been approved by the Food and Drug Administration (FDA) to treat relapsing forms of multiple
sclerosis, and pegylated formulations of interferon alfa-2a and interferon alfa-2b have been approved
by the FDA to treat hepatitis B and hepatitis C virus infections. Several interferons, including interferon
alfa, beta, and lambda, have been evaluated for the treatment of COVID-19. Interferon lambda is not
currently approved or authorized by the FDA for any use.
Recommendations
• For nonhospitalized patients with mild to moderate COVID-19, the COVID-19 Treatment
Guidelines Panel (the Panel) recommends against the use of interferon alfa or beta, except in a
clinical trial (AIIa).
• For hospitalized patients with COVID-19, the Panel recommends against the use of systemic
interferon alfa, except in a clinical trial (AIIa).
• For hospitalized patients with COVID-19, the Panel recommends against the use of systemic
interferon beta (AI).
• The Panel is unable to recommend either for or against the use of interferon lambda because this
product is not currently available for clinical use.
Rationale
Interferon Alfa and Beta
Many of the studies that evaluated the use of systemic interferons for the treatment of hospitalized
adults with COVID-19 were conducted in early 2020, before the widespread use of remdesivir or
corticosteroids and other immunomodulators. In addition, these studies administered interferons with
other drugs that have since been shown to have no clinical benefit in people with COVID-19, such as
lopinavir/ritonavir and hydroxychloroquine.1-3
More recent studies have shown no benefit of using interferon beta-1a to treat patients with COVID-19,
and some of the trials have suggested that interferon beta-1a can cause harm in patients with severe
disease, such as those who require high-flow oxygen, noninvasive ventilation, or mechanical
ventilation.4,5 In a large randomized controlled trial of hospitalized patients with COVID-19, the
combination of interferon beta-1a plus remdesivir showed no clinical benefit when compared to
remdesivir alone.4 Similarly, the World Health Organization Solidarity trial did not show a benefit
of administering interferon beta-1a to hospitalized patients, approximately 50% of whom were on
corticosteroids.5
Systemic interferon alfa and inhaled interferons have also been evaluated in patients with COVID-19.
The trials that have evaluated the use of interferon alfa have generally been small or moderate in size
and have not been adequately powered to assess whether this agent provides a clinical benefit for
patients with COVID-19.6-8
Interferon Lambda
Pegylated interferon lambda was studied in a randomized, double-blind, adaptive clinical trial that
COVID-19 Treatment Guidelines 228
Considerations in Children
There are insufficient data on the use of interferons to treat respiratory viral infections in children to
make any recommendations for treating children with COVID-19.
References
1. Alavi Darazam I, Hatami F, Mahdi Rabiei M, et al. An investigation into the beneficial effects of high-dose
interferon beta 1-a, compared to low-dose interferon beta 1-a in severe COVID-19: The COVIFERON II
randomized controlled trial. Int Immunopharmacol. 2021;99:107916. Available at: https://www.ncbi.nlm.nih.
gov/pubmed/34224994.
2. Hung IF, Lung KC, Tso EY, et al. Triple combination of interferon beta-1b, lopinavir-ritonavir, and ribavirin
in the treatment of patients admitted to hospital with COVID-19: an open-label, randomised, Phase 2 trial.
Lancet. 2020;395(10238):1695-1704. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32401715.
3. Rahmani H, Davoudi-Monfared E, Nourian A, et al. Interferon beta-1b in treatment of severe COVID-19: a
randomized clinical trial. Int Immunopharmacol. 2020;88:106903. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32862111.
4. Kalil AC, Mehta AK, Patterson TF, et al. Efficacy of interferon beta-1a plus remdesivir compared with
remdesivir alone in hospitalised adults with COVID-19: a double-bind, randomised, placebo-controlled, Phase
3 trial. Lancet Respir Med. 2021;9(12):1365-1376. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34672949.
5. WHO Solidarity Trial Consortium. Repurposed antiviral drugs for COVID-19—interim WHO Solidarity trial
results. N Engl J Med. 2021;384(6):497-511. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33264556.
6. Yu J, Lu X, Tong L, et al. Interferon-alpha-2b aerosol inhalation is associated with improved clinical outcomes
in patients with coronavirus disease–2019. Br J Clin Pharmacol. 2021;87(12):4737-4746. Available at: https://
pubmed.ncbi.nlm.nih.gov/33982806.
7. Wang B, Li D, Liu T, Wang H, Luo F, Liu Y. Subcutaneous injection of IFN alpha-2b for COVID-19:
COVID-19 Treatment Guidelines 229
The clinical trials described in this table do not represent all the trials that the Panel reviewed while developing the recommendations
for interferons. The studies summarized below are the randomized controlled trials that have had the greatest impact on the Panel’s
recommendations.
ACTT-3: Multinational, Double-Blind RCT of Interferon Beta-1a and Remdesivir in Hospitalized Adults With COVID-191
Key Inclusion Criteria Participant Characteristics Key Limitation
• Evidence of pneumonia (radiographic infiltrates, SpO2 • Mean age 59 years; 38% were aged ≥65 years • After 270 patients were enrolled, OS6
≤94% on room air, or supplemental oxygen) • 58% men; 32% Latinx, 60% White, 17% Black patients were excluded because of an
• No MV required increased frequency of AEs in this group.
• Mean of 8.6 days of symptoms before enrollment
Key Exclusion Criteria • 90% had ≥1 comorbidity; 58% with HTN; 58% with Interpretation
• AST or ALT >5 times ULN obesity; 37% with DM • There was no clinical benefit of adding
IFN beta-1a to RDV in hospitalized
• Impaired renal function Primary Outcome patients with COVID-19.
• Hospital discharge or transfer anticipated within 72 • Median time to recovery: 5 days in both arms (rate ratio • The use of IFN beta-1a was associated
hours 0.99; 95% CI, 0.87–1.13; P = 0.88) with worse outcomes among patients
Interventions • In patients on high-flow oxygen or NIV (OS6) at who were OS6 at baseline.
baseline, median time to recovery: >28 days in IFN
• RDV 200 mg IV on Day 1, then RDV 100 mg IV once daily
beta-1a arm vs. 9 days in placebo arm (rate ratio 0.40;
for 9 days plus IFN beta-1a 44 µg SUBQ every other day
95% CI, 0.22–0.75; P = 0.0031)
for up to 4 doses (n = 487)
• RDV 200 mg IV on Day 1, then RDV 100 mg IV once daily Secondary Outcomes
for 9 days plus placebo (n = 482) • No difference between arms in clinical status at Day 14
(OR 1.01; 95% CI, 0.79–1.28)
Primary Endpoint
• No difference between IFN beta-1a arm and placebo arm
• Time to recovery by Day 28
in mortality by Day 28 in:
Key Secondary Endpoints • All patients: 5% vs. 3% (HR 1.33; 95% CI, 0.69–2.55)
• Clinical status at Day 14, as measured by an OS • Patients who were OS6 at baseline: 21% vs. 12% (HR
• Mortality by Day 28 1.74; 95% CI, 0.51–5.93)
TOGETHER: Double-Blind, Adaptive RCT of Pegylated Interferon Lambda in Nonhospitalized Patients With COVID-19 in Brazil and Canada4
Key Inclusion Criteria Participant Characteristics Key Limitations
• Positive SARS-CoV-2 antigen test result • Median age 43 years; 57.1% women; 95.1% self- • Health care facility capacity may have
• Within 7 days of symptom onset identified as mixed race influenced the number and duration of
• 1,919 (98.5%) from Brazil, 30 (1.5%) from Canada ED observations.
• ≥1 high-risk factor for disease progression (e.g., age ≥50
years, comorbidities, immunosuppression) • 50% with obesity • As this was an adaptive platform trial
where multiple investigational treatments
• Up to 25% of patients could have no high-risk factors. • 59.4% were randomized within 3 days of symptom or placebos were being evaluated
onset. simultaneously, not all patients in the
Key Exclusion Criteria
• 83% received ≥1 COVID-19 vaccine dose. placebo arm received a placebo that was
• Need for hospitalization
Primary Outcome matched to PEG-IFN lambda.
• SpO2 ≤93% on room air
• Composite of ED observation >6 hours or hospitalization Interpretation
Interventions
for COVID-19 by Day 28 (ITT): 25 (2.7%) in PEG-IFN • In outpatients with COVID-19 who were
• Single dose of PEG-IFN lambda 180 μg SUBQ (n = 931) lambda arm vs. 57 (5.6%) in placebo arm (relative risk within 7 days of symptom onset, PEG-
• Placebo (n = 1,018; 825 received single SUBQ injection, 0.49; 95% Bayesian CrI, 0.30–0.76) IFN lambda reduced the need for ED
193 received PO placebo) • 61 events (74%) were hospitalizations (ITT). observations >6 hours or hospitalization
when compared with placebo.
Primary Endpoint Secondary Outcomes
• Composite of ED observation >6 hours or hospitalization • Composite of COVID-19–related hospitalization or
for COVID-19 by Day 28 death by Day 28: 22 (2.4%) in PEG-IFN lambda arm vs.
Key Secondary Endpoints 40 (3.9%) in placebo arm (relative risk 0.61; 95% CrI,
0.36–0.99)
• Composite of COVID-19–related hospitalization or death
by Day 28 • SARS-CoV-2 viral clearance at Day 7 among the 15%
of patients with VL >192 million copies/mL at baseline:
• SARS-CoV-2 viral clearance at Day 7 50.5% in PEG-IFN lambda arm vs. 32.9% in placebo arm
• Occurrence of AEs (OR 2.13; 95% CrI, 1.14–4.00)
• Occurrence of AEs: 141 (15.1%) in PEG-IFN lambda arm
vs. 172 (16.9%) in placebo arm (relative risk 0.90; 95%
CrI, 0.73–1.10)
Single-Blind RCT of Pegylated Interferon Lambda-1a for Treatment of Outpatients With Uncomplicated COVID-19 in the United States5
Key Inclusion Criteria Participant Characteristics Key Limitation
• Aged 18–65 years • Median age 36 years; 42% women; 63% Latinx, 28% • Small sample size
• Asymptomatic or symptomatic White
Interpretation
• Positive SARS-CoV-2 RT-PCR result within 72 hours of • 7% were asymptomatic.
• PEG-IFN lambda-1a provided no virologic
enrollment • Median of 5 days of symptoms before randomization or clinical benefit compared to placebo
Key Exclusion Criteria Primary Outcome among outpatients with uncomplicated
COVID-19.
• Current or imminent hospitalization • Median time to cessation of viral shedding: 7 days in
• Respiratory rate >20 breaths/min both arms (aHR 0.81; 95% CI, 0.56–1.19; P = 0.29)
• SpO2 <94% on room air Secondary Outcomes
• Decompensated liver disease • No difference between PEG-IFN lambda-1a and placebo
arms in:
Interventions
• Proportion of patients hospitalized by Day 28: 3.3% for
• Single dose of PEG-IFN lambda-1a 180 µg SUBQ (n = 60) each arm
• Placebo (n = 60) • Time to resolution of symptoms: 8 days vs. 9 days (HR
Primary Endpoint 0.94; 95% CI, 0.64–1.39)
• Time to first negative SARS-CoV-2 RT-PCR result Other Outcome
Key Secondary Endpoints • Patients who received PEG-IFN lambda-1a were more
• Hospitalization by Day 28 likely to have elevations of transaminase concentrations
than patients who received placebo (25% vs. 8%; P =
• Time to complete symptom resolution 0.027).
Double-Blind RCT of Pegylated Interferon Lambda in Outpatients With Laboratory-Confirmed COVID-19 in Canada6
Key Inclusion Criteria Participant Characteristics Key Limitation
• Positive SARS-CoV-2 PCR result • Median age 46 years; 58% women; 52% White • Small sample size
• Patients were within 7 days of symptom onset, or, if • 19% were asymptomatic. Interpretation
asymptomatic, were within 7 days of first positive SARS- • Mean of 4.5 days of symptoms before randomization • PEG-IFN lambda may accelerate VL
CoV-2 test result.
Primary Outcome decline and clearance in outpatients
Key Exclusion Criterion with COVID-19; however, the clinical
• 80% in PEG-IFN lambda arm vs. 63% in placebo arm significance of this finding is unclear.
• Immunosuppression or condition that could be worsened were negative for SARS-CoV-2 RNA at Day 7 (P = 0.15).
by PEG-IFN lambda
Secondary Outcomes
Interventions
• VL decline by Day 7 was greater in PEG-IFN lambda arm
• Single dose of PEG-IFN lambda 180 µg SUBQ (n = 30) than in placebo arm (P = 0.0041).
• Placebo (n = 30) • 1 participant in each arm hospitalized by Day 14
Primary Endpoint Other Outcome
• Proportion of patients with negative SARS-CoV-2 test • 3 participants in each arm had mild elevations of
result on nasal mid-turbinate swab at Day 7 aminotransferase concentrations. Increase was greater
Key Secondary Endpoints in PEG-IFN lambda arm.
• Quantitative change in SARS-CoV-2 RNA over time
• Hospitalization by Day 14
Key: AE = adverse event; ALT = alanine transaminase; AST = aspartate aminotransferase; DM = diabetes mellitus; ECMO = extracorporeal membrane oxygenation; ED
= emergency department; HCQ = hydroxychloroquine; HTN = hypertension; IFN = interferon; ITT = intention-to-treat; IV = intravenous; LPV/RTV = lopinavir/ritonavir;
MV = mechanical ventilation; NIV = noninvasive ventilation; OS = ordinal scale; the Panel = the COVID-19 Treatment Guidelines Panel; PCR = polymerase chain
reaction; PEG-IFN = pegylated interferon; PO = oral; RCT = randomized controlled trial; RDV = remdesivir; RT-PCR = reverse transcription polymerase chain reaction;
SOC = standard of care; SpO2 = oxygen saturation; SUBQ = subcutaneous; ULN = upper limit of normal; VL = viral load; WHO = World Health Organization
References
1. Kalil AC, Mehta AK, Patterson TF, et al. Efficacy of interferon beta-1a plus remdesivir compared with remdesivir alone in hospitalised adults with
COVID-19: a double-bind, randomised, placebo-controlled, Phase 3 trial. Lancet Respir Med. 2021;9(12):1365-1376. Available at: https://www.ncbi.
nlm.nih.gov/pubmed/34672949.
2. WHO Solidarity Trial Consortium. Repurposed antiviral drugs for COVID-19—interim WHO Solidarity trial results. N Engl J Med. 2021;384(6):497-
511. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33264556.
3. Ader F, Peiffer-Smadja N, Poissy J, et al. An open-label randomized controlled trial of the effect of lopinavir/ritonavir, lopinavir/ritonavir plus
COVID-19 Treatment Guidelines 236
Hospitalized Patients
• 5 days or until hospital discharge
• If a patient does not clinically
improve, clinicians may extend the
treatment course for ≤5 additional
days, for a total duration of 10 days.
References
1. Ritonavir-boosed nirmatrelvir (Paxlovid) [package insert]. Food and Drug Administration. 2023. Available at: https://www.accessdata.fda.gov/
drugsatfda_docs/label/2023/217188s000lbl.pdf.
2. Food and Drug Administration. Fact sheet for healthcare providers: Emergency Use Authorization for Paxlovid. 2023. Available at:
https://www.fda.gov/media/155050/download.
3. BC COVID Therapeutics Committee COVID Therapy Review and Advisory Working Group. Therapeutic brief: crushing nirmatrelvir/ritonavir
(Paxlovid). 2022. Available at: http://www.bccdc.ca/Health-Professionals-Site/Documents/COVID-treatment/Crushing_Paxlovid.pdf.
4. Remdesivir (Veklury) [package insert]. Food and Drug Administration. 2023. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/214787s019lbl.pdf.
5. Food and Drug Administration. Fact sheet for healthcare providers: Emergency Use Authorization for Lagevrio (molnupiravir) capsules. 2023.
Available at: https://www.fda.gov/media/155054/download.
6. Food and Drug Administration. Fact sheet for health care providers: Emergency Use Authorization (EUA) of COVID-19 convalescent plasma for
treatment of coronavirus disease 2019 (COVID-19). 2021. Available at: https://www.fda.gov/media/141478/download.
Summary Recommendations
• The hyperactive inflammatory response to SARS-CoV-2 infection plays a central role in the pathogenesis of COVID-19.
See Therapeutic Management of Hospitalized Adults With COVID-19 and Therapeutic Management of Hospitalized
Children With COVID-19 for the COVID-19 Treatment Guidelines Panel’s (the Panel) recommendations on the use of
the following immunomodulators in hospitalized patients with COVID-19 according to their disease severity (listed in
alphabetical order):
• Abatacept
• Baricitinib (or tofacitinib)
• Dexamethasone
• Infliximab
• Tocilizumab (or sarilumab)
• There is insufficient evidence for the Panel to recommend either for or against the use of the following
immunomodulators for the treatment of COVID-19:
• Anakinra
• Inhaled corticosteroids
• Vilobelimab
• The Panel recommends against the use of canakinumab for the treatment of COVID-19, except in a clinical trial
(BIIa).
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
The results of several randomized trials indicate that systemic corticosteroid therapy improves clinical
outcomes and reduces mortality in hospitalized patients with COVID-19 who require supplemental
oxygen,1,2 presumably by mitigating the COVID-19–induced systemic inflammatory response that
can lead to lung injury and multisystem organ dysfunction. In contrast, in hospitalized patients with
COVID-19 who do not require supplemental oxygen, the use of systemic corticosteroids provided
no benefit and increased mortality.3,4 The COVID-19 Treatment Guidelines Panel’s (the Panel)
recommendations for the use of systemic corticosteroids in hospitalized patients with COVID-19 are
based on results from these clinical trials (see Table 5a). There are no data to support the use of systemic
corticosteroids in nonhospitalized patients with COVID-19.
Recommendations
• The Panel recommends against the use of dexamethasone or other systemic corticosteroids
to treat outpatients with mild to moderate COVID-19 who do not require hospitalization or
supplemental oxygen (AIIb).
• See Therapeutic Management of Hospitalized Adults With COVID-19 for the Panel’s
recommendations on the use of dexamethasone or other systemic corticosteroids in certain
hospitalized patients with COVID-19.
• Patients with COVID-19 who are receiving dexamethasone or another corticosteroid for an
underlying condition should continue this therapy as directed by their health care provider (AIII).
Rationale
Nonhospitalized Adults
There are no data to support the use of systemic corticosteroids in nonhospitalized patients with
COVID-19. Therefore, the safety and efficacy of using systemic corticosteroids in this population have
not been established. Generally, the use of systemic corticosteroids is associated with adverse events
(e.g., hyperglycemia, neuropsychiatric symptoms, secondary infections), which may be difficult to
detect and monitor in an outpatient setting. For more information, see Therapeutic Management of
Nonhospitalized Adults With COVID-19.
Hospitalized Adults
The RECOVERY trial was a multicenter, open-label trial in the United Kingdom that randomly
assigned 6,425 hospitalized patients to receive up to 10 days of dexamethasone 6 mg once daily plus
standard care or standard care alone.3 Mortality at 28 days was lower among the patients who received
dexamethasone than among those who received standard care alone. This benefit of dexamethasone
was observed in patients who were mechanically ventilated or who required supplemental oxygen at
enrollment. In contrast, no benefit was seen in patients who did not require supplemental oxygen at
enrollment.
Several clinical trials have evaluated the use of systemic corticosteroids in critically ill patients with
COVID-19 who were on supplemental oxygen with or without mechanical ventilation. These trials
reported that all-cause mortality at 28 days was lower in those who received systemic corticosteroids
Dexamethasone Dose
The RECOVERY platform trial studied the use of dexamethasone 6 mg once daily for up to 10
days,3 which is currently the recommended dose for hospitalized adults with COVID-19 who require
supplemental oxygen. Several other randomized controlled trials evaluated the role of higher doses
of dexamethasone or other corticosteroids in hospitalized patients with different levels of respiratory
support. The results of some key studies are summarized below.
Considerations in Children
Dexamethasone is recommended for hospitalized children with COVID-19 who require supplemental
oxygen. See Therapeutic Management of Hospitalized Children With COVID-19 for the Panel’s
recommendations. Methylprednisolone or another corticosteroid is recommended for the treatment of
multisystem inflammatory syndrome in children (MIS-C). See Therapeutic Management of Hospitalized
Children With MIS-C, Plus a Discussion on MIS-A for the Panel’s recommendations.
References
1. WHO Rapid Evidence Appraisal for COVID-19 Therapies (REACT) Working Group. Association between
administration of systemic corticosteroids and mortality among critically ill patients with COVID-19: a meta-
analysis. JAMA. 2020;324(13):1330-1341. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32876694.
2. Li H, Yan B, Gao R, Ren J, Yang J. Effectiveness of corticosteroids to treat severe COVID-19: a systematic
review and meta-analysis of prospective studies. Int Immunopharmacol. 2021;100:108121. Available at:
The clinical trials described in this table do not represent all the trials that the Panel reviewed while developing the recommendations for
systemic corticosteroids. The studies summarized below are those that have had the greatest impact on the Panel’s recommendations. Unless
stated otherwise, the clinical trials listed below only included participants aged ≥18 years.
CoDEX: Open-Label RCT of Dexamethasone in Patients With Moderate to Severe ARDS and COVID-19 in Brazil2
Key Inclusion Criteria Participant Characteristics Key Limitations
• Suspected or laboratory-confirmed • Mean age 61 years; 63% men • Open-label study
SARS-CoV-2 infection • Comorbidities in DEX arm vs. SOC arm: • Underpowered; enrollment stopped after release
• Received MV within 48 hours of meeting • Obesity: 31% vs. 24% of data from the RECOVERY trial.
criteria for moderate to severe ARDS • Since no follow-up data were collected after
(PaO2/FiO2 ≤200 mm Hg) • DM: 38% vs. 47%
hospital discharge for patients who were
• Vasopressor use: 66% in DEX arm vs. 68% in SOC arm discharged before Day 28, no data on deaths or
Key Exclusion Criteria
• Mean PaO2/FiO2: 131 mm Hg in DEX arm vs. 133 mm Hg in rehospitalization were available for these patients
• Received immunosuppressive drugs in SOC arm between day of discharge and Day 28.
past 21 days
• Median of 10 days of DEX therapy • The high mortality in this study may limit the
• Death expected within 24 hours
• No patients received RDV or tocilizumab. generalizability of results to populations with a
Interventions lower baseline mortality.
• 35% in SOC arm received corticosteroids for indications such
• DEX 20 mg IV once daily for 5 days, then as bronchospasm or septic shock. • More than a third of those randomized to receive
DEX 10 mg IV once daily for 5 days or SOC also received corticosteroids.
until ICU discharge (n = 151) Primary Outcome
Interpretation
• SOC alone (n = 148) • Mean number of days alive and free from MV by Day 28: 7 in
DEX arm vs. 4 in SOC arm (P = 0.04) • Compared with SOC alone, DEX increased the
Primary Endpoint mean number of days alive and free of MV by Day
Secondary Outcomes 28 in patients with COVID-19 and moderate to
• Number of days alive and free from MV
by Day 28 • No differences between arms by Day 28 in all-cause mortality severe ARDS.
(56% in DEX arm vs. 62% in SOC arm), number of ICU-free
Key Secondary Endpoints days, or duration of MV
• All-cause mortality by Day 28 • No difference between arms at Day 15 in score on 6-point OS
• Number of ICU-free days by Day 28 • Mean SOFA score at Day 7: 6.1 in DEX arm vs. 7.5 in SOC arm
• Duration of MV by Day 28 (P = 0.004)
• Score on 6-point OS at Day 15 Other Outcome
• SOFA score at Day 7 • Post hoc analysis of probability of death or MV at Day 15: 68%
Key Exploratory Analysis in DEX arm vs. 80% in SOC arm (OR 0.46; P = 0.01)
• Death or MV at Day 15
Observational Cohort Study of Dexamethasone in Hospitalized Patients With COVID-19 Who Were Not on Intensive Respiratory Support in the United States3
Key Inclusion Criterion Participant Characteristics Key Limitations
• Within 14 days of a positive SARS-CoV-2 • Mean age 71 years; 95% men; 27% Black, 55% White • Retrospective observational study
test result • 77% did not receive IRS within 48 hours. • Because nearly all patients on MV or HFNC oxygen
Key Exclusion Criteria • 83% were hospitalized within 1 day of a positive SARS-CoV-2 received DEX, analysis was restricted to patients
test result. who did not receive IRS (i.e., those who received
• Recent receipt of corticosteroids
no supplemental oxygen or only low-flow nasal
• Receipt of IRS (defined as HFNC oxygen, • Median duration of DEX for patients who did not receive IRS: cannula oxygen).
NIV, or MV) within 48 hours 5 days for those not on supplemental oxygen at baseline vs. 6
days for those on low-flow nasal cannula oxygen • There were differences between the arms in other
• Hospital LOS <48 hours therapies received. The investigators attempted to
• Received RDV: 43% of those who received DEX vs. 13% of account for this using different approaches (e.g.,
Interventions those who did not propensity scoring, weighted analyses, subgroup/
• Corticosteroids (95% received DEX) • Received anticoagulants: 46% of those who received DEX vs. sensitivity analyses).
administered within 48 hours of 10% of those who did not
admission (n = 7,507) Interpretation
Primary Outcome • In hospitalized patients with COVID-19, the use
• No corticosteroids administered (n =
7,433) • Risk of all-cause mortality at 90 days was higher in those who of DEX was not associated with a reduction in
received DEX. mortality among those who received low-flow
Primary Endpoint nasal cannula oxygen during the first 48 hours
• Combination of those not on supplemental oxygen and
• All-cause mortality at 90 days those on low-flow nasal cannula oxygen: HR 1.59; 95% CI, after hospital admission, but it was associated
1.39–1.81 with increased mortality among those who
received no supplemental oxygen during the first
• Those not on supplemental oxygen: HR 1.76; 95% CI,
48 hours after admission.
1.47–2.12
• Those on low-flow nasal cannula oxygen: HR 1.08; 95% CI,
0.86–1.36
Key: AE = adverse event; ARDS = acute respiratory distress syndrome; BMI = body mass index; CPAP = continuous positive airway pressure; CT = computed
tomography; CYP = cytochrome P450; DEX = dexamethasone; DM = diabetes mellitus; ECMO = extracorporeal membrane oxygenation; FiO2 = fraction of inspired
oxygen; HFNC = high-flow nasal cannula; ICU = intensive care unit; IL = interleukin; IRS = intensive respiratory support; IV = intravenous; JAK = Janus kinase; LOS =
length of stay; MV = mechanical ventilation; NIV = noninvasive ventilation; OS = ordinal scale; the Panel = the COVID-19 Treatment Guidelines Panel; PaO2 = arterial
partial pressure of oxygen; PEEP = positive end-expiratory pressure; PO = oral; RCT = randomized controlled trial; RDV = remdesivir; SAE = serious adverse event; SOC
= standard of care; SOFA = sequential organ failure assessment; SpO2 = oxygen saturation; TB = tuberculosis
COVID-19 Treatment Guidelines 258
Inhaled corticosteroids have been identified as potential COVID-19 therapeutic agents because of
their targeted anti-inflammatory effects on the lungs. In addition, certain inhaled corticosteroids have
been shown to impair viral replication of SARS-CoV-21 and downregulate the expression of the
receptors used for cell entry.2,3 Several trials provide additional insights regarding the role of inhaled
corticosteroids in treating outpatients with COVID-19. These trials are described below and in Table 5b.
Recommendations
• There is insufficient evidence for the COVID-19 Treatment Guidelines Panel (the Panel) to
recommend either for or against the use of inhaled corticosteroids for the treatment of COVID-19.
• There is insufficient evidence for the Panel to recommend either for or against the use of
the combination of inhaled budesonide plus fluvoxamine for the treatment of COVID-19 in
nonhospitalized patients.
• Patients with COVID-19 who are receiving an inhaled corticosteroid for an underlying condition
should continue this therapy as directed by their health care provider (AIII).
Rationale
Compared to usual care, inhaled corticosteroid therapy decreased the time to recovery in 2 open-label
randomized controlled trials in outpatients with mild symptoms of COVID-19.4,5 However, subsequent
placebo-controlled, double-blind trials have shown that corticosteroid therapy did not reduce the
duration of COVID-19 symptoms.6-8 The available evidence does not show that inhaled corticosteroid
therapy reduces the risk of hospitalization or death due to COVID-19. However, the Panel acknowledges
that there are areas of uncertainty. Studies conducted predominantly among unvaccinated patients have
reported mixed results.
ACTIV-6 is the only randomized controlled trial of inhaled corticosteroid monotherapy that was
conducted in a predominantly vaccinated population.8 In this study, treatment with inhaled fluticasone
did not reduce the number of hospitalizations or health care visits or the time to sustained recovery.
However, this study included patients who were at modest risk for complications from COVID-19. The
median age of the patients was 45 years, and patients were not required to have a comorbidity to be
included in the study.
The mixed results from these studies make it difficult to draw definitive conclusions about the benefit
of using inhaled corticosteroids in people who are at high risk of disease progression. See Therapeutic
Management of Nonhospitalized Adults With COVID-19 for recommendations regarding therapies for
high-risk outpatients.
The combination of inhaled budesonide plus oral fluvoxamine was studied in a large, double-blind,
placebo-controlled, adaptive randomized trial in Brazil.9 Over 90% of the patients had received at least
2 doses of a COVID-19 vaccine. Treatment with this combination significantly reduced the incidence of
the primary outcome, which was a composite of hospitalization or retention in an emergency setting for
>6 hours. The proportion of patients who were hospitalized was the same in the treatment and placebo
arms (0.9% vs. 1.1%), and the treatment did not significantly impact secondary outcomes such as health
care attendance or the need for an emergency setting visit. It is unclear how the >6-hour emergency
Considerations in Children
There is insufficient evidence for the Panel to recommend either for or against the use of inhaled
corticosteroids for the treatment of COVID-19 in children. Children with COVID-19 who are receiving
an inhaled corticosteroid for an underlying condition should continue this therapy as directed by their
health care provider (AIII).
References
1. Matsuyama S, Kawase M, Nao N, et al. The inhaled steroid ciclesonide blocks SARS-CoV-2 RNA replication
by targeting the viral replication-transcription complex in cultured cells. J Virol. 2020;95(1):e01648-20.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/33055254.
2. Finney LJ, Glanville N, Farne H, et al. Inhaled corticosteroids downregulate the SARS-CoV-2 receptor ACE2
in COPD through suppression of type I interferon. J Allergy Clin Immunol. 2021;147(2):510-519.e5. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/33068560.
3. Peters MC, Sajuthi S, Deford P, et al. COVID-19-related genes in sputum cells in asthma. Relationship to
demographic features and corticosteroids. Am J Respir Crit Care Med. 2020;202(1):83-90. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32348692.
4. Ramakrishnan S, Nicolau DV Jr, Langford B, et al. Inhaled budesonide in the treatment of early COVID-19
(STOIC): a Phase 2, open-label, randomised controlled trial. Lancet Respir Med. 2021;9(7):763-772. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/33844996.
5. Yu LM, Bafadhel M, Dorward J, et al. Inhaled budesonide for COVID-19 in people at high risk of
complications in the community in the UK (PRINCIPLE): a randomised, controlled, open-label, adaptive
platform trial. Lancet. 2021;398(10303):843-855. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34388395.
6. Clemency BM, Varughese R, Gonzalez-Rojas Y, et al. Efficacy of inhaled ciclesonide for outpatient treatment
of adolescents and adults with symptomatic COVID-19: a randomized clinical trial. JAMA Intern Med.
2022;182(1):42-49. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34807241.
7. Ezer N, Belga S, Daneman N, et al. Inhaled and intranasal ciclesonide for the treatment of COVID-19 in adult
The clinical trials described in this table do not represent all the trials that the Panel reviewed while developing the recommendations for
inhaled corticosteroids. The studies summarized below are those that have had the greatest impact on the Panel’s recommendations.
PRINCIPLE: Open-Label RCT of Inhaled Budesonide in Nonhospitalized Patients With COVID-19 in the United Kingdom1
Key Inclusion Criteria Participant Characteristics Key Limitations
• Aged ≥65 years or aged ≥50 years with comorbidities • Mean age 64.2 years; 52% women; 92% • Open-label trial
• PCR-confirmed or suspected COVID-19 White • Primary endpoint of time to recovery
• ≤14 days of COVID-19 symptoms • 81% with comorbidities was based on patient self-report.
• Median of 6 days from symptom onset to Interpretation
Key Exclusion Criteria
randomization
• Already taking inhaled or systemic corticosteroids • Inhaled budesonide reduced the time to
Primary Outcomes reported recovery but not the incidence
• Unable to use an inhaler of COVID-19–related hospitalization or
• COVID-19–related hospitalization or death by
• Contraindication for inhaled budesonide Day 28: 6.8% in budesonide arm vs. 8.8% in death.
Interventions usual care arm (OR 0.75; 95% CrI, 0.55–1.03) • The clinical significance of self-reported
• Usual care plus inhaled budesonide 800 µg twice daily for 14 days • Median time to reported recovery: 11.8 days time to recovery in an open-label study
(n = 1,069) in budesonide arm vs. 14.7 days in usual care is unclear.
• Usual care (n = 787) arm (HR 1.21; 95% CrI, 1.08–1.36)
Primary Endpoints
• COVID-19–related hospitalization or death by Day 28
• Time to reported recovery up to 28 days from randomization
STOIC: Open-Label, Phase 2 RCT of Inhaled Budesonide in Nonhospitalized Adults With Early COVID-19 in the United Kingdom2
Key Inclusion Criteria Participant Characteristics Key Limitations
• Aged ≥18 years • Mean age 45 years; 58% women • Small, open-label trial
• ≤7 days of COVID-19 symptoms • 9% with CVD; 5% with DM • Trial was terminated early after
• 95% with positive SARS-CoV-2 RT-PCR result statistical analysis determined that
Key Exclusion Criteria
additional patients would not alter study
• Use of inhaled or systemic glucocorticoids in past 7 days • Median of 3 days from symptom onset to outcome.
randomization
• Known allergy or contraindication to budesonide
STOIC: Open-Label, Phase 2 RCT of Inhaled Budesonide in Nonhospitalized Adults With Early COVID-19 in the United Kingdom2, continued
Interventions Primary Outcome • Secondary endpoint of time to recovery
• Usual care plus inhaled budesonide 800 µg twice daily until • COVID-19–related urgent care visit: 1% in was based on patient self-report.
symptom resolution (n = 70) budesonide arm vs. 14% in usual care arm Interpretation
• Usual care (n = 69) (difference in proportion 0.131; 95% CI, 0.043–
• In adult outpatients with mild COVID-19,
0.218; P = 0.004)
Primary Endpoint inhaled budesonide may reduce the
Secondary Outcome need for urgent care, ED visit, or
• COVID-19–related urgent care visit, including ED visit or hospitalization.
hospitalization • Median time to clinical recovery: 7 days in
budesonide arm vs. 8 days in usual care arm • The clinical significance of self-reported
Key Secondary Endpoint time to recovery in an open-label study
• Time to clinical recovery is unclear.
Phase 3, Double-Blind, Placebo-Controlled RCT of Inhaled Ciclesonide in Nonhospitalized Patients With COVID-19 in the United States3
Key Inclusion Criteria Participant Characteristics Key Limitations
• Aged ≥12 years • Mean age 43.3 years; 55.3% women; 86.3% White • ED visit or hospital admission outcome
• Positive SARS-CoV-2 molecular or antigen diagnostic test • Mean BMI 29.4 was based on a small number of
result in previous 72 hours events.
• 22.3% with HTN; 7.5% with type 2 DM
• ≥1 symptoms of COVID-19 (i.e., fever, cough, dyspnea) • Primary endpoint of time to alleviation
• Higher rates of DM and asthma in ciclesonide arm of all symptoms was based on patient
Key Exclusion Criteria Primary Outcome self-report.
• Use of inhaled or intranasal corticosteroid within 14 days • Median of 19 days in both arms for alleviation of Interpretation
of enrollment or systemic corticosteroid within 90 days of all COVID-19–related symptoms (HR 1.08; 95% CI,
enrollment • Inhaled ciclesonide did not reduce
0.84–1.38) the time to reported recovery in
• Unable to use an inhaler nonhospitalized patients with COVID-19.
Secondary Outcomes
Interventions • Alleviation of COVID-19–related symptoms by Day • The robustness of the conclusion that
• Ciclesonide MDI 160 µg/actuation, administered as 2 30: 70.6% in ciclesonide arm vs. 63.5% in placebo inhaled ciclesonide reduced COVID-19-
actuations twice daily for 30 days (n = 197) arm (OR 1.28; 95% CI, 0.84–19.7) related ED visits or hospital admissions
• Placebo MDI twice daily for 30 days (n = 203) • ED visit or hospital admission for COVID-19 by Day is uncertain. The small number of
30: 1.0% in ciclesonide arm vs. 5.4% in placebo arm events is most likely due to the
Primary Endpoint relatively low rate of comorbidities in
(OR 0.18; 95% CI, 0.04–0.85)
• Time to alleviation of all COVID-19–related symptoms by Day the study population.
• Hospital admission or death by Day 30: 1.5% in
30
ciclesonide arm vs. 3.4% in placebo arm (OR 0.45;
Key Secondary Endpoints 95% CI, 0.11–1.84)
• Alleviation of COVID-19–related symptoms by Day 30 • No deaths by Day 30 in either arm
Phase 3, Double-Blind, Placebo-Controlled RCT of Inhaled Ciclesonide in Nonhospitalized Patients With COVID-19 in the United States3, continued
• ED visit or hospital admission for COVID-19 by Day 30
• Hospital admission or death by Day 30
CONTAIN: Double-Blind RCT of Inhaled Ciclesonide Plus Intranasal Ciclesonide in Nonhospitalized Patients With COVID-19 in Canada4
Key Inclusion Criteria Participant Characteristics Key Limitation
• Aged ≥18 years • Median age 35 years; 54% women; 61% White • Small study with a relatively young,
• Positive SARS-CoV-2 molecular diagnostic test result • 20% with comorbidities healthy population
• ≥1 symptoms of COVID-19 (i.e., fever, cough, shortness of Primary Outcome Interpretation
breath)
• Resolution of fever and all respiratory symptoms at • The use of inhaled ciclesonide plus
• ≤5 days of COVID-19 symptoms Day 7: 40% in ciclesonide arm vs. 35% in placebo intranasal ciclesonide did not improve
arm (adjusted risk difference 5.5%; 95% CI, -7.8% resolution of fever and respiratory
Key Exclusion Criteria symptoms in nonhospitalized patients
to 18.8%)
• Receiving an inhaled corticosteroid or received a PO or IM with COVID-19.
corticosteroid within 7 days of enrollment Secondary Outcomes
• Unable to use an inhaler • Resolution of fever and all respiratory symptoms
• Has only nonrespiratory symptoms at Day 14: 66% in ciclesonide arm vs. 58% in
placebo arm (adjusted risk difference 7.5%; 95%
• Use of oxygen at home CI, -5.9% to 20.8%)
• Vaccinated against COVID-19 • Hospital admission by Day 14: 6% in ciclesonide
Interventions arm vs. 3% in placebo arm (adjusted risk
difference 2.3%; 95% CI, -3.0% to 7.6%)
• Ciclesonide MDI 600 µg/actuation plus intranasal ciclesonide
100 µg, both twice daily for 14 days (n = 105)
• Saline placebo MDI plus intranasal saline, both twice daily for
14 days (n = 98)
Primary Endpoint
• Resolution of fever and all respiratory symptoms at Day 7
Key Secondary Endpoints
• Resolution of fever and all respiratory symptoms at Day 14
• Hospital admission by Day 14
COVERAGE: Open-Label RCT of Inhaled Ciclesonide in Nonhospitalized Adults With COVID-19 in France5
Key Inclusion Criteria Participant Characteristics Key Limitation
• Aged ≥60 years or aged ≥50 years with comorbidities • Median age 63 years; 51% women • Small, open-label study
• Positive SARS-CoV-2 nasopharyngeal RT-PCR result or • 72% with ≥1 comorbidities Interpretation
antigen test result • 14% received ≥1 COVID-19 vaccine doses. • In adult outpatients with mild COVID-19,
• ≤7 days of COVID-19 symptoms inhaled ciclesonide did not reduce the
Primary Outcome
Key Exclusion Criteria proportion of patients who died, were
• Composite of hospitalization from any cause, need hospitalized, or required COVID-19–
• Chronic use of inhaled corticosteroid therapy for COVID-19–related oxygen therapy at home, or related oxygen therapy at home.
• Unable to use an inhalation chamber death by Day 14: 16% in ciclesonide arm vs. 12%
in control arm
• Ongoing therapy with a potent CYP3A4 inhibitor
Secondary Outcome
Interventions
• Sustained alleviation of symptoms by Day 14: 54%
• Ciclesonide 160 µg via inhalation chamber, 2 puffs twice daily in ciclesonide arm vs. 57% in control arm
for 10 days (n = 110)
• Vitamin and trace element supplement, 2 capsules PO once or
twice daily for 10 days (n = 107)
Primary Endpoint
• Composite of hospitalization from any cause, need for
COVID-19–related oxygen therapy at home, or death by Day
14
Key Secondary Endpoint
• Sustained alleviation of symptoms by Day 14
ACTIV-6: Decentralized, Placebo-Controlled, Platform RCT of Inhaled Fluticasone in Outpatients With COVID-19 in the United States6
Key Inclusion Criteria Participant Characteristics Key Limitations
• Aged ≥30 years • Median age 45 years; 63% women • Low numbers of some clinical endpoints
• Positive SARS-CoV-2 nasopharyngeal RT-PCR result or • 39% with BMI >30; 26% with HTN limited the ability to assess the effect of
antigen test result inhaled fluticasone on the key secondary
• 65% received ≥2 COVID-19 vaccine doses. endpoints.
• ≤7 days of ≥2 COVID-19 symptoms
Primary Outcome • Not all patients in the placebo arm
Key Exclusion Criterion • No difference between arms in time to sustained received a matched placebo.
• Use of inhaled or systemic corticosteroids in preceding 30 recovery (HR 1.01; 95% CrI, 0.91–1.12) Interpretation
days
Secondary Outcomes • In adult outpatients with mild COVID-19,
Interventions • Hospitalization or death by Day 28: 0.5% in inhaled fluticasone did not reduce the
• Inhaled fluticasone 200 µg once daily for 14 days (n = 656) fluticasone arm vs. 0.5% in placebo arm time to sustained symptom recovery or
• Matching inhaled placebo (n = 350) or placebo from a • Urgent care visit, ED visit, or hospitalization by Day the occurrence of urgent care visits, ED
different study (n = 271) 28: 3.7% in fluticasone arm vs. in 2.1% placebo visits, or hospitalizations.
arm (HR 1.9; 95% CrI, 0.8–3.5)
Primary Endpoint
• Mean number of days unwell with ongoing
• Time to sustained recovery (i.e., the last of 3 consecutive days
symptoms: 11.2 in fluticasone arm vs. 11.3 in
without symptoms)
placebo arm
Key Secondary Endpoints
• Hospitalization or death by Day 28
• Urgent care visit, ED visit, or hospitalization by Day 28
• Number of days unwell with ongoing symptoms
TOGETHER: Placebo-Controlled, Platform RCT of Oral Fluvoxamine and Inhaled Budesonide in Adults With Early-Onset COVID-19 in Brazil7
Key Inclusion Criteria Participant Characteristics Key Limitation
• Aged ≥50 years or aged ≥18 years with comorbidities • Median age 51 years; 61% women • Multiple investigational treatments or
• Laboratory-confirmed SARS-CoV-2 infection • 42% with BMI >30 placebos were evaluated simultaneously.
Not all patients in the placebo arm
• ≤7 days of COVID-19 symptoms • 44% with HTN; 68% with ≥2 comorbidities received a matched placebo.
Key Exclusion Criteria • 94% received ≥2 COVID-19 vaccine doses.
Interpretation
• Use of an SSRI Primary Outcome • Adult outpatients with mild COVID-19 who
• Severe mental illness • Composite of ED observation >6 hours or received a combination of fluvoxamine
• Cirrhosis, recent seizures, or severe ventricular cardiac hospitalization by Day 28: 1.8% in fluvoxamine and and inhaled budesonide had fewer ED
arrythmia budesonide arm vs. 3.7% in placebo arm (relative observations >6 hours or hospitalizations
risk 0.50; 95% CrI, 0.25–0.92) for COVID-19 by Day 28 than those who
Interventions received placebo.
Secondary Outcomes
• Fluvoxamine 100 mg PO twice daily plus inhaled budesonide • The use of fluvoxamine plus inhaled
800 mcg twice daily for 10 days (n = 738) • Hospitalization by Day 28: 0.9% in fluvoxamine
plus budesonide arm vs. 1.1% in placebo arm budesonide did not reduce the risk of
• Placebo (n = 738; route, dosing frequency, and duration may hospitalization, health care attendance, or
have differed from fluvoxamine arm) • Health care attendance by Day 28: 2.6% in ED visits.
fluvoxamine plus budesonide arm vs. 4.1% in
Primary Endpoint placebo arm (relative risk 0.64; 95% CrI, 0.36– • It is difficult to define the clinical
1.11) relevance of the >6-hour ED observation
• Composite of ED observation >6 hours or hospitalization for
endpoint and apply it to practice settings
COVID-19 by Day 28 • Any ED visit by Day 28: 12.2% in fluvoxamine plus in different countries.
Key Secondary Endpoints budesonide arm vs. 13.0% in placebo arm
• More AEs occurred with the use of
• Hospitalization by Day 28 • Occurrence of treatment-emergent AEs: 17.6% fluvoxamine plus inhaled budesonide than
in fluvoxamine plus budesonide arm vs. 12.9% in with placebo.
• Health care attendance by Day 28 placebo arm (relative risk 1.37; 95% CrI, 1.07–
• Any ED visit by Day 28 1.75)
• Occurrence of treatment-emergent AEs • Most AEs were grade 2 events.
Key: AE = adverse event; BMI = body mass index; CVD = cardiovascular disease; CYP = cytochrome P450; DM = diabetes mellitus; ED = emergency department; HTN
= hypertension; IM = intramuscular; MDI = metered dose inhaler; the Panel = the COVID-19 Treatment Guidelines Panel; PCR = polymerase chain reaction; PO = oral;
RCT = randomized controlled trial; RT-PCR = reverse transcription polymerase chain reaction; SSRI = selective serotonin reuptake inhibitor
Recommendations
• See Therapeutic Management of Hospitalized Adults With COVID-19 for the COVID-19
Treatment Guidelines Panel’s (the Panel) recommendations on the use of tocilizumab in
combination with dexamethasone in hospitalized patients who require conventional oxygen,
high-flow nasal cannula (HFNC) oxygen, NIV, or mechanical ventilation.
Additional Considerations
• If none of the recommended immunomodulatory therapies discussed in Therapeutic Management
of Hospitalized Adults With COVID-19 are available or feasible to use, IV sarilumab can be used
in combination with dexamethasone (CIIa). Sarilumab is only commercially available as a SUBQ
injection; see Table 5e for information regarding the preparation of an IV infusion using the SUBQ
product.
• Tocilizumab and sarilumab should be used with caution in patients with COVID-19 who belong
to populations that have not been adequately represented in clinical trials. This includes patients
who are significantly immunosuppressed, such as those who have recently received other biologic
immunomodulators, and patients with any of the following:
• Alanine transaminase levels >5 times the upper limit of normal
• A high risk for gastrointestinal perforation
• An uncontrolled serious bacterial, fungal, or non–SARS-CoV-2 viral infection
• Absolute neutrophil counts <500 cells/µL
• Platelet counts <50,000 cells/µL
Rationale
The results of the RECOVERY and REMAP-CAP trials provide consistent evidence that tocilizumab,
when administered as a second immunomodulatory agent in combination with a corticosteroid, offers
a survival benefit in certain patients with COVID-19.7,8 Specifically, the patients who may benefit are
those who are severely ill and require HFNC oxygen or NIV, those who are rapidly deteriorating and
have increasing oxygen needs, or those who are having a significant inflammatory response. In the
REMAP-CAP trial, a long-term follow-up through 180 days confirmed that treatment with an anti–IL-6
receptor mAb improved survival among patients with severe to critical COVID-19.12 However, the Panel
found it challenging to determine which patients with COVID-19 who are receiving low-flow oxygen
would benefit from receiving tocilizumab or sarilumab plus dexamethasone.
If none of the recommended immunomodulatory therapies are available or feasible to use, sarilumab
may be used because the REMAP-CAP trial demonstrated that the use of tocilizumab and the use of
sarilumab improved survival and reduced the duration of organ support.12,13 Sarilumab is currently only
approved for use in the United States as a SUBQ injection.
Considerations in Children
See Therapeutic Management of Hospitalized Children With COVID-19 for the Panel’s
Drug Availability
On December 21, 2022, the FDA approved the use of IV tocilizumab for the treatment of COVID-19 in
hospitalized adults who are receiving systemic corticosteroids and require supplemental oxygen, NIV,
mechanical ventilation, or ECMO.5 In June 2021, the FDA issued an Emergency Use Authorization
for the use of tocilizumab in combination with corticosteroids for the treatment of COVID-19 in
hospitalized children aged ≥2 years who require supplemental oxygen, NIV, mechanical ventilation,
or ECMO.6 If a patient’s clinical signs or symptoms worsen or do not improve after the first dose of
tocilizumab, 1 additional IV infusion of tocilizumab may be administered at least 8 hours after the initial
infusion.
The IV administration of sarilumab is not approved by the FDA, but in clinical trials, single SUBQ
sarilumab doses were modified to enable IV administration. See Table 5e for additional details.
Clinical Data
Two large randomized controlled trials, REMAP-CAP and RECOVERY, evaluated the use of
tocilizumab in combination with standard of care corticosteroids.7,8 Both studies reported a statistically
significant survival benefit from the use of tocilizumab in certain patients, including patients who
exhibited rapid respiratory decompensation associated with an inflammatory response.
REMAP-CAP enrolled critically ill patients who were within 24 hours of receiving respiratory support
in an intensive care unit.7 At baseline, 29% of these patients were receiving HFNC oxygen, 42% were
receiving NIV, and 29% were receiving mechanical ventilation. The patients were randomized to receive
open-label tocilizumab or usual care. In-hospital mortality was 28% in the tocilizumab arm and 36% in
the usual care arm. A follow-up analysis confirmed these findings.12 At 180 days, mortality was 36% in
the tocilizumab arm and 40% in the usual care arm.
The RECOVERY trial enrolled hospitalized patients with COVID-19 into an open-label platform
trial that included several treatment options.8 A subset of all trial participants who had hypoxemia and
CRP levels ≥75 mg/L were offered enrollment into a second randomization that compared the use of
tocilizumab to usual care. In this subgroup, the 28-day mortality was 31% in the tocilizumab arm and
35% in the usual care arm.
In contrast to the REMAP-CAP and RECOVERY trials, other randomized trials, including the
REMDACTA and EMPACTA trials, found that tocilizumab did not reduce all-cause mortality.18,19 In
those trials, >80% of participants received corticosteroids as part of standard care, and most participants
in the REMDACTA trial required NIV or HFNC oxygen.18
In the REMAP-CAP trial, the efficacy results for sarilumab were similar to those for tocilizumab.13
When compared with patients in the standard of care arm (n = 406), patients in the sarilumab arm
(n = 485) had more organ support-free days (OR 1.50; 95% CrI, 1.13–2.00) and a greater likelihood
of survival while hospitalized (OR 1.51; 95% CrI, 1.06–2.20). In-hospital mortality was 33% for
the sarilumab arm and 37% for the standard of care arm, and mortality at 180 days was 33% for the
sarilumab arm and 40% for the standard of care arm.12 A notable limitation to the sarilumab findings in
the REMAP-CAP trial is that patients in the standard of care arm were enrolled earlier in the pandemic
than those in the sarilumab arm.13 Randomization closed in November 2020 for the standard of care arm
and continued through April 2021 for the sarilumab arm.
An adaptive, multinational, double-blind, randomized (2:2:1) trial compared the efficacy and safety of
sarilumab 400 mg IV and sarilumab 200 mg IV to placebo in hospitalized patients with COVID-19.20
COVID-19 Treatment Guidelines 272
References
1. Yoshikawa T, Hill T, Li K, Peters CJ, Tseng CTK. Severe acute respiratory syndrome (SARS) coronavirus-
induced lung epithelial cytokines exacerbate SARS pathogenesis by modulating intrinsic functions of
monocyte-derived macrophages and dendritic cells. J Virol. 2009;83(7):3039-3048. Available at: https://www.
ncbi.nlm.nih.gov/pubmed/19004938.
2. Zhou F, Yu T, Du R, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in
Wuhan, China: a retrospective cohort study. Lancet. 2020;395(10229):1054-1062. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32171076.
3. Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan,
China. Lancet. 2020;395(10223):497-506. Available at: https://www.ncbi.nlm.nih.gov/pubmed/31986264.
4. Wang Z, Yang B, Li Q, Wen L, Zhang R. Clinical features of 69 cases with coronavirus disease 2019 in
Wuhan, China. Clin Infect Dis. 2020;71(15):769-777. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32176772.
5. Tocilizumab (Actemra) [package insert]. Food and Drug Administration. 2022. Available at:
https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/125276s138lbl.pdf.
6. Food and Drug Administration. Fact sheet for healthcare providers: Emergency Use Authorization for Actemra
(tocilizumab). 2021. Available at: https://www.fda.gov/media/150321/download.
7. REMAP-CAP Investigators. Interleukin-6 receptor antagonists in critically ill patients with COVID-19. N
Engl J Med. 2021;384(16):1491-1502. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33631065.
8. RECOVERY Collaborative Group. Tocilizumab in patients admitted to hospital with COVID-19
(RECOVERY): a randomised, controlled, open-label, platform trial. Lancet. 2021;397(10285):1637-1645.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/33933206.
9. Lier AJ, Tuan JJ, Davis MW, et al. Case report: disseminated strongyloidiasis in a patient with COVID-19. Am
J Trop Med Hyg. 2020;103(4):1590-1592. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32830642.
10. Marchese V, Crosato V, Gulletta M, et al. Strongyloides infection manifested during immunosuppressive
therapy for SARS-CoV-2 pneumonia. Infection. 2021;49(3):539-542. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32910321.
11. Stauffer WM, Alpern JD, Walker PF. COVID-19 and dexamethasone: a potential strategy to avoid steroid-
related Strongyloides hyperinfection. JAMA. 2020;324(7):623-624. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32761166.
12. Writing Committee for the REMAP-CAP Investigators. Long-term (180-day) outcomes in critically ill patients
with COVID-19 in the REMAP-CAP randomized clinical trial. JAMA. 2023;329(1):39-51. Available at:
https://pubmed.ncbi.nlm.nih.gov/36525245.
13. REMAP-CAP Investigators. Effectiveness of tocilizumab, sarilumab, and anakinra for critically ill patients
with COVID-19: the REMAP-CAP COVID-19 immune modulation therapy domain randomized clinical trial.
medRxiv. 2021;Preprint. Available at: https://www.medrxiv.org/content/10.1101/2021.06.18.21259133v2.
COVID-19 Treatment Guidelines 273
RECOVERY: Open-Label RCT of Tocilizumab and Usual Care in Hospitalized Adults With COVID-19 in the United Kingdom1
Key Inclusion Criteria Participant Characteristics Key Limitations
• Evidence of COVID-19 progression ≤21 days • Mean age 64 years; 67% men; 76% White • Arbitrary CRP ≥75 mg/L cutoff for
after initial randomization to an intervention • 95% with PCR-confirmed SARS-CoV-2 infection enrollment
within the RECOVERY protocol, defined as: • Difficult to define exact subset of
• At baseline:
• SpO2 <92% on room air or receipt of patients in RECOVERY cohort who were
supplemental oxygen; and • 45% on conventional oxygen subsequently selected for secondary
• CRP ≥75 mg/L • 41% on HFNC oxygen or NIV randomization/tocilizumab trial
• 14% on MV Interpretation
Key Exclusion Criterion
• 82% receiving corticosteroids • Among hospitalized patients with
• Presence of non-SARS-CoV-2 infection
Primary Outcomes COVID-19, hypoxemia, and elevated
Interventions CRP levels, the use of tocilizumab was
• 28-day all-cause mortality: 31% in tocilizumab arm vs. 35% in usual
• 1 weight-based dose of tocilizumab associated with a reduction in all-cause
care arm (rate ratio 0.85; 95% CI, 0.76–0.94; P = 0.003)
(maximum 800 mg) with possible second mortality and a shorter time to hospital
dose (n = 2,022) • 28-day all-cause mortality among those who required MV at baseline: discharge.
49% in tocilizumab arm vs. 51% in usual care arm (risk ratio 0.93;
• Usual care (n = 2,094) 95% CI, 0.74–1.18)
Primary Endpoint Secondary Outcomes
• 28-day all-cause mortality • Proportion discharged from hospital within 28 days: 57% in
Key Secondary Endpoints tocilizumab arm vs. 50% in usual care arm (rate ratio 1.22; 95% CI,
• Time to hospital discharge within 28 days 1.12–1.33; P < 0.0001)
• Among those not on MV at baseline, death • Median time to hospital discharge: 19 days in tocilizumab arm vs. 28
or receipt of MV (including ECMO) within 28 days in usual care arm
days • Proportion not on MV at baseline who died or required MV within 28
days: 35% in tocilizumab arm vs. 42% in usual care arm (rate ratio
0.84; 95% CI, 0.77–0.92; P < 0.0001)
REMAP-CAP: Open-Label, Adaptive-Platform RCT of Tocilizumab and Sarilumab in Adults With COVID-19 in 21 Countries in Europe and North America2,4
Key Inclusion Criteria Participant Characteristics Key Limitation
• ICU admission • Mean age 60 years; 69% men; 75% White • The SOC arm closed in November 2020,
• Suspected or laboratory-confirmed SARS- • 86% with PCR-confirmed SARS-CoV-2 infection after which patients were randomized
CoV-2 infection to active drug arms only; enrollment in
• Median of 14 hours between ICU admission and enrollment the tocilizumab and sarilumab arms was
• Receipt of MV, NIV, or cardiovascular support • At baseline: partially nonconcurrent with the SOC
Key Exclusion Criteria • 68% on HFNC oxygen or NIV arm. Although comparisons to the SOC
• >24 hours after ICU admission arm were adjusted for this time period,
• 32% on MV
there is a possibility of bias.
• Death imminent • Receiving corticosteroids: 67% in SOC arm, 82% in tocilizumab
arm, 89% in sarilumab arm Interpretation
• Immunosuppression
• Among patients with respiratory
• ALT >5 times ULN Primary Outcomes
failure who were within 24 hours of
Interventions Tocilizumab vs. SOC ICU admission, the tocilizumab and
• SOC plus 1 of the following (drug selection • Median number of organ support-free days: 7 in tocilizumab arm vs. 0 sarilumab arms had higher rates of in-
based on provider preference, availability, or in SOC arm hospital survival and shorter durations of
adaptive probability): • Improvement in OS score by Day 21 for composite outcome was more organ support than the SOC arm.
• 1 dose of tocilizumab 8 mg/kg IV with likely in tocilizumab arm (median aOR 1.46; 95% CrI, 1.13–1.87). • The use of IL-6 receptor antagonists
possible second dose in 12–24 hours (n • Highest CRP tercile: aOR 1.87 (95% CrI, 1.35–2.59) reduced all-cause mortality at 180 days.
= 952) • Outcomes were consistent across subgroups according to oxygen • The treatment effect appeared to be
• Single dose of sarilumab 400 mg IV (n = requirement at baseline. strongest in the highest CRP tercile.
485) • Tocilizumab and sarilumab were
Sarilumab vs. SOC similarly effective in these patients.
• SOC alone (n = 406)
• Median number of organ support-free days: 9 in sarilumab arm vs. 0
Primary Endpoint in SOC arm
• Composite of in-hospital mortality or • Improvement in OS score by Day 21 for composite outcome was more
number of organ support-free days by Day likely in sarilumab arm (median aOR 1.50; 95% CrI, 1.13–2.00).
21, as measured by an OS • Highest CRP tercile: aOR 1.85 (95% CrI, 1.24–2.69)
Key Secondary Endpoints • Outcomes were consistent across subgroups according to oxygen
• In-hospital survival requirement at baseline.
• All-cause mortality at 180 days Secondary Outcomes
Tocilizumab vs. SOC
• In-hospital survival: 66% in tocilizumab arm vs. 63% in SOC arm (aOR
1.42; 95% CrI, 1.05–1.93)
EMPACTA: Double-Blind RCT of Tocilizumab in Hospitalized Adults With COVID-19 in 6 Countries in North America, South America, and Africa6
Key Inclusion Criteria Participant Characteristics Key Limitation
• PCR-confirmed SARS-CoV-2 infection • Mean age 56 years; 59% men; 56% Hispanic/Latinx, 15% Black/ • Moderate sample size
• COVID-19 pneumonia African American, 13% American Indian/Alaska Native
Interpretation
• 84% with elevated CRP
Key Exclusion Criteria • In patients with COVID-19 pneumonia,
• Concomitant medications: tocilizumab reduced the likelihood of
• Death imminent
• Corticosteroids: 80% in tocilizumab arm vs. 88% in placebo arm progression to MV, ECMO, or death by
• Receiving NIV or MV Day 28 but did not reduce all-cause
• RDV: 53% in tocilizumab arm vs. 59% in placebo arm
Interventions mortality by Day 28.
Primary Outcome
• 1 dose of tocilizumab 8 mg/kg with possible
second dose, plus SOC (n = 249) • Proportion who progressed to MV, ECMO, or death by Day 28: 12%
in tocilizumab arm vs. 19% in placebo arm (HR 0.56; 95% CI, 0.33–
• Placebo plus SOC (n = 128)
0.97; P = 0.04)
Primary Endpoint
Secondary Outcomes
• Progression to MV, ECMO, or death by Day
• Median time to hospital discharge or readiness for discharge: 6.0
28
days in tocilizumab arm vs. 7.5 days in placebo arm (HR 1.16; 95%
Key Secondary Endpoints CI, 0.91–1.48)
• Time to hospital discharge or readiness for • All-cause mortality by Day 28: 10.4% in tocilizumab arm vs. 8.6% in
discharge, as measured by an OS placebo arm (95% CI, -5.2 to 7.8)
• All-cause mortality by Day 28
BACC Bay: Double-Blind RCT of Tocilizumab in Hospitalized Adults With COVID-19 in the United States7
Key Inclusion Criteria Participant Characteristics Key Limitations
• Laboratory-confirmed SARS-CoV-2 infection • Median age 60 years; 58% men; 45% Hispanic/Latinx, 43% White • Wide confidence intervals due to small
• ≥2 of the following conditions: • 50% with BMI ≥30; 49% with HTN; 31% with DM sample size and low event rates
• Fever >38°C • 80% receiving oxygen ≤6 L/min; 4% on HFNC oxygen; 16% receiving • Few patients received RDV or
no supplemental oxygen corticosteroids.
• Pulmonary infiltrates
• Need for supplemental oxygen • Concomitant medications: Interpretation
• ≥1 of the following laboratory criteria: • Corticosteroids: 11% in tocilizumab arm vs. 6% in placebo arm • The use of tocilizumab did not prevent
• RDV: 33% in tocilizumab arm vs. 29% in placebo arm MV or death, reduce the risk of clinical
• CRP ≥50 mg/L worsening, or reduce the time to
• D-dimer >1,000 ng/mL Primary Outcome discontinuation of oxygen. This could
• LDH ≥250 U/L • Progression to MV or death by Day 28: 11% in tocilizumab arm vs. be due to the low rate of concomitant
12% in placebo arm (HR 0.83; 95% CI, 0.38–1.81; P = 0.64) corticosteroid use among the study
• Ferritin >500 ng/mL participants.
Key Exclusion Criteria Secondary Outcomes
• Receipt of supplemental oxygen at rate >10 • Proportion with clinical worsening of disease by Day 28: 19% in
L/min tocilizumab arm vs. 17% in placebo arm (HR 1.11; 95% CI, 0.59–
2.10; P = 0.73)
• Recent use of biologic agents or small-
molecule immunosuppressive therapy • Median time to discontinuation of supplemental oxygen: 5.0 days in
tocilizumab arm vs. 4.9 days in placebo arm (P = 0.69)
• Receipt of immunosuppressive therapy that
increased risk for infection
Interventions
• Tocilizumab 8 mg/kg plus usual care (n =
161)
• Placebo plus usual care (n = 81)
Primary Endpoint
• Progression to MV or death by Day 28
Key Secondary Endpoints
• Clinical worsening of disease by Day 28, as
measured by an OS
• Discontinuation of supplemental oxygen
among patients receiving it at baseline
Double-Blind RCT of Sarilumab in Hospitalized Adults With Severe to Critical COVID-19 in 11 Countries in Europe, North America, South America, and Asia8
Key Inclusion Criteria Participant Characteristics Key Limitation
• COVID-19 pneumonia • Median age 59 years; 63% men; 77% White, 36% Hispanic/Latinx • Moderate sample size
• Need for supplemental oxygen or intensive • 39% on HFNC oxygen, MV, or NIV Interpretation
care • 42% with BMI ≥30; 43% with HTN; 26% with type 2 DM • The use of sarilumab did not reduce
Key Exclusion Criteria • 20% received systemic corticosteroids before receiving intervention; mortality or time to clinical improvement
• Low probability of surviving or remaining at 63% received ≥1 doses of corticosteroids during the study. in hospitalized adults with COVID-19.
study site Primary Outcomes
• Dysfunction of ≥2 organ systems and need • Median time to clinical improvement: 10 days in sarilumab 200 mg
for ECMO or renal replacement therapy arm vs. 10 days in sarilumab 400 mg arm vs. 12 days in placebo arm
Interventions • Sarilumab 200 mg arm vs. placebo arm: HR 1.03; 95% CI, 0.75–
• Sarilumab 200 mg IV (n = 159) 1.40; P = 0.96
• Sarilumab 400 mg IV (n = 173) • Sarilumab 400 mg arm vs. placebo arm: HR 1.14; 95% CI, 0.84–
1.54; P = 0.34
• Placebo (n = 84)
Secondary Outcome
Primary Endpoint
• Survival to Day 29: 92% in placebo arm; 90% in sarilumab 200 mg
• Time to clinical improvement of ≥2 points
arm (P = 0.63 vs. placebo); 92% in sarilumab 400 mg arm (P = 0.85
on a 7-point OS
vs. placebo)
Key Secondary Endpoint
• Survival to Day 29
Janus kinase (JAK) inhibitors interfere with phosphorylation of the signal transducer and activator of
transcription (STAT) proteins1,2 that are involved in vital cellular functions, including signaling, growth,
and survival. JAK inhibitors are used to treat COVID-19 because they can prevent phosphorylation of
key proteins involved in the signal transduction that leads to immune activation and inflammation (e.g.,
the cellular response to proinflammatory cytokines such as interleukin [IL]-6).3 Several JAK inhibitors
are available for clinical use, but only baricitinib and tofacitinib have been studied for the treatment of
COVID-19.
In May 2022, the Food and Drug Administration (FDA) approved the use of baricitinib for the treatment
of COVID-19 in hospitalized adults who require supplemental oxygen, noninvasive ventilation (NIV),
mechanical ventilation, or extracorporeal membrane oxygenation.4 Baricitinib is an oral JAK inhibitor
that is selective for JAK1 and JAK2. It can modulate downstream inflammatory responses via JAK1/
JAK2 inhibition and has exhibited dose-dependent inhibition of IL-6–induced STAT3 phosphorylation.5
Tofacitinib is a JAK inhibitor that is predominantly selective for JAK1 and JAK3. However, it also has
modest activity against JAK2; thus, it can block signaling from gamma-chain cytokines (e.g., IL-2,
IL-4) and glycoprotein 130 proteins (e.g., IL-6, IL-11, interferons). It is an oral agent that was first
approved by the FDA for the treatment of rheumatoid arthritis and has been shown to decrease levels of
IL-6 in patients with this disease.6 Tofacitinib is also approved by the FDA for the treatment of psoriatic
arthritis, juvenile idiopathic arthritis, and ulcerative colitis.7
Recommendation
• See Therapeutic Management of Hospitalized Adults With COVID-19 for the COVID-19
Treatment Guidelines Panel’s (the Panel) recommendations on the use of baricitinib in
combination with dexamethasone in hospitalized patients who require conventional oxygen,
high-flow nasal cannula oxygen, NIV, or mechanical ventilation.
Additional Consideration
• If none of the recommended immunomodulatory therapies discussed in Therapeutic Management
of Hospitalized Adults With COVID-19 are available or feasible to use, oral tofacitinib can be
used in combination with dexamethasone (CIIa).
Rationale
Several large randomized controlled trials have demonstrated that some patients who require
supplemental oxygen and most patients who require oxygen through a high-flow device, NIV, or
mechanical ventilation benefit from the use of dexamethasone in combination with a JAK inhibitor.
In the RECOVERY trial, baricitinib was associated with a survival benefit among hospitalized patients,
with a treatment effect that was most pronounced among patients who were receiving NIV or oxygen
supplementation through a high-flow device.8 The COV-BARRIER trial also demonstrated a survival
benefit for baricitinib that was most pronounced among patients who were receiving high-flow oxygen
or NIV.9 In the addendum to the COV-BARRIER trial, the benefit extended to patients who were
receiving mechanical ventilation.10 Data from the ACTT-211 and ACTT-412 trials support the overall
Considerations in Children
See Therapeutic Management of Hospitalized Children With COVID-19 for the Panel’s
recommendations regarding the use of baricitinib or tofacitinib in children with COVID-19.
References
1. Babon JJ, Lucet IS, Murphy JM, Nicola NA, Varghese LN. The molecular regulation of Janus kinase (JAK)
activation. Biochem J. 2014;462(1):1-13. Available at: https://www.ncbi.nlm.nih.gov/pubmed/25057888.
2. Bousoik E, Montazeri Aliabadi H. “Do we know jack” about JAK? A closer look at JAK/STAT signaling
pathway. Front Oncol. 2018;8:287. Available at: https://www.ncbi.nlm.nih.gov/pubmed/30109213.
3. Zhang W, Zhao Y, Zhang F, et al. The use of anti-inflammatory drugs in the treatment of people with severe
The clinical trials described in this table do not represent all the trials that the Panel reviewed while developing the recommendations for JAK
inhibitors. The studies summarized below are those that have had the greatest impact on the Panel’s recommendations.
STOP-COVID: Double-Blind, Placebo-Controlled, Randomized Trial of Tofacitinib in Hospitalized Patients With COVID-19 Pneumonia in Brazil6
Key Inclusion Criteria Participant Characteristics Key Limitations
• Laboratory-confirmed SARS-CoV-2 infection • Mean age 56 years; 35% women • Small sample size
• COVID-19 pneumonia on CXR or CT • Median of 10 days from symptom onset to randomization • RDV not available during trial
• Hospitalized for <72 hours • At baseline: Interpretation
Key Exclusion Criteria • 75% on supplemental oxygen
• When compared with placebo, use of
• Receiving NIV, MV, or ECMO at baseline • 13% on HFNC oxygen tofacitinib led to a lower risk of mortality
• History of or current thrombosis • Use of glucocorticoids: 79% at baseline, 89% during hospitalization or respiratory failure among hospitalized
adults with COVID-19 pneumonia, most
• Immunosuppression or active cancer Primary Outcome
of whom received glucocorticoids.
treatment • Mortality or respiratory failure by Day 28: 18% in tofacitinib arm vs.
Interventions 29% in placebo arm (risk ratio 0.63; 95% CI, 0.41–0.97; P = 0.04)
• Tofacitinib 10 mg PO twice daily for up to 14 Secondary Outcome
days or until hospital discharge (n = 144) • Mortality by Day 28: 2.8% in tofacitinib arm vs. 5.5% in placebo arm
• Placebo (n = 145) (HR 0.49; 95% CI, 0.15–1.63)
Primary Endpoint
• Mortality or respiratory failure by Day 28
Key Secondary Endpoint
• Mortality by Day 28
Key: AE = adverse event; ALC = absolute lymphocyte count; ALT = alanine transaminase; ANC = absolute neutrophil count; AST = aspartate aminotransferase;
BAR = baricitinib; CCP = COVID-19 convalescent plasma; CRP = C-reactive protein; CT = computed tomography; CXR = chest X-ray; DEX = dexamethasone;
ECMO = extracorporeal membrane oxygenation; eGFR = estimated glomerular filtration rate; HFNC = high-flow nasal cannula; IV = intravenous; IVIG = intravenous
immunoglobulin; JAK = Janus kinase; LDH = lactate dehydrogenase; MV = mechanical ventilation; NIV = noninvasive ventilation; OS = ordinal scale; the Panel = the
COVID-19 Treatment Guidelines Panel; PCR = polymerase chain reaction; PO = oral; RCT = randomized controlled trial; RDV = remdesivir; SAE = serious adverse event;
SOC = standard of care; SpO2 = oxygen saturation; TB = tuberculosis; ULN = upper limit of normal
References
1. RECOVERY Collaborative Group. Baricitinib in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label,
platform trial and updated meta-analysis. Lancet. 2022;400(10349):359-368. Available at: https://pubmed.ncbi.nlm.nih.gov/35908569.
2. Marconi VC, Ramanan AV, de Bono S, et al. Efficacy and safety of baricitinib for the treatment of hospitalised adults with COVID-19 (COV-
BARRIER): a randomised, double-blind, parallel-group, placebo-controlled Phase 3 trial. Lancet Respir Med. 2021;9(12):1407-1418. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34480861.
Recommendation
• See Therapeutic Management of Hospitalized Adults With COVID-19 for the COVID-19 Treatment
Guidelines Panel’s (the Panel) recommendations on the use of abatacept in hospitalized patients
who require conventional oxygen, high-flow nasal cannula (HFNC) oxygen, or noninvasive
ventilation (NIV).
Rationale
The ACTIV-1 immunomodulator trial was a double-blind, multi-arm, randomized trial in moderately to
severely ill adults who were hospitalized with COVID-19.6 The trial separately evaluated treatment with
abatacept, cenicriviroc, and infliximab versus placebo. All arms received standard care, and the separate
analyses included data from a shared placebo arm. One substudy compared the use of a single dose of
intravenous abatacept 10 mg/kg to placebo. The primary endpoint was time to recovery by Day 28. Key
secondary endpoints included clinical status at Day 14 and mortality through Days 28 and 60.
The study concluded that use of abatacept in patients with COVID-19 did not have a significant effect on the
time to recovery. A reduction in 28-day mortality, a secondary endpoint, was found. Patients who required
mechanical ventilation or extracorporeal membrane oxygenation did not benefit from the use of abatacept.
Considerations in Children
The intravenous formulation of abatacept is approved by the Food and Drug Administration for the
treatment of juvenile idiopathic arthritis and acute graft-versus-host disease in children aged ≥2 years.
It is not approved for the treatment of COVID-19 in children, and there are no published reports on the
efficacy of using abatacept in this population. No patients aged <18 years were included in the ACTIV-1
trial.
Clinical Data
In the ACTIV-1 trial, the modified intention-to-treat analysis for the abatacept substudy included
509 patients in the abatacept arm and 510 patients in the placebo arm. At baseline, 53% of patients
required conventional oxygen supplementation, and 33% required HFNC oxygen or NIV. As part of
their standard care before or during the study, 93% of patients received remdesivir, and 91% received
corticosteroids.
Results
• The median time to recovery was 9 days in both the infliximab and placebo arms (recovery rate
ratio 1.12; 95% CI, 0.98–1.28; P = 0.09), and there was no differential effect across subgroups
based on disease severity (interaction P = 0.66).
• Mortality by Day 28 was lower among patients who received abatacept (56 of 509 patients
[11.0%]) than among those who received placebo (77 of 510 patients [15.1%]; OR 0.62; 95% CI,
0.41–0.94).
• Subgroup analyses showed reduced mortality only among patients in the abatacept arm who
required HFNC oxygen or NIV (OR 0.48; 95% CI, 0.28–0.84).
• Among patients who required mechanical ventilation or extracorporeal membrane oxygenation,
there was no difference in mortality by Day 28 (OR 1.63; 95% CI, 0.66–4.05).
• There were no differences in secondary infections or in the number or severity of serious adverse
events between the abatacept and placebo arms.
Limitations
• Each of the 3 active agents was compared to a shared placebo group without adjusting for multiple
comparisons.
• Mortality was a secondary endpoint. Although the treatment difference for mortality by Day 28
was nominally significant, no adjustment was made to the analysis to assess multiple outcomes
(primary outcome and mortality).
• The study was not powered to analyze differences within disease severity subgroups.
References
1. Alegre ML, Frauwirth KA, Thompson CB. T-cell regulation by CD28 and CTLA-4. Nat Rev Immunol.
2001;1(3):220-228. Available at: https://pubmed.ncbi.nlm.nih.gov/11905831.
2. Oyewole-Said D, Konduri V, Vazquez-Perez J, Weldon SA, Levitt JM, Decker WK. Beyond T-cells:
functional characterization of CTLA-4 expression in immune and non-immune cell types. Front Immunol.
2020;11:608024. Available at: https://pubmed.ncbi.nlm.nih.gov/33384695.
3. Chen Z, Wherry EJ. T cell responses in patients with COVID-19. Nat Rev Immunol. 2020;20:529-536.
Recommendation
• See Therapeutic Management of Hospitalized Adults With COVID-19 for the COVID-19
Treatment Guidelines Panel’s (the Panel) recommendations on the use of infliximab in
hospitalized patients who require conventional oxygen, high-flow nasal cannula (HFNC) oxygen,
or noninvasive ventilation (NIV).
Rationale
The ACTIV-1 immunomodulator trial was a double-blind, multi-arm, randomized trial in moderately to
severely ill adults who were hospitalized with COVID-19.4 The trial separately evaluated treatment with
abatacept, cenicriviroc, and infliximab versus placebo. All arms received standard care, and the separate
analyses included data from a shared placebo arm. One substudy compared the use of a single dose of
intravenous infliximab 5 mg/kg to placebo. The primary endpoint was time to recovery by Day 28. Key
secondary endpoints included clinical status at Day 14 and mortality through Days 28 and 60.
The study concluded that use of infliximab in patients with COVID-19 did not have a significant effect
on the time to recovery. A reduction in 28-day mortality, a secondary endpoint, was found. Patients who
required mechanical ventilation or extracorporeal membrane oxygenation did not benefit from the use of
infliximab.
Considerations in Children
Infliximab is approved for the treatment of inflammatory bowel disease in children and is often used
to treat juvenile idiopathic arthritis. The Food and Drug Administration has not approved the use of
infliximab for the treatment of COVID-19 in children, and there are no published reports on the efficacy
of using infliximab in this population. No patients aged <18 years were included in the ACTIV-1 trial.
See Therapeutic Management of Hospitalized Children With MIS-C, Plus a Discussion on MIS-A for
the Panel’s recommendations regarding the use of infliximab in pediatric patients with multisystem
inflammatory syndrome in children (MIS-C).
Clinical Data
In the ACTIV-1 trial, the modified intention-to-treat analysis for the infliximab substudy included
517 patients in the infliximab arm and 516 patients in the placebo arm. At baseline, 52% of patients
required conventional oxygen supplementation, and 33% required HFNC oxygen or NIV. As part of
their standard care before or during the study, 93% of patients received remdesivir, and 92% received
corticosteroids.
Results
• The median time to recovery was 8 days in the infliximab arm versus 9 days in the placebo arm
(recovery rate ratio 1.12; 95% CI, 0.99–1.28; P = 0.08), and there was no differential effect across
subgroups based on disease severity (interaction P = 0.36).
• Mortality by Day 28 was lower among patients who received infliximab (52 of 517 patients
[10.1%]) than among those who received placebo (75 of 516 patients [14%]; OR 0.59; 95% CI,
0.39–0.90).
• Subgroup analyses showed reduced mortality only among patients in the infliximab arm who
required HFNC oxygen or NIV (OR 0.52; 95% CI, 0.29–0.91).
• Among patients who required mechanical ventilation or extracorporeal membrane oxygenation,
there was no difference in mortality by Day 28 (OR 1.11; 95% CI, 0.45–2.72).
• There were no differences in secondary infections or in the number or severity of serious adverse
events between the infliximab and placebo arms.
Limitations
• Each of the 3 active agents was compared to a shared placebo group without adjusting for multiple
comparisons.
• Mortality was a secondary endpoint. Although the treatment difference for mortality by Day 28
was nominally significant, no adjustment was made to the analysis to assess multiple outcomes
(primary outcome and mortality).
• The study was not powered to analyze differences within disease severity subgroups.
References
1. Del Valle DM, Kim-Schulze S, Huang HH, et al. An inflammatory cytokine signature predicts COVID-19
severity and survival. Nat Med. 2020;26(10):1636-1643. Available at: https://pubmed.ncbi.nlm.nih.
gov/32839624.
2. Curtis JR, Zhou X, Rubin DT, et al. Characteristics, comorbidities, and outcomes of SARS-CoV-2 infection in
COVID-19 Treatment Guidelines 297
Recommendations
• There is insufficient evidence for the COVID-19 Treatment Guidelines Panel (the Panel) to
recommend either for or against the use of anakinra for the treatment of COVID-19.
• The Panel recommends against the use of canakinumab for the treatment of COVID-19, except
in a clinical trial (BIIa).
Rationale
In the SAVE-MORE trial, 594 hospitalized patients who had moderate or severe COVID-19 pneumonia
and plasma suPAR levels ≥6 ng/mL were randomized to receive either anakinra or placebo.7 The study
found that patients who received anakinra had a lower risk of clinical progression of COVID-19 than
those who received placebo. REMAP-CAP, an open-label, adaptive platform trial that evaluated the use
of several immunomodulators in patients with COVID-19 who required organ support, found no clinical
benefit of anakinra in these patients. In addition, among patients who received anakinra, no reduction
in mortality was observed during a 180-day follow up.8 Several smaller trials that evaluated the use of
anakinra in people with COVID-19 were either stopped early due to futility or did not detect a benefit
of anakinra in these patients.9-11 Meta-analyses of the available data have not detected a benefit of using
anakinra to treat COVID-19.12,13
The SAVE-MORE study population was restricted to participants with high levels of suPAR (≥6 ng/
mL) based on the hypothesis that this group is most likely to benefit from IL-1 inhibition. However,
the laboratory assay that is used to assess suPAR levels is not currently available in many countries,
including the United States. Using data from the SAVE-MORE and SAVE trials (both a priori,
open-label, single-arm prospective studies), the FDA developed a scoring system that uses common
COVID-19 Treatment Guidelines 299
Considerations in Children
Anakinra has been used to treat severely ill children with rheumatologic conditions, including systemic
juvenile idiopathic arthritis and macrophage activation syndrome. The data on the use of anakinra in
pediatric patients with acute respiratory distress syndrome or sepsis are limited. Information on the
use of anakinra in pediatric patients with acute COVID-19 is limited to small case series.22,23 However,
anakinra has been used in approximately 10% of cases of multisystem inflammatory syndrome in
children (MIS-C).24-29 Anakinra is often included in institutional protocols for the treatment of MIS-C in
the United States, and it is an option for second-line therapy for refractory MIS-C in national consensus
guidelines, including the COVID-19 Treatment Guidelines.30-32 For more information, see Therapeutic
Management of Hospitalized Children With MIS-C, Plus a Discussion on MIS-A.
Data on using canakinumab in pediatric patients are limited to use in patients with periodic fever
syndromes and systemic juvenile idiopathic arthritis. There are no data on its use in pediatric patients
COVID-19 Treatment Guidelines 300
Clinical Data
SAVE-MORE
SAVE-MORE was a randomized controlled trial in 594 hospitalized patients with moderate to severe
COVID-19 pneumonia and plasma suPAR levels ≥6 ng/mL.7 Patients who required noninvasive
ventilation (NIV) or mechanical ventilation were excluded from the study. Patients were randomized
2:1 to receive anakinra 100 mg subcutaneously once daily for 10 days or placebo. The primary endpoint
was clinical status at Day 28 on the 11-point World Health Organization Clinical Progression Scale
(WHO-CPS). Additional analyses assessed outcomes at Days 60 and 90.33
Results
• Patients who were randomized to receive anakinra had lower odds of a worse WHO-CPS score at
Day 28 (OR 0.36; 95% CI, 0.26–0.50; P < 0.0001).
• The secondary endpoints also favored anakinra, including the absolute decrease in WHO-CPS
scores from baseline at Days 14 and 28, the absolute decrease in sequential organ failure
assessment (SOFA) scores from baseline at Day 7, the median time to hospital discharge, and the
median duration of ICU stays.
• A smaller proportion of patients in the anakinra arm experienced secondary infections, including
ventilator-associated pneumonias, than in the placebo arm (8.4% vs. 15.9%; P = 0.01).
• Twenty-eight–day mortality was lower among patients who received anakinra than among those
who received placebo (3.2% vs. 6.9%; HR 0.45; 95% CI, 0.21–0.98; P = 0.045).
• Additional analyses performed at Days 60 and 90 showed a sustained survival benefit for
anakinra.
Limitations
The laboratory assay that is used to assess suPAR levels is not currently available in many countries,
including the United States. The FDA worked with the SAVE-MORE investigators to develop a scoring
system that predicts whether a patient has suPAR levels ≥6 ng/mL using baseline data from patients who
were randomized during the trial and a subset of patients who were screened but not randomized. The
FDA’s surrogate for suPAR levels ≥6 ng/mL is called SCORE 2, and it includes the following patient
characteristics:
• Aged ≥75 years
• Severe pneumonia, as determined by WHO criteria
• Current or past smoker
• SOFA score ≥3
• Neutrophil to lymphocyte ratio ≥7
• Hemoglobin ≤10.5 g/dL
• Medical history of ischemic stroke
• Blood urea ≥50 mg/dL and/or medical history of renal disease
Patients who met ≥3 of these criteria were considered positive for SCORE 2 and likely to have a suPAR
level ≥6 ng/mL. SCORE 2 had a positive predictive value of 0.95, a sensitivity of 0.41, and specificity
of 0.96 when retrospectively applied to the SAVE-MORE trial, and it had similar characteristics
when applied to the SAVE trial, an open-label, single-arm prospective study that served as an external
COVID-19 Treatment Guidelines 301
REMAP-CAP
The REMAP-CAP trial is an open-label, adaptive platform trial in which eligible participants are
randomized to several domains, including the Immune Modulation Therapy domain, which consists of 2
IL-6 inhibitors, anakinra, interferon beta-1a, and a control group.8 Participants are eligible for enrollment
if they are within 24 hours of receiving respiratory or cardiovascular organ support in the ICU and they
have suspected or microbiologically confirmed COVID-19. This population had more advanced disease
than the population enrolled in the SAVE-MORE trial.
Anakinra 300 mg was given intravenously (IV) as a loading dose, followed by anakinra 100 mg IV
every 6 hours for 14 days until patients were either free from mechanical ventilation for >24 hours
or discharged from the ICU. The primary outcome was measured using an ordinal scale that included
a composite of in-hospital mortality and duration of respiratory and cardiovascular organ support at
21 days; all deaths up to 90 days were assigned the worst outcome. The trial used a Bayesian design
that allowed the authors to compare nonconcurrently randomized interventions across time periods.
Additional analyses assessed outcomes at 180 days.3,8
Results
• Of the 2,274 participants who were randomized to 1 of the arms in the Immune Modulation
Therapy domain, 365 individuals were assigned to receive anakinra and included in the analysis,
406 were assigned to the usual care (control) arm, 943 were assigned to receive tocilizumab, and
483 were assigned to receive sarilumab.
• Of those assigned to receive anakinra, 37% were receiving mechanical ventilation at study entry
compared with 32% of patients in the other arms. The other patients received oxygen through a
high-flow nasal cannula or NIV, with a few exceptions.
• The median number of organ support-free days was similar for patients who received anakinra and
those who received usual care (0 days [IQR -1 to 15 days] vs. 0 days [IQR -1 to 15 days]). The
aOR for organ support-free days was 0.99 for anakinra (95% CrI, 0.74–1.35), with a 47% posterior
probability of superiority to control. Sixty percent of those who were assigned to receive anakinra
survived compared with 63% of those who were assigned to the control arm, with a 44% posterior
probability that anakinra was superior to usual care.
• Additional analyses performed at 180 days showed no reduction in mortality among patients who
received anakinra.3,8
• The risk of experiencing serious adverse events was similar between the arms.
Limitations
Patients were not randomized contemporaneously to receive anakinra or usual care; the treatment
effect was estimated from an overarching model that mostly included patients who were randomized to
receive an IL-6 inhibitor (tocilizumab or sarilumab) or usual care, and patients who were randomized to
receive an IL-6 inhibitor or anakinra. Thus, the estimate of the treatment effect is not fully protected by
randomization. This study also had an open-label design.
CORIMUNO-ANA-1
The CORIMUNO-ANA-1 trial randomized 116 hospitalized patients with COVID-19 pneumonia 1:1
to receive either usual care plus anakinra (200 mg IV twice daily on Days 1–3, 100 mg IV twice on Day
COVID-19 Treatment Guidelines 302
COV-AID
The COV-AID trial enrolled 342 hospitalized patients with COVID-19, hypoxia, and signs of
hyperinflammation.10 This trial had an open-label, 2 x 2 factorial design to compare IL-1 inhibition
to no IL-1 inhibition and IL-6 inhibition to no IL-6 inhibition. The primary outcome was the time to
clinical improvement, which was defined as an increase of 2 or more points on a 6-point ordinal scale or
discharge from the hospital.
Results
• There was no difference between the anakinra arm and the usual care arm in the occurrence of
the primary outcome. The estimated median time to clinical improvement was 12 days (95% CI,
10–16 days) in the anakinra arm and 12 days (95% CI, 10–15 days) in the usual care arm (HR
0.94; 95% CI, 0.73–1.21).
• Fifty-five patients died during the study, and no statistically significant differences in mortality
were found between the study arms.
• The risk of experiencing serious adverse events was similar between the arms.
Limitations
The limitations of this study include the open-label design and the fact that many patients did not
receive current standard of care therapy (e.g., corticosteroids, remdesivir). In addition, the 2 x 2 factorial
structure was underpowered to detect interactions between treatment arms, because some patients
received both an IL-1 inhibitor and an IL-6 inhibitor.
ANA-COVID-GEAS
ANA-COVID-GEAS was a multicenter, randomized, open-label, Phase 2/3 clinical trial of 179
hospitalized patients with severe COVID-19 pneumonia and hyperinflammation.11 Patients were
COVID-19 Treatment Guidelines 303
CAN-COVID
CAN-COVID was a double-blind, placebo-controlled randomized trial of 454 hospitalized patients with
COVID-19 who were hypoxemic but not mechanically ventilated and had elevated levels of C-reactive
protein (≥20 mg/L) or ferritin (≥600 µg/L).14 Patients were randomized 1:1 to receive a single dose of
IV canakinumab (450 mg for a body weight of 40 kg to <60 kg, 600 mg for 60–80 kg, and 750 mg for
>80 kg) or placebo. The primary outcome was survival without the need for mechanical ventilation from
Days 3 through Day 29.
Results
• There was no statistical difference between the canakinumab arm and placebo arm in the
proportion of patients who survived without mechanical ventilation (88.8% vs. 85.7%; P = 0.29).
• The number of COVID-19–related deaths at 4 weeks was similar for the 2 arms (11 of 223 patients
[4.9%] in the canakinumab arm vs. 16 of 222 patients [7.2%] in the placebo arm; OR 0.67; 95%
CI, 0.30–1.50).
• Forty-one percent of patients in the canakinumab arm and 32% in the placebo arm received
dexamethasone.
• Serious adverse events occurred in 16% of patients who received canakinumab and in 21% of
patients who received placebo.
Limitations
The use of corticosteroids was unbalanced in this study, with more patients receiving dexamethasone at
baseline in the canakinumab arm than in the placebo arm. More patients received dexamethasone after
the trial was underway in the placebo arm than in the canakinumab arm (22.5% vs. 14.5%), and more
patients received tocilizumab in the placebo arm than in the canakinumab arm (8.8% vs. 2.2%).
CanCovDia
CanCovDia was a double-blind, placebo-controlled, randomized trial of 116 hospitalized patients with
type 2 diabetes, a body mass index >25, and COVID-19.15 Most patients (65.8%) required oxygen
supplementation. Patients were randomized 1:1 to receive a single dose of canakinumab (using weight-
adapted dosing between 450 and 750 mg) or placebo. The primary outcome was reported as a win
COVID-19 Treatment Guidelines 304
References
1. Shakoory B, Carcillo JA, Chatham WW, et al. Interleukin-1 receptor blockade is associated with reduced
mortality in sepsis patients with features of macrophage activation syndrome: reanalysis of a prior Phase III
trial. Crit Care Med. 2016;44(2):275-281. Available at: https://www.ncbi.nlm.nih.gov/pubmed/26584195.
2. Monteagudo LA, Boothby A, Gertner E. Continuous intravenous anakinra infusion to calm the cytokine storm
in macrophage activation syndrome. ACR Open Rheumatol. 2020;2(5):276-282. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32267081.
3. Writing Committee for the REMAP-CAP Investigators. Long-term (180-day) outcomes in critically ill patients
with COVID-19 in the REMAP-CAP randomized clinical trial. JAMA. 2023;329(1):39-51. Available at:
https://pubmed.ncbi.nlm.nih.gov/36525245.
4. Anakinra (Kineret) [package insert]. Food and Drug Administration. 2020. Available at: https://www.
accessdata.fda.gov/drugsatfda_docs/label/2020/103950s5189lbl.pdf.
5. Food and Drug Administration. Fact sheet for healthcare providers: Emergency Use Authorization for Kineret.
2022. Available at: https://www.fda.gov/media/163075/download.
6. Canakinumab (Ilaris) [package insert]. Food and Drug Administration. 2020. Available at: https://www.
accessdata.fda.gov/drugsatfda_docs/label/2020/125319s100lbl.pdf.
7. Kyriazopoulou E, Poulakou G, Milionis H, et al. Early treatment of COVID-19 with anakinra guided by
soluble urokinase plasminogen receptor plasma levels: a double-blind, randomized controlled Phase 3 trial.
Nat Med. 2021;27(10):1752-1760. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34480127.
8. REMAP-CAP Investigators. Effectiveness of tocilizumab, sarilumab, and anakinra for critically ill patients
with COVID-19: the REMAP-CAP COVID-19 immune modulation therapy domain randomized clinical trial.
medRxiv. 2021;Preprint. Available at: https://www.medrxiv.org/content/10.1101/2021.06.18.21259133v2.
9. CORIMUNO-19 Collaborative Group. Effect of anakinra versus usual care in adults in hospital with
Vilobelimab is an anti-C5a monoclonal antibody. High concentrations of C5a have been reported
in patients with severe COVID-19.1 C5a activates innate immune system responses, including
inflammation and the release of histamines, and can increase damage to local tissues.2 A study in mice
demonstrated that an anti-C5a monoclonal antibody reduced immune system activation and inhibited
lung injury.3 Vilobelimab targets C5a, which is a product of complement activation, and preserves
membrane attack complex function.4 Vilobelimab is not approved by the Food and Drug Administration
(FDA) for any indication.
On April 4, 2023, the FDA issued an Emergency Use Authorization for the use of vilobelimab for the
treatment of COVID-19 in hospitalized adults when it is administered within 48 hours of mechanical
ventilation or extracorporeal membrane oxygenation.5
Recommendation
• There is insufficient evidence for the COVID-19 Treatment Guidelines Panel (the Panel) to
recommend either for or against the use of vilobelimab for the treatment of COVID-19.
Rationale
Results from the PANAMO trial were used to support the FDA Emergency Use Authorization.5
However, the prespecified analysis that stratified by study site showed that 28-day mortality among
patients with COVID-19 who received vilobelimab was not significantly different from 28-day mortality
among those who received placebo. The initially proposed primary study analysis did not stratify
by study site. In the second phase of the study, the primary analysis was changed to stratify by site
based on a recommendation from the FDA. The analysis that did not stratify by site demonstrated that
all-cause mortality through Day 28 was significantly lower in the vilobelimab arm than in the placebo
arm. Concomitant use of corticosteroids (97%) and antithrombotic agents (98%) was high in this study
population. Prior or concomitant use of additional immunomodulators, such as tocilizumab (17% in
the vilobelimab arm, 16% in the placebo arm) and baricitinib (3% in each arm), was low. The Panel
determined that the results from the PANAMO trial were insufficient to recommend either for or against
the use of vilobelimab for the treatment of COVID-19.
Considerations in Children
There are no data on the use of vilobelimab in children. Vilobelimab is not authorized by the FDA for
the treatment of COVID-19 in pediatric patients.
Clinical Data
The small (n = 30) Phase 2 portion of the Phase 2/3 PANAMO trial was too underpowered to draw any
conclusions about study outcomes, including physiologic improvement at 5 days and mortality.7
The Phase 3 portion of the trial was a double-blind, randomized trial performed at 46 hospitals in
Western Europe (i.e., Netherlands, France, Germany, Belgium), Brazil, Mexico, Russia, Peru, and
South Africa from October 1, 2020, to October 4, 2021.6 The trial compared the use of vilobelimab
plus standard of care with placebo plus standard of care in patients aged ≥18 years who had laboratory-
confirmed SARS-CoV-2 infection, were receiving mechanical ventilation (and were within 48 hours of
intubation), and had a ratio of arterial partial pressure of oxygen to fraction of inspired oxygen of 60 to
200 mm Hg at study entry. Vilobelimab 800 mg was administered intravenously on Days 1, 2, 4, 8, 15,
and 22, if the patient remained hospitalized, for a maximum of 6 doses.
The primary outcome was all-cause mortality at 28 days. Secondary outcomes included all-cause
mortality at 60 days, the proportion of patients who improved on a World Health Organization 8-point
ordinal scale, the proportion of patients who developed acute kidney failure by Day 28, and the
proportion of patients free from renal replacement therapy at Day 28.
Results
• The trial enrolled 369 patients; 368 patients were included in the analysis that did not stratify by
study site (177 in the vilobelimab arm, 191 in the placebo arm).
• In the prespecified analysis that stratified by study site (n = 307), 28-day mortality was not
significantly different between the vilobelimab and placebo arms (HR 0.73; 95% CI, 0.50–1.06;
P = 0.094). The analysis for 28-day mortality that stratified by study site excluded the 61 patients
(16.6%) from sites that had no deaths or had only 1 treatment group.
• In the analysis that did not stratify by study site (n = 368), 28-day mortality was lower in the
vilobelimab arm than in the placebo arm (54 of 177 patients [31%] vs. 77 of 191 patients [44%]),
and the difference between arms was statistically significant (HR 0.67; 95% CI, 0.48–0.96; P =
0.027).
• Prespecified subgroup analyses identified a significant reduction in 28-day mortality in the
vilobelimab arm for subgroups of patients with severe acute respiratory distress syndrome (HR
0.55; 95% CI, 0.30–0.98; P = 0.044), patients with an estimated glomerular filtration rate of <60
mL/min (HR 0.55; 95% CI, 0.31–0.96; P = 0.036), and patients receiving mechanical ventilation
and additional organ support (category 7 on the World Health Organization 8-point ordinal scale;
HR 0.62; 95% CI, 0.40–0.95; P = 0.028).
• In a prespecified analysis of the Western Europe subgroup (i.e., Netherlands, France, Germany,
Belgium), the vilobelimab arm had significantly lower 28-day mortality than the placebo arm (HR
0.51; 95% CI, 0.30–0.87; P = 0.014).
• For the secondary outcomes:
• The analysis that stratified by study site showed no significant difference between the arms for
Limitations
• The results for the study’s site-stratified, prespecified analysis were not significant.
• The analysis for 28-day mortality that stratified by study site excluded the 61 patients (16.6%)
from sites that had no deaths or had only 1 treatment group.
• Very few patients received a second immunomodulator (tocilizumab or baricitinib), which makes
the study results difficult to apply to current practice.
• Compared to other studies that have evaluated the use of immunomodulators for the treatment of
COVID-19, Phase 3 of the PANAMO trial had a relatively small sample size.
References
1. Cugno M, Meroni PL, Gualtierotti R, et al. Complement activation in patients with COVID-19: a novel
therapeutic target. J Allergy Clin Immunol. 2020;146(1):215-217. Available at: https://www.ncbi.nlm.nih.gov/
pubmed/32417135.
2. Manthey HD, Woodruff TM, Taylor SM, Monk PN. Complement component 5a (C5a). Int J Biochem Cell
Biol. 2009;41(11):2114-2117. Available at: https://www.ncbi.nlm.nih.gov/pubmed/19464229.
3. Carvelli J, Demaria O, Vély F, et al. Association of COVID-19 inflammation with activation of the
C5a-C5aR1 axis. Nature. 2020;588(7836):146-150. Available at: https://pubmed.ncbi.nlm.nih.gov/32726800.
4. Chouaki Benmansour N, Carvelli J, Vivier E. Complement cascade in severe forms of COVID-19: recent
advances in therapy. Eur J Immunol. 2021;51(7):1652-1659. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33738806.
5. Food and Drug Administration. Fact sheet for healthcare providers: Emergency Use Authorization for Gohibic.
2023. Available at: https://www.fda.gov/media/166824/download.
6. Vlaar APJ, Witzenrath M, van Paassen P, et al. Anti-C5a antibody (vilobelimab) therapy for critically ill,
invasively mechanically ventilated patients with COVID-19 (PANAMO): a multicentre, double-blind,
randomised, placebo-controlled, Phase 3 trial. Lancet Respir Med. 2022;10(12):1137-1146. Available at:
https://pubmed.ncbi.nlm.nih.gov/36087611.
7. Vlaar APJ, de Bruin S, Busch M, et al. Anti-C5a antibody IFX-1 (vilobelimab) treatment versus best
supportive care for patients with severe COVID-19 (PANAMO): an exploratory, open-label, Phase 2
randomised controlled trial. Lancet Rheumatol. 2020;2(12):e764-e773. Available at: https://pubmed.ncbi.nlm.
nih.gov/33015643.
Key: AE = adverse event; ALC = absolute lymphocyte count; ALT = alanine transaminase; ANC = absolute neutrophil count; AST = aspartate aminotransferase; BAR =
baricitinib; BMP = basic metabolic panel; BP = blood pressure; CBC = complete blood count; CD4 = CD4 T lymphocyte; COPD = chronic obstructive pulmonary disease;
CrCl = creatinine clearance; CVA = cerebral vascular accident; CYP = cytochrome P450; DEX = dexamethasone; DILI = drug-induced liver injury; DVT = deep vein
thrombosis; ECMO = extracorporeal membrane oxygenation; eGFR = estimated glomerular filtration rate; EUA = Emergency Use Authorization; FDA = Food and Drug
Administration; GDH-PQQ = glucose dehydrogenase pyrroloquinoline quinone; GI = gastrointestinal; HBV = hepatitis B virus; Hgb = hemoglobin; HSR = hypersensitivity
reaction; HSV = herpes simplex virus; HTN = hypertension; ICU = intensive care unit; IL = interleukin; IV = intravenous; JIA = juvenile idiopathic arthritis; MDI =
metered dose inhaler; MI = myocardial infarction; MV = mechanical ventilation; NaCl = sodium chloride; NIV = noninvasive ventilation; NMBA = neuromuscular blocking
agent; OAT = organic anion transporter; the Panel = the COVID-19 Treatment Guidelines Panel; PE = pulmonary embolism; PLT = platelet count; PO = oral; SUBQ =
subcutaneous; TB = tuberculosis; TNF = tumor necrosis factor
Summary Recommendations
Chronic Anticoagulant and Antiplatelet Therapy
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends that patients with COVID-19 who are receiving
anticoagulant or antiplatelet therapies for underlying conditions continue these medications unless significant bleeding
develops or other contraindications are present (AIII).
• Before prescribing ritonavir-boosted nirmatrelvir (Paxlovid) to patients who are receiving anticoagulant or antiplatelet
therapy, clinicians should carefully review the patient's concomitant medications to evaluate potential drug-drug
interactions. It may be necessary to modify the dosage of the antithrombotic agent, switch to another antithrombotic
agent, or prescribe an alternative COVID-19 therapy. See Drug-Drug Interactions Between Ritonavir-Boosted
Nirmatrelvir (Paxlovid) and Concomitant Medications for more information.
Screening and Evaluation for Venous Thromboembolism
• There is insufficient evidence for the Panel to recommend either for or against routine screening for venous
thromboembolism (VTE) in patients with COVID-19 who do not have signs or symptoms of VTE, regardless of the status
of their coagulation markers.
• For hospitalized patients with COVID-19 who experience rapid deterioration of pulmonary, cardiac, or neurological
function or sudden, localized loss of peripheral perfusion, the Panel recommends evaluating the patients for
thromboembolic disease (AIII).
Antithrombotic Therapy for Nonhospitalized Adults Without Evidence of Venous Thromboembolism
• In nonhospitalized patients with COVID-19, the Panel recommends against the use of anticoagulant and antiplatelet
therapy (i.e., aspirin, P2Y12 inhibitors) for the prevention of VTE or arterial thrombosis, except in a clinical trial (AIIa).
This recommendation does not apply to patients with other indications for antithrombotic therapy.
Antithrombotic Therapy for Hospitalized, Nonpregnant Adults Without Evidence of Venous Thromboembolism
• The Panel recommends against using anticoagulant or antiplatelet therapy to prevent arterial thrombosis outside of
the usual standard of care for patients without COVID-19 (AIII).
• In hospitalized patients, low-molecular-weight heparin (LMWH) or unfractionated heparin (UFH) is preferred over
oral anticoagulants (AIII). Because these types of heparin have shorter half-lives, their effects can be reversed quickly.
They can also be administered intravenously or subcutaneously, and they have fewer drug-drug interactions than oral
anticoagulants.
• When heparin is used, LMWH is preferred over UFH.
For adults who require low-flow oxygen and do not require intensive care unit (ICU)-level care:
• The Panel recommends the use of a therapeutic dose of heparin for patients with D-dimer levels above the upper
limit of normal who require low-flow oxygen and who do not have an increased risk of bleeding (CIIa).
• Contraindications for the use of therapeutic anticoagulation in patients with COVID-19 include a platelet count
<50,000 cells/µL, hemoglobin <8 g/dL, the need for dual antiplatelet therapy, bleeding within the past 30 days
that required an emergency department visit or hospitalization, a history of a bleeding disorder, or an inherited or
active acquired bleeding disorder. This list is based on the exclusion criteria from clinical trials; patients with these
conditions have an increased risk of bleeding
• In patients without VTE who have started treatment with therapeutic doses of heparin, treatment should continue until 1
of the following occurs, whichever comes first:
• The patient receives 14 days of treatment, at which time, they should be switched to prophylactic anticoagulation
until hospital discharge;
• The patient is transferred to the ICU, and prophylactic anticoagulation should be administered for the remainder of
the hospitalization period; or
References
1. Han H, Yang L, Liu R, et al. Prominent changes in blood coagulation of patients with SARS-CoV-2 infection.
Clin Chem Lab Med. 2020;58(7):1116-1120. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32172226.
2. Driggin E, Madhavan MV, Bikdeli B, et al. Cardiovascular considerations for patients, health care workers,
and health systems during the COVID-19 pandemic. J Am Coll Cardiol. 2020;75(18):2352-2371. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32201335.
3. Guan WJ, Ni ZY, Hu Y, et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med.
2020;382(18):1708-1720. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32109013.
The clinical trials described in this table do not represent all the trials that the Panel reviewed while developing the recommendations for
anticoagulant therapy. The studies summarized below are those that have had the greatest impact on the Panel’s recommendations.
RAPID: Open-Label RCT of Therapeutic Heparin in Moderately Ill, Hospitalized Patients With COVID-19 in 6 Countries2
Key Inclusion Criteria Participant Characteristics Key Limitations
• Hospitalized with COVID-19 and D-dimer level • Median age 60 years; 57% men; mean BMI 30 • Open-label study
≥2 times ULN or any elevated D-dimer level and • 48% with HTN; 34% with DM; 7% with CVD • Trial only enrolled 12% of screened patients.
SpO2 ≤93% on room air
• 91% with hypoxia; 6% received HFNC oxygen. Interpretation
• Hospitalized <5 days
• D-dimer level: • Compared to prophylactic heparin, therapeutic
Key Exclusion Criteria • 49% <2 times ULN heparin reduced mortality (a secondary endpoint)
• Need for therapeutic anticoagulation • 51% ≥2 times ULN but had no effect on the primary composite
• Receiving dual antiplatelet therapy endpoint of ICU admission, the need for NIV or MV,
• 69% on corticosteroids or death up to 28 days.
• High bleeding risk
Primary Outcome • There were no differences between the arms in
Interventions • Composite of ICU admission, NIV or MV, or death at 28 the percentages of patients who experienced VTE
• Therapeutic UFH or LMWH for 28 days or until days: 16% in therapeutic arm vs. 22% in prophylactic or major bleeding events.
hospital discharge or death (n = 228) arm (OR 0.69; 95% CI, 0.43–1.10)
• Prophylactic UFH or LMWH for 28 days or until Secondary Outcomes
hospital discharge or death (n = 237)
• All-cause death at 28 days: 2% in therapeutic arm vs.
Primary Endpoint 8% in prophylactic arm (OR 0.22; 95% CI, 0.07–0.65)
• Composite of ICU admission, NIV or MV, or death • Mean number of organ support-free days: 26 in
at 28 days therapeutic arm vs. 24 in prophylactic arm (OR 1.41;
Key Secondary Endpoints 95% CI, 0.9–2.21)
• All-cause death at 28 days • No difference between arms for VTE (1% in therapeutic
arm vs. 3% in prophylactic arm) or major bleeding
• Mean number of organ support-free days events (1% in therapeutic arm vs. 2% in prophylactic
• VTE arm)
• Major bleeding events • Mean number of hospital-free days alive: 20 in
• Mean number of hospital-free days alive therapeutic arm vs. 18 in prophylactic arm (OR 1.09;
95% CI, 0.79–1.50)
HEP-COVID: Open-Label RCT of Therapeutic Heparin in High-Risk, Hospitalized Patients With COVID-19 in the United States3
Key Inclusion Criteria Participant Characteristics Key Limitations
• Hospitalized with COVID-19 and required • Median age 67 years; 54% men; mean BMI 30 • Open-label study
supplemental oxygen • 60% with HTN; 37% with DM; 75% with CVD • Trial only enrolled 2% of screened patients.
• D-dimer level >4 times ULN or sepsis-induced • 64% received oxygen via nasal cannula; 15% received Interpretation
coagulopathy score of ≥4 HFNC oxygen or NIV; 5% received MV.
• Compared to usual care, therapeutic LMWH
• Hospitalized <72 hours • 80% on corticosteroids reduced the incidence of VTE, ATE, and death.
Key Exclusion Criteria Primary Outcomes • Among patients who were not in the ICU,
• Need for therapeutic anticoagulation • Composite of VTE, ATE, or death within 32 days: 29% in therapeutic LMWH significantly reduced the
• Receiving dual antiplatelet therapy therapeutic arm vs. 42% in usual care arm (relative risk percentage of patients who experienced
0.68; 95% CI, 0.49–0.96) thrombotic events and did not increase the
• High bleeding risk percentage of patients who experienced major
• CrCl <15 mL/min • Thrombotic events: 11% in therapeutic arm vs. bleeding events.
29% in usual care arm (relative risk 0.37; 95% CI,
Interventions 0.21–0.66)
• Therapeutic LMWH until hospital discharge or • Death: 19% in therapeutic arm vs. 25% in usual care
primary endpoint met (n = 129) arm (relative risk 0.78; 95% CI, 0.49–1.23)
• Usual care of prophylactic or intermediate- • Non-ICU stratum composite of VTE, ATE, or death within
dose LMWH until hospital discharge or primary 32 days: 17% in therapeutic arm vs. 36% in usual care
endpoint met (n = 124) arm (relative risk 0.46; 95% CI, 0.27–0.81)
Primary Endpoint Safety Outcomes
• Composite of VTE, ATE, or death from any cause • Major bleeding events within 32 days: 5% in
within 32 days of randomization therapeutic arm vs. 2% in usual care arm (relative risk
Key Safety Endpoint 2.88; 95% CI, 0.59–14.02)
• Major bleeding events within 32 days • Non-ICU stratum major bleeding events within 32
days: 2% in both arms
ACTION: Open-Label RCT of Therapeutic Rivaroxaban in Hospitalized Patients With COVID-19 in Brazil4
Key Inclusion Criteria Participant Characteristics Key Limitations
• Hospitalized with COVID-19 and elevated • Median age 57 years; 60% men; mean BMI 30 • Open-label study
D-dimer level • 49% with HTN; 24% with DM; 5% with CAD • Trial only enrolled 18% of screened patients.
• Symptoms for ≤14 days • Critically ill: 7% in therapeutic arm vs. 5% in usual care • Therapeutic rivaroxaban was administered for a
Key Exclusion Criteria arm longer duration than prophylactic anticoagulation
• 75% required oxygen: 60% received low-flow oxygen; (30 days vs. a mean duration of 8 days).
• Need for therapeutic anticoagulation
8% received HFNC oxygen; 1% received NIV; 6% Interpretation
• CrCl <30 mL/min
received MV.
• Receiving P2Y12 inhibitor therapy or aspirin • When compared with usual care, therapeutic
• 83% on corticosteroids rivaroxaban did not reduce mortality, hospital
>100 mg
Primary Outcome duration, oxygen use duration, or the percentage
• High bleeding risk of patients who experienced thrombosis.
• No difference between arms in the composite of time to
Interventions
death, hospital duration, or oxygen use duration by Day • Patients who received therapeutic rivaroxaban had
• Therapeutic anticoagulation for 30 days: 30 (win ratio 0.86; 95% CI, 0.59–1.22) more clinically relevant, nonmajor bleeding events
rivaroxaban 15 mg or 20 mg once daily; if than those who received usual care.
clinically unstable, enoxaparin 1 mg/kg twice Secondary Outcomes
• The longer duration of therapy in the therapeutic
daily or UFH (n = 311) • No difference between therapeutic and usual care arms arm may have influenced the difference in
• Usual care prophylactic anticoagulation with in: bleeding events.
enoxaparin or UFH during hospitalization (n = • Thrombosis: 7% vs. 10%
304) • Death by Day 30: 11% vs. 8%
Primary Endpoint • Any bleeding events: 12% in therapeutic arm vs. 3% in
• Hierarchical composite of time to death, hospital usual care arm
duration, or oxygen use duration by Day 30 • Major bleeding events: 3% in therapeutic arm vs. 1% in
Key Secondary Endpoints usual care arm
• Thrombosis, with and without all-cause death • Clinically relevant, nonmajor bleeding events: 5% in
therapeutic arm vs. 1% in usual care arm
• Death by Day 30
• Bleeding events
FREEDOM: RCT of Anticoagulation Strategies in Noncritically Ill Patients Who Were Hospitalized With COVID-19 in 10 Countries5
Key Inclusion Criterion Participant Characteristics Key Limitations
• Hospitalized with symptomatic COVID-19 for <48 • Median age 52 years; 59% men; mean BMI 26 • Open-label study
hours • 32% with HTN; 19% with DM • Trial was terminated early due to slow recruitment
Key Exclusion Criteria • 22% on corticosteroids; 10% on RDV (3,452 of 3,600 planned patients recruited).
• Need for therapeutic anticoagulation • Minimal treatment with RDV or DEX as SOC for
Primary Outcome
COVID-19
• CrCl <30 mL/min • 30-day composite outcome: 11.3% in combined
• Receiving P2Y12 inhibitor therapy or aspirin therapeutic arms vs. 13.2% in prophylactic arm (HR Interpretation
>100 mg per day 0.85; 95% CI, 0.69–1.04; P = 0.11) • When compared with prophylactic enoxaparin,
• Anticipated hospitalization for <72 hours • Primary endpoint was not statistically significant when therapeutic apixaban and therapeutic enoxaparin
therapeutic enoxaparin or apixaban were compared to did not reduce 30-day mortality, the need for ICU-
Interventions level care, or the occurrence of thromboembolism
prophylactic enoxaparin.
• Therapeutic apixaban 5 mg twice daily (n = or ischemic stroke.
1,121) Secondary Outcomes
• Fewer patients died in the therapeutic enoxaparin
• Therapeutic enoxaparin 1 mg/kg twice daily (n = • 30-day all-cause mortality: 4.9% in therapeutic and therapeutic apixaban arms than in the
1,136) enoxaparin arm vs. 7.0% in prophylactic enoxaparin prophylactic enoxaparin arm.
arm (HR 0.69; 95% CI, 0.49–0.99)
• Usual care prophylactic enoxaparin (n = 1,141) • There were no statistically significant differences
• 30-day all-cause mortality: 5.0% in therapeutic between the arms in the percentages of patients
Primary Endpoint apixaban arm vs. 7.0% in prophylactic enoxaparin arm who experienced severe bleeding events.
• 30-day composite of all-cause mortality, need (HR 0.7; 95% CI, 0.49–0.99)
for ICU-level care, systemic thromboembolism, • BARC type 3 or 5 bleeding events: 0.4% in combined
or ischemic stroke. Endpoint assessed for the therapeutic arms vs. 0.1% in prophylactic arm (IRR
combined therapeutic arms vs. the prophylactic 3.96; 95% CI, 0.50–31.27)
arm.
Key Secondary Endpoints
• 30-day all-cause mortality
• BARC type 3 or 5 bleeding events
INSPIRATION: Open-Label RCT of Intermediate-Dose Versus Prophylactic-Dose Anticoagulation in Patients With COVID-19 in Intensive Care Units in Iran8
Key Inclusion Criteria Participant Characteristics Key Limitations
• Admitted to ICU • Median age 62 years; 58% men; median BMI 27 • Open-label study
• Hospitalized <7 days • 44% with HTN; 28% with DM; 14% with CAD • Not all patients received ICU-level care.
Key Exclusion Criteria • 32% received NIV; 20% received MV. Interpretation
• Life expectancy <24 hours • 23% on vasopressors; 93% on corticosteroids; 60% on • Intermediate-dose anticoagulation did not
RDV significantly reduce the occurrence of VTE and
• Need for therapeutic anticoagulation
Primary Outcome ATE, the need for ECMO, or mortality.
• Bleeding or high bleeding risk
• Composite of adjudicated acute VTE, ATE, the need • Although the difference was not significant,
Interventions patients who received intermediate-dose
for ECMO, or all-cause mortality at 30 days: 46% in
• Intermediate-dose anticoagulation: enoxaparin 1 intermediate-dose arm vs. 44% in prophylactic arm (OR anticoagulation had more bleeding events
mg/kg once daily (n = 276) 1.06; 95% CI, 0.76–1.48) than patients who received prophylactic-dose
• Prophylactic-dose anticoagulation (n = 286) anticoagulation.
Secondary Outcomes
Primary Endpoint • No difference between intermediate-dose arm and
• Composite of adjudicated acute VTE, ATE, the prophylactic arm in:
need for ECMO, or all-cause mortality at 30 days • All-cause mortality at 30 days: 43% vs. 41%
Key Secondary Endpoints • VTE: 3% in both arms
• All-cause mortality at 30 days • Major bleeding events and clinically relevant,
• VTE nonmajor bleeding events: 6.3% vs. 3.1% (OR 2.02;
95% CI, 0.89–4.61)
• Bleeding events
ACTIV-4B: Double-Blind RCT of Anticoagulant and Antiplatelet Therapy in Symptomatic, Nonhospitalized Patients With COVID-19 in the United States10
Key Inclusion Criteria Participant Characteristics Key Limitation
• Aged 40–80 years • Listed characteristics are for all 657 randomized • Initial target sample size was 7,000 patients, but
• Symptomatic SARS-CoV-2 infection patients. trial was terminated after enrolling only 9% of
• Median age 54 years; 59% women; median BMI 30; target because of a low event rate.
• CrCl >30 mL/min
13% Black, 28% Hispanic Interpretation
• PLT >100,000 cells/µL
• 18% with DM; 20% with a history of smoking; 35% • Among symptomatic outpatients with COVID-19
Key Exclusion Criteria with HTN who were clinically stable, treatment with aspirin
• Previously hospitalized with COVID-19 Primary Outcome or a therapeutic or prophylactic dose of apixaban
• Acute leukemia, recent major bleeding events, or • Composite of all-cause mortality, symptomatic VTE, did not reduce the risk of death, symptomatic VTE
indication or contraindication for anticoagulant or or ATE, or hospitalization for cardiovascular or
ATE, MI, stroke, or hospitalization at 45 days: 1 (0.7%) pulmonary causes compared to placebo.
antiplatelet therapy in aspirin arm vs. 1 (0.7%) in apixaban 2.5 mg arm vs.
Interventions 2 (1.4%) in apixaban 5 mg arm vs. 1 (0.7%) in placebo
• 558 of 657 randomized patients received study arm
drugs. • Risk differences compared with placebo:
• Aspirin 81 mg PO once daily for 45 days (n = • 0.0% (95% CI, not calculable) in aspirin arm
144) • 0.7% (95% CI, -2.1% to 4.1%) in 2.5 mg apixaban
• Prophylactic-dose anticoagulation: apixaban 2.5 arm
mg PO twice daily for 45 days (n = 135) • 1.4% (95% CI, -1.5% to 5.0%) in 5 mg apixaban arm
• Therapeutic-dose anticoagulation: apixaban 5 • Cumulative incidence of primary outcomes did not
mg PO twice daily for 45 days (n = 143) significantly differ across treatment arms (log-rank P =
• Placebo (n = 136) 0.78).
Primary Endpoint Secondary Outcome
• Composite of all-cause mortality, symptomatic • No differences between all treatment arms and placebo
VTE, ATE, MI, stroke, or hospitalization for arm in occurrence of individual components of the
cardiovascular or pulmonary cause at 45 days primary endpoint
Key Secondary and Safety Endpoints Safety Outcome
• Component events of primary endpoint • No major bleeding events occurred.
• Major bleeding
ACTIV-4C: Double-Blind RCT of 30 Days of Apixaban After Hospital Discharge in Patients With COVID-19 in the United States13
Key Inclusion Criteria Participant Characteristics Key Limitation
• Hospitalized >48 hours with confirmed SARS- • Median age 54 years; 50% men; 27% Black, 17% • Trial was terminated early due to a low event
CoV-2 infection within 2 weeks of admission Hispanic rate and because the decreasing number of
• PLT >50,000 cells/µL and Hgb >8 g/dL • 15% on antiplatelet therapy hospitalizations for people with COVID-19 made
recruitment difficult.
Key Exclusion Criteria • At hospital discharge, 16% were prescribed antiplatelet
therapy; 93% received aspirin. Interpretation
• Need for therapeutic or prophylactic
anticoagulation at hospital discharge Primary Outcome • Incidence of death or thromboembolism was low
in this cohort of patients.
• Ischemic stroke, intracranial bleed, or • Composite of death, ATE, or VTE by Day 30: 13 (2.1%)
neurosurgery within 3 months in apixaban arm vs. 14 (2.3%) in placebo arm (relative • Because the trial was terminated early, the results
risk 0.92; 95% CI, 0.44–1.95; P = 0.85) were imprecise, and the study was inconclusive.
• Bleeding events within past 30 days
• Major surgery within 14 days Safety Outcomes
• Inherited or active acquired bleeding disorder • Major bleeding events: 2 (0.4%) in apixaban arm vs.
1 (0.2%) in placebo arm (relative risk 2.00; 95% CI,
Interventions 0.18–22.03)
• Apixaban 2.5 mg PO twice daily for 30 days, • Clinically relevant, nonmajor bleeding events: 3 (0.6%)
starting at hospital discharge (n = 610) in apixaban arm vs. 6 (1.1%) in placebo arm (relative
• Placebo (n = 607) risk 0.50; 95% CI, 0.13–1.99)
Primary Endpoint
• Composite of death, ATE, or VTE by Day 30
Key Safety Endpoint
• Bleeding events
The clinical trials described in this table do not represent all the trials that the Panel reviewed while developing the recommendations for
antiplatelet therapy. The studies summarized below are those that have had the greatest impact on the Panel’s recommendations.
RECOVERY: Open-Label RCT of Aspirin in Hospitalized Patients With COVID-19 in Indonesia, Nepal, and the United Kingdom2
Key Inclusion Criterion Participant Characteristics Key Limitation
• Clinically suspected or laboratory-confirmed • Mean age 59 years; 62% men; 75% White • Because of the open-label design, reporting of
SARS-CoV-2 infection • 97% had laboratory-confirmed SARS-CoV-2 infection major bleeding and thrombotic events may have
been influenced by the treatment allocation.
Key Exclusion Criteria • At baseline:
• Hypersensitivity to aspirin • 5% on MV Interpretation
• Recent history of major bleeding events • 28% on NIV • In hospitalized patients with COVID-19, the use
of aspirin was not associated with reductions in
• Currently receiving aspirin or another antiplatelet • 34% received intermediate- or therapeutic-dose 28-day mortality or the risk of progressing to MV
treatment LMWH. or death.
Interventions • 60% received standard-dose LMWH.
• Aspirin 150 mg once daily until hospital • 7% received no thromboprophylaxis.
discharge (n = 7,351) • 94% received corticosteroids; 26% received RDV; 13%
• SOC alone (n = 7,541) received tocilizumab; 6% received baricitinib.
Primary Endpoint Primary Outcome
• All-cause mortality at 28 days • All-cause mortality at 28 days: 17% in both arms (rate
ratio 0.96; 95% CI, 0.89–1.04; P = 0.35)
Key Secondary Endpoints
• Composite of progression to MV or death at 28 Secondary Outcomes
days • Composite of progression to MV or death at 28 days:
• Major bleeding events at 28 days 21% in aspirin arm vs. 22% in SOC arm (risk ratio 0.96;
95% CI, 0.90–1.03)
• Thrombotic events at 28 days
• Major bleeding events at 28 days: 1.6% in aspirin arm
vs. 1.0% in SOC arm (P = 0.0028)
• Thrombotic events at 28 days: 4.6% in aspirin arm vs.
5.3% in SOC arm (P = 0.07)
REMAP-CAP: Open-Label, Adaptive RCT of Antiplatelet Therapy in Critically Ill Patients With COVID-19 in 8 Countries in Europe and Asia3
Key Inclusion Criteria Participant Characteristics Key Limitations
• Clinically suspected or laboratory-confirmed • Mean age 57 years; 34% women; 77% White • Open-label study
SARS-CoV-2 infection • At baseline, 98% were receiving LMWH: • Different P2Y12 inhibitors were used.
• Within 48 hours of ICU admission • 19% received low-dose LMWH. • Trial was stopped for futility. Because
Key Exclusion Criteria • 59% received intermediate-dose LMWH. equivalence for aspirin and P2Y12 inhibitor
arms was reached, these arms were pooled for
• Bleeding risk sufficient to contraindicate • 12% received therapeutic-dose LMWH. analyses.
antiplatelet therapy • 98% received steroids; 21% received RDV; 44% received
• CrCl <30 mL/min tocilizumab; 11% received sarilumab. Interpretation
• Receiving antiplatelet therapy or NSAIDs • In P2Y12 inhibitor arm, 88.5% received clopidogrel, 1.3% • In critically ill patients with COVID-19, the use
received ticagrelor, 1.3% received prasugrel, and 8.8% of aspirin or a P2Y12 inhibitor did not reduce
Interventions the number of organ support-free days or in-
received an unknown P2Y12 inhibitor.
• 1 of the following plus anticoagulation for 14 hospital mortality.
days or until hospital discharge, whichever came Primary Outcome • Patients in the pooled antiplatelet arm had
first: • Data from aspirin and P2Y12 inhibitor arms were pooled more major bleeding events than those in the
• Aspirin 75–100 mg once daily (n = 565) and reported as “pooled antiplatelet arm” in final analysis: control arm, but they had improved survival
• P2Y12 inhibitor (n = 455) • Median number of organ support-free days by Day 21: over 90 days.
7 in pooled antiplatelet arm vs. 7 in control arm (aOR
• No antiplatelet therapy (control arm; n = 529)
1.02; 95% CrI, 0.86–1.23; posterior probability of futility
Primary Endpoint 96%)
• Number of organ support-free days by Day 21 Secondary Outcomes
Key Secondary Endpoints • Survival to hospital discharge: 71.5% in pooled antiplatelet
• Survival to hospital discharge arm vs. 67.9% in control arm (median-adjusted OR 1.27;
95% CrI, 0.99–1.62; adjusted absolute difference 5%;
• Survival to Day 90 95% CrI, -0.2% to 9.5%; 97% posterior probability of
• Major bleeding events by Day 14 efficacy)
• Survival to Day 90: 72% in pooled antiplatelet arm vs.
68% in control arm (HR with pooled antiplatelets 1.22;
95% CrI, 1.06–1.40; 99.7% posterior probability of
efficacy)
• Major bleeding events by Day 14: 2.1% in pooled
antiplatelet arm vs. 0.4% in control arm (aOR 2.97; 95%
CrI, 1.23–8.28; posterior probability of harm 99.4%)
COVID-PACT: Open-Label RCT of Clopidogrel in Adults With COVID-19 Who Were Receiving Intensive Care Unit-Level Care in the United States4
Key Inclusion Criteria Participant Characteristics Key Limitations
• Aged ≥18 years • Median age 58 years; 41% women; 71% White • Open-label study (adjudication committee
• Acute SARS-CoV-2 infection • At baseline, 99% on HFNC oxygen, NIV, or MV; 15% on members were blinded to the study arms).
• Required ICU-level care for ≤96 hours prior to MV • Trial was stopped early because the
randomization • 37% required MV during the study. decreasing number of ICU admissions for
patients with COVID-19 made recruitment
Key Exclusion Criteria Primary Outcome difficult.
• Ongoing or planned use of a therapeutic dose of • Composite of VTE or ATE events by hospital discharge • 31% discontinued clopidogrel.
anticoagulation or dual antiplatelet therapy or Day 28: 10% in both arms (win ratio 1.04; 95% CI,
0.54–2.01; P = 0.90) Interpretation
• High risk of bleeding
• In patients with COVID-19 who required
• History of HIT Secondary Outcome
ICU-level care, clopidogrel did not reduce the
• Ischemic stroke within 2 weeks • Composite of clinically evident VTE or ATE events by incidence of thrombotic complications.
hospital discharge or Day 28: 7% in clopidogrel arm
Interventions vs. 9% in no clopidogrel arm (win ratio 0.79; 95% CI,
• Clopidogrel 300 mg at randomization, then 0.38–1.65; P = 0.53)
clopidogrel 75 mg once daily until hospital discharge
or Day 28, whichever came first (n = 152) Safety Outcomes
• No clopidogrel therapy (n = 140) • Fatal or life-threatening bleeding events: 1.3% in
clopidogrel arm vs. 1.4% in no clopidogrel arm (P =
• Some patients were also randomized to receive a 1.00)
therapeutic or prophylactic dose of anticoagulation (n
= 290) • Moderate or severe bleeding events: 4.0% in
clopidogrel arm vs. 6.4% in no clopidogrel arm (P =
Primary Endpoint 0.83)
• Composite of VTE or ATE events by hospital discharge
or Day 28. Events included death due to VTE or
ATE, PE, clinically evident DVT, MI, ischemic stroke,
systemic embolic events or acute limb ischemia, and
clinically silent DVT.
Secondary Endpoint
• Composite of clinically evident VTE or ATE events by
hospital discharge or Day 28
Safety Endpoints
• Fatal or life-threatening bleeding events
• Moderate or severe bleeding events
References
1. Berger JS, Kornblith LZ, Gong MN, et al. Effect of P2Y12 inhibitors on survival free of organ support among non-critically ill hospitalized patients
with COVID-19: a randomized clinical trial. JAMA. 2022;327(3):227-236. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35040887.
2. RECOVERY Collaborative Group. Aspirin in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label,
platform trial. Lancet. 2022;399(10320):143-151. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34800427.
3. REMAP-CAP Writing Committee for the REMAP-CAP Investigators. Effect of antiplatelet therapy on survival and organ support-free days in
critically ill patients with COVID-19: a randomized clinical trial. JAMA. 2022;327(13):1247-1259. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35315874.
4. Bohula EA, Berg DD, Lopes MS, et al. Anticoagulation and antiplatelet therapy for prevention of venous and arterial thrombotic events in critically ill
patients with COVID-19: COVID-PACT. Circulation. 2022;146(18):1344-1356. Available at: https://www.ncbi.nlm.nih.gov/pubmed/36036760.
Summary Recommendations
Fluvoxamine
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends against the use of fluvoxamine for the treatment
of COVID-19 in nonhospitalized patients (AIIa).
• There is insufficient evidence for the Panel to recommend either for or against the use of the combination of inhaled
budesonide plus fluvoxamine for the treatment of COVID-19 in nonhospitalized patients.
• Patients with COVID-19 who are receiving fluvoxamine for an underlying condition should continue this therapy as
directed by their health care provider (AIII).
Intravenous Immunoglobulin
• The Panel recommends against the use of intravenous immunoglobulin (IVIG) for the treatment of acute COVID-19
in adults and children, except in a clinical trial (AIII). This recommendation should not preclude the use of IVIG when
it is otherwise indicated for the treatment of underlying conditions or complications that arise during the course of
COVID-19.
• For the Panel’s recommendations on the use of IVIG in people with multisystem inflammatory syndrome in children
(MIS-C) or multisystem inflammatory syndrome in adults (MIS-A) and a discussion of the clinical data that support those
recommendations, see Therapeutic Management of Hospitalized Children With MIS-C, Plus a Discussion on MIS-A.
Ivermectin
• The Panel recommends against the use of ivermectin for the treatment of COVID-19 (AIIa).
Metformin
• There is insufficient evidence for the Panel to recommend either for or against the use of metformin for the treatment of
COVID-19 in nonhospitalized patients.
• The Panel recommends against the use of metformin for the treatment of COVID-19 in hospitalized patients, except
in a clinical trial (BIII).
• Patients with COVID-19 who are receiving metformin for an underlying condition should continue this therapy as
directed by their health care provider (AIII).
The Panel reviewed clinical trials that evaluated the use of the anti-inflammatory drug colchicine for the treatment of
COVID-19; however, these trials failed to show a benefit of using colchicine in patients with COVID-19.
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
Fluvoxamine is a selective serotonin reuptake inhibitor (SSRI) that is approved by the Food and
Drug Administration (FDA) for the treatment of obsessive-compulsive disorder and is used for other
conditions, including depression. Fluvoxamine is not approved by the FDA for the treatment of any
infection.
In a murine sepsis model, fluvoxamine was found to bind to the sigma-1 receptor on immune cells,
resulting in reduced production of inflammatory cytokines.1 In an in vitro study of human endothelial
cells and macrophages, fluvoxamine reduced the expression of inflammatory genes.2
Recommendations
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends against the use of
fluvoxamine for the treatment of COVID-19 in nonhospitalized patients (AIIa).
• There is insufficient evidence for the Panel to recommend either for or against the use of
the combination of inhaled budesonide plus fluvoxamine for the treatment of COVID-19 in
nonhospitalized patients.
• Patients with COVID-19 who are receiving fluvoxamine for an underlying condition should
continue this therapy as directed by their health care provider (AIII).
Rationale
Six randomized trials have studied the use of fluvoxamine for the treatment of nonhospitalized patients
with COVID-19.3-8 The TOGETHER and STOP COVID 2 trials enrolled unvaccinated patients with
COVID-19 who had at least 1 risk factor for disease progression.3,5 These studies did not identify
a consistent benefit of using fluvoxamine in these patients, although STOP COVID 2 was stopped
early due to low primary outcome rates. Other outpatient therapies (i.e., ritonavir-boosted nirmatrelvir
[Paxlovid], remdesivir) have been shown to be effective in preventing hospitalization or death in
unvaccinated patients who are at high risk of disease progression. In subsequent trials where the
majority of enrolled patients were vaccinated against COVID-19, fluvoxamine did not significantly
reduce the risk of hospitalization or death, the time to recovery, or health care utilization.6-8 In several of
these studies, fluvoxamine was associated with decreased adherence and/or an increase in the occurrence
of nonserious adverse effects, primarily gastrointestinal symptoms.3,5,6
The TOGETHER trial was a large, double-blind, placebo-controlled, adaptive randomized trial in Brazil
that evaluated the use of inhaled budesonide plus oral fluvoxamine in patients with COVID-19.9 Over
90% of the patients had received at least 2 doses of a COVID-19 vaccine. Treatment with this combination
significantly reduced the incidence of the primary outcome, which was a composite of hospitalization or
retention in an emergency setting for >6 hours. The proportion of patients who were hospitalized was the
same in the treatment and placebo arms (0.9% vs. 1.1%), and the treatment did not significantly impact
secondary outcomes such as health care attendance or the need for an emergency setting visit. It is unclear
how the >6-hour emergency setting outcome translates to other settings. In addition, treatment with
budesonide plus fluvoxamine was associated with significantly more adverse events.
Summaries of the studies that informed the Panel’s recommendations can be found in Table 7a.
Considerations in Children
Fluvoxamine is approved by the FDA for the treatment of obsessive-compulsive disorder in children
aged ≥8 years.14 The adverse effects of SSRI use seen in children are similar to those seen in adults,
although children and adolescents appear to have higher rates of activation and vomiting than adults.15
There are no data on the use of fluvoxamine for the prevention or treatment of COVID-19 in children.
References
1. Rosen DA, Seki SM, Fernández-Castañeda A, et al. Modulation of the sigma-1 receptor-IRE1 pathway
is beneficial in preclinical models of inflammation and sepsis. Sci Transl Med. 2019;11(478):eaau5266.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/30728287.
2. Rafiee L, Hajhashemi V, Javanmard SH. Fluvoxamine inhibits some inflammatory genes expression in LPS/
stimulated human endothelial cells, U937 macrophages, and carrageenan-induced paw edema in rat. Iran J
Basic Med Sci. 2016;19(9):977-984. Available at: https://www.ncbi.nlm.nih.gov/pubmed/27803785.
3. Reis G, Dos Santos Moreira-Silva EA, Medeiros Silva DC, et al. Effect of early treatment with fluvoxamine
on risk of emergency care and hospitalisation among patients with COVID-19: the TOGETHER randomised,
platform clinical trial. Lancet Glob Health. 2022;10(1):e42-e51. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34717820.
4. Lenze EJ, Mattar C, Zorumski CF, et al. Fluvoxamine vs placebo and clinical deterioration in outpatients with
symptomatic COVID-19: a randomized clinical trial. JAMA. 2020;324(22):2292-2300. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33180097.
5. Reiersen AM, Mattar C, Bender Ignacio RA, et al. The STOP COVID 2 study: fluvoxamine vs placebo for
outpatients with symptomatic COVID-19, a fully remote randomized controlled trial. Open Forum Infect Dis.
2023;10(8):ofad419. Available at: https://www.ncbi.nlm.nih.gov/pubmed/37622035.
6. Stewart TG, Rebolledo PA, Mourad A, et al. Higher-dose fluvoxamine and time to sustained recovery in
outpatients with COVID-19: the ACTIV-6 randomized clinical trial. JAMA. 2023;Published online ahead of
print. Available at: https://www.ncbi.nlm.nih.gov/pubmed/37976072.
7. McCarthy MW, Naggie S, Boulware DR, et al. Effect of fluvoxamine vs placebo on time to sustained recovery
in outpatients with mild to moderate COVID-19: a randomized clinical trial. JAMA. 2023;329(4):296-305.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/36633838.
The clinical trials described in this table do not represent all the trials thatResults
Methods the Panel reviewed while developing the recommendations
Limitations and Interpretation
for fluvoxamine. The studies summarized below are the randomized clinical trials that have had the greatest impact on the Panel’s1
ACTIV-6: Decentralized, Randomized, Placebo-Controlled, Platform Trial of Low-Dose Fluvoxamine in Patients With Mild to Moderate COVID-19
recommendations.
Key Inclusion Criteria Participant Characteristics Key Limitation
• Aged ≥30 years • Mean age 47 years; 57% women; 81% White • Low number of some clinical events, such as
• Positive SARS-CoV-2 nasopharyngeal RT-PCR result or • 36% with BMI ≥30; 24% with HTN hospitalization
antigen test result • 67% received ≥2 doses of a SARS-CoV-2 vaccine. Interpretation
• ≥2 COVID-19 symptoms for ≤7 days • Median of 5 days from symptom onset to receipt of • In outpatients with mild to moderate
Key Exclusion Criterion study drug COVID-19, fluvoxamine 50 mg twice daily for
10 days did not reduce the time to recovery
• Receipt of fluvoxamine in past 14 days Primary Outcome or the incidence of clinical events such as
Interventions • Median time to recovery: 12 days in fluvoxamine hospitalization, urgent care visits, or ED
arm vs. 13 days in placebo arm (HR 0.96; 95% CrI, visits.
• Fluvoxamine 50 mg PO twice daily for 10 days (n = 674)
0.86–1.06)
• Placebo (n = 614; 326 received matching placebo, 288
received placebo from another study arm) Secondary Outcomes
• Hospitalization or death by Day 28: 0.2% in
Primary Endpoint
fluvoxamine arm vs. 0.3% in placebo arm (3 events
• Time to recovery, defined as time to third day of 3 total)
consecutive days without symptoms
• Urgent care visit, ED visit, or hospitalization by Day
Key Secondary Endpoints 28: 3.9% in fluvoxamine arm vs. 3.8% in placebo
• Hospitalization or death by Day 28 arm (HR 1.1; 95% CrI, 0.5–1.8)
• Urgent care visit, ED visit, or hospitalization by Day 28
ACTIV-6: Decentralized, Randomized, Placebo-Controlled, Platform Trial of High-Dose Fluvoxamine in Patients With Mild to Moderate COVID-192
Key Inclusion Criteria Participant Characteristics Key Limitation
• Aged ≥30 years • Median age 50 years; 66% women; 73% White • Low number of some clinical events, such as
• Positive SARS-CoV-2 nasopharyngeal RT-PCR result or • 36% with BMI ≥30; 26% with HTN hospitalization
antigen test result • 77% received ≥2 doses of a SARS-CoV-2 vaccine. Interpretation
• ≥2 COVID-19 symptoms for ≤7 days • Median of 5 days from symptom onset to receipt of • In outpatients with mild to moderate
Key Exclusion Criterion study drug COVID-19, fluvoxamine 100 mg twice daily
did not reduce the time to symptom recovery
• Receipt of fluvoxamine or other selective serotonin or Primary Outcome or the incidence of clinical events such as
norepinephrine reuptake inhibitors in past 14 days • Median time to recovery: 10 days in fluvoxamine hospitalization, urgent care visits, or ED
Interventions arm vs. 10 days in placebo arm (HR 0.99; 95% CrI, visits.
0.89–1.09)
• Fluvoxamine 50 mg PO twice daily for 1 day, then
fluvoxamine 100 mg PO twice daily for 12 days (n = 589) Secondary Outcomes
• Placebo (n = 586) • Hospitalization or death by Day 28: 0.2% in
fluvoxamine arm vs. 0.3% in placebo arm (3 events
Primary Endpoint
total)
• Time to recovery, defined as time to third day of 3
• Urgent care visit, ED visit, or hospitalization by Day
consecutive days without symptoms
28: 2.4% in fluvoxamine arm vs. 3.6% in placebo
Key Secondary Endpoints arm (HR 0.69; 95% CrI, 0.27–1.21)
• Hospitalization or death by Day 28
• Urgent care visit, ED visit, or hospitalization by Day 28
COVID-OUT: Randomized Trial of Metformin, Ivermectin, and Fluvoxamine in Patients With COVID-193
Key Inclusion Criteria Participant Characteristics Key Limitation
• Aged 30–85 years • Median age 43–46 years; 54% women; 82% • In this trial, the study arms that did not include
• BMI ≥25 or ≥23 if Asian or Latinx White metformin were underpowered to detect differences
• 27% with CVD; 47% with BMI ≥30 in the primary endpoint.
• Laboratory-confirmed SARS-CoV-2 infection within 3
days of randomization • 56% received primary vaccination series. Interpretation
• <7 days of symptoms • Mean of 5 days from symptom onset to • Fluvoxamine did not impact the incidence
randomization of COVID-19–related complications such as
Key Exclusion Criteria hospitalization.
• Immunocompromised Primary Outcome
• Fluvoxamine did not impact symptom severity.
• Hepatic impairment, severe kidney disease • Composite of hypoxemia, ED visit,
hospitalization, or death by Day 14: 24% in
Interventions fluvoxamine arm vs. 25% in control arm (aOR
• Fluvoxamine 50 mg PO twice daily for 14 days (n = 334) 0.94; 95% CI, 0.66–1.36)
• Control (n = 327) Secondary Outcomes
Primary Endpoint • Hospitalization by Day 14: 1.8% in
• Composite of hypoxemia (as measured by a home pulse fluvoxamine arm vs. 1.5% in control arm (aOR
oximeter), ED visit, hospitalization, or death by Day 14 1.11; 95% CI, 0.33–3.76).
• Composite of ED visit, hospitalization, or
Key Secondary Endpoints death: 5.5% in fluvoxamine arm vs. 4.6% in
• Individual components of the composite endpoint control arm (aOR 1.17; 95% CI, 0.57–2.40)
• Total symptom severity score • No deaths occurred in either arm.
• Drug interruption or discontinuation • No difference between arms in total symptom
severity score over 14 days
• Drug interruption or discontinuation: 30% in
those who only received fluvoxamine vs. 25%
in those who only received placebo
TOGETHER: Double-Blind, Adaptive RCT of Fluvoxamine in Nonhospitalized Patients With COVID-19 in Brazil4
Key Inclusion Criteria Participant Characteristics Key Limitations
• Aged ≥50 years or aged ≥18 years with comorbidities • Median age 50 years; 58% women; 95% self- • The >6-hour ED observation endpoint has not
• Laboratory-confirmed SARS-CoV-2 infection identified as mixed race been used in other studies of interventions for
• 13% with uncontrolled HTN; 13% with type 2 nonhospitalized patients who are at high risk of
• ≤7 days of symptoms hospitalization and death.
DM; 50% with BMI ≥30
Key Exclusion Criteria • Hospitalization or ED observation for >24 hours
• Mean of 3.8 days from symptom onset to
• Use of an SSRI randomization was analyzed in a post hoc analysis.
• Severe mental illness • As this was an adaptive platform trial where
Primary Outcome
• Cirrhosis, recent seizures, severe ventricular cardiac multiple investigational treatments or placebos
• Composite of ED observation >6 hours or were being evaluated simultaneously, not all
arrhythmia hospitalization by Day 28: 11% in fluvoxamine patients in the placebo arm received a placebo
Interventions arm vs. 16% in placebo arm (relative risk 0.68; that was matched to fluvoxamine by route of
• Fluvoxamine 100 mg PO twice daily for 10 days (n = 741) 95% CrI, 0.52–0.88) administration, dosing frequency, or duration of
therapy.
• Placebo (n = 756; route, dosing frequency, and duration Secondary Outcomes
of placebo may have differed from fluvoxamine for some • 87% of clinical events were hospitalizations. • PP analyses are not randomized comparisons, and
patients) they introduce bias when adherence is associated
• No difference between arms in COVID-19– with factors that influence the outcome.
Primary Endpoint related hospitalizations: 10% in fluvoxamine
• Composite of ED observation >6 hours or hospitalization arm vs. 13% in placebo arm (OR 0.77; 95% CI, • Adherence was self-reported and not verified.
for COVID-19 by Day 28 0.55–1.05) Interpretation
• Lower risk of hospitalization or ED observation • Fluvoxamine reduced the proportion of patients
Key Secondary Endpoints >24 hours in fluvoxamine arm than in placebo who met the composite endpoint of COVID-19–
• COVID-19–related hospitalization by Day 28 arm (relative risk 0.74; 95% CI, 0.56–0.98)5 related hospitalization or retention in an ED for >6
• Composite of hospitalization or ED observation >24 hours • No difference between arms in time to hours.
• Time to symptom resolution symptom resolution • The use of fluvoxamine did not impact the
• Adherence to study drugs, defined as receiving >80% of • Adherence: 74% in fluvoxamine arm vs. 82% incidence of COVID-19-related hospitalizations
possible doses in placebo arm (OR 0.62; 95% CI, 0.48–0.81) but did reduce the need for hospitalization or ED
• 11% in fluvoxamine arm vs. 8% in observations >24 hours.
• Mortality in both the primary ITT population and a PP
population that included patients who took >80% of the placebo arm stopped drug due to issues of • It is difficult to define the clinical relevance of the
study medication doses tolerability. >6-hour ED observation endpoint and apply it to
• Mortality (ITT): 2% in fluvoxamine arm vs. 3% practice settings in different countries.
in placebo arm (OR 0.68; 95% CI, 0.36–1.27) • Fluvoxamine did not have a consistent impact on
• Mortality (PP): <1% in fluvoxamine arm vs. 2% mortality.
in placebo arm (OR 0.09; 95% CI, 0.01–0.47) • Fluvoxamine did not impact the time to symptom
resolution.
COVID-19 Treatment Guidelines 363
STOP COVID 2: Fully Remote RCT of Fluvoxamine Versus Placebo in Outpatients With Symptomatic COVID-196
Key Inclusion Criteria Participant Characteristics Key Limitations
• Aged ≥30 years • Median age 47 years; 62% women; 27% • Small sample size compared to other trials
• Positive SARS-CoV-2 PCR result per patient self-report non-White • Short follow-up period
• ≤7 days of symptoms • 44% with obesity; 21% with HTN
Interpretation
• ≥1 risk factor for clinical deterioration • Median of 5 days from symptom onset to
• Fluvoxamine did not reduce the proportion of patients
randomization
Key Exclusion Criteria who experienced clinical deterioration by Day 15.
Primary Outcome
• Unstable medical comorbidities
• Clinical deterioration: 4.8% in
• Significant interacting medications fluvoxamine arm vs. 5.5% in placebo arm
Interventions (absolute difference 0.68%; 95% CI, -3.0
• Fluvoxamine 50 mg PO for 1 dose, then fluvoxamine 100 to 4.4)
mg PO twice daily through Day 15 (n = 272) Secondary Outcomes
• Placebo (n = 275) • GI AEs were significantly more common
Primary Endpoint in fluvoxamine arm
• Clinical deterioration within 15 days of randomization.
Clinical deterioration was defined as:
• Having dyspnea or being hospitalized for dyspnea or
pneumonia; and
• Having SpO2 <92% on room air or requiring
supplemental oxygen to attain SpO2 ≥92%
Key Secondary Endpoints
• Occurrence of AEs
STOP COVID: Double-Blind RCT of Fluvoxamine in Nonhospitalized Patients With COVID-19 in the United States7
Key Inclusion Criteria Participant Characteristics Key Limitations
• Aged ≥18 years • Mean age 46 years; 72% women; 25% • Small sample size
• Positive SARS-CoV-2 PCR result Black • Short follow-up period
• ≤7 days of symptoms • 56% with obesity; 20% with HTN; 17% • Ascertaining clinical deterioration was challenging
with asthma because all assessments were done remotely.
Key Exclusion Criteria
• Median of 4 days from symptom onset to • 24% of patients stopped responding to follow-up prior
• Immunocompromised randomization to Day 15 but were included in the final analysis.
• Unstable medical comorbidities Primary Outcome Interpretation
Interventions • Clinical deterioration: 0% in fluvoxamine • Fluvoxamine reduced the proportion of patients who
• Fluvoxamine 50 mg PO for 1 dose, then fluvoxamine 100 arm vs. 8.3% in placebo arm (absolute experienced clinical deterioration.
mg PO twice daily, then fluvoxamine 100 mg PO 3 times difference 8.7%; 95% CI, 1.8% to 16.4%)
• Due to significant limitations, it is difficult to draw
daily through Day 15 (n = 80) Secondary Outcome definitive conclusions about the efficacy of using
• Placebo (n = 72) • No patients in fluvoxamine arm fluvoxamine to treat COVID-19.
Primary Endpoint and 4 patients in placebo arm were
• Clinical deterioration within 15 days of randomization. hospitalized.
Clinical deterioration was defined as:
• Having dyspnea or being hospitalized for dyspnea or
pneumonia; and
• Having SpO2 <92% on room air or requiring
supplemental oxygen to attain SpO2 ≥92%
Key Secondary Endpoint
• Hospitalization by Day 15
TOGETHER: Randomized Platform Trial of Oral Fluvoxamine Plus Inhaled Budesonide for the Treatment of Early Onset COVID-198
Key Inclusion Criteria Participant Characteristics Key Limitation
• Aged ≥50 years or aged ≥18 years with comorbidities • Median age 51 years; 61% women • Multiple investigational treatments or placebos
• Laboratory-confirmed SARS-CoV-2 infection • 42% with BMI >30; 44% with HTN; 68% with were evaluated simultaneously. Not all patients in
multiple comorbidities the placebo arm received a matched placebo.
• ≤7 days of symptoms
• 94% received ≥2 doses of a COVID-19 vaccine. Interpretation
Key Exclusion Criteria
Primary Outcome • In adult outpatients with mild COVID-19,
• Use of an SSRI fluvoxamine plus inhaled budesonide reduced
• Severe mental illness • Composite of ED observation >6 hours or the need for ED observations >6 hours or
hospitalization by Day 28: 1.8% in fluvoxamine hospitalization when compared with placebo.
• Cirrhosis, recent seizures, severe ventricular cardiac plus inhaled budesonide arm vs. 3.7% in
arrhythmia placebo arm (relative risk 0.50; 95% CrI, • The use of fluvoxamine plus inhaled budesonide did
Interventions 0.25–0.92) not reduce hospitalization, health care attendance,
or the occurrence of any ED visit.
• Fluvoxamine 100 mg PO twice daily plus budesonide Secondary Outcomes
800 µg inhaled twice daily for 10 days (n = 738) • It is difficult to define the clinical relevance of the
• Hospitalization by Day 28: 0.9% in fluvoxamine >6-hour ED observation endpoint and apply it to
• Placebo (n = 738; route, dosing frequency, and duration plus inhaled budesonide arm vs. 1.1% in practice settings in different countries.
for some patients may have differed from treatment placebo arm
group) • The use of fluvoxamine plus inhaled budesonide
• Health care attendance by Day 28: 2.6% in resulted in more AEs than placebo.
Primary Endpoint fluvoxamine plus inhaled budesonide arm vs.
• Composite of ED observation >6 hours or hospitalization 4.1% in placebo arm (relative risk 0.64; 95%
for COVID-19 by Day 28 CrI, 0.36–1.11)
• Any ED visit by Day 28: 12.2% in fluvoxamine
Key Secondary Endpoints plus inhaled budesonide arm vs. 13.0% in
• Hospitalization by Day 28 placebo arm
• Health care attendance by Day 28 • Treatment-emergent AEs: 17.6% in fluvoxamine
• Any ED visit by Day 28 plus inhaled budesonide arm vs. 12.9% in
placebo arm (relative risk 1.37; 95% CrI,
• Occurrence of AEs
1.07–1.75)
• Most AEs were grade 2.
Key: AE = adverse event; BMI = body mass index; CVD = cardiovascular disease; DM = diabetes mellitus; ED = emergency department; GI = gastrointestinal; HTN =
hypertension; ITT = intention-to-treat; the Panel = the COVID-19 Treatment Guidelines Panel; PCR = polymerase chain reaction; PO = oral; PP = per protocol; RCT =
randomized controlled trial; RT-PCR = reverse transcription polymerase chain reaction; SpO2 = oxygen saturation; SSRI = selective serotonin reuptake inhibitor
Recommendations
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends against the use of
intravenous immunoglobulin (IVIG) for the treatment of acute COVID-19 in adults and
children, except in a clinical trial (AIII). This recommendation should not preclude the use of
IVIG when it is otherwise indicated for the treatment of underlying conditions or complications
that arise during the course of COVID-19.
• For the Panel’s recommendations on the use of IVIG in people with multisystem inflammatory
syndrome in children (MIS-C) or multisystem inflammatory syndrome in adults (MIS-A) and a
discussion of the clinical data that support those recommendations, see Therapeutic Management
of Hospitalized Children With MIS-C, Plus a Discussion on MIS-A.
Rationale
It is unknown whether IVIG products derived from pooled donor plasma contain high titers of antibodies
that neutralize SARS-CoV-2. Information on SARS-CoV-2 antibody titer was not reported in the
clinical trials that evaluated the use of IVIG for the treatment of COVID-19. The levels of SARS-CoV-2
antibodies in IVIG products likely vary depending on which SARS-CoV-2 variant was dominant when
the plasma products were collected, and different lots of IVIG may have different titers of antibodies.
Although IVIG preparations may have general immunomodulatory effects, these theoretical effects do
not appear to benefit patients with COVID-19.1
Considerations in Children
No comparative studies have evaluated the use of IVIG in pediatric patients with acute COVID-19.
IVIG is used in combination with glucocorticoids to treat MIS-C in pediatric patients.3-6 However,
because there is no clear evidence that IVIG is an effective treatment for acute COVID-19 in adults, the
Panel recommends against the use of IVIG for the treatment of acute COVID-19 in children, except
in a clinical trial (AIII). This recommendation should not preclude the use of IVIG when it is otherwise
indicated.
For the Panel’s recommendations for children with MIS-C, see Therapeutic Management of Hospitalized
Children With MIS-C, Plus a Discussion on MIS-A.
Clinical Data
In a meta-analysis of 6 randomized controlled trials that enrolled hospitalized patients with COVID-19,
the use of non-SARS-CoV-2–specific IVIG was not associated with a survival benefit.1 All of the
COVID-19 Treatment Guidelines 368
References
1. Lai CC, Chen WC, Chen CY, Wei YF. The effect of intravenous immunoglobulins on the outcomes of patients
with COVID-19: a systematic review and meta-analysis of randomized controlled trials. Expert Rev Anti Infect
Ther. 2022;20(10):1333-1340. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35786174.
2. Lakkaraja M, Berkowitz RL, Vinograd CA, et al. Omission of fetal sampling in treatment of subsequent
pregnancies in fetal-neonatal alloimmune thrombocytopenia. Am J Obstet Gynecol. 2016;215(4):471.e1-471.
e9. Available at: https://www.ncbi.nlm.nih.gov/pubmed/27131591.
3. Belhadjer Z, Auriau J, Méot M, et al. Addition of corticosteroids to immunoglobulins is associated with
recovery of cardiac function in multi-inflammatory syndrome in children. Circulation. 2020;142(23):2282-
2284. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33112651.
4. Ouldali N, Toubiana J, Antona D, et al. Association of intravenous immunoglobulins plus methylprednisolone
vs immunoglobulins alone with course of fever in multisystem inflammatory syndrome in children. JAMA.
2021;325(9):855-864. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33523115.
5. Son MBF, Murray N, Friedman K, et al. Multisystem inflammatory syndrome in children—initial therapy and
outcomes. N Engl J Med. 2021;385(1):23-34. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34133855.
6. McArdle AJ, Vito O, Patel H, et al. Treatment of multisystem inflammatory syndrome in children. N Engl J
Med. 2021;385(1):11-22. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34133854.
7. Upasani V, Townsend K, Wu MY, et al. Commercial immunoglobulin products contain neutralising antibodies
Ivermectin is a Food and Drug Administration (FDA)-approved antiparasitic drug used to treat several
neglected tropical diseases, including onchocerciasis, helminthiases, and scabies.1 For these indications,
ivermectin has been widely used and is generally well tolerated.1,2 Ivermectin is not approved by the
FDA for the treatment of any viral infection, including COVID-19. See the FDA webpage Why You
Should Not Use Ivermectin to Treat or Prevent COVID-19 for more information.
Ivermectin has been shown to inhibit the replication of SARS-CoV-2 in cell cultures.3 However,
pharmacokinetic and pharmacodynamic studies suggest that achieving the plasma concentrations
necessary for the antiviral efficacy detected in vitro would require administration of doses up to 100-fold
higher than those approved for use in humans.4,5
The safety and efficacy of ivermectin for the prevention and treatment of COVID-19 have been
evaluated in clinical trials and observational cohorts. Summaries of the studies that informed the
COVID-19 Treatment Guidelines Panel’s (the Panel) recommendation can be found in Table 7b. The
Panel reviewed additional studies, but these studies are not summarized in Table 7b because they have
study design limitations or results that make them less definitive and informative.
Recommendation
• The Panel recommends against the use of ivermectin for the treatment of COVID-19 (AIIa).
Rationale
The Panel’s recommendation is primarily informed by adequately powered, randomized trials of
ivermectin that reported clinical outcomes. Studies that randomized participants to receive ivermectin or
a matched placebo had the greatest impact on the Panel’s recommendation.6-13
Trials have failed to find a clinical benefit of using ivermectin to treat COVID-19 in outpatients. In
TOGETHER, an adaptive platform trial conducted in Brazil, there was no apparent difference between
the ivermectin and placebo arms for the primary outcome of risk of emergency department visits
or hospitalization (14.7% vs. 16.4%).14 In addition, there was no statistically significant difference
between the ivermectin and placebo arms in mortality (3.1% vs. 3.5%). In COVID-OUT, a randomized
factorial trial, the use of ivermectin did not reduce the occurrence of a composite outcome of emergency
department visits, hospitalization, or death when compared with a matched control (5.7% vs. 4.1%).6
The ACTIV-6 trial was an adaptive platform trial conducted in outpatients with mild to moderate
COVID-19 in the United States.15,16 Participants were randomized to receive an ivermectin regimen
(either 400 μg/kg for 3 days or 600 μg/kg for 6 days) or a matching placebo. In the 400 μg/kg phase of
the study, the median time to sustained recovery was 12 days for the ivermectin arm and 13 days for the
placebo arm. In the 600 μg/kg phase of the study, the median time to sustained recovery was 11 days for
both arms.
I-TECH, an open-label trial conducted in Malaysia, found no difference between the ivermectin
and standard of care arms in the occurrence of the primary outcome of risk of progression to severe
COVID-19 (21.6% vs. 17.3%).17 Patients in the ivermectin arm had a lower risk of mortality than those
in the standard of care arm (relative risk 0.31; 95% CI, 0.09–1.11; P = 0.09), but this difference was not
statistically significant.
COVID-19 Treatment Guidelines 371
References
1. Omura S, Crump A. Ivermectin: panacea for resource-poor communities? Trends Parasitol. 2014;30(9):445-
455. Available at: https://www.ncbi.nlm.nih.gov/pubmed/25130507.
2. Kircik LH, Del Rosso JQ, Layton AM, Schauber J. Over 25 years of clinical experience with ivermectin: an
overview of safety for an increasing number of indications. J Drugs Dermatol. 2016;15(3):325-332. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/26954318.
3. Caly L, Druce JD, Catton MG, Jans DA, Wagstaff KM. The FDA-approved drug ivermectin inhibits the
replication of SARS-CoV-2 in vitro. Antiviral Res. 2020;178:104787. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32251768.
4. Chaccour C, Hammann F, Ramón-García S, Rabinovich NR. Ivermectin and COVID-19: keeping rigor in
times of urgency. Am J Trop Med Hyg. 2020;102(6):1156-1157. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32314704.
5. Guzzo CA, Furtek CI, Porras AG, et al. Safety, tolerability, and pharmacokinetics of escalating high doses of
ivermectin in healthy adult subjects. J Clin Pharmacol. 2002;42(10):1122-1133. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12362927.
6. Bramante CT, Huling JD, Tignanelli CJ, et al. Randomized trial of metformin, ivermectin, and fluvoxamine
for COVID-19. N Engl J Med. 2022;387(7):599-610. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/36070710.
7. Vallejos J, Zoni R, Bangher M, et al. Ivermectin to prevent hospitalizations in patients with COVID-19
(IVERCOR-COVID19) a randomized, double-blind, placebo-controlled trial. BMC Infect Dis.
2021;21(1):635. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34215210.
8. López-Medina E, López P, Hurtado IC, et al. Effect of ivermectin on time to resolution of symptoms among
adults with mild COVID-19: a randomized clinical trial. JAMA. 2021;325(14):1426-1435. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33662102.
9. Buonfrate D, Chesini F, Martini D, et al. High-dose ivermectin for early treatment of COVID-19 (COVER
study): a randomised, double-blind, multicentre, phase II, dose-finding, proof-of-concept clinical trial. Int J
Antimicrob Agents. 2022;59(2):106516. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34999239.
10. Abd-Elsalam S, Noor RA, Badawi R, et al. Clinical study evaluating the efficacy of ivermectin in COVID-19
treatment: a randomized controlled study. J Med Virol. 2021;93(10):5833-5838. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34076901.
11. Ravikirti, Roy R, Pattadar C, et al. Evaluation of ivermectin as a potential treatment for mild to moderate
COVID-19: a double-blind randomized placebo controlled trial in Eastern India. J Pharm Pharm Sci.
2021;24:343-350. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34265236.
12. Mohan A, Tiwari P, Suri TM, et al. Single-dose oral ivermectin in mild and moderate COVID-19 (RIVET-
COV): a single-centre randomized, placebo-controlled trial. J Infect Chemother. 2021;27(12):1743-1749.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/34483029.
13. Bermejo Galan LE, Dos Santos NM, Asato MS, et al. Phase 2 randomized study on chloroquine,
COVID-19 Treatment Guidelines 372
The clinical trials described in this table are the RCTs that had the greatest impact on the Panel’s recommendation. The Panel reviewed other
clinical studies that evaluated the use of IVM for the treatment of COVID-19.1-26 However, those studies have limitations that make them less
definitive and informative than the studies summarized in this table.
ACTIV-6: Double-Blind RCT of Ivermectin 600 μg/kg in Outpatients With Mild to Moderate COVID-19 in the United States27
Key Inclusion Criteria Participant Characteristics Key Limitation
• Aged ≥30 years • Median age 48 years; 59.1% women • The low number of events limited
• Not hospitalized • 38.1% with BMI >30; 9.2% with DM; 26.8% with HTN the power to determine an effect on
hospitalization and death.
• Positive SARS-CoV-2 test result within past 10 days • 83.6% received ≥2 COVID-19 vaccine doses.
• ≥2 COVID-19 symptoms for ≤7 days • Median of 5 days from symptom onset to receipt of Interpretation
study drug • Among outpatients with COVID-19, IVM
Key Exclusion Criteria 600 μg/kg PO once daily for 6 days did
• End-stage kidney disease Primary Outcome not shorten time to sustained recovery
• Liver failure or decompensated cirrhosis • Median time to sustained recovery: 11 days in IVM arm or reduce incidence of hospitalization or
vs. 11 days in placebo arm (HR 1.02; 95% CrI, 0.92– death.
Interventions 1.13)
• IVM 600 μg/kg PO once daily for 6 days (n = 602) Secondary Outcome
• Placebo (n = 604) • Hospitalization or death by Day 28: 5 (0.8%) in IVM arm
Primary Endpoint vs. 2 (0.3%) in placebo arm
• Time to sustained recovery (i.e., ≥3 consecutive days Safety Outcomes
without symptoms) • Occurrence of AEs: 52 of 566 patients (9.2%) in IVM arm
Key Secondary Endpoint vs. 41 of 576 patients (7.1%) in placebo arm
• Hospitalization or death by Day 28 • Occurrence of SAEs: 5 of 566 patients (0.9%) in IVM arm
vs. 3 of 576 patients (0.5%) in placebo arm
Safety Endpoint
• Occurrence of AEs and SAEs
ACTIV-6: Double-Blind RCT of Ivermectin 400 μg/kg Once Daily in Outpatients With Mild to Moderate COVID-19 in the United States28
Key Inclusion Criteria Participant Characteristics Key Limitation
• Aged ≥30 years • Mean age 48 years; 59% women • The low number of events limited
• Not hospitalized • 41% with BMI >30; 11.5% with DM; 26% with HTN the power to determine an effect on
hospitalization and death.
• Positive SARS-CoV-2 test result within past 10 days • 47% received ≥2 COVID-19 vaccine doses.
• ≥2 COVID-19 symptoms for ≤7 days • Median of 6 days from symptom onset to receipt of study Interpretation
drug • Among outpatients with COVID-19, IVM
Key Exclusion Criteria 400 μg/kg PO once daily for 3 days did
• End-stage kidney disease Primary Outcome not shorten time to sustained recovery
• Liver failure or decompensated cirrhosis • Median time to sustained recovery: 12 days in IVM arm or reduce incidence of hospitalization or
vs. 13 days in placebo arm (HR 1.07; 95% CrI, 0.96– death.
Interventions 1.17)
• IVM 400 μg/kg PO once daily for 3 days (n = 817) Secondary Outcome
• Placebo (n = 774) • Hospitalization or death by Day 28: 10 (1.2%) in IVM arm
Primary Endpoint vs. 9 (1.2%) in placebo arm
• Time to sustained recovery (i.e., ≥3 consecutive days Safety Outcomes
without symptoms) • Occurrence of AEs: 24 of 766 patients (3.1%) in IVM arm
Key Secondary Endpoint vs. 27 of 724 patients (3.7%) in placebo arm
• Hospitalization or death by Day 28 • Occurrence of SAEs: 9 of 766 patients (1.2%) in IVM arm
vs. 9 of 724 patients (1.2%) in placebo arm
Safety Endpoint
• Occurrence of AEs and SAEs
TOGETHER: Double-Blind, Adaptive RCT of Ivermectin in Nonhospitalized Patients With COVID-19 in Brazil29
Key Inclusion Criteria Participant Characteristics Key Limitations
• Positive SARS-CoV-2 antigen test result • Median age 49 years; 46% aged ≥50 years; 58% • Health care facility capacity may have
• Within 7 days of symptom onset women; 95% self-identified as mixed race influenced the number and duration of
• Most prevalent risk factor: 50% with obesity ED visits and hospitalizations.
• ≥1 high-risk factor for disease progression (e.g., aged
>50 years, comorbidities, immunosuppression) • 44% within 3 days of symptom onset at enrollment • No details on safety outcomes (e.g., type
of treatment-emergent AEs) other than
Interventions Primary Outcome grading were reported.
• IVM 400 μg/kg PO once daily for 3 days (n = 679) • Composite of ED observation >6 hours or hospitalization Interpretation
• Placebo (n = 679; not all patients received IVM placebo) for COVID-19 by Day 28 (ITT): 100 (14.7%) in IVM arm vs.
111 (16.4%) in placebo arm (relative risk 0.90; 95% CrI, • In outpatients with recent SARS-CoV-2
Primary Endpoint 0.70–1.16) infection, IVM did not reduce the need
• Composite of ED observation >6 hours or hospitalization for ED visits or hospitalization when
• 171 (81%) of events were hospitalizations (ITT) compared with placebo.
for COVID-19 by Day 28
Secondary Outcomes
Key Secondary Endpoints
• No difference between IVM arm and placebo arm in:
• Viral clearance at Day 7
• Viral clearance at Day 7 (relative risk 1.00; 95% CrI,
• All-cause mortality 0.68–1.46)
• Occurrence of AEs • All-cause mortality: 21 (3.1%) vs. 24 (3.5%) (relative
risk 0.88; CrI, 0.49–1.55)
• Occurrence of AEs
COVID-OUT: RCT of Metformin, Ivermectin, and Fluvoxamine in Nonhospitalized Adults With COVID-19 in the United States30
Key Inclusion Criteria Participant Characteristics Key Limitations
• Aged 30–85 years • Median age 46 years; 56% women; 82% White • Study included SpO2 measurements
• BMI ≥25 or ≥23 if Asian or Latinx • Median BMI 30 using home pulse oximeters as 1 of
the composite measures of the primary
• Laboratory-confirmed SARS-CoV-2 infection within 3 days • 27% with CVD endpoint. However, the FDA has issued
of randomization • 52% received primary COVID-19 vaccination series. a statement concerning the accuracy
• ≤7 days of COVID-19 symptoms • Mean of 4.8 days of symptoms of these home pulse oximeters, making
Key Exclusion Criteria this study endpoint less reliable.
• Approximately 68% enrolled while Delta was the
• Immunocompromised dominant variant; approximately 29% enrolled while • SpO2 data were incomplete or missing
Omicron was dominant. for 30% of the patients.
• Hepatic impairment
Primary Outcomes • The low number of events limited
• Stage 4–5 chronic kidney disease or eGFR <45 mL/ the power to determine the effect on
min/1.73 m² • Composite of hypoxemia, ED visit, hospitalization, or hospitalization and death.
death by Day 14: 105 (25.8%) in IVM arm vs. 96 (24.6%)
Interventions Interpretations
in control arm (aOR 1.05; 95% CI, 0.76–1.45, P = 0.78)
• IVM 390–470 ug/kg PO once daily for 3 days (n = 410) in • IVM did not prevent the composite
• No difference between IVM alone arm and placebo alone
the following arms: endpoint of hypoxemia, ED visit,
arm in occurrence of primary endpoint (aOR 1.06; 95%
• IVM alone (n = 206) CI, 0.67–1.67) hospitalization, or death.
• Metformin plus IVM (n = 204) • ED visit, hospitalization, or death by Day 14 in a • No primary, secondary, or subgroup
• IVM control (n = 398), which included the following arms: prespecified secondary analysis: 23 (5.7%) in IVM arm analysis demonstrated a benefit for the
vs. 16 (4.1%) in control arm (aOR 1.39; 95% CI, 0.72– use of IVM over placebo.
• Placebo alone (n = 203)
2.69)
• Metformin alone (n = 195)
• Hospitalization or death by Day 14 in a prespecified
Primary Endpoints secondary analysis: 4 (1.0%) in IVM arm vs. 5 (1.3%) in
• Composite of hypoxemia (SpO2 ≤93%, as measured by a control arm (aOR 0.73; 95% CI, 0.19–2.77); 1 death in
home pulse oximeter), ED visit, hospitalization, or death IVM arm vs. 0 deaths in control arm
by Day 14 Secondary Outcomes
• A prespecified secondary analysis evaluated the • No difference between arms in total symptom severity
occurrence of ED visits, hospitalization, or death by Day score by Day 14
14.
• Drug discontinuation or interruption: 20% in IVM arm vs.
Key Secondary Endpoints 25% in placebo alone arm
• Total symptom severity score by Day 14, as measured by
a symptom severity scale
• Drug discontinuation or interruption
IVERCOR-COVID19: Double-Blind, Placebo-Controlled, Randomized Trial of Ivermectin in Nonhospitalized Patients With COVID-19 in Argentina31
Key Inclusion Criterion Participant Characteristics Key Limitation
• Positive SARS-CoV-2 RT-PCR result within 48 hours of • Mean age 42 years; 8% aged ≥65 years; 47% women • Study enrolled a young population with
screening • 24% with HTN; 10% with DM; 58% with ≥1 comorbidity few of the comorbidities that predict
disease progression.
Key Exclusion Criteria • Median of 4 days from symptom onset
• Required supplemental oxygen or hospitalization Interpretation
Primary Outcome
• Concomitant use of CQ or HCQ • Among patients who had recently
• Hospitalization for any reason: 5.6% in IVM arm vs. 8.3% acquired SARS-CoV-2 infection, there
Interventions in placebo arm (OR 0.65; 95% CI, 0.32–1.31; P = 0.23) was no evidence that IVM provided any
• Weight-based dose of IVM PO at enrollment and 24 Secondary Outcomes clinical benefit.
hours later for a maximum total dose of 48 mg (n = • Need for MV: 2% in IVM arm vs. 1% in placebo arm (P =
250) 0.7)
• Placebo (n = 251) • All-cause mortality: 2% in IVM arm vs. 1% in placebo
Primary Endpoint arm (P = 0.7)
• Hospitalization for any reason • Occurrence of AEs: 18% in IVM arm vs. 21% in placebo
arm (P = 0.6)
Key Secondary Endpoints
• Need for MV
• All-cause mortality
• Occurrence of AEs
Double-Blind, Placebo-Controlled, Randomized Trial of Ivermectin in Patients With Mild COVID-19 in Colombia32
Key Inclusion Criteria Participant Characteristics Key Limitations
• Positive SARS-CoV-2 RT-PCR or antigen test result • Median age 37 years; 4% aged ≥65 years in IVM arm, 8% • Due to low event rates, the primary
• ≤7 days of COVID-19 symptoms in placebo arm; 39% men in IVM arm, 45% in placebo endpoint changed from the proportion of
arm patients with clinical deterioration to the
• Mild disease time to symptom resolution during the
• 79% with no known comorbidities
Key Exclusion Criteria trial.
• Median of 5 days from symptom onset to randomization
• Asymptomatic disease • The study enrolled younger, healthier
Primary Outcome patients, a population that does not
• Severe pneumonia
• Median time to symptom resolution: 10 days in IVM arm typically develop severe COVID-19.
• Hepatic dysfunction vs. 12 days in placebo arm (HR 1.07; P = 0.53) Interpretation
Interventions • Symptoms resolved by Day 21: 82% in IVM arm vs. • In patients with mild COVID-19, IVM
• IVM 300 μg/kg PO once daily for 5 days (n = 200) 79% in placebo arm 300 μg/kg once daily for 5 days did not
• Placebo PO (n = 198) Secondary Outcomes improve the time to symptom resolution.
Primary Endpoint • No difference between arms in proportion of patients who
• Time to symptom resolution within 21 days showed clinical deterioration or required escalation of
care
Key Secondary Endpoints • Occurrence of AEs:
• Clinical deterioration • Discontinued treatment due to AEs: 8% in IVM arm vs.
• Escalation of care 3% in placebo arm
• Occurrence of AEs • No SAEs related to intervention
I-TECH: Open-Label RCT of Ivermectin in Patients With Mild to Moderate COVID-19 in Malaysia33
Key Inclusion Criteria Participant Characteristics Key Limitation
• Positive SARS-CoV-2 RT-PCR or antigen test result • Mean age 63 years; 55% women • Open-label study
within 7 days of symptom onset • 68% received ≥1 COVID-19 vaccine dose; 52% received Interpretation
• Aged ≥50 years 2 doses.
• In patients with mild to moderate
• ≥1 comorbidities • Most common comorbidities: 75% with HTN; 54% with COVID-19, there was no evidence
DM; 24% with dyslipidemia that IVM provided any clinical benefit,
Key Exclusion Criteria
• Mean of 5 days symptom duration including no evidence that IVM reduced
• Required supplemental oxygen the risk of progression to severe disease.
• Severe hepatic impairment (ALT >10 times the ULN) Primary Outcome
• Progression to severe COVID-19 (mITT): 52 (21.6%)
Interventions
in IVM plus SOC arm vs. 43 (17.3%) in SOC alone arm
• IVM 400 μg/kg PO once daily for 5 days plus SOC (n = (relative risk 1.25; 95% CI, 0.87–1.80; P = 0.25)
241)
Secondary Outcomes
• SOC (n = 249)
• No difference between IVM plus SOC arm and SOC alone
Primary Endpoint arm in:
• Progression to severe COVID-19 (i.e., hypoxemia • In-hospital, all-cause mortality by Day 28: 3 (1.2%) vs.
requiring supplemental oxygen to maintain SpO2 ≥95%) 10 (4.0%) (relative risk 0.31; 95% CI, 0.09–1.11; P =
Key Secondary Endpoints 0.09)
• In-hospital, all-cause mortality by Day 28 • MV: 4 (1.7%) vs. 10 (4.0%) (relative risk 0.41; 95% CI,
0.13–1.30; P = 0.17)
• MV or ICU admission
• ICU admission: 6 (2.5%) vs. 8 (3.2%) (relative risk 0.78;
• Occurrence of AEs 95% CI, 0.27–2.20; P = 0.79)
• Occurrence of AEs: 33 (13.7%) in IVM plus SOC arm vs.
11 (4.4%) in SOC alone arm; most with diarrhea (14 vs. 4)
COVER: Phase 2, Double-Blind RCT of Ivermectin in Nonhospitalized Patients With COVID-19 in Italy34
Key Inclusion Criteria Participant Characteristics Key Limitations
• Asymptomatic or oligosymptomatic disease • Median age 47 years; 58% men • Small, Phase 2 study
• SARS-CoV-2 infection confirmed by RT-PCR result • 86% with COVID-19 symptoms • 90% of subjects screened were not
• Not hospitalized or receiving supplemental oxygen • 2.2% received a COVID-19 vaccine. enrolled for various reasons.
• Recruitment stopped early because of a
Key Exclusion Criteria Primary Outcomes
decline in the number of COVID-19 cases.
• CNS disease • No SAEs related to intervention
Interpretations
• Receiving dialysis • Mean log10 reduction in VL at Day 7: 2.9 in IVM 1,200 μg/
kg arm vs. 2.5 in IVM 600 μg/kg arm vs. 2.0 in placebo • A high dose of IVM (1,200 μg/kg) appears
• Severe medical condition with <6 months survival to be safe but not well tolerated; 34% of
prognosis arm (IVM 1,200 μg/kg vs. placebo, P = 0.099; IVM 600
μg/kg vs. placebo, P = 0.122) patients discontinued therapy due to AEs.
• Use of warfarin, antiviral agents, CQ, or HCQ • There was no significant difference in
Other Outcomes
Interventions reduction of VL between IVM and placebo
• 14 (15.1%) discontinued treatment: 11 (34.4%) in IVM arms.
• IVM 1,200 μg/kg PO once daily for 5 days (n = 32) 1,200 μg/kg arm vs. 2 (6.9%) in IVM 600 μg/kg arm vs. 1
• IVM 600 μg/kg plus placebo PO once daily for 5 days (n (3.1%) in placebo arm
= 29) • All discontinuations in IVM 1,200 μg/kg arm were due to
• Placebo PO (n = 32) tolerability
Primary Endpoints
• Number of SAEs
• Change in VL at Day 7
Other Endpoint
• Drug discontinuation or interruption
Double-Blind, Placebo-Controlled, Randomized Trial of Ivermectin in Patients With Mild to Moderate COVID-19 in India36
Key Inclusion Criteria Participant Characteristics Key Limitations
• Positive SARS-CoV-2 RT-PCR or antigen test result • Mean age 53 years; 28% women • Although the primary endpoint was a
• Hospitalized with mild to moderate COVID-19 • 35% with HTN; 36% with DM negative SARS-CoV-2 RT-PCR result
on Day 6, no RT-PCR result or an
Interventions • 79% with mild COVID-19 inconclusive RT-PCR result was reported
• IVM 12 mg PO once daily for 2 days (n = 55) • Mean of 6.9 days from symptom onset for 42% of patients in the IVM arm and
• Placebo PO (n = 57) • 100% received HCQ, steroids, and antibiotics; 21% 23% in the placebo arm.
received RDV; 6% received tocilizumab. • The time to discharge was not reported,
Primary Endpoint and outcomes after discharge were not
Primary Outcome
• Negative SARS-CoV-2 RT-PCR result on Day 6 evaluated.
• Negative SARS-CoV-2 RT-PCR result on Day 6: 24% in
Key Secondary Endpoints IVM arm vs. 32% in placebo arm (rate ratio 0.8; P = Interpretation
• Symptom resolution by Day 6 0.348) • IVM provided no significant virologic or
• Discharge by Day 10 Secondary Outcomes clinical benefit for patients with mild to
moderate COVID-19.
• Need for ICU admission or MV • Symptom resolution by Day 6: 84% in IVM arm vs. 90% in
• In-hospital mortality placebo arm (rate ratio 0.9; P = 0.36)
• Discharge by Day 10: 80% in IVM arm vs. 74% in placebo
arm (rate ratio 1.1; P = 0.43)
• No difference between arms in need for ICU admission or
MV
• In-hospital mortality: 0 in IVM arm (0%) vs. 4 in placebo
arm (7%)
RIVET-COV: Double-Blind, Placebo-Controlled, Randomized Trial of Ivermectin in Patients With Mild to Moderate COVID-19 in India37
Key Inclusion Criteria Participant Characteristics Key Limitation
• Positive SARS-CoV-2 RT-PCR or antigen test result • Mean age 35 years; 89% men • Small sample size
• Nonsevere COVID-19 • 60% to 68% with mild COVID-19 (including asymptomatic Interpretation
patients); 33% to 40% with moderate COVID-19
Key Exclusion Criteria • The use of IVM did not affect the
• Median of 4–5 days symptom duration; similar across proportion of patients with negative
• CrCl <30 mL/min
arms SARS-CoV-2 RT-PCR results at Day 5 or
• Transaminases >5 times ULN the clinical outcomes.
• 10% in each arm received concurrent antivirals (RDV,
• MI, heart failure, QTc interval prolongation favipiravir, or HCQ).
• Severe comorbidity Primary Outcomes
Interventions • Negative SARS-CoV-2 RT-PCR result at Day 5: 48% in
• Single dose of IVM 24 mg PO (n = 51) IVM 24 mg arm vs. 35% in IVM 12 mg arm vs. 31% in
• Single dose of IVM 12 mg PO (n = 49) placebo arm (P = 0.30)
• Placebo (n = 52) • No significant difference between arms in decline of VL at
Day 5
Primary Endpoints
Secondary Outcomes
• Negative SARS-CoV-2 RT-PCR result at Day 5
• No difference between arms in time to symptom
• Decline of VL at Day 5 resolution
Key Secondary Endpoints • Clinical worsening at Day 14: 8% in IVM 24 mg arm vs.
• Time to symptom resolution 5% in IVM 12 mg arm vs. 11% in placebo arm (P = 0.65)
• Clinical worsening at Day 14 • No difference between arms in number of hospital-free
days at Day 28
• Number of hospital-free days at Day 28
• No difference between arms in frequency of AEs; no SAEs
• Frequency of AEs
were reported
Double-Blind RCT of Ivermectin, Chloroquine, or Hydroxychloroquine in Hospitalized Adults With Severe COVID-19 in Brazil38
Key Inclusion Criteria Participant Characteristics Key Limitations
• Hospitalized with laboratory-confirmed SARS-CoV-2 • Mean age 53 years; 58% men • Small sample size
infection • Most common comorbidities: 43% with HTN; 28% with • No clearly defined primary endpoint
• ≥1 of the following severity criteria: DM; 38% with BMI >30
Interpretation
• Dyspnea • 76% with respiratory failure on admission
• Compared to CQ or HCQ, IVM did not
• Tachypnea (>30 breaths/min) Outcomes reduce the proportion of hospitalized
• SpO2 <93% • No difference between IVM, CQ, and HCQ arms in: patients with severe COVID-19 who died
• PaO2/FiO2 <300 mm Hg or who required supplemental oxygen,
• Need for supplemental oxygen: 88% vs. 89% vs. 90% ICU admission, or MV
• Involvement of >50% of lungs confirmed by CXR or • Need for MV: 24% vs. 21% vs. 21%
CT scan
• ICU admission: 28% vs. 22% vs. 21%
Key Exclusion Criterion • Mortality: 23% vs. 21% vs. 22%
• Cardiac arrhythmia • Mean number of days of supplemental oxygen: 8 days
Interventions in each arm
• IVM 14 mg once daily for 3 days (n = 53) • No difference between arms in occurrence of AEs
• CQ 450 mg twice daily on Day 0, then once daily for 4 • Baseline characteristics significantly associated with
days (n = 61) mortality:
• HCQ 400 mg twice daily on Day 0, then once daily for 4 • Aged >60 years (HR 2.4)
days (n = 54) • DM (HR 1.9)
Endpoints • BMI >33 (HR 2.0)
• Need for supplemental oxygen, MV, or ICU admission • SpO2 <90% (HR 5.8)
• Occurrence of AEs
• Mortality
Key: AE = adverse event; ALT = alanine transaminase; BMI = body mass index; CNS = central nervous system; CQ = chloroquine; CrCl = creatinine clearance; CT =
computed tomography; CVD = cardiovascular disease; CXR = chest X-ray; DM = diabetes mellitus; ED = emergency department; eGFR = estimated glomerular filtration
rate; FDA = Food and Drug Administration; HCQ = hydroxychloroquine; HTN = hypertension; ICU = intensive care unit; ITT = intention-to-treat; IVM = ivermectin; LOS
= length of stay; MI = myocardial infarction; mITT = modified intention-to-treat; MV = mechanical ventilation; the Panel = the COVID-19 Treatment Guidelines Panel;
PaO2/FiO2 = ratio of arterial partial pressure of oxygen to fraction of inspired oxygen; PO = oral; RCT = randomized controlled trial; RDV = remdesivir; RT-PCR = reverse
transcriptase polymerase chain reaction; SAE = severe adverse event; SOC = standard of care; SpO2 = oxygen saturation; ULN = upper limit of normal; VL = viral load
Metformin has been identified as a potential COVID-19 therapeutic agent because of its possible
action against the proteins that are involved in translation, its antiviral activity in vitro, and its anti-
inflammatory and antithrombotic activities.1-4 Data from observational studies have suggested that
patients who were receiving metformin as treatment for diabetes at the time of their COVID-19
diagnosis had a lower risk of progressing to severe COVID-19.5-7 Randomized controlled trials have
provided insight into the role of metformin in treating nonhospitalized patients with COVID-19. These
trials are described below and in Table 7c.
Recommendations
• There is insufficient evidence for the COVID-19 Treatment Guidelines Panel (the Panel)
to recommend either for or against the use of metformin for the treatment of COVID-19 in
nonhospitalized patients.
• The Panel recommends against the use of metformin for the treatment of COVID-19 in
hospitalized patients, except in a clinical trial (BIII).
• Patients with COVID-19 who are receiving metformin for an underlying condition should
continue this therapy as directed by their health care provider (AIII).
Rationale
Two randomized controlled trials (the TOGETHER and COVID-OUT trials) assessed the efficacy of
using metformin in nonhospitalized patients with COVID-19. In these trials, the use of metformin did
not reduce the risk of hospitalization or death in these patients. The Panel’s recommendations are based
on the results of these trials.
Other outpatient therapies (i.e., ritonavir-boosted nirmatrelvir [Paxlovid], remdesivir, molnupiravir) have
been shown to be effective in preventing hospitalization or death in unvaccinated patients who are at
high risk of disease progression.
Considerations in Children
Although metformin is approved by the Food and Drug Administration for the treatment of type 2
diabetes mellitus in children aged >10 years, clinical trials that have evaluated its use for the treatment
of COVID-19 have not included people aged <18 years. Given the lack of clear evidence of efficacy in
adults, the Panel recommends against the use of metformin for the treatment of COVID-19 in pediatric
patients, except in a clinical trial (AIII).
Clinical Data
TOGETHER Trial
The TOGETHER trial was a placebo-controlled platform clinical trial that was conducted in Brazil.8 The
study enrolled nonhospitalized patients who had symptomatic SARS-CoV-2 infection for ≤7 days, no
history of COVID-19 vaccination, and an increased risk of progressing to severe disease. Patients were
randomized to receive extended-release metformin 750 mg (n = 215) or placebo (n = 203) twice daily
for 10 days.
The primary endpoint was a composite of retention in an emergency setting for >6 hours or
hospitalization within 28 days of randomization. Secondary endpoints included viral clearance at Days
3 and 7, clinical improvement at Day 28, time to hospitalization or death, and the occurrence of adverse
events. The study was stopped by the data and safety monitoring board for futility, as there was a low
probability of demonstrating a difference between the study arms. Overall, there was no difference
between the arms in the number of adverse events; however, the proportion of patients who experienced
grade 3 events was higher in the metformin arm (9.8%) than in the placebo arm (4.4%).
COVID-OUT Trial
The COVID-OUT trial was a Phase 3, double-blind, placebo-controlled 2 x 3 factorial trial that
evaluated the effectiveness of metformin, ivermectin, or fluvoxamine in patients with COVID-19.9
Patients were randomized to receive metformin or placebo in 1 factor and ivermectin, fluvoxamine,
or placebo in the other factor. The study enrolled nonhospitalized adults within 3 days of a confirmed
diagnosis of COVID-19 and ≤7 days from symptom onset. Patients were aged 30 to 85 years and
overweight. Those with stage 4 or 5 chronic kidney disease or an estimated glomerular filtration rate of
<45 mL/min/1.73 m2 were excluded. The metformin arm included those assigned to receive immediate-
release oral metformin (titrated over several days to a final daily dose of 1,500 mg) alone or in
combination with ivermectin or fluvoxamine. The control arm included those who received placebo with
or without ivermectin or fluvoxamine.
The primary endpoint was a composite of development of hypoxemia (defined as oxygen saturation
≤93%, as measured by a home pulse oximeter), emergency department visit, hospitalization, or death by
Day 14. While this study was underway, the Food and Drug Administration raised concerns about the
accuracy of home pulse oximeters. Approximately 50% of the patients received a primary COVID-19
vaccine series. The analyses showed no benefit for any of the 3 investigational agents in preventing the
primary endpoint. In addition, the use of these agents did not lower the severity of COVID-19 symptoms
over 14 days. A prespecified secondary analysis determined that, over 14 days of follow-up, those who
received metformin had a lower risk of an emergency department visit, hospitalization, or death than
those who did not receive metformin (adjusted OR 0.58; 95% CI, 0.35–0.94). A key secondary endpoint
in the analysis was a composite of hospitalization or death by Day 28. Eight of 596 patients (1.3%) who
received metformin met this endpoint compared with 19 of 601 patients (3.2%) who did not receive
COVID-19 Treatment Guidelines 390
References
1. Karam BS, Morris RS, Bramante CT, et al. mTOR inhibition in COVID-19: a commentary and review of
efficacy in RNA viruses. J Med Virol. 2021;93(4):1843-1846. Available at: https://www.ncbi.nlm.nih.gov/
pubmed/33314219.
2. Del Campo JA, García-Valdecasas M, Gil-Gómez A, et al. Simvastatin and metformin inhibit cell growth in
hepatitis C virus infected cells via mTOR increasing PTEN and autophagy. PLoS One. 2018;13(1):e0191805.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/29385181.
3. Postler TS, Peng V, Bhatt DM, Ghosh S. Metformin selectively dampens the acute inflammatory response
through an AMPK-dependent mechanism. Sci Rep. 2021;11(1):18721. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34548527.
4. Xin G, Wei Z, Ji C, et al. Metformin uniquely prevents thrombosis by inhibiting platelet activation and
mtDNA release. Sci Rep. 2016;6:36222. Available at: https://www.ncbi.nlm.nih.gov/pubmed/27805009.
5. Li Y, Yang X, Yan P, Sun T, Zeng Z, Li S. Metformin in patients with COVID-19: a systematic review and
meta-analysis. Front Med (Lausanne). 2021;8:704666. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34490296.
6. Bramante CT, Buse J, Tamaritz L, et al. Outpatient metformin use is associated with reduced severity of
COVID-19 disease in adults with overweight or obesity. J Med Virol. 2021;93(7):4273-4279. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33580540.
7. Luo P, Qiu L, Liu Y, et al. Metformin treatment was associated with decreased mortality in COVID-19 patients
with diabetes in a retrospective analysis. Am J Trop Med Hyg. 2020;103(1):69-72. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32446312.
8. Reis G, Dos Santos Moreira Silva EA, Medeiros Silva DC, et al. Effect of early treatment with metformin
on risk of emergency care and hospitalization among patients with COVID-19: the TOGETHER randomized
platform clinical trial. Lancet Reg Health Am. 2022;6:100142. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34927127.
9. Bramante CT, Huling JD, Tignanelli CJ, et al. Randomized trial of metformin, ivermectin, and fluvoxamine
for COVID-19. N Engl J Med. 2022;387(7):599-610. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/36070710.
10. Bramante CT, Buse JB, Liebovitz DM, et al. Outpatient treatment of COVID-19 and incidence of post-
COVID-19 condition over 10 months (COVID-OUT): a multicentre, randomised, quadruple-blind,
parallel-group, Phase 3 trial. Lancet Infect Dis. 2023;23(10):1119-1129. Available at: https://www.ncbi.nlm.
nih.gov/pubmed/37302406.
The Panel’s recommendations for metformin are based on data from the clinical trials described in this table.
References
1. Reis G, Dos Santos Moreira Silva EA, Medeiros Silva DC, et al. Effect of early treatment with metformin on risk of emergency care and hospitalization
among patients with COVID-19: the TOGETHER randomized platform clinical trial. Lancet Reg Health Am. 2022;6:100142. Available at: https://
www.ncbi.nlm.nih.gov/pubmed/34927127.
2. Bramante CT, Huling JD, Tignanelli CJ, et al. Randomized trial of metformin, ivermectin, and fluvoxamine for COVID-19. N Engl J Med.
2022;387(7):599-610. Available at: https://www.ncbi.nlm.nih.gov/pubmed/36070710.
• The information in this table is derived from data on the use of these drugs for FDA-approved indications or in investigational trials. It
is supplemented with data on the use of these drugs in patients with COVID-19 or MIS-C, when available.
• For dose modifications for patients with organ failure or those who require extracorporeal devices, please refer to product labels, when
available.
• There are currently not enough data to determine whether certain medications can be safely coadministered with therapies for the
treatment of COVID-19. When using concomitant medications with similar toxicity profiles, consider performing additional safety
monitoring.
• The potential additive, antagonistic, or synergistic effects and the safety of using certain combination therapies for the treatment of
COVID-19 are unknown. Clinicians are encouraged to report AEs to the FDA MedWatch program.
• For drug-drug interaction information, please refer to product labels and visit the Liverpool COVID-19 Drug Interactions website.
Monitoring
Dosing Regimens Adverse Events Drug-Drug Interaction Potential Comments
Parameters
Fluvoxamine
Not recommended by the Panel for the treatment of COVID-19 in nonhospitalized patients.
Doses for COVID-19 in Clinical • Nausea • Hepatic function • CYP2D6 substrate • Fluvoxamine may enhance
Trials • Diarrhea • Drug-drug • Fluvoxamine inhibits several CYP the anticoagulant effects
• Fluvoxamine 50 mg PO twice daily interactions isoenzymes (CYP1A2, CYP2C9, of antiplatelets and
• Dyspepsia anticoagulants. Consider
• Fluvoxamine 100 mg PO twice daily • Withdrawal CYP3A4, CYP2C19, CYP2D6).
• Asthenia additional monitoring
• Fluvoxamine 100 mg PO 3 times symptoms • Coadministration of tizanidine, when these drugs are used
• Insomnia during dose thioridazine, alosetron, or
daily concomitantly with fluvoxamine.
• Somnolence tapering pimozide with fluvoxamine is
contraindicated. • The use of MAOIs concomitantly
• Sweating
with fluvoxamine or within
• Suicidal ideation (rare) 14 days of treatment
with fluvoxamine is
contraindicated.
Intravenous Immunoglobulin
Primarily used for the treatment of MIS-C. Currently under investigation in clinical trials.
Dose for MIS-C • Allergic reactions, including • Transfusion-related • Not a CYP substrate; no drug- • Rapid infusion should be
• 1 dose of IVIG 2 g/kg IBW IV, up to anaphylaxis reactions drug interactions expected avoided in patients with renal
a maximum total dose of 100 g • Renal failure • Vital signs at dysfunction or those who are
baseline and during at risk of thromboembolic
• In the event of cardiac • Thromboembolic events events.
dysfunction or fluid overload, and after infusion
• Aseptic meningitis syndrome
consider administering IVIG in • Renal function;
divided doses (i.e., IVIG 1 g/ • Hemolysis discontinue
kg IBW IV, up to 50 g daily for 2 • TRALI treatment if
doses). • Transmission of infectious renal function
pathogens deteriorates.
• AEs may vary by formulation.
• Risk and severity of AEs may
increase with high dose or
rapid infusion.
Metformin
There is insufficient evidence for the Panel to recommend either for or against the use of metformin in nonhospitalized patients. Not recommended by the Panel for
the treatment of COVID-19 in hospitalized patients, except in a clinical trial.
Doses for COVID-19 in Clinical • Diarrhea • Renal function • OCT1 and OCT2 substrate • Alcohol intake may increase
Trials • Nausea and vomiting • Hepatic function • Drugs that interfere with OCT the risk of lactic acidosis.
• Immediate-release metformin • Headache • Drug-drug systems (e.g., cimetidine,
500 mg PO on Day 1; 500 mg interactions dolutegravir, ranolazine,
twice daily on Days 2–5; and 500 • Lactic acidosis vandetanib) could increase
mg in morning and 1,000 mg in • Alcohol use disorder systemic exposure to metformin.
evening on Days 6–14 • Concomitant use with carbonic
• Extended-release metformin 750 anhydrase inhibitors (e.g.,
mg PO twice daily for 10 days acetazolamide, topiramate,
zonisamide) may increase the
risk of lactic acidosis.
Key: AE = adverse event; CYP = cytochrome P450; FDA = Food and Drug Administration; IBW = ideal body weight; IV = intravenous; IVIG = intravenous
immunoglobulin; MAOI = monoamine oxidase inhibitor; MIS-C = multisystem inflammatory syndrome in children; OCT = organic cation transporter; the Panel = the
COVID-19 Treatment Guidelines Panel; PO = oral; TRALI = transfusion-related acute lung injury
Summary Recommendations
Vitamin C
• There is insufficient evidence for the COVID-19 Treatment Guidelines Panel (the Panel) to recommend either for or
against the use of vitamin C for the treatment of COVID-19 in nonhospitalized patients.
• The Panel recommends against the use of vitamin C for the treatment of COVID-19 in hospitalized patients (AIIa).
Vitamin D
• There is insufficient evidence for the Panel to recommend either for or against the use of vitamin D for the prevention or
treatment of COVID-19.
Zinc
• There is insufficient evidence for the Panel to recommend either for or against the use of zinc for the treatment of
COVID-19.
• The Panel recommends against using zinc supplementation above the recommended dietary allowance (i.e., zinc
11 mg daily for men, zinc 8 mg daily for nonpregnant women) for the prevention of COVID-19, except in a clinical trial
(BIII).
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
Vitamin C (ascorbic acid) is a water-soluble vitamin that has been considered for potential beneficial
effects in patients with varying degrees of illness severity. It is an antioxidant and free radical scavenger
that has anti-inflammatory properties, influences cellular immunity and vascular integrity, serves as a
cofactor in endogenous catecholamine generation, and has been studied in many disease states, including
COVID-19.1,2
Rationale
Because patients who are not critically ill with COVID-19 are less likely to experience oxidative stress
or severe inflammation, the role of vitamin C in this setting is unknown.
Rationale
Randomized clinical trials have failed to demonstrate benefit from vitamin C as a therapeutic
COVID-19 Treatment Guidelines 398
Other Consideration
High concentrations of circulating vitamin C may affect the accuracy of point-of-care glucometers.8,9
References
1. Fisher BJ, Seropian IM, Kraskauskas D, et al. Ascorbic acid attenuates lipopolysaccharide-induced acute lung
injury. Crit Care Med. 2011;39(6):1454-1460. Available at: https://www.ncbi.nlm.nih.gov/pubmed/21358394.
2. Wei XB, Wang ZH, Liao XL, et al. Efficacy of vitamin C in patients with sepsis: an updated meta-analysis.
Eur J Pharmacol. 2020;868:172889. Available at: https://www.ncbi.nlm.nih.gov/pubmed/31870831.
3. Thomas S, Patel D, Bittel B, et al. Effect of high-dose zinc and ascorbic acid supplementation vs usual care
on symptom length and reduction among ambulatory patients with SARS-CoV-2 infection: the COVID A to Z
randomized clinical trial. JAMA Netw Open. 2021;4(2):e210369. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33576820.
4. LOVIT-COVID Investigators on behalf of the Canadian Critical Care Trials Group and the REMAP-CAP
Investigators. Intravenous vitamin C for patients hospitalized with COVID-19: two harmonized randomized
clinical trials. JAMA. 2023;330(18):1745-1759. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/37877585.
5. Kumari P, Dembra S, Dembra P, et al. The role of vitamin C as adjuvant therapy in COVID-19. Cureus.
2020;12(11):e11779. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33409026.
6. Zhang J, Rao X, Li Y, et al. Pilot trial of high-dose vitamin C in critically ill COVID-19 patients. Ann
Intensive Care. 2021;11(1):5. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33420963.
7. Coppock D, Violet PC, Vasquez G, et al. Pharmacologic ascorbic acid as early therapy for hospitalized patients
with COVID-19: a randomized clinical trial. Life (Basel). 2022;12(3):453. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35330204.
8. Hager DN, Martin GS, Sevransky JE, Hooper MH. Glucometry when using vitamin C in sepsis: a note of
caution. Chest. 2018;154(1):228-229. Available at: https://www.ncbi.nlm.nih.gov/pubmed/30044741.
9. Food and Drug Administration. Blood glucose monitoring devices. 2019. Available at: https://www.fda.gov/
medical-devices/in-vitro-diagnostics/blood-glucose-monitoring-devices. Accessed November 21, 2023.
Vitamin D is critical for bone and mineral metabolism. Because the vitamin D receptor is present
on immune cells such as B cells, T cells, and antigen-presenting cells, and because these cells can
synthesize the active vitamin D metabolite, vitamin D also has the potential to modulate innate and
adaptive immune responses.1 It is postulated that these immunomodulatory effects of vitamin D could
potentially protect against SARS-CoV-2 infection or decrease the severity of COVID-19.
Vitamin D deficiency (defined as a serum concentration of 25-hydroxyvitamin D ≤20 ng/mL) is
common in the United States, particularly among people who identified as Hispanic or non-Hispanic
Black.2 These groups are overrepresented among cases of COVID-19 in the United States.3 Vitamin
D deficiency is also more common in older patients and patients with obesity and hypertension; these
factors have been associated with worse outcomes in patients with COVID-19.4 High levels of vitamin D
may cause hypercalcemia and nephrocalcinosis.5
Recommendation
• There is insufficient evidence for the COVID-19 Treatment Guidelines Panel (the Panel)
to recommend either for or against the use of vitamin D for the prevention or treatment of
COVID-19.
Rationale
The results from several cohort studies, clinical trials, and meta-analyses on the use of vitamin D for
the prevention or treatment of COVID-19 have been published in peer-reviewed journals or have been
made available as manuscripts ahead of peer review. However, most of these studies had significant
limitations, such as small sample sizes or a lack of randomization and/or blinding. In addition, these
studies used varying doses and formulations of vitamin D, enrolled participants with a range of
COVID-19 severities, included different concomitant medications, and measured different study
outcomes. All these factors make it difficult to compare results across studies. The studies summarized
below are those that have had the greatest impact on the Panel's recommendations.
Although multiple observational cohort studies suggest that people with low vitamin D levels are
at increased risk of SARS-CoV-2 infection and worse clinical outcomes after infection (e.g., higher
mortality), clear evidence that vitamin D supplementation provides protection against infection or
improves outcomes in patients with COVID-19 is still lacking.6,7
References
1. Aranow C. Vitamin D and the immune system. J Investig Med. 2011;59(6):881-886. Available at: https://www.
ncbi.nlm.nih.gov/pubmed/21527855.
Increased intracellular zinc concentrations efficiently impair the replication of a number of RNA
viruses.1 Zinc has been shown to enhance cytotoxicity and induce apoptosis when used in vitro with a
zinc ionophore (e.g., chloroquine). Chloroquine has also been shown to enhance intracellular zinc uptake
in vitro.2 Zinc levels are difficult to measure accurately, as zinc is distributed as a component of various
proteins and nucleic acids.3
The recommended dietary allowance for elemental zinc is 11 mg daily for men and 8 mg daily for
nonpregnant women.4 Long-term zinc supplementation can cause copper deficiency with subsequent
reversible hematologic defects (i.e., anemia, leukopenia) and potentially irreversible neurologic
manifestations (i.e., myelopathy, paresthesia, ataxia, spasticity).5-7 The use of zinc supplementation for
durations as short as 10 months has been associated with copper deficiency.3 In addition, oral zinc can
decrease the absorption of medications that bind with polyvalent cations (e.g., fluoroquinolones, HIV
integrase inhibitors, tetracyclines).4
Recommendations
• There is insufficient evidence for the COVID-19 Treatment Guidelines Panel (the Panel) to
recommend either for or against the use of zinc for the treatment of COVID-19.
• The Panel recommends against using zinc supplementation above the recommended dietary
allowance (i.e., zinc 11 mg daily for men, zinc 8 mg daily for nonpregnant women) for the
prevention of COVID-19, except in a clinical trial (BIII).
Rationale
The results from some cohort studies and clinical trials that evaluated the use of zinc in patients with
COVID-19 have been published in peer-reviewed journals or have been made available as manuscripts
ahead of peer review. However, most of these studies have significant limitations, such as small
sample sizes or a lack of randomization or blinding. In addition, these studies used varying doses and
formulations of zinc, enrolled participants with a range of COVID-19 severities, included different
concomitant medications, and measured different study outcomes. All of these factors make it difficult
to compare results across studies. Because zinc has not been shown to have a clinical benefit and may
be harmful, the Panel recommends against using zinc supplementation above the recommended dietary
allowance for the prevention of COVID-19, except in a clinical trial (BIII).
The studies summarized below are those that have had the greatest impact on the Panel’s
recommendations.
Clinical Data
In a double-blind, multicenter trial in Tunisia, nonhospitalized and hospitalized adults with COVID-19
were randomized within 7 days of symptom onset to receive elemental zinc 25 mg orally twice daily (n
= 231) or matching placebo (n = 239) for 15 days.8 Approximately 20% of these patients had received a
COVID-19 vaccine prior to enrollment. During the study, none of the patients received antiviral drugs,
and <40% received corticosteroids.
The primary outcome in the study was a composite of death due to COVID-19 or intensive care unit
References
1. te Velthuis AJW, van den Worm SHE, Sims AC, et al. Zn(2+) inhibits coronavirus and arterivirus RNA
polymerase activity in vitro and zinc ionophores block the replication of these viruses in cell culture. PLoS
Pathog. 2010;6(11):e1001176. Available at: https://www.ncbi.nlm.nih.gov/pubmed/21079686.
2. Xue J, Moyer A, Peng B, et al. Chloroquine is a zinc ionophore. PLoS One. 2014;9(10):e109180. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25271834.
3. Hambridge K. The management of lipohypertrophy in diabetes care. Br J Nurs. 2007;16(9):520-524. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/17551441.
4. Office of Dietary Supplements, National Institutes of Health. Zinc fact sheet for health professionals. 2022.
Available at: https://ods.od.nih.gov/factsheets/Zinc-HealthProfessional. Accessed November 21, 2023.
5. Myint ZW, Oo TH, Thein KZ, Tun AM, Saeed H. Copper deficiency anemia: review article. Ann Hematol.
2018;97(9):1527-1534. Available at: https://www.ncbi.nlm.nih.gov/pubmed/29959467.
6. Kumar N. Copper deficiency myelopathy (human swayback). Mayo Clin Proc. 2006;81(10):1371-1384.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/17036563.
COVID-19 Treatment Guidelines 405
Summary Recommendations
• Patients with COVID-19 who are receiving concomitant medications (e.g., angiotensin-converting enzyme [ACE]
inhibitors, angiotensin receptor blockers [ARBs], HMG-CoA reductase inhibitors [statins], systemic or inhaled
corticosteroids, nonsteroidal anti-inflammatory drugs, acid-suppressive therapy) for underlying medical conditions
should not discontinue ACE inhibitors and ARBs (AIIa) or other medications (AIII) unless discontinuation is otherwise
warranted by their clinical condition.
• The COVID-19 Treatment Guidelines Panel recommends against using medications off-label to treat COVID-19 if they
have not been shown to be safe and effective for this indication in a clinical trial (AIII).
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
Individuals with underlying medical conditions, such as cardiovascular disease, pulmonary disease,
diabetes, and malignancy, and those who receive chronic immunosuppressive therapy are at higher risk
of severe illness with COVID-19. These patients are often prescribed medications to treat their
underlying medical conditions.
Early in the pandemic, some of these medications, such as angiotensin-converting enzyme (ACE)
inhibitors, angiotensin receptor blockers (ARBs),1 HMG-CoA reductase inhibitors (statins),2,3 and
histamine-2 receptor antagonists,4 were hypothesized to offer potential as COVID-19 therapeutic agents.
Others, such as nonsteroidal anti-inflammatory agents, were postulated to have negative impacts.5
Currently, there is no evidence that discontinuing medication for underlying medical conditions offers
a clinical benefit for patients with COVID-19.6-8 For example, the Food and Drug Administration stated
that there is no evidence linking the use of nonsteroidal anti-inflammatory agents with worsening of
COVID-19 and advised patients to use them as directed.9 Additionally, the American Heart Association,
the Heart Failure Society of America, and the American College of Cardiology issued a joint statement
that renin-angiotensin-aldosterone system antagonists, such as ACE inhibitors and ARBs, should be
continued as prescribed in those with COVID-19.10
Therefore, patients with COVID-19 who are treated with concomitant medications for an underlying
medical condition should not discontinue ACE inhibitors and ARBs (AIIa) or other medications
(AIII) unless discontinuation is otherwise warranted by their clinical condition. For patients with
COVID-19 who require nebulized medications, precautions should be taken to minimize the potential
for transmission of SARS-CoV-2 in the home and in health care settings.11,12
The COVID-19 Treatment Guidelines Panel recommends against using medications off-label to
treat COVID-19 if they have not been shown to be safe and effective for this indication in a clinical
trial (AIII). Clinicians should refer to the Therapies section of the Guidelines for information on the
medications that have been studied as potential therapeutic options for patients with COVID-19.
When prescribing medications to treat COVID-19, clinicians should always assess the patient’s
current medications for potential drug-drug interactions and additive adverse effects. The decision to
continue or change a patient’s medications should be individualized based on their specific clinical
condition. Clinicians can refer to product labels and visit the Liverpool COVID-19 Drug Interactions
website for guidance on identifying and managing drug-drug interactions. It is also worth noting that
COVID-19 Treatment Guidelines 407
References
1. Patel AB, Verma A. COVID-19 and angiotensin-converting enzyme inhibitors and angiotensin receptor
blockers: what is the evidence? JAMA. 2020;323(18):1769-1770. Available at: https://www.ncbi.nlm.nih.gov/
pubmed/32208485.
2. Lee KCH, Sewa DW, Phua GC. Potential role of statins in COVID-19. Int J Infect Dis. 2020;96:615-617.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/32502659.
3. Kashour T, Halwani R, Arabi YM, et al. Statins as an adjunctive therapy for COVID-19: the biological and
clinical plausibility. Immunopharmacol Immunotoxicol. 2021;43(1):37-50. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33406943.
4. Mather JF, Seip RL, McKay RG. Impact of famotidine use on clinical outcomes of hospitalized patients with
COVID-19. Am J Gastroenterol. 2020;115(10):1617-1623. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32852338.
5. Yousefifard M, Zali A, Zarghi A, Madani Neishaboori A, Hosseini M, Safari S. Non-steroidal anti-
inflammatory drugs in management of COVID-19; a systematic review on current evidence. Int J Clin Pract.
2020;74(9):e13557. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32460369.
6. Lopes RD, Macedo AVS, de Barros E Silva PGM, et al. Effect of discontinuing vs continuing angiotensin-
converting enzyme inhibitors and angiotensin II receptor blockers on days alive and out of the hospital in
patients admitted with COVID-19: a randomized clinical trial. JAMA. 2021;325(3):254-264. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33464336.
7. Cohen JB, Hanff TC, William P, et al. Continuation versus discontinuation of renin-angiotensin system
inhibitors in patients admitted to hospital with COVID-19: a prospective, randomised, open-label trial. Lancet
Respir Med. 2021;9(3):275-284. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33422263.
8. Bauer A, Schreinlechner M, Sappler N, et al. Discontinuation versus continuation of renin-angiotensin-system
inhibitors in COVID-19 (ACEI-COVID): a prospective, parallel group, randomised, controlled, open-label
trial. Lancet Respir Med. 2021;9(8):863-872. Available at: https://www.ncbi.nlm.nih.gov/pubmed/34126053.
9. Food and Drug Administration. FDA advises patients on use of non-steroidal anti-inflammatory drugs
(NSAIDs) for COVID-19. 2020. Available at: https://www.fda.gov/drugs/drug-safety-and-availability/fda-
advises-patients-use-non-steroidal-anti-inflammatory-drugs-nsaids-covid-19. Accessed October 19, 2023.
10. Bozkurt B, Kovacs R, Harrington B. Joint HFSA/ACC/AHA statement addresses concerns re: using RAAS
antagonists in COVID-19. J Card Fail. 2020;26(5):370. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32439095.
11. Cazzola M, Ora J, Bianco A, Rogliani P, Matera MG. Guidance on nebulization during the current COVID-19
pandemic. Respir Med. 2021;176:106236. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33248363.
12. Sethi S, Barjaktarevic IZ, Tashkin DP. The use of nebulized pharmacotherapies during the COVID-19
pandemic. Ther Adv Respir Dis. 2020;14:1753466620954366. Available at: https://www.ncbi.nlm.nih.gov/
pubmed/33167796.
Summary Recommendations
Prevention of COVID-19
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends COVID-19 vaccination for everyone who is eligible,
including those who are immunocompromised, according to the Centers for Disease Control and Prevention’s Advisory
Committee on Immunization Practices (AI).
• Vaccine response rates may be lower in patients who are moderately or severely immunocompromised. Specific
guidance on administering vaccines to these individuals is provided by the Centers for Disease Control and Prevention.
• Clinicians should strongly encourage all household members and close contacts of patients who are
immunocompromised to be vaccinated against COVID-19 (AI).
• Currently, antibody tests are not recommended for assessing SARS-CoV-2 immunity following COVID-19 vaccination or
for assessing the need for vaccination in a person who is unvaccinated.
Management of Patients With COVID-19 Who Are Immunocompromised
• Clinicians should consult with the appropriate specialists when making decisions about stopping or adjusting the doses
of immunosuppressive drugs in patients with COVID-19.
• When selecting treatments for COVID-19, clinicians should consider factors such as the underlying disease; the specific
immunosuppressants being used; the severity of COVID-19; and the potential for drug-drug interactions, overlapping
toxicities, and secondary infections.
• For nonhospitalized patients with mild to moderate COVID-19 who are immunocompromised, the Panel recommends
prompt treatment with antiviral drugs at the doses and durations recommended for the general population (AIII). For
more information, see Therapeutic Management of Nonhospitalized Adults With COVID-19.
• For most hospitalized patients with severe or critical COVID-19 who are immunocompromised, the Panel recommends
using antiviral drugs and immunomodulatory therapies at the doses and durations recommended for the general
population (AIII). For more information, see Therapeutic Management of Hospitalized Adults With COVID-19.
• For patients who are immunocompromised and have prolonged COVID-19 symptoms and evidence of ongoing viral
replication, the optimal management is unknown. The data for these approaches are not definitive, but some Panel
members would use 1 or more of the following treatment options:
• Longer and/or additional courses of ritonavir-boosted nirmatrelvir (Paxlovid)
• Longer and/or additional courses of remdesivir
• High-titer COVID-19 convalescent plasma from a vaccinated donor who recently recovered from COVID-19 likely
caused by a SARS-CoV-2 variant similar to the variant causing the patient’s illness
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
Introduction
Approximately 3% of people in the United States have immunocompromising conditions.1 People who
are immunocompromised are a heterogeneous population, and the severity of COVID-19 can vary
significantly in this group. Some individuals who are immunocompromised may have a higher risk of
hospitalization, complications, or death, and some may have outcomes that are comparable to those in
the general population.
This section pertains to people who are moderately or severely immunocompromised, which includes
those who:
Pre-Exposure Prophylaxis
As of February 2024, no biomedical intervention other than vaccines prevents COVID-19 disease.
Previously, the Food and Drug Administration (FDA) authorized the use of the anti-SARS-CoV-2
monoclonal antibodies tixagevimab plus cilgavimab (Evusheld) as pre-exposure prophylaxis (PrEP)
of COVID-19 in certain people who were not expected to mount an adequate immune response to
COVID-19 vaccination and in people with COVID-19 vaccine contraindications. Because the current
Omicron subvariants are not susceptible to tixagevimab plus cilgavimab, this combination is not
currently authorized by the FDA for use as PrEP of COVID-19.31
Therapeutic Management of Patients Who Are Hospitalized for Reasons Other Than
COVID-19
People who are immunocompromised and have COVID-19 but were hospitalized for conditions other
than COVID-19 should receive the same treatments as nonhospitalized patients (AIII). See Therapeutic
Management of Nonhospitalized Adults With COVID-19 for more information.
Prevention
The Panel recommends COVID-19 vaccination for everyone who is eligible, including pregnant and
lactating individuals, according to the CDC’s Advisory Committee on Immunization Practices (AI).
COVID-19 vaccination is strongly recommended for pregnant individuals due to their increased risk
for severe disease.106,107 Vaccination is especially important for pregnant people with concomitant
risk factors such as underlying immunocompromising conditions (including those who are receiving
immunosuppressive medications), as the risk for severe disease is likely additive.100
Treatment
Although pregnant patients have been excluded from the majority of the clinical trials that evaluated
the use of COVID-19 therapeutics, pregnant patients with COVID-19 can be treated the same as
nonpregnant patients, with a few exceptions. Pregnant patients who are immunocompromised or who
have other risk factors likely have an even higher risk of severe COVID-19 and should be prioritized
for treatment. Providers should refer to Pregnancy, Lactation, and COVID-19 Therapeutics for the
Panel’s guidance on treating COVID-19 in pregnant and lactating patients. Pregnant people who
are immunocompromised are a heterogeneous group of patients, ranging from those who are mildly
immunocompromised to those who are severely immunocompromised. Evaluating and managing
pregnant patients require collaboration from a multidisciplinary team. This team should include a
COVID-19 Treatment Guidelines 418
Considerations in Children
Although the overall risk of critical illness and death related to COVID-19 is lower in children than in
adults, severe disease does occur, particularly in children with risk factors such as moderate or severe
immunocompromising conditions. See Special Considerations in Children for a discussion of the
risk factors for severe COVID-19 in children, and see Therapeutic Management of Nonhospitalized
Children With COVID-19 for the Panel’s framework for assessing a child’s risk of progression to severe
COVID-19 based on vaccination status, comorbidities, and age.
Prevention
The Panel recommends COVID-19 vaccination for everyone who is eligible, including children,
according to the CDC’s Advisory Committee on Immunization Practices (AI).
Treatment
The majority of children with mild to moderate COVID-19 will not progress to more severe illness;
therefore, the Panel recommends managing these patients with supportive care alone (AIII). Few
children, if any, have been enrolled in clinical trials of treatments for COVID-19. Among the children
who were enrolled, very few were immunocompromised. Therefore, clinicians should be cautious when
applying recommendations based on adult data to children. Clinicians need to consider the potential
risks and benefits of therapy, the severity of the patient’s disease, and underlying risk factors. See
Therapeutic Management of Hospitalized Children With COVID-19 and Therapeutic Management
of Nonhospitalized Children With COVID-19 for the Panel’s treatment recommendations in these
scenarios.
References
1. Wallace BI, Kenney B, Malani PN, et al. Prevalence of immunosuppressive drug use among commercially
insured US adults, 2018–2019. JAMA Netw Open. 2021;4(5):e214920. Available at: https://www.ncbi.nlm.nih.
gov/pubmed/34014329.
2. Vijenthira A, Gong IY, Fox TA, et al. Outcomes of patients with hematologic malignancies and COVID-19: a
systematic review and meta-analysis of 3377 patients. Blood. 2020;136(25):2881-2892. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33113551.
3. Conway R, Grimshaw AA, Konig MF, et al. SARS-CoV-2 infection and COVID-19 outcomes in rheumatic
diseases: a systematic literature review and meta-analysis. Arthritis Rheumatol. 2022;74(5):766-775. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/34807517.
4. Song Q, Bates B, Shao YR, et al. Risk and outcome of breakthrough COVID-19 infections in vaccinated
patients with cancer: real-world evidence from the National COVID Cohort Collaborative. J Clin Oncol.
2022;40(13):1414-1427. Available at: https://www.ncbi.nlm.nih.gov/pubmed/35286152.
5. Ao G, Wang Y, Qi X, et al. The association between severe or death COVID-19 and solid organ
transplantation: a systematic review and meta-analysis. Transplant Rev (Orlando). 2021;35(3):100628.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/34087553.
6. Wang Y, Feng R, Xu J, et al. An updated meta-analysis on the association between HIV infection and
COVID-19 mortality. AIDS. 2021;35(11):1875-1878. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/34397487.
7. MacKenna B, Kennedy NA, Mehrkar A, et al. Risk of severe COVID-19 outcomes associated with immune-
mediated inflammatory diseases and immune-modifying therapies: a nationwide cohort study in the
Summary Recommendations
• COVID-19 vaccination remains the most effective way to prevent serious outcomes and death from SARS-CoV-2
infection. The COVID-19 Treatment Guidelines Panel (the Panel) recommends COVID-19 vaccination for everyone who is
eligible, including patients with active cancer and patients receiving treatment for cancer, according to the Centers for
Disease Control and Prevention’s (CDC) Advisory Committee on Immunization Practices (AI).
• See the CDC webpage COVID-19 Vaccines for People Who Are Moderately or Severely Immunocompromised for the
current COVID-19 vaccination schedule.
• Vaccinating household members, close contacts, and health care providers who provide care to patients with cancer
is important to protect these patients from infection. Clinicians should strongly encourage all household members and
close contacts of patients with cancer to be vaccinated against COVID-19 (AI).
• The Panel defers to CDC recommendations for diagnostic molecular or antigen testing for SARS-CoV-2 infection in
patients with cancer who develop signs and symptoms that suggest acute COVID-19. The Panel also defers to CDC
recommendations for testing of asymptomatic patients before procedures and hospital admissions.
• For patients with cancer and COVID-19, clinicians should follow COVID-19 evaluation and management guidelines
for patients who do not have cancer. See Therapeutic Management of Nonhospitalized Adults With COVID-19 and
Therapeutic Management of Hospitalized Adults With COVID-19 for more information.
• Decisions about administering cancer-directed therapy to patients with acute COVID-19 and those who are recovering
from COVID-19 should be made on a case-by-case basis; clinicians should consider the indication for chemotherapy,
the goals of care, and the patient’s history of tolerance to the cancer treatment (BIII).
• Clinicians should consult with a hematologist or oncologist when making decisions about stopping or adjusting the
doses of cancer-directed medications in patients with cancer and COVID-19.
• Clinicians should pay careful attention to potential overlapping toxicities and drug-drug interactions between drugs
used to treat COVID-19 (e.g., ritonavir-boosted nirmatrelvir [Paxlovid], dexamethasone) and cancer-directed therapies,
prophylactic antimicrobials, and other medications (AIII).
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
People being treated for cancer may be at increased risk of severe COVID-19, and clinical outcomes of
COVID-19 are generally worse in people with cancer than in people without cancer.1-4 A meta-analysis of
46,499 patients with COVID-19 showed that all-cause mortality was higher in patients with cancer (risk
ratio 1.66; 95% CI, 1.33–2.07), and that patients with cancer were more likely to be admitted to intensive
care units (risk ratio 1.56; 95% CI, 1.31–1.87).5 A patient’s risk of immunosuppression and susceptibility
to SARS-CoV-2 infection depend on the type of cancer, the treatments administered, and the stage of
disease (e.g., patients actively being treated compared to those in remission). In a study that used data
from the COVID-19 and Cancer Consortium registry, patients with cancer who were in remission or who
had no evidence of disease had a lower risk of death from COVID-19 than those who were receiving
active treatment.6 Cancer survivors may also have an increased risk of severe COVID-19.7
This section of the COVID-19 Treatment Guidelines focuses on testing for SARS-CoV-2 and managing
COVID-19 and cancer-directed therapies in people with cancer and COVID-19.
Febrile Neutropenia
Patients with cancer and febrile neutropenia should undergo diagnostic molecular or antigen testing
for SARS-CoV-2 and evaluation for other infectious agents. They should also be given empiric
antimicrobial therapy per the standard of care.39
Drug-Drug Interactions
The use of antiviral or immune-based therapies to treat COVID-19 can present additional challenges
in patients with cancer. Clinicians should pay careful attention to potential overlapping toxicities and
drug-drug interactions between drugs used to treat COVID-19 (e.g., ritonavir-boosted nirmatrelvir
[Paxlovid], dexamethasone) and cancer-directed therapies, prophylactic antimicrobials, and other
medications (AIII).
A 5-day course of ritonavir-boosted nirmatrelvir is 1 of the preferred therapies for treating mild to
moderate COVID-19 in nonhospitalized patients who are at risk for disease progression. However,
this regimen has the potential for significant and complex drug-drug interactions with concomitant
COVID-19 Treatment Guidelines 430
References
1. Dai M, Liu D, Liu M, et al. Patients with cancer appear more vulnerable to SARS-CoV-2: a multicenter study
during the COVID-19 outbreak. Cancer Discov. 2020;10(6):783-791. Available at: https://www.ncbi.nlm.nih.
gov/pubmed/32345594.
2. Shah V, Ko Ko T, Zuckerman M, et al. Poor outcome and prolonged persistence of SARS-CoV-2 RNA in
COVID-19 patients with haematological malignancies; King’s College Hospital experience. Br J Haematol.
2020;190(5):e279-e282. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32526039.
3. Yang K, Sheng Y, Huang C, et al. Clinical characteristics, outcomes, and risk factors for mortality in patients
with cancer and COVID-19 in Hubei, China: a multicentre, retrospective, cohort study. Lancet Oncol.
2020;21(7):904-913. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32479787.
4. Robilotti EV, Babady NE, Mead PA, et al. Determinants of COVID-19 disease severity in patients with cancer.
Nat Med. 2020;26(8):1218-1223. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32581323.
5. Giannakoulis VG, Papoutsi E, Siempos II. Effect of cancer on clinical outcomes of patients with COVID-19: a
Summary Recommendations
COVID-19 Vaccination
• COVID-19 vaccination remains the most effective way to prevent serious outcomes and death from SARS-CoV-2
infection. The COVID-19 Treatment Guidelines Panel (the Panel) recommends COVID-19 vaccination for everyone who
is eligible, including transplant and cellular immunotherapy candidates and recipients, according to the Centers for
Disease Control and Prevention’s Advisory Committee on Immunization Practices (AI).
• See the Centers for Disease Control and Prevention webpage COVID-19 Vaccines for People Who Are Moderately
or Severely Immunocompromised for the current COVID-19 vaccination schedule for transplant and cellular
immunotherapy recipients.
• Clinicians should strongly encourage all household members, close contacts, and health care providers who provide
care to transplant and cellular immunotherapy candidates and recipients to be vaccinated against COVID-19 (AI).
• Clinicians should strongly encourage all potential organ and hematopoietic cell donors to get vaccinated against
COVID-19 (AI).
Potential Transplant and Cellular Immunotherapy Candidates
• The Panel recommends performing diagnostic molecular or antigen testing for SARS-CoV-2 in all potential solid organ
transplant, hematopoietic cell transplant, and cellular immunotherapy candidates with signs and symptoms that
suggest acute COVID-19 (AIII). Additional guidance is available from medical professional organizations. See the text
below for more information.
• If SARS-CoV-2 is detected or infection is strongly suspected in a potential transplant or cellular immunotherapy
candidate, transplantation or immunotherapy should be deferred, if possible (BIII).
• The optimal management and therapeutic approach to COVID-19 in these populations is unknown. At this time, the
procedures for evaluating and managing COVID-19 in transplant candidates are the same as those for nontransplant
patients (AIII).
Potential Transplant Donors
• The Panel recommends performing diagnostic molecular testing for SARS-CoV-2 and assessing for symptoms of
COVID-19 in all potential solid organ transplant and hematopoietic cell transplant donors prior to donation (AIII).
Additional guidance is available from medical professional organizations. See the text below for more information.
Transplant and Cellular Immunotherapy Recipients With COVID-19
• For transplant and cellular immunotherapy recipients with COVID-19, clinicians should follow COVID-19 evaluation and
management guidelines for nontransplant patients. See Therapeutic Management of Hospitalized Adults With COVID-19,
Therapeutic Management of Nonhospitalized Adults With COVID-19, and Special Considerations in People Who Are
Immunocompromised for more information.
• For nonhospitalized patients with mild to moderate COVID-19 who are transplant or cellular immunotherapy recipients,
the Panel recommends prompt treatment with antiviral drugs at the doses and durations recommended for the general
population (AIII).
• Clinicians should consult with a transplant specialist when making decisions about stopping or adjusting the doses of
immunosuppressive drugs in transplant or cellular immunotherapy recipients with COVID-19.
• Clinicians should pay special attention to the potential for drug-drug interactions and overlapping toxicities between
treatments for COVID-19 and concomitant medications, such as immunosuppressants used to prevent or treat allograft
rejection and antimicrobials used to prevent or treat opportunistic infections.
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
COVID-19 Vaccination
The clinical trials that evaluated the safety and efficacy of the COVID-19 vaccines excluded
patients who were severely immunocompromised.1,2 Authorized and approved COVID-19 vaccines
in the United States are not live-virus vaccines and can be safely administered to patients who are
immunocompromised. However, solid organ transplant recipients have reduced immunological antibody
responses following a primary 2-dose or 3-dose series of the mRNA COVID-19 vaccines.3-6
COVID-19 vaccination remains the most effective way to prevent serious outcomes and death
from SARS-CoV-2 infection. The COVID-19 Treatment Guidelines Panel (the Panel) recommends
COVID-19 vaccination for everyone who is eligible, including transplant and cellular immunotherapy
candidates and recipients, according to the Centers for Disease Control and Prevention’s (CDC)
Advisory Committee on Immunization Practices (AI). See the CDC webpage COVID-19 Vaccines for
People Who Are Moderately or Severely Immunocompromised for the current COVID-19 vaccination
schedule for transplant and cellular immunotherapy recipients.
When determining the timing of COVID-19 vaccination in solid organ transplant, HCT, and cellular
immunotherapy recipients, clinicians should consider the following factors:
• Ideally, solid organ transplant candidates should receive COVID-19 vaccines while they wait for
transplant.
• In general, vaccination should be completed at least 2 weeks before a solid organ transplant, or
vaccination should be started 1 month after a solid organ transplant.
COVID-19 Treatment Guidelines 436
Concomitant Medications
Clinicians should pay special attention to the potential for drug-drug interactions and overlapping
toxicities between treatments for COVID-19 and concomitant medications, such as immunosuppressants
used to prevent or treat allograft rejection and antimicrobials used to prevent or treat opportunistic
infections. Dose modifications may be necessary for drugs used to treat COVID-19 in transplant
recipients with pre-existing organ dysfunction. Adjustments to the immunosuppressive regimen
should be individualized based on disease severity, the specific immunosuppressants used, the type
of transplant, the time since transplantation, the drug concentration, and the risk of graft rejection.23
Clinicians should consult with a transplant specialist when making decisions about stopping or
adjusting the doses of immunosuppressive drugs in transplant or cellular immunotherapy recipients with
COVID-19.
Drug-Drug Interactions
Calcineurin inhibitors (e.g., cyclosporine, tacrolimus) and mammalian target of rapamycin (mTOR)
inhibitors (e.g., everolimus, sirolimus), which are commonly used to prevent allograft rejection, have
narrow therapeutic indices. Medications that inhibit or induce cytochrome P450 (CYP) enzymes or
P-glycoprotein may put patients who receive these drugs at risk of clinically significant drug-drug
interactions, increasing the need for therapeutic drug monitoring and the need to assess for signs of
toxicity or rejection.35
COVID-19 Treatment Guidelines 439
References
1. Baden LR, El Sahly HM, Essink B, et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N
Engl J Med. 2021;384(5):403-416. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33378609.
2. Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine. N
Engl J Med. 2020;383(27):2603-2615. Available at: https://www.ncbi.nlm.nih.gov/pubmed/33301246.
3. Boyarsky BJ, Werbel WA, Avery RK, et al. Antibody response to 2-dose SARS-CoV-2 mRNA vaccine series
in solid organ transplant recipients. JAMA. 2021;325(21):2204-2206. Available at: https://www.ncbi.nlm.nih.
gov/pubmed/33950155.
Summary Recommendations
• Pregnant people should be counseled about the increased risk for severe disease from SARS-CoV-2 infection and the
measures they can take to protect themselves and their families from infection.
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends against withholding COVID-19 vaccination from
pregnant or lactating individuals specifically because of pregnancy or lactation (AIII).
• If hospitalization for COVID-19 is indicated for a pregnant patient, care should be provided in a facility that can conduct
maternal and fetal monitoring, when appropriate.
• General management of COVID-19 in pregnant patients should include:
• Fetal and uterine contraction monitoring based on gestational age, when appropriate
• Individualized delivery planning
• A multispecialty, team-based approach that may include consultation with obstetric, maternal-fetal medicine,
infectious disease, pulmonary-critical care, and pediatric specialists, as appropriate
• The Panel recommends against withholding COVID-19 treatment from pregnant or lactating individuals specifically
because of pregnancy or lactation (AIII).
• In general, the therapeutic management of pregnant patients with COVID-19 should be the same as for nonpregnant
patients, with a few exceptions (AIII). Notable exceptions include:
• The Panel recommends against the use of molnupiravir for the treatment of COVID-19 in pregnant patients unless
there are no other options and therapy is clearly indicated (AIII).
• There is insufficient evidence for the Panel to recommend either for or against the use of therapeutic anticoagulation
in pregnant patients with COVID-19 who do not have evidence of venous thromboembolism. See Antithrombotic
Therapy in Patients With COVID-19 for more information.
• For details regarding therapeutic recommendations and pregnancy considerations, see Therapeutic Management of
Nonhospitalized Adults With COVID-19; Therapeutic Management of Hospitalized Adults With COVID-19; Pregnancy,
Lactation, and COVID-19 Therapeutics; and the individual drug sections.
• Data on the use of COVID-19 therapeutic agents in pregnant and lactating people are limited. When making decisions
about treatment, pregnant or lactating people and their clinical teams should use a shared decision-making process
and consider several factors, including the severity of COVID-19, the risk of disease progression, and the safety of
specific medications for the fetus, infant, or pregnant or lactating individual. For detailed guidance on using the Panel-
recommended COVID-19 therapeutic agents during pregnancy, see Pregnancy, Lactation, and COVID-19 Therapeutics.
• The decision to feed the infant breast milk while the lactating patient is receiving therapeutic agents for COVID-19
should be a collaborative effort between the patient and the clinical team, including infant care providers. The patient
and the clinical team should discuss the potential benefits of the therapeutic agent and evaluate the potential risk
of pausing lactation on future breast milk delivery to the infant. For more information, see Pregnancy, Lactation, and
COVID-19 Therapeutics.
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
COVID-19 Vaccines
The COVID-19 Treatment Guidelines Panel (the Panel) recommends against withholding COVID-19
vaccination from pregnant or lactating individuals specifically because of pregnancy or lactation (AIII).
COVID-19 Treatment Guidelines 446
Timing of Delivery
The ACOG provides detailed guidance on the timing of delivery and the risk of vertical transmission of
SARS-CoV-2.
In most cases, the timing of delivery should be dictated by obstetric indications rather than maternal
diagnosis of COVID-19. For people who had suspected or confirmed COVID-19 early in pregnancy and
who recovered, no alteration to the usual timing of delivery is indicated.
After Delivery
Therapeutic management in postpartum patients should follow guidelines for nonpregnant patients.
However, the use of anticoagulant therapy in the immediate postpartum period should be individualized,
as there may be an increased risk of bleeding, especially after an operative delivery.
The majority of studies have not demonstrated the presence of SARS-CoV-2 in breast milk; therefore,
breastfeeding is not contraindicated for people with laboratory-confirmed or suspected SARS-CoV-2
infection.22 Precautions should be taken to avoid transmission to the infant, including practicing
appropriate hand hygiene, wearing face coverings, and performing proper pump cleaning before and
after breast milk expression.
The decision to feed the infant breast milk while the lactating patient is receiving therapeutic agents for
COVID-19 should be a collaborative effort between the patient and the clinical team, including infant
care providers. The patient and the clinical team should discuss the potential benefits of the therapeutic
agent and evaluate the potential risk of pausing lactation on future breast milk delivery to the infant.
Specific guidance on the postdelivery management of infants born to individuals with known or
suspected SARS-CoV-2 infection, including breastfeeding recommendations, is provided by the
American Academy of Pediatrics.
References
1. Zambrano LD, Ellington S, Strid P, et al. Update: characteristics of symptomatic women of reproductive age
with laboratory-confirmed SARS-CoV-2 infection by pregnancy status—United States, January 22–October
3, 2020. MMWR Morb Mortal Wkly Rep. 2020;69(44):1641-1647. Available at: https://pubmed.ncbi.nlm.nih.
gov/33151921.
2. Clinical manifestations, risk factors, and maternal and perinatal outcomes of coronavirus disease 2019 in
pregnancy: living systematic review and meta-analysis. BMJ. 2020;370:m3320. Available at: https://pubmed.
ncbi.nlm.nih.gov/32873575.
General Considerations
The COVID-19 Treatment Guidelines Panel (the Panel) recommends against withholding COVID-19
treatments or vaccination from pregnant or lactating individuals specifically because of pregnancy or
lactation (AIII). Pregnant patients should be offered the opportunity to participate in the COVID-19
International Drug Pregnancy Registry or other pregnancy registries.
The decision to feed the infant breast milk while the lactating patient is receiving therapeutic agents for
COVID-19 should be a collaborative effort between the patient and the clinical team, including infant
care providers. The patient and the clinical team should consider the benefits of breastfeeding, the
postnatal age of the infant, the need for the medication, any underlying risks of exposing the infant to the
drug, and the potential adverse outcomes of COVID-19.
If a patient is receiving a treatment for COVID-19 that prevents them from feeding breast milk to their
infant for a limited time, the clinical team should still encourage pumping breast milk and provide
lactation support. This ensures that lactation can continue after the patient stops receiving the treatment.
While a person with COVID-19 is breastfeeding, prevention measures should be taken to avoid
transmitting SARS-CoV-2 to the infant. These measures include practicing appropriate hand hygiene,
wearing face coverings, and performing proper pump cleaning before and after breast milk expression.
Table A: Recommendations for the Use of COVID-19 Therapeutics in Pregnant and Lactating
People
For the Panel’s recommendations on when to use the medications listed below, refer to Therapeutic
Management of Nonhospitalized Adults With COVID-19 and Therapeutic Management of Hospitalized
Adults With COVID-19. Pregnant patients should be offered the opportunity to participate in the
COVID-19 International Drug Pregnancy Registry or other pregnancy registries. For additional
information on the use of these medications during pregnancy and lactation, see the section text below.
Drug Name
(in alphabetical Pregnancy Lactation
order)
Abatacept Recommended in hospitalized patients, if indicated. Should be offered to patients who qualify
Pregnant patients and their health care providers for this therapy. There is minimal data on
should jointly decide whether to use abatacept during the transfer of abatacept to breast milk.
pregnancy, and the decision-making process should Breastfeeding may be considered while a
include a discussion of the potential risks and benefits. patient receives abatacept.
Baricitinib Recommended in hospitalized patients, if indicated. Feeding breast milk should be avoided
Pregnant patients and their health care providers while taking baricitinib and for 4 days after
should jointly decide whether to use baricitinib during the last dose. Lactation support should be
pregnancy, and the decision-making process should provided during this time.a
include a discussion of the potential risks and benefits.
Dexamethasone Recommended in hospitalized patients, if indicated. Should be offered to patients who qualify
for this therapy. Breastfeeding can continue
while a patient receives dexamethasone.
Rationale
Abatacept
Pregnancy
As there are no data on the use of abatacept during pregnancy in hospitalized patients with COVID-19,
this drug should be used only if baricitinib and tocilizumab are not available or feasible to use. When
deciding whether to prescribe abatacept to a pregnant individual, clinicians need to consider the severity
of the patient’s COVID-19, the patient’s comorbidities, and the gestational age of the fetus.
There is a paucity of data on the use of abatacept in pregnant individuals. It is currently not known
whether abatacept can cross the human placenta; however, abatacept has crossed the placenta in animal
studies. One study reported alterations to the immune systems of the offspring of animals that received
supratherapeutic doses of abatacept throughout pregnancy.2 It is not known whether the immune systems
of infants who were exposed to a single dose of abatacept in utero might be impacted. Abatacept should
only be used during pregnancy if the benefits clearly outweigh the potential risks.
Baricitinib
Pregnancy
When deciding whether to prescribe baricitinib to a pregnant individual, clinicians need to consider the
severity of the patient’s COVID-19, the patient’s comorbidities, and the gestational age of the fetus.
Baricitinib is a Janus kinase inhibitor. As a small-molecule drug, baricitinib is likely to pass through
the placenta; therefore, fetal risk cannot be ruled out.3 In animal studies, baricitinib doses that exceeded
the therapeutic human dose were associated with embryofetal developmental abnormalities. Pregnancy
registries provide some data on the use of tofacitinib, another Janus kinase inhibitor, during pregnancy
for other conditions (e.g., ulcerative colitis, rheumatoid arthritis, psoriasis).4-6 Pregnancy outcomes
among the participants who received tofacitinib were similar to those among the general population.
Lactation
There is no information on the use of baricitinib in lactating people or on the effects of baricitinib on
breastfed infants; however, baricitinib has been detected in the breast milk of lactating rats.7 Feeding
breast milk should be avoided for 4 days (approximately 5–6 elimination half-lives) after baricitinib is
discontinued.
Dexamethasone
Pregnancy
A short course of betamethasone or dexamethasone, which are both known to cross the placenta, is
routinely used to decrease neonatal complications of prematurity in people who are at risk of imminent
preterm birth.8,9 Treating COVID-19 with a short course of dexamethasone can lower the risk of death in
pregnant individuals. In addition, dexamethasone carries a low risk of fetal adverse effects.
Lactation
Dexamethasone should be offered to lactating patients with COVID-19 who qualify for this therapy.
Breast milk can be fed to the infant while the lactating patient is receiving dexamethasone. Although
there are limited data on the use of dexamethasone in lactating patients, some published reports about
a related antenatal corticosteroid (betamethasone) reported a time-limited decrease in the volume of
breast milk production.10,11 Given the benefits of breast milk, additional lactation support has been
recommended if needed.
Infliximab
Pregnancy
As there are no data on the use of infliximab during pregnancy in hospitalized patients with COVID-19,
infliximab should be used only if baricitinib and tocilizumab are not available or feasible to use. When
deciding whether to prescribe infliximab to a pregnant individual, clinicians need to consider the
severity of the patient’s COVID-19, the patient’s comorbidities, and the gestational age of the fetus.
There are limited data on the use of infliximab to treat COVID-19 in pregnant patients. It has been used
to treat autoimmune diseases in pregnant individuals when the benefits outweigh the potential risks.
Infliximab crosses the placenta and has been detected in the serum of infants born to patients treated
with infliximab during pregnancy. No adverse effects have been reported in these infants.
Lactation
Infants who are breastfed by people receiving infliximab show minimal absorption of this agent.12 No
adverse effects have been reported in these infants. Therefore, infliximab should be offered to patients
who qualify. Breastfeeding can continue while a patient receives infliximab.
Molnupiravir
Pregnancy
The Panel recommends against the use of molnupiravir for the treatment of COVID-19 in pregnant
patients unless there are no other options and therapy is clearly indicated (AIII).
The Food and Drug Administration (FDA) Emergency Use Authorization states that molnupiravir is not
recommended for use in pregnant patients because fetal toxicity has been reported in animal studies of
molnupiravir.1 However, when other therapies are not available, pregnant people with COVID-19 who
are at high risk of progressing to severe disease may reasonably choose molnupiravir therapy after being
fully informed of the potential risks, particularly if they are beyond the time of embryogenesis (i.e., >10
weeks’ gestation).
Lactation
There is no data on the use of molnupiravir in lactating people; however, molnupiravir has been
detected in the offspring of lactating rats. Molnupiravir is not authorized for use in children aged <18
years. Because the risk of adverse effects in infants is currently unknown, the FDA Emergency Use
Authorization fact sheet does not recommend feeding an infant breast milk from a patient who is taking
molnupiravir for the duration of the treatment course and until 4 days after the final dose.1
Remdesivir
Pregnancy
While pregnant individuals were excluded from the clinical trials that evaluated the safety and efficacy
of remdesivir for the treatment of COVID-19, subsequent reports on the use of remdesivir in pregnant
individuals have been reassuring. Among 95 pregnant patients with moderate, severe, or critical
COVID-19 who were included in a secondary analysis of data from a COVID-19 pregnancy registry
in Texas, the composite maternal and neonatal outcomes were similar between those who received
Tocilizumab
Pregnancy
Pregnant individuals have been excluded from clinical trials that evaluated the use of the anti-
interleukin-6 receptor monoclonal antibody tocilizumab for the treatment of COVID-19. An analysis of
data from a global safety database reported pregnancy outcomes from 288 women who were exposed
to tocilizumab during their pregnancies.26 Eighty-nine percent of these women received tocilizumab
as ongoing treatment for rheumatoid arthritis, and most were exposed to tocilizumab during their first
trimester. The rates of congenital abnormalities among the infants born to these women were not higher
than the rates seen in the general population. However, an increased rate of preterm birth was observed
among these individuals when compared with the general population. A retrospective report of 61
pregnant women who were exposed to tocilizumab at conception or during their first trimesters showed
no increased rates of congenital abnormalities or spontaneous abortion.27
As pregnancy progresses, monoclonal antibodies are actively transported across the placenta, with the
greatest transfer occurring during the third trimester. This may affect immune responses in the exposed
fetus. If a pregnant patient receives tocilizumab after 20 weeks’ gestation, clinicians should delay
administering live viral vaccines to the infant for at least 6 months.
Lactation
There is limited information on the use of tocilizumab in lactating patients. Based on case report data,
the amount of tocilizumab transferred to the infant via breast milk appears to be very low, with no
reports of adverse effects.28
References
1. Food and Drug Administration. Fact sheet for healthcare providers: Emergency Use Authorization for
Lagevrio (molnupiravir) capsules. 2023. Available at: https://www.fda.gov/media/155054/download.
2. Abatacept (Orencia) [package insert]. Food and Drug Administration. 2021. Available at: https://www.
accessdata.fda.gov/drugsatfda_docs/label/2021/125118s240lbl.pdf.
3. Sammaritano LR, Bermas BL, Chakravarty EE, et al. 2020 American College of Rheumatology guideline
for the management of reproductive health in rheumatic and musculoskeletal diseases. Arthritis Rheumatol.
2020;72(4):529-556. Available at: https://pubmed.ncbi.nlm.nih.gov/32090480.
4. Clowse ME, Feldman SR, Isaacs JD, et al. Pregnancy outcomes in the tofacitinib safety databases for
rheumatoid arthritis and psoriasis. Drug Saf. 2016;39(8):755-762. Available at:
https://pubmed.ncbi.nlm.nih.gov/27282428.
5. Mahadevan U, Dubinsky MC, Su C, et al. Outcomes of pregnancies with maternal/paternal exposure in the
tofacitinib safety databases for ulcerative colitis. Inflamm Bowel Dis. 2018;24(12):2494-2500. Available at:
https://pubmed.ncbi.nlm.nih.gov/29982686.
6. Wieringa JW, van der Woude CJ. Effect of biologicals and JAK inhibitors during pregnancy on health-
related outcomes in children of women with inflammatory bowel disease. Best Pract Res Clin Gastroenterol.
2020;44-45:101665. Available at: https://pubmed.ncbi.nlm.nih.gov/32359679.
7. Baricitinib (Olumiant) [package insert]. Food and Drug Administration. 2022. Available at: https://www.
COVID-19 Treatment Guidelines 457
Summary Recommendations
Influenza Vaccination
• People with acute COVID-19 who have not received an influenza vaccine during influenza season should be vaccinated
after they recover from acute illness and are no longer in isolation (BIII).
• Patients may be vaccinated while they are still in isolation if they are in a health care setting.
• An influenza vaccine and a COVID-19 vaccine may be administered concurrently at different injection sites. The
Advisory Committee on Immunization Practices and the Centers for Disease Control and Prevention (CDC) provide more
information on COVID-19 and influenza vaccines.
Diagnosis of Influenza and COVID-19 When Influenza Viruses and SARS-CoV-2 Are Cocirculating
• Only testing can distinguish between SARS-CoV-2 and influenza virus infections and identify SARS-CoV-2 and influenza
virus coinfection.
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends performing influenza testing in addition to SARS-
CoV-2 testing in outpatients with acute respiratory illness if the results will change the clinical management strategy for
the patient (e.g., administering antiviral treatment for influenza) (BIII).
• The Panel recommends testing for both viruses in all hospitalized patients with acute respiratory illness (AIII).
• Clinicians should consider performing additional testing in specific clinical circumstances. Secondary bacterial infection
is more common with influenza than with COVID-19, so additional testing for bacterial pathogens is important in
patients with influenza who have clinical signs that suggest bacterial superinfection, especially for those who are
immunocompromised or intubated.
• See the CDC webpage Information for Clinicians on Influenza Virus Testing and the Infectious Diseases Society of
America (IDSA) clinical practice guidelines for more information.
Antiviral Treatment of Influenza When Influenza Viruses and SARS-CoV-2 Are Cocirculating
• Antiviral treatment for influenza is the same for all patients regardless of SARS-CoV-2 coinfection (AIII).
• For information on using antiviral drugs to treat influenza in hospitalized and nonhospitalized patients, see the CDC
and IDSA recommendations.
• There are no clinically significant drug-drug interactions between the antiviral agents used to treat influenza and the
antiviral agents or immunomodulators used to treat COVID-19.
• The Panel recommends starting hospitalized patients who are suspected of having influenza on empiric treatment for
influenza with oseltamivir as soon as possible regardless of their COVID-19 status and without waiting for influenza
test results (AIIb).
• Oseltamivir treatment should be continued until nucleic acid detection assay results rule out influenza. For patients
who are not intubated, assays should be performed on upper respiratory tract specimens. For patients who are
intubated, assays should be performed on both upper and lower respiratory tract specimens.
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
Introduction
Clinicians should monitor local influenza and SARS-CoV-2 activities during influenza season to inform
the evaluation and management of patients with acute respiratory illness. This can be done by tracking
local and state public health surveillance data, assessing the results of testing performed at health care
facilities, and reviewing the Centers for Disease Control and Prevention (CDC) Weekly U.S. Influenza
Surveillance Report.
References
1. Grohskopf LA, Blanton LH, Ferdinands JM, et al. Prevention and control of seasonal influenza with vaccines:
recommendations of the Advisory Committee on Immunization Practices—United States, 2023–24 influenza
season. MMWR Recomm Rep. 2023;72(2):1-25. Available at: https://www.cdc.gov/mmwr/volumes/72/rr/
rr7202a1.htm.
2. Centers for Disease Control and Prevention. Contraindications and precautions. 2023. Available at: https://
www.cdc.gov/vaccines/hcp/acip-recs/general-recs/contraindications.html. Accessed December 18, 2023.
3. Izikson R, Brune D, Bolduc JS, et al. Safety and immunogenicity of a high-dose quadrivalent influenza
vaccine administered concomitantly with a third dose of the mRNA-1273 SARS-CoV-2 vaccine in adults aged
≥65 years: a Phase 2, randomised, open-label study. Lancet Respir Med. 2022;10(4):392-402. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35114141.
4. Lazarus R, Baos S, Cappel-Porter H, et al. Safety and immunogenicity of concomitant administration
of COVID-19 vaccines (ChAdOx1 or BNT162b2) with seasonal influenza vaccines in adults in the UK
(ComFluCOV): a multicentre, randomised, controlled, Phase 4 trial. Lancet. 2021;398(10318):2277-2287.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/34774197.
5. Hause AM, Zhang B, Yue X, et al. Reactogenicity of simultaneous COVID-19 mRNA booster and influenza
vaccination in the US. JAMA Netw Open. 2022;5(7):e2222241. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/35838667.
6. Janssen C, Mosnier A, Gavazzi G, et al. Coadministration of seasonal influenza and COVID-19 vaccines: a
systematic review of clinical studies. Hum Vaccin Immunother. 2022;18(6):2131166. Available at:
https://pubmed.ncbi.nlm.nih.gov/36256633.
7. Hashemi SA, Safamanesh S, Ghasemzadeh-Moghaddam H, Ghafouri M, Azimian A. High prevalence of
SARS-CoV-2 and influenza A virus (H1N1) coinfection in dead patients in Northeastern Iran. J Med Virol.
2021;93(2):1008-1012. Available at: https://www.ncbi.nlm.nih.gov/pubmed/32720703.
8. Huang BR, Lin YL, Wan CK, et al. Co-infection of influenza B virus and SARS-CoV-2: a case report from
Taiwan. J Microbiol Immunol Infect. 2021;54(2):336-338. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32646801.
9. Yue H, Zhang M, Xing L, et al. The epidemiology and clinical characteristics of co-infection of SARS-CoV-2
and influenza viruses in patients during COVID-19 outbreak. J Med Virol. 2020;92(11):2870-2873. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/32530499.
Summary Recommendations
COVID-19 Vaccination
• The COVID-19 Treatment Guidelines Panel (the Panel) recommends that people with HIV receive COVID-19 vaccines,
regardless of their CD4 T lymphocyte (CD4) cell count or HIV viral load, because the potential benefits outweigh the
potential risks (AIIb).
• For people with untreated or advanced HIV, the Panel recommends following the most recent COVID-19 vaccination
schedule from the Centers for Disease Control and Prevention (CDC) for people who are moderately or severely
immunocompromised. The CDC defines advanced HIV as CD4 counts <200 cells/mm3, a history of an AIDS-defining
illness without immune reconstitution, or clinical manifestations of symptomatic HIV.
Diagnosis of SARS-CoV-2 Infection
• The Panel defers to CDC recommendations for diagnostic molecular or antigen testing for SARS-CoV-2 infection in
people with HIV who develop signs and symptoms that suggest acute COVID-19.
Managing COVID-19 in People With HIV
• The recommendations for the triage, management, and treatment of COVID-19 in people with HIV are the same as
those for the general population (AIII).
• Nonhospitalized people with HIV and mild to moderate COVID-19 may be eligible to receive the therapies that are
currently recommended for treatment (see Therapeutic Management of Nonhospitalized Adults With COVID-19).
However, in situations where there are logistical constraints for administering these therapies, priority should be given
to those with untreated or advanced HIV (AIII). See Prioritization of Anti-SARS-CoV-2 Therapies for the Treatment of
COVID-19 in Nonhospitalized Patients When There Are Logistical Constraints for details.
• People with HIV who are receiving a 5-day course of ritonavir-boosted nirmatrelvir (Paxlovid) to treat COVID-19 can
continue using their antiretroviral therapy (ART) doses of ritonavir or cobicistat without alteration or interruption.
• In patients with advanced HIV who have suspected or laboratory-confirmed SARS-CoV-2 infection, clinicians should
consider HIV-associated opportunistic infections in the differential diagnosis of clinical symptoms and consider
consulting an HIV specialist.
• When starting treatment for COVID-19 in patients with HIV, clinicians should pay careful attention to potential drug-drug
interactions and overlapping toxicities among COVID-19 treatments, antiretroviral (ARV) medications, antimicrobial
therapies, and other medications (AIII).
Managing HIV in People With COVID-19
• People with HIV who develop COVID-19, including those who require hospitalization, should continue their ART and their
medications for the treatment or prevention of opportunistic infections whenever possible.
• Clinicians treating COVID-19 in people with HIV should consult an HIV specialist before adjusting or switching a patient’s
ARV medications.
• An ARV regimen should not be modified for the purpose of preventing or treating SARS-CoV-2 infection.
• Clinicians should consult with an HIV specialist about the timing of ART in people who present with COVID-19 and
untreated HIV.
Each recommendation in the Guidelines receives a rating for the strength of the recommendation (A, B, or C) and a rating
for the evidence that supports it (I, IIa, IIb, or III). See Guidelines Development for more information.
Introduction
Approximately 1.2 million people in the United States are living with HIV. Most of these individuals
are in care, and many are receiving antiretroviral therapy (ART) and have well-controlled disease.1
Similar to COVID-19, HIV disproportionately affects racial and ethnic minorities and people living in
COVID-19 Treatment Guidelines 466
Prevention of COVID-19
The COVID-19 Treatment Guidelines Panel (the Panel) recommends that people with HIV receive
COVID-19 vaccines, regardless of their CD4 count or HIV viral load, because the potential benefits
outweigh the potential risks (AIIb). People with HIV were included in the clinical trials of the 2 mRNA
vaccines (Pfizer and Moderna) and the glycoprotein vaccine (Novavax) that are currently available
through Emergency Use Authorizations and/or approval from the Food and Drug Administration.21-23
Typically, people with HIV who are receiving ART and who have achieved virologic suppression
respond well to licensed vaccines. Data from studies that used COVID-19 vaccines in people with HIV
confirm that people who are receiving ART and have normal CD4 counts have good immunologic
responses to the vaccines.24-26 However, vaccine response rates are generally lower in people with lower
CD4 counts (e.g., <200 cells/mm3).19,20,27
For people with untreated or advanced HIV, the Panel recommends following the most recent
COVID-19 vaccination schedule from the Centers for Disease Control and Prevention (CDC) for
people who are moderately or severely immunocompromised. The CDC defines advanced HIV as CD4
counts <200 cells/mm3, a history of an AIDS-defining illness without immune reconstitution, or clinical
manifestations of symptomatic HIV. Patients who have poor adherence or who experience virologic
failure while on ART may have a similar risk of severe COVID-19 as those with untreated HIV. For
additional considerations regarding vaccination in people who are immunocompromised, see Special
Considerations in People Who Are Immunocompromised.
There is currently no clear evidence that antiretroviral (ARV) medications can prevent SARS-CoV-2
infection. Some studies suggested that tenofovir disoproxil fumarate/emtricitabine may play a role in
preventing SARS-CoV-2 acquisition or hospitalization or death associated with COVID-19; however,
the significance of these findings is unclear.28-30 These studies may not have adequately controlled for
confounding variables such as age and comorbidities. In addition, most of these studies were conducted
in unvaccinated patients.
Considerations in Children
In general, children appear less likely to become severely ill with COVID-19 than adults. In the few
publications that have described cases of COVID-19 among children or adolescents with HIV, most
cases were mild, and HIV did not appear to be an independent predictor of severe COVID-19.44-47
Children with HIV who are eligible should receive COVID-19 vaccines and booster doses regardless
of their CD4 count or viral load. Children with HIV and COVID-19 or multisystem inflammatory
syndrome in children (MIS-C) should receive the same treatment as children without HIV. See
Therapeutic Management of Hospitalized Children With COVID-19, Therapeutic Management of
Nonhospitalized Children With COVID-19, and Therapeutic Management of Hospitalized Children With
MIS-C, Plus a Discussion on MIS-A for more information.
Parents of children with HIV and COVID-19 should be advised to continue their child’s ART without
interruption if the child is being managed at home. For children with HIV who are hospitalized for
COVID-19, ART should be continued for the duration of hospitalization.
References
1. Harris NS, Johnson AS, Huang YA, et al. Vital signs: status of human immunodeficiency virus testing, viral
suppression, and HIV preexposure prophylaxis—United States, 2013–2018. MMWR Morb Mortal Wkly Rep.
2019;68(48):1117-1123. Available at: https://pubmed.ncbi.nlm.nih.gov/31805031.
2. Meyerowitz EA, Kim AY, Ard KL, et al. Disproportionate burden of coronavirus disease 2019 among racial
COVID-19 Treatment Guidelines 470
Name Affiliation
Co-Chairs
Roy M. Gulick, MD, MPH Weill Cornell Medicine, New York, NY
H. Clifford Lane, MD National Institutes of Health, Bethesda, MD
Henry Masur, MD National Institutes of Health, Bethesda, MD
Executive Secretary
Alice K. Pau, PharmD National Institutes of Health, Bethesda, MD
Members
Judith Aberg, MD Icahn School of Medicine at Mount Sinai, New York, NY
Adaora Adimora, MD, MPH University of North Carolina School of Medicine, Chapel Hill, NC
Jason Baker, MD, MS Hennepin Healthcare and University of Minnesota, Minneapolis, MN
Lisa Baumann Kreuziger, MD, MS Versiti and Medical College of Wisconsin, Milwaukee, WI
Roger Bedimo, MD, MS University of Texas Southwestern and Veterans Affairs North Texas Health Care
System, Dallas, TX
Pamela S. Belperio, PharmD Department of Veterans Affairs, Los Angeles, CA
Anoopindar Bhalla, MD Children’s Hospital Los Angeles and University of Southern California, Los
Angeles, CA
Stephen V. Cantrill, MD Denver Health, Denver, CO
Kara Chew, MD, MS University of California, Los Angeles, Los Angeles, CA
Kathleen Chiotos, MD, MSCE Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
Craig Coopersmith, MD Emory University School of Medicine, Atlanta, GA
Eric Daar, MD Harbor-UCLA Medical Center, Torrance, CA
Amy L. Dzierba, PharmD New York-Presbyterian Hospital, New York, NY
Gregory Eschenauer, PharmD University of Michigan, Ann Arbor, MI
Laura Evans, MD, MSc University of Washington, Seattle, WA
John J. Gallagher, DNP, RN Case Western Reserve University School of Nursing, Cleveland, OH
Rajesh Gandhi, MD Massachusetts General Hospital and Harvard Medical School, Boston, MA
David V. Glidden, PhD University of California, San Francisco, San Francisco, CA
Neil Goldenberg, MD, PhD Johns Hopkins All Children’s Institute for Clinical and Translational Research,
St. Petersburg, FL
Birgit Grund, PhD University of Minnesota, Minneapolis, MN
Erica J. Hardy, MD, MMSc Warren Alpert Medical School of Brown University, Providence, RI
Lauren Henderson, MD, MMSc Boston Children’s Hospital and Harvard Medical School, Boston, MA
Carl Hinkson, MSRC Providence Health & Services, Everett, WA
Brenna L. Hughes, MD, MSc Duke University School of Medicine, Durham, NC
Steven Johnson, MD University of Colorado School of Medicine, Aurora, CO
Marla J. Keller, MD Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY
Arthur Kim, MD Massachusetts General Hospital and Harvard Medical School, Boston, MA
Jeffrey L. Lennox, MD Emory University School of Medicine, Atlanta, GA
Mitchell M. Levy, MD Warren Alpert Medical School of Brown University, Providence, RI
Jonathan Li, MD, MMSc Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
Financial Disclosure
Panel Member
Company Relationship
Judith Aberg, MD Emergent Research support
Gilead Research support
GSK/ViiV Healthcare Advisory board, research support
Janssen Research support
Kintor DSMB chair/member
Merck Advisory board, research support
Pfizer Research support
Regeneron Research support, protocol reviewer
Adaora Adimora, MD, MPH Gilead Consultant, honoraria
Merck Advisory board, consultant, honoraria, research support
Jason Baker, MD, MS None N/A
Lisa Baumann Kreuziger, MD, MS 3M Spouse is an employee, stockholder
CSL Behring Research support
Takeda Research support
Versiti Employee
Roger Bedimo, MD, MS Gilead Advisory board
GSK/ViiV Healthcare Advisory board
Janssen Advisory board
Merck Advisory board, research support
Shionogi Advisory board
Theratechnologies Advisory board
Pamela S. Belperio, PharmD None N/A
Anoopindar Bhalla, MD None N/A
Timothy Burgess, MD AstraZeneca Research support
Danielle M. Campbell, MPH Gilead Advisory board
GSK/ViiV Healthcare Attendee at a community stakeholder meeting, honoraria
Stephen V. Cantrill, MD None N/A
Kara Chew, MD, MS Pardes Biosciences Consultant
Kathleen Chiotos, MD, MSCE None N/A
Craig Coopersmith, MD None N/A
Eric Daar, MD Gilead Consultant, research support
GSK/ViiV Healthcare Consultant, research support
Merck Consultant
Theratechnologies Consultant
Richard T. Davey, Jr., MD None N/A
Key: DSMB = data and safety monitoring board; N/A = not applicable